Open Access

Exploring the complex relationship between metabolomics and breast cancer early detection (Review)

  • Authors:
    • Omar Mahmoud Alshajrawi
    • Tengku Ahmad Damitri Al Astani Tengku Din Tengku Din
    • Shahira Sofea Binti Marzuki
    • Maulidiani Maulidiani
    • Nurul Anaqi Rukaini Binti Mohd Rusli
    • Norul Faezzatul Ain Binti Badrol Hisham
    • Lim Hui Ying
    • Maya Mazuwin Binti Yahya
    • Wan Norlina Binti Wan Azman
    • Ras A. Ramli
    • Wan Faiziah Wan Abdul Rahman
  • View Affiliations

  • Published online on: February 20, 2025     https://doi.org/10.3892/mco.2025.2830
  • Article Number: 35
  • Copyright: © Alshajrawi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

An overview of metabolomics in cancer research, focusing on the identification of biomarkers, pharmacological targets and therapeutic agents, is provided in the present review. The fundamentals of metabolomics, the role of metabolites in cancer emergence and the methods used in metabolomic analysis, are reviewed. The applications of metabolomics in cancer therapy and diagnostics, as well as the challenges encountered in metabolomic research, are discussed. Finally, the potential clinical uses of metabolomics in cancer research and its future possibilities are explored, emphasising the importance of non‑invasive diagnostic and monitoring techniques. The present review highlights the significance of metabolite‑based metabolomics as a specialised tool for illuminating disease processes and identifying treatment potentials. The malfunctioning of metabolomic pathways and metabolite accumulation or depletion is caused by metabolomics abnormalities. Metabolite signatures close to a subject's phenotypic informative dimension can be used to monitor therapies and disease prediction diagnosis and prognosis. Non‑invasive diagnostic and monitoring techniques with high specificity and selectivity are urgently needed. Metabolite‑based metabolomics is a specialised metabolic biomarker and pathway‑analysis technique, illuminating the putative processes of numerous human illnesses and determining treatment potentials. Locating biochemical pathway modifications that are early warning signs of pathological malfunction and illness is possible by identifying functional biomarkers linked to phenotypic variance. Scientists generated numerous metabolomics profiles to disclose the underlying processes and metabolomics networks for therapeutic target research in biomedicine. The metabolomic analysis of the potential utility of metabolites as biomarkers for clinical events is summarised in the present review. The significance of metabolite‑based metabolomics as a specialised tool for illuminating disease processes and identifying treatment potentials is highlighted.

1. Introduction

The metabolome, a final product of the transcriptome, genome and proteome, contains small-molecule metabolites correlated with specific metabolic phenotypes. It provides insights into the pathophysiology and therapeutic targets of numerous illnesses (1). The metabolome has previously shown considerable advantages for identifying biomarkers, diagnosing and treating illnesses, and defining metabolic-control mechanisms. Comprehensive metabolic fingerprints can identify treatment targets and infer potential illness mechanisms. In the rapidly expanding science of metabolomics, tiny molecules in biological processes known as metabolites undergo comprehensive investigation. As a potential method of identifying new biomarkers, pharmacological targets and therapeutic agents, metabolomics in cancer research has attracted considerable attention (2).

The results of protein translation, gene transcription, or structural modifications to the proteome, genome, or transcriptome are called metabolites. Metabolites have the potential to play a significant role in the interaction between genotype and environment and give a clearer picture of the final phenotype. A publicly available human metabolome primarily includes comprehensive data on 41,993 small-molecule metabolites (1-3). In addition to acting as cofactors, energy producers' storage units, signalling molecules, metabolites may also start regulatory processes. Compared with other omic methods, metabolomics focuses on metabolites and has several benefits. Although metabolomics may directly identify the biochemical reaction to a stimulus, genomics may not have considerable influence on how a protein's expression induces its function (3). The present review aims to cover various aspects of metabolomics in the context of cancer research, including fundamentals, the role of metabolites in cancer development, analysis methods, applications in cancer detection and diagnostics, comparison of metabolomic analysis instruments, and potential clinical uses in cancer and breast cancer (BC) research (4). Overall, a comprehensive overview of metabolomics in cancer research is provided, highlighting its potential as a powerful tool for understanding cancer biology and improving clinical outcomes through early detection and personalised treatment strategies. Finally, we discuss metabolomics' possible clinical uses in cancer and BC research. It is aimed to identify and discuss the possibility of metabolites' early detection through metabolomics research.

2. Discussion

BC

Malignant tumours are divided into locally malignant at the same organ or tissue without spreading and tumours spreading to other organs or parts of the body, which is called metastasis. Not all tumours arrive at the metastatic stage, especially if diagnosed early. The tumour derives nutrients from other surrounding healthy cells. As a result, the healthy cells die, which allows the tumour cells to grow even faster. The process of spreading the cancer cells to other body parts and growing continuously in those locations is known as metastasis. BC is a type of cancer affecting one in every eight women in high-income nations by the age of 85, and it will continue to be the primary source of disease burden for women (5). BC remains a severe health issue despite significant advancements in the field of cancer research and is now a high focus for biomedical research. The most frequent disease amongst women worldwide is BC, and its incidence and mortality rates are predicted to rise sharply in the coming years (6). With over 1,700,000 new cases each year, the frequency of this aggressive illness remains disturbingly high, and these numbers point to decreased progress in the preventative field (7). The estimated number of deaths globally in 2020 according to Globocan 2020 (WHO) is 684,996 cases, which comprises 15.5% of the total worldwide death percentage. Genes, the fundamental building blocks of inheritance, can change in ways that lead to cancer (8). Genetic changes that cause cancer can happen because of errors that occur as cells divide or DNA damage inflicted by harmful environmental substances (such as the chemicals in tobacco smoke and ultraviolet rays from the sun). Such changes can also be inherited from parents (9). In elderly people, whose bodies become less capable of eliminating damaged and old cells, the chance of developing cancer later increases. The genetic mutations in every individual cancer differ from one another. Further changes occur when the cancer spreads. Several cells in the same tumour may have distinct genetic changes (10).

Types of BC. BC is categorized into several types based on the characteristics of the cancer cells and their behaviour (11-14). The major types of BC are as follows: i) Ductal carcinoma in situ (DCIS); DCIS is a non-invasive cancer where abnormal cells are found in the lining of a breast duct. While it is not life-threatening, it can increase the risk of developing invasive BC later. ii) invasive ductal carcinoma (IDC); IDC is the most common type of BC, accounting for ~70-80% of cases. It begins in the milk ducts and invades surrounding breast tissue. Symptoms may include a lump or changes in breast shape. iii) invasive lobular carcinoma (ILC); This type starts in the lobules (milk-producing glands) and accounts for ~10-20% of invasive BCs. ILC may present as a thickening or swelling rather than a distinct lump, making it harder to detect via mammograms. iv) human epidermal growth factor receptor 2 (HER2)-positive BC; HER2-positive BC tests positive for excess HER2 proteins, which promote cell proliferation. This type tends to be more aggressive but responds well to targeted therapies that inhibit HER2. A total of ~15-20% of BCs are HER2-positive. v) triple-negative BC (TNBC); TNBC lacks three common receptors: Estrogen, progesterone and HER2. This type is more prevalent among younger women and tends to be more aggressive with fewer treatment options available compared with other types.

Metabolites

The metabolism or metabolic reaction can be defined as the sum of all biochemical reactions carried out by an organism. Metabolites have various roles, including those related to energy, structure, signalling, catalysis, defence and interactions with other organisms. Plants, humans and microbes, all produce metabolites. Metabolites can be divided into two different types, namely, primary metabolites and secondary metabolites. Metabolites are the intermediates or final products of metabolic reactions, which are typically limited to small molecules and are catalysed by several enzymes that naturally exist within cells (15,16). The cell produces primary metabolites and typically participates in respiration and photosynthesis, the two main metabolic activities. Primary metabolites can keep the body's physiological processes running smoothly. Considering this function, it is often referred to as the central metabolite. Amino acids, alcohols, polyols, organic acids, vitamins (B2 and B12), inosine-5'-monophosphate, and guanosine-5'-monophosphate are notable examples of primary metabolites. Ethanol, citric acid, lactic acid and acetic acid are primary metabolites necessary for healthy development, growth and reproduction. Cells utilise primary metabolites, intermediate by-products of anabolic metabolism, to create necessary macromolecules (17).

Secondary metabolites are substances an organism produces that are not necessary for primary metabolic activities but may serve crucial ecological and other purposes. Secondary metabolites are not involved in cell proliferation and development and are synthesised at or near the end of the stationary growth phase (18). Given that secondary metabolites are produced by the same metabolic pathways that primary metabolites use, secondary metabolites are known as the final products of primary metabolites. Primary metabolites are present in every living cell with the ability to divide. Secondary metabolites are present merely incidentally and are not crucial to an organism's survival (16). However, secondary metabolites are produced from primary metabolites, which do not constitute the organism's fundamental molecular structure. Primary metabolites' absence does not immediately shorten an organism's lifespan; instead, survival is compromised to a greater extent. Within a phylogenetic group, its existence and synthesis are found in ecologically disadvantageous species (19). Drugs, flavours, scents, dyes, pigments, insecticides and food additives are examples of secondary metabolites used in pharmaceuticals, industries and agriculture (20).

Numerous intermediates in primary metabolism overlap with the intermediates of secondary metabolites, thus distinguishing between primary and secondary metabolites is not easy. Amino acids, considered primary metabolites, are also unquestionably secondary metabolites (16), in contrast to the claim that sterols are secondary metabolites essential to numerous cellular structural frameworks. The mosaic structure of an intermediate suggests that primary and secondary metabolism share the same metabolic pathway. Adding extra nitrogen and carbon can be directed into the secondary metabolites, which operate as a buffer zone to produce an inactive primary metabolism. When needed, the metabolic disintegration of secondary metabolites can convert the stored carbon and nitrogen back into primary metabolites. The primary and secondary metabolisms are dynamic and in a delicate balance with the growth, tissue differentiation, and development of the cell or organism, as well as external influences, all impacting. Secondary metabolites, also known as natural products or heterogeneous groups of natural metabolic products, are considered to play adaptive roles in ecological interactions, symbiosis, metal transport, competition and other processes even though they are not required for the vegetative growth of the producing organisms (21). For instance, they may act as defence compounds or signalling molecules.

According to Jones et al (23), a comprehensive analysis reveals that a typical human body contains ~2,500 metabolites. Arachidonic acid is a metabolite of prostaglandin, and the two compounds share numerous of the same functional groups, physical characteristics and formulae. Additionally, a specific sequence of enzyme-catalysed reactions that follow a rational path of chemical change connects both chemicals (24). Tyrosine is an amino acid that produces catecholamines, whereas cholesterol creates steroid hormones. By making only minor modifications to the cholesterol ring's superstructure, steroid hormones that differ biochemically from the cholesterol source molecule can be produced (2). Tyrosine is the starting point for an irreversible route that leads to catecholamines, such as norepinephrine or dopamine. Moreover, all precursors of catecholamine must pass through a tyrosine intermediate owing to biochemical principles (3). According to the free-energy exchange theory, inosine-5'-monophosphate is a metabolite that develops from the one-way condensation of two or more intermediates, specifically glutamine and phosphoribosyl-pyrophosphate (4). Small molecules are complex to define precisely because they quickly diverge from their parent structure. A metabolite may also be a component of a larger structure or a degraded product that needs to be disposed. A freely available electronic database including comprehensive data on metabolites discovered in the human body is known as the Human Metabolome Database (3-5).

BC metabolomics

In attempts to discover potential biomarkers that can be used to detect cancer cells in their earliest stages, numerous studies have been performed on the biological samples of patients with BC. Samples from patients such as tissues, blood and urine have been collected and examined to obtain the best results that can benefit individuals. Tumour DNA is the element that has been most thoroughly evaluated, including DNA concentrations, integrity, mutations and methylation status. The main aim is to gauge its potential clinical relevance (24,25). Cancer cells also have the exact needs and capacities for energy as regular cells. It has been demonstrated that most cancer cells produce energy through cytoplasm glycolysis. Energy generation is typically utilised by several contemporary technologies to detect malignancy. The rate of protein turnover and lipolysis, which is the breakdown of fat stored in fat cells, increases in cancer cells (26).

Cancer cells undergo significant metabolic changes compared with normal cells. These changes are critical to cancer cells' survival and proliferation, providing a unique opportunity to differentiate cancer cells from normal cells. Metabolomics can be used to identify these metabolic changes and thus help diagnose and treat cancer. It can also help in the discovery of new biomarkers and therapeutic targets. Some researchers have focused on potential indicators found in urine samples of patients with BC. The metabolomics approach is used for the test and research, which involves running tests on technologies such as nuclear magnetic resonance (NMR), high-performance liquid chromatography (HPLC), gas chromatography (GC)-mass spectrometry (MS), or other suitable analytical tools to obtain the most accurate results. In total, 44 pair-wise rates of RNA metabolites exist for BC urinary tests. Numerous different indicators or biomarkers can be found in the urine samples of patients with BC. Based on a study by Nam et al (27), homovanillate, 4-hydroxyphenylacetate, 5-hydroxyindoleacetate and urea are all found in the urine samples of patients with BC.

The main contributing compounds in the urinary metabolomics for BC include formate, succinate and nucleoside uracil. Succinate, a metabolite of the tricarboxylic acid (TCA) cycle and a marker for the Warburg effect, is also highlighted by another MS investigation (28). With reasonable specificity and sensitivity, the panel of succinic acid and dimethyl-heptanoyl-carnitine is used to distinguish between BC and healthy controls. According to research looking at nucleosides in urine, 5-hydroxymethyl-2'-deoxyuridine, 8-hydroxy-2-deoxyguanosine and succinyl adenosine are all shown to be more common in patients with BC (29-31).

Patients with BC have higher amounts of glucose, creatinine, glutamine, glutamate, arginine, lysine and valine than healthy controls. These metabolisms are closely linked to a higher risk of BC. Moreover, it has been found that those with greater levels of 5-amino valeric acid, tryptophan, phenylalanine, y-glutamyl threonine, valine, or iso-glutamine are more likely to be diagnosed with BC (23,33). A recent study predicted that 2-o-methylcytidine and 5-methylthioadenosine levels in patients with BC will rise (34). Based on the same research, hierarchical analysis reveals 71 out of 168 differentially expressed metabolites.

Analysing urine metabolomics biomarkers often uses analytical techniques such as NMR and MS (35). By identifying the distinctive electrochemical environment of each constituent proton, the urine NMR readings of molecules can be identified using NMR. Low levels of several metabolites including succinate have been found in the urine of patients with epithelial ovarian cancer and BC according to research on urinary-metabolite modifications (36,37). A total of nine metabolites significantly differ in a study comparing the urinary proton NMR metabolomic profiles of BC (n=48) and ovarian cancer (n=50) based on Wilcoxon's rank-sum test. The metabolites involved are acetone, allantoin, carnitine, urea, 1-methyl nicotinamide and levoglucosan. Slupsky et al (39) discovered that the amount of several high-level metabolites including glucose and creatine, which are high in cancer tissue, decreases in the urine of patients with BC (38).

Moreover, patients with BC have lower urine succinate levels compared with healthy controls (39). The urine samples of patients with BC have decreased glutamine level, which is typically high in breast tissue. This discovery is also validated by additional research that produces comparable outcomes (38). Urine of patients with BC has lower threonine levels than controls as well (40). Changes can further be observed in metabolites such as choline and 2-hydroxybutyrate. These two metabolites have higher levels in BC samples than in healthy control samples (41,42). Valine and lysine also rise (43). Furthermore, patients with BC have lower amounts of melatonin and indole-3-acetate in their urine tests.

A study on BC indicators in urine examines metabolic differences between patients with BC and healthy volunteers. The investigation identified12 metabolites including amino acids, organic acids and nucleosides as possible biomarkers (30). In a separate study, (27) used a LC-ion trap MS to analyse urine samples from 85 patients with BC and corresponding controls. A total of 44 pairwise ratios of metabolite characteristics were effectively examined by computational analysis, with a sensitivity and specificity of 83.5 and 90.6%, respectively, for the best BC prediction. S-Adenosylhomocysteine and a few other methylated nucleosides significantly dominate the classification performance. In another study, a capillary electrophoresis (CE) MS was used to examine urine samples from 21 patients with advanced BC before and after receiving chemotherapy, as well as samples from the general population (44). The aforementioned study found that metabolite levels decrease by 30% in chemotherapy-sensitive patients compared with the control group. Specifically, glycine, cysteine, histidine, cysteine, and tryptophan levels are affected. Those who are resistant to treatment have 9% changes in metabolite levels. Meanwhile, the amounts of succinate increases and the levels of chromium considerably drop, whereas most amino and organic acids do not show any apparent alterations. In another study, urine samples from 22 healthy controls were compared with those from 10 patients with BC, 9 with ovarian cancer and 12 with cervical cancer. The cancer biomarkers were found to comprise 5-hydroxymethyl-2-deoxyuridine and 8-hydroxy-2-deoxyguanosine (45).

The identification of BC biomarkers in urine samples of patients with BC is also influenced by environmental factors. Cadmium is markedly more prevalent in urine of patients with BC (46). The same applies to increasing chromium and arsenic. Moreover, it was revealed that patients with BC have a general decrease in amino acids, nucleotides and TCA cycle intermediates (40). The marker results from previous studies based on different sample types, such as tissue, serum, plasma and urine samples, are included in Table I.

Table I

Metabolomics in studies of human BC: Comparison between blood, tissue and urine samples.

Table I

Metabolomics in studies of human BC: Comparison between blood, tissue and urine samples.

First author/yearSample type and sample sizeMethod Results/MarkersPopulation(Refs.)
Asiago et al, 2010257 serial blood serum samples: 116 samples with recurrent BC, 141 samples with no sign of recurrenceNMR and GC-MS11 markers (NMR: formate, His, Pro, Cho, Tyr, 3-HB, Lact; GC-MS: Glu, N-acetyl-Gly, nonanedioic acid, 3-hydroxy-2-methyl-butanoic acidHouston, Texas(48)
Oakman et al, 2011Pre- and post-operative blood serum samples from 44 patients with early BC and 51 meta-static patientsNMRMetastatic samples: Higher values of Pro, Phe, Gluc, Lys and N-acetyl-Cys and lower values of lipids.Prato, Italy(49)
Tenori et al, 2012Blood serum samples from 579 women with metastatic BC randomized to paclitaxel plus either anti-HER2 (lapatinib) or placeboNMRGluc higher in the patients with longer time to progression. Glutamate and Phe higher in patients with shorter time to progression in on-treatment samplesItaly(50)
Wei et al, 2013Serum samples from 28 patients with different response rates to NACNMR/LC-MSThree metabolites (Ile, Thr, Gln) from NMR and linolenic acid from LC-MS were significantly different when comparing response to chemotherapyGermany(51)
Jobard et al, 2014Blood serum from 197 patients with early BC and 90 metastatic patientsNMRAla, His and betaine were higher in the serum of patients with early BC; end products of lipid degradation and β-oxidation (Acac and 3-HB) (glycerol), Pyr, NAC glycoproteins, lipids, or Phe, Glu and mannose concentrations increased for metastatic BCFrance(52)
Tenori et al, 2015Blood serum from 80 patients with early-stage BC and 95 patients with metastatic BCNMRSignificantly lower levels of His and higher serum levels of Gluc, Tyr, Lact and lipids in metastatic patientsNew York and Italy(53)
Henneges et al, 2009Urine samples from 85 patients with BC and 85 HCLC-MS44 pairwise ratios of metabolite features had distinct predictive capacity; S-adenosylhomo-cysteine as main identifiers; various methylated nucleosidesGermany(28)
Nam et al, 2009Urine samples from 50 patients with BC and 50 HCGC-MSHomovanillate, 5-hydroxyindoleacetate, 4-hydroxyphenylacetate and urea were identified to be different in normal subjects and cancerKorea(27)
Woo et al, 2009Urine samples from 10 patients with BC, 9 patients with OV, 12 patients with cervical cancer and 22 normal controlsGC-MS/LC-MSBC samples contain 2-hydroxymethyl-2-deoxyuridine and 8-hydroxy-2-deoxyguanosineKorea(46)
Kim et al, 2010Urine samples from 50 patients with BC and 50 controlsGC-MSFive potential urinary biomarkers for BC; meta-bolites were not identifiedKorea(54)
Slupsky et al, 2010Urine samples from 48 patients with BC, 50 patients with OV and 73 healthy volunteersNMR67 metabolites identified; amino acids, tricarboxylic acid cycle and metabolites relating to energy metabolism, and gut microbial metabolismEdmonton, Canada(39)
Yu et al, 2013Urine samples from 21 patients with advanced or locally advanced BC before and after chemotherapy and 21 healthy volunteersCE-MSIn chemotherapy-sensitive patients: Cys, Gly, cystine, His and Trp were significantly decreased after chemotherapy; In chemotherapy-insensitive patients: few obvious differences between patients before and after chemotherapy (Succ increased while Cr decreased)China(45)
Chen et al, 2009Urine samples from 20 patients withBC and 18 HCLC-MS12 metabolites as potential biomarkers including organic acids, amino acids, and nucleosides; elevated Trp and nucleosides metabolism and protein degradation in patients with BCChina(31)
Bathen et al, 2013228 BC tissuesNMRThe loading profiles from both PCA and PLS-DA analyses revealed choline-containing compounds as the key indicators for tumour content, with phosphocholine being more abundant in tumour tissue. Glycine, taurine and glucose are also suggestive metabolites.Trondheim (Norway)(55)
Borgan et al, 201046 BC tissuesNMROne of the categories, A2, had samples with markedly lower glucose and higher alanine levels than the other luminal A samples, indicating that these tumours were more glycolytically active. Additionally, this group was enriched for genes having Gene Ontology concepts associated with DNA repair and cell cycle.Trondheim (Norway)(56)
Debik et al, 2019118 BC tissues and serumNMRPLS-DA multilevel analysis revealed significant changes in blood metabolite levels following therapy (P=0.001), including unfavorable alterations in lipid levels. PLS-DA detected metabolic differences between survivors and non-survivors in tissue samples received 12 weeks into therapy with an accuracy of 72% (P=0.005), but not in serum samplesOslo (Norway)(57)
Haukaas et al, 2016228BC tissuesNMRAmong the most notable changes were Mc1's high amounts of GPC and phosphocholine (PCho), Mc2's high levels of glucose, and Mc3's high levels of lactate and alanineOslo (Norway)(58)
Chae et al, 201660 BC tissuesNMRThe GPC/PC ratio, as well as the concentrations of myo-inositol and succinate, were greater in the pure DCIS group than in the DCIS with invasive cancer group (P=0.004, Bonferroni-corrected P=0.064). The OPLS-DA models generated using HRMAS MR metabolic profiles could clearly distinguish between pure DCIS and DCIS of myo-inositol and succinate with concomitant invasive cancer using multivariate analysis.Seoul (South Korea)(59)
Euceda et al, 2019122 BC tissuesNMRLinear mixed-effects models revealed a significant interaction between time and bevacizumab for glutathione, indicating higher levels of this antioxidant in chemotherapy-only patients than in bevacizumab receivers after treatmentTrondheim(60)
Cala et al, 2019Plasma 58 (29BC; 29HC)NMRParticularly, the understanding of the up regulation of long chain fatty acyl carnitines and the downregulation of cyclic phosphatidic acid. In addition, the mapped metabolic signatures in BC were similar but not identical to those reported for non-Hispanic women, despite racial differences.Bogotà (Colombia)(61)
Lécuyer et al, 2018Plasma 602 (206BC; 396HC)NMRWomen characterized by higher fasting plasma levels of valine, lysine, arginine, glutamine, creatine, creatinine and glucose, and lower plasma levels of lipoproteins, lipids, glycoproteins, acetone, glycerol-derived compounds, and unsaturated lipids had a higher risk of developing BC.France(62)
Suman et al, 2018Plasma 122 (72BC;50HC)NMRThe levels of hydroxybutyrate, lysine, glutamate, glucose, NAC glycoprotein and lactate were highly distinguished in BC stages and showed a favorable biomarker potential using receiver-operating curves based diagnostic models. Furthermore, the significant modulation and favorable diagnostic performances of glutamate, NAC glycoprotein and Lactate in LBC as compared with EBC give their significance in the BC progression.Lucknow (India)(63)
Louis et al, 2015Plasma 145 (73BC;72HC)NMRThe levels of hydroxybutyrate, lysine, gluta-mate, glucose, NAC glycoprotein and lactate were highly distinguished in BC stages and showed a favorable biomarker potential using receiver-operating curves based diagnostic models. Furthermore, the significant modulation and favorable diagnostic performances of glutamate, NAC glycoprotein and lactate in LBC as compared with EBC give their significance in the BC progression.Hasselt (Belgium)(64)
Vignoli et al, 2020Plasma 43 BCNMRER status in patients with HER2-positive BC was found to induce significant changes in the host circulatory metabolome with important implications for the pCR to NACT and for the overall clinical outcomeAviano (Italy)(65)
Jobard et al, 2021Plasma 1582 (791BC;791HC)NMRThe concentration of NAC glycoproteins, ethanol, hypoxanthine and dimethylamine, were positively associated with BC. The concentration of 10 metabolites were increase in premenopausal group after FDR adjustment. The strongest association: histidine. Borderline inversely associated with BC are LDL and VLDL (fatty acids)Lyon (France)(1)
McCartney et al, 2019115 Serum BCNMRMetabolomic signature between patients with early BC and metastatic BC is similar and would be predictive of cancer recurrence. Low Random Forest score: Disease free at follow-up. High Random Forest score: One relapse case among seven patients.NewYork (USA)(66)
Jiang et al, 201829 Serum BCNMRNMR spectra containing signal from a variety of amino acids (isoleucine, valine, leucine, alanine, threonine, lysine, glutamine, glycine, ornithine, phenylalanine, tyrosine, histidine), amino acid derivative (creatinine, creatine, betaine), variety moieties of lipids, ketone bodies (acetone, 3-D-hydroxybutyrate, acetoacetate), choline metabolites, carbohydrate metabolism related metabolites, NAC glycoproteins and organic acids.Singapore(67)
Wojtowicz et al, 2020Serum 95 (9BC;86HC)NMR31 metabolites, four unknown signals, and nine ranges of chemical shift regions assigned to different lipid types. Levels of citrate, glutamine, creatinine, acetoacetate, acetate, glucose, betaine, glycerol, leucine, choline and lysine were upregulated in TNBC Lipid levels, lactate, acetone, alanine, glutamate, tyrosine, pyruvate and isoleucine were down regulated in TNBC.Wroclaw (Poland)(68)
Men et al, 2020Urine 144 (106BC;38HC)NMRHeavy metals in urine samples. Cd has been detected in BC tissue at high concentrations. The Cd was markedly increased in the urine of patients with BC compared with the control population (~2-fold). Numerous small molecule metabolites were altered in the urine of patients with BC compared with the control population.Tengzhou (China)(69)
Wang et al, 2017Urine 78 (40BC;38HC)NMRA total of 10 metabolites exhibited the highest contribution towards discriminating patients with BC from HXs (variable importance in projection (VIP) >1, P<0.05). The metabolomic pathway analysis indicated several metabolism pathway disruptions, including amino acid and carbohydrate metabolisms, in patients with BC, namely, glycine and butanoate metabolisms.Funchal (Portugal)(2)
Slupsky et al, 2010Urine 170 (48BC;50OC;72 HC)NMRAll metabolites that were significantly different between the cancers and normal controls were lower in concentration in both the EOC and BC groups as compared with normal. Intermediates of the tricarboxylic acid cycle and metabolites relating to energy metabolism, amino acids and gut microbial metabolism were perturbed.Edmonton (Canada)(71)

[i] BC, breast cancer; TNBC, triple-negative HC, healthy control; OV, ovarian cancer; NMR, nuclear magnetic resonance; LC-MS liquid chromatography-mass spectrometry; NAC, N-acetylcysteine; GPC, glycerol-phosphocholine; DCIS, ductal carcinoma in situ.

Role of metabolites in cancer development

A complex network of chemical processes is responsible for metabolism within cells, which supports healthy development and reproduction. Metabolism involves catabolism and anabolism. The former provides energy and generates the cellular building blocks required for cell division. Uncontrolled cell proliferation and a diverse microenvironment are characteristics of cancer. According to Cairns et al (71), cancer cells alter their preferred metabolic pathway to balance their energy requirements with their need to produce biosynthesis precursors for development (69) and to survive in low nutrient areas and low oxygen concentrations (72). By changing the functions of current metabolic pathways or rewiring new connections, cancer cells experience widespread metabolic modifications, notably in glycolysis, mitochondrial biogenesis, lipid metabolism and the pentose phosphate pathway (73). Through various processes, metabolic reprogramming in cancer cells causes the accumulation or depletion of intermediate metabolites (74). The first and foremost one is an alteration in the activity of metabolic enzymes. Since the 1920s, the Warburg effect has been recognised as a distinctive feature of cancer. It is a change in metabolic state wherein cells show an enhanced conversion of glucose into lactate even in highly oxygenated areas (75-77). For instance, activating glycolysis-related enzymes results in the build-up of several glycolytic intermediates during glycolysis, the preferred method by which cancer cells receive energy and biosynthesis building blocks. Conversely, the build-up of succinate and fumarate is caused by a decrease in succinate dehydrogenase and fumarate hydratase activities, respectively.

Since the discovery of oncogenic functions of various mitochondrial metabolites such as 2-HG, succinate and fumarate, researchers have become increasingly interested in the functions of these ‘oncometabolites’ in cancer. Oncometabolites affect signal transduction, post-transcriptional modifications, and epigenetic changes. The inactivation of tumour-suppressor genes and the promotion of carcinogenesis are caused by metabolic remodelling, which can encourage DNA hypermethylation and histone hyperacetylation (78,79). Numerous intermediate metabolites, in addition to oncometabolites, can bind directly to proteins or nucleotides and cause them to malfunction. These intermediate metabolites can also function as transmembrane receptor ligands, triggering subsequent signalling cascades.

The phenomenon in which cancer cells enhance their intake of glucose and the formation of lactate with a significant reliance on aerobic glycolysis is described as the Warburg effect (75). Cancer cells can produce only minimal ATP during this metabolic state, and they may start to rely on glutamine as a fuel source (80). Thus, cancer therapies are intensively researching the suppression of glucose and glutamine metabolism (80,81). Under normoxic conditions, the contribution of lactate to oxidative respiration has attracted newfound attention (82). The finding that lactate is a waste product and a crucial energy source for tumours raises the possibility that metabolites other than glucose and glutamine may support an environment favourable for the proliferation and multiplication of cancer cells. Asparagine, arginine, cysteine, serine and glycine are examples of downstream amino acid by-products that have been studied for their role in the survival of cancer cells. Further research into medicines targeting each metabolic pathway is required, even if the deprivation of these nutrients is beneficial in some situations. As an alternative, several amino acids and essential vitamins such as vitamins A, B, C, D, E and K function as antitumorigenic agents and slow the spread of cancer. The same study emphasises the roles of lactate, vitamins and amino acids in advancing, inhibiting, and preventing cancer by drawing attention to these generally underestimated metabolites (Table II).

Table II

Metabolite contribution to tumour survival according to cancer types.

Table II

Metabolite contribution to tumour survival according to cancer types.

First author, yearMetabolitesCancer typeRole in tumour progression(Refs.)
Wang et al, 2021Vitamin ABreast4-HPR induces cell death(83)
Sullivan et al, 2016  Vitamin A and retinol reduce risk(75)
Wang et al, 2021 Colon/Colorectal Prostate4-HPR induces cell death 4-HPR induces cell death(83)
Sullivan et al, 2016Vitamin B1BreastIntermediate concentrations promote Ehrlich's ascites proliferation in thiamine-deficient patients; high concentrations inhibit proliferation Patients exhibit decreased expression of SLC9A3 transporter gene(75)
Doldo et al, 2015Vitamin CBreastLow concentrations induce cell invasiveness; high doses restrict EMT(84)
Sullivan et al, 2016Vitamin DBreast Colon/ColorectalCalcitriol and D3 analogs suppress MMP-2 and -9 and VCAM-1; low serum D3 levels are associated with high incidence Low serum D3 levels are associated with high incidence(75)
Zeng et al, 2019Vitamin EBreast Colon/Colorectal ProstateTocotrienols exhibit chemotherapeutic and antitumour properties Tocotrienols exhibit antitumour properties Tocotrienols exhibit chemotherapeutic properties(85)
Miyazawa et al, 2020Vitamin KBreastK2 induces nonapoptotic cell death(87)
 ArginineBreastLow plasma levels act as a prognostic biomarker(87)
Miyazawa et al, 2020; Qiu et al, 2014  Arginine starvation is used to treat arginosuccinate synthase-deficient patients(87,88)
Cheng et al, 2018 OvarianCancer cells are deficient in arginosuccinate synthase-1; ADI-PED-20 is used to degrade arginine(89)
Ji et al, 2020AsparagineBreastMaintains health of glutamine-independent cells(90)
Sullivan et al, 2016Cysteine Lactate SerineBreast Colon/Colorectal Breast BreastInhibition of histone deacetylase-6 sensitizes TNBC cells to cysteine deprivation via cystine/glutamate antiporter-targeted therapies Starvation induces a reduction in liver-metastatic cell proliferation 10 mM L-lactate acts as chemoattractant and facilitates migration Cells prefer serine over glycine and exhibit a decrease in nucleic acid synthesis when starved of serine(75)
Metabolomic techniques

Under certain specific circumstances, any metabolite can be broken down into smaller product ions. Specific pressure, temperature and collisional energy are required to break down the metabolites. These processes of breaking down produce a distinctive pattern of fragmentation used as identification information. Each chemical has a unique fragmentation pattern crucial to determining a compound's retention time for intensity quantification. The methodology used in metabolomics is unique and has its own set of procedures. Each approach has a similar set-up procedure. Importantly, the samples needed must be prepared according to the desired test. Following the metabolic extraction, the collected samples are sent to metabolomics equipment for compound separation, detection and analysis (91).

Fresh tissue and cells from in vitro cultures are the two common sample types used to extract metabolomics data. For fresh tissue models, the tissue is collected, immediately snap-frozen in nitrogenous liquid N2, and then homogenised in an identical mixture of solvents. This phase must maintain the effectiveness of the extraction and the biochemical integrity. Samples are centrifuged several times to guarantee that all precipitated proteins and other macromolecules are wholly removed using chemicals to help in this function. The pallets are preserved for protein concentration analysis to normalise the metabolite levels. These supernatants are collected, and then the methanol-chloroform-water mixture is removed using a speed vacuum and lyophilisation. The result is the formation of the powdered metabolites. Then, prior to metabolomics acquisition with metabolomics devices, metabolites are resuspended in a solvent combination (92-95).

Numerous options are available for selecting metabolomic equipment. Separation, detection and hyphenated techniques are the three common strategies used for categorising the instruments. Techniques including GC, CE, HPLC, ultra-performance LC and ion chromatography can be used to separate distinct metabolites that elute at varying retention durations. Regarding the method of detection, MS equipment is frequently utilised. MS equipment includes quadrupole time-of-flight (TOF) chromatography, triple quadrupole and Fourier transforms (FT) orbitrap. NMR spectroscopy is another detection method not requiring separation techniques. It is also commonly used to determine the structures of organic compounds. HPLC-MS, FT, ion cyclotron resonance (ICR)-MS and GC-MS (91).

Software programs for metabolomic analysis are required to analyse experimental metabolomic data. MS-based equipment can identify metabolites by using an internal compound standard database and MS/MS fragmentation capture under the same conditions. The sample's fragmentation should match the database's fragmentation to verify one's structure. NMR-based methods can be used to investigate the structure of compounds and isotopomers. Analyses based on NMR and MS can cross-validate and cover more metabolites overall. Planning and conducting a metabolomics study involves four significant steps. These steps include sample collection or generation, data acquisition, bioinformatics and interpretation. Based on the results, it is recommended that a hypothesis be formed or the newly discovered biomarkers to be tested in further studies. Adding quality control to obtain reproducible outcomes and generate meaningful metabolomics data during data acquisition is optimal.

MS-based metabolomics. One of the most popular analytical tools used in metabolomics applications is the MS. The primary goal of MS is the structural characterisation of significant metabolites in the search for biomarkers (96). Metabolic fingerprinting can be acquired by MS direct injection, although it has several limitations such as co-suppression and low ionisation efficiency. To avoid these issues, MS-based metabolomic techniques such as CE-MS, GC-MS, LC-MS (97) and CE are used. These tools can eliminate co-suppression whilst reducing the complexity of biological material. Adding MS to these methods increases the accuracy of compound identification, detection and quantification and shows great sensitivity, selectivity, speed and efficiency (42). The samples prepared are infused directly or by chromatography before being analysed in a MS. Data are recorded, analysed, processed and interpreted before being compared with the theoretical data.

GC-MS. GC-MS has emerged as a crucial and trusted analytical technique for the metabolomic study of separation, detection and identification (98,99). The collected samples are subjected to metabolite extraction before being injected in split-less mode. Afterwards, the high-resolution capillary column is used to propel and release the carrier gas through the sample (42). GC analysis must be performed under certain circumstances (for example, high temperatures and in an oven), and the metabolites must be volatile and thermally stable (for example, metabolites such as alkenes, organic acids, ketones and aldehydes). Non-volatile metabolites including lipids, amines, amino acids, phosphorylated metabolites and sugars must first go through derivatization (42). The samples can be ionised by electro-impact (EI) or chemical ionisation for MS detection. The EI approach is frequently used in ionisation. The mass spectra can be revealed by molecular-ion fragmentation, which EI can offer. The three techniques most frequently used in metabolomics are quadrupole, TOF and ion trap.

Salivary volatiles are screened for potential BC by GC-quadrupole MS (qMS) as part of an exploratory investigation; geographically remote communities are included (100). It has been claimed that the metabolomic signature of human BC cell lines can be established using GC-qMS (98). Based on the urinary volatomic biosignature, it can also be utilised to distinguish amongst various cancer types (101). In contrast to GC-qMS, GC-TOFMS can assess glutamate enrichment as a potential new method of diagnosing BC. Patients with BC with oestrogen receptor (ER)-positive (ER+) and ER-negative (ER-) cells can be compared metabolically using a GC-TOF-MS framework (102). In a pilot investigation on patients with BC, it was found that GC-MS can be used to assess the detectability, reliability and distribution of metabolites obtained in pre-diagnostic plasma samples (103). The sensitivity, specificity, reproducibility and high-throughput technology of GC-MS-based metabolomics to handle a huge volume of samples renders it preferable to use. However, GC-MS is limited in its mass range, and because of fragmentation, molecule ions are frequently undetected. Determining unknown metabolites is difficult because of these limitations. Additionally, the required metabolites must be thermally stable and volatile (104).

HPLC-MS. HPLC-MSis a simple method of separating and characterising various metabolites, including acids, bases, salts and hydrophobic and hydrophilic metabolites. Owing to its capability to accommodate separation processes and various mass analysers, LC-MS or HPLC-MS is preferred over MS-based metabolomics because it is not restricted to volatile and thermally stable metabolites (105). Ahad and Nissar (106) used the fundamental principles of HPLC-MS, eluting the metabolites through a column based on the partition between a stationary phase and mobile liquid phase. The kind of stationary phase that the metabolites should elute through depends on their charge, size, hydrophobicity and molecular weight (106). To achieve a quicker separation of metabolites, the current HPLC technology focuses on smaller columns, miniaturisation and low solvent volumes. Thus, ultra-high-performance LC (UHPLC) replaces HPLC. UHPLC does not require large amounts of solvent and speeds up resolution within short analysis times.

NMR-based metabolomics. NMR-based metabolomics is an alternative to MS-based metabolomics. NMR spectroscopy, commonly known as NMR, is acknowledged as a promising metabolomic approach. Despite having lesser intrinsic sensitivity than MS, NMR offers a thorough metabolite fingerprinting, profiling and metabolic study under particular conditions. This drawback has limited its ability to deal with metabolites at the trace level. NMR-based metabolomics has the benefits of automation, minor or no sample preparation requirements, non-destructive, non-selectivity in metabolite detection, excellent repeatability, and the capacity to quantify numerous classes of metabolites simultaneously (106). The foundation of NMR spectroscopy is the radiation that numerous isotopes' nuclei absorb at a particular frequency when exposed to a magnetic field (104).

An NMR spectrum has been demonstrated to correspond with a particular metabolite pattern. Additionally, it offers structural details to enable easier identification of unknown metabolites, which can be accelerated by combining spin-spin coupling, chemical shift and relaxation or diffusion data. In contrast to localised early disease (EBC), a 1H NMR-based metabolic phenotyping study to identify metabolic serum abnormalities connected with advanced metastatic BC (MBC) is conducted (51). The MBC and EBC groups are distinguished by the metabolite's acetoacetate, histidine, pyruvate, glutamate, glycoproteins (N-acetylcysteine), mannose, glycerol and phenylalanine.

The general flowchart of the details of the in vitro and ex vivo NMR spectroscopy methodology in BC study is shown in Fig. 1. The samples obtained from the patients and controls can be analysed and studied through in vitro or ex vivo NMR spectroscopy based on the suitability of the samples. This research primarily focuses on urinary samples, thus the method used is in vitro. Based on previous studies, if the samples used are urine, they should be collected in the morning pre-prandial period under sterile conditions after overnight fasting (107). Then, the samples should be placed on ice and frozen in liquid nitrogen before being stored at -40˚C or lower. Next, to perform NMR analysis, the samples must be diluted with sodium phosphate buffer prepared in ddH2O at 1:2 (prepared buffer/sample). The pH of the urine sample needs to be adjusted to 7.4 constantly because it can lead to changes in the chemical shift of the samples. A total of ~3 mM sodium azide is added to prevent bacterial growth in the solution. Then, 0.5 mM TSP is added for chemical-shift referencing and concentration quantification. TSP is an internal reference for metabolites' chemical-shift calibration and quantification in tissue and urine.

Hyphenated techniques metabolomics. Hyphenated approaches, along with MS-based and NMR-based metabolomics, are eliciting attention in metabolomic investigations owing to their ability to simultaneously detect hundreds of metabolites. This is because this method can simultaneously detect hundreds of metabolites. GC-GC-MS, LC-LC-MS, LC-FT-ICR-MS, LC-MS-NMR and MALDI-FT-ICR-MS are a few examples of analytical techniques. Two-dimensional liquid-LC and gas-GC are gaining increased attention in the metabolomics field because metabolite overlapping can be avoided by redirecting each peak from one GC or LC column to a second column. These methods also increase sensitivity and complementary selectivity (108).

Comparison between MS-based and NMR-based metabolomics study

NMR and MS are the most often applied metabolomic techniques for metabolomic profiling. NMR and MS may be utilised to detect and identify metabolites whilst precisely measuring the concentration, regardless of whether the study focuses on targeted or untargeted analysis. However, each method has advantages and disadvantages. Using several complementary technology platforms to obtain the best results is optimal.

NMR is quantitative and reproducible and does not require extensive sample preparation procedures such as separation or derivation (109-111). This method supports the simultaneous measurement of routine lipids, lipoprotein subclass profiling with lipid concentrations within 14 subclasses, fatty acid composition, and various low-molecular metabolites, including amino acids, ketone bodies and metabolites related to gluconeogenesis, in molar concentration units. Considering that no sample preparation is required, it is a quick analysis that requires ~5 min. The outcomes can be enhanced by running more scans and using a stronger magnetic field (111). Additionally, NMR requires a larger sample volume than MS analysis. However, the high scalability and thorough coverage of numerous chemical pathways of NMR render it ideal for the biomarker detection for chronic diseases.

A small sample quantity can be used to evaluate numerous metabolites through the compassionate MS technique. Additionally, it can quantify molecular concentrations as low as nanomolar and picomolar (109). MS can be utilised to find hundreds of metabolites in a sample when used in conjunction with chromatography, including GC and LC. If combined with chromatography (109), MS can investigate secondary metabolites even when the detection level is lower. However, a sample in MS cannot be recovered after analysis (111). In addition to requiring sample preparation and separation, MS is more expensive than NMR (112). MS is a favourable option for achieving comprehensive metabolome coverage in metabolomic profiling.

Biomarker identification. Biomarkers

A biomarker is a term that refers to a trait that is objectively assessed as an indication of normal biological processes, pathological processes, or pharmacological reactions to a therapeutic intervention (113), anticipating sickness occurrence or outcome (114). Biomarkers are used to convey information about human biology, and the discovery of new oncological biomarkers is at the top of the list of translation research goals. Diagnostic biomarkers are used to differentiate sick from healthy persons. Conversely, predictive, prognostic and therapeutic biomarkers may affect therapeutic decision-making and management techniques with the goal of personalising illness therapy (115). Prognostic biomarkers aim to predict the likelihood of a clinical event in the context of illness. Unfortunately, prognostic biomarkers are occasionally a blunt measure of stratifying outcomes, and their reliability is limited by interindividual variability (that is, varying values for a range of patients), intraindividual variability (that is, varying scoring by histopathologists providing Ki-67 measurement), and sensitivity and specificity implications (116).

BC biomarkers. Currently, biomarkers are crucial to managing patients with BC, particularly when choosing the kind of systemic treatment to be used (117). Cell receptors, one of the several varieties of biomarkers, show significant value as diagnostic, prognostic and predictive biomarkers in cancer research and therapy. Accordingly, they are incorporated into drug-development trials (118). ER, PR and HER2/neu receptors are two excellent examples of biomarkers that are prognostic of outcomes and predictive of responsiveness to specific therapy in BC (8). ERs and progesterone receptors (PR) should be assessed on all newly diagnosed invasive BCs to select patients likely to respond to endocrine therapy (117).

ER and PR. PR is a steroid receptor superfamily member that mediates progesterone's action in its target tissues. Particularly, in the mammary gland, the luminal epithelial cell compartment is the only place where PR is expressed (118). The development of sex organs, pregnancy, bone density, cholesterol mobilisation, brain function, cardiovascular system and other biological processes are only a few of the functions regulated by steroid hormones and their receptors (119). They are essential for the development and spread of BC. Hormone receptors exist in >70% of breast tumours (120). Their cells exhibit positive ER and PR expression, which is linked to the development and spread of cancer cells. The development and spread of BC are significantly influenced by oestrogen and its receptor, ER. PR can influence how ER functions because it is an ER-upregulated target gene whose expression is regulated by oestrogen (119). In BC, PR is a useful predictive indicator of overall survival or disease-free survival (121).

The primary physiological actions of progesterone, a 21-carbon steroid, are mediated by binding to PRs A and B (PR-A and PR-B), which trigger the transcription of specific genes and change proliferative endometrium in an oestrogen-primed uterus into secretory endometrium (121). Progesterone's physiological function is essentially limited to pregnancy, the peri- and post-ovulatory periods of the menstrual cycle. The corpus luteum starts producing progesterone in the early post-ovulatory phase of the menstrual cycle (119). In the later stages of breast growth, side branching and amelogenesis, the receptor activator of nuclear factor kappa B ligand (RANKL) acts as a paracrine mediator of PR-B (119). By autocrine activation through the RANKL pathway and the activation of the downstream target Cyclin D1, the intrinsic proliferation of PR-negative luminal epithelial cells of the breast can be induced by progesterone and PR (121).

Experiments on a breast mouse model, normal human breast tissue, and clinical trials have all shown that progesterone and oestrogen are the two main proliferative steroid hormones in the mammary epithelium that signal mammary gland development (119). Early puberty requires ductal elongation but not progesterone/PR; it requires oestradiol and epithelial ER signalling (122). PR signalling is necessary for ductal elongation and side branching in the epithelial compartment in response to elevated oestrogen levels (8). Early in pregnancy, PR signalling can cause the epithelial compartment to expand rapidly. In mid-to-late pregnancy, progesterone is necessary for alveolar differentiation (117). Progesterone changes from promoting terminal differentiation to inhibiting it at term, and it must be withdrawn for lactation (119).

Progesterone has been linked to the development of BC in mechanistic investigations. However, weak epidemiologic evidence does not indicate a link between circulating levels and the risk of the disease (119). Progesterone metabolites may exert pro- and anti-carcinogenic effects, and the balance amongst these factors may affect BC risk according to data primarily from the Wiebe laboratory (120). However, population-based research pays little attention to this hypothesis primarily because assays are insufficient (117). Lastly, research links progesterone signalling to the development of BC in BRCA1 mutation carriers, raising the possibility that using chemotherapy to block downstream signalling can be beneficial (120).

3. Conclusion

According to previous studies in the manuscripts and their associations with cancer pathways and treatment, metabolomics can be used to identify new biomarkers or be one for cancer diagnosis and treatment stages and the effectivity of the medications. By comparing metabolites in patients with BC and healthy individuals, researchers may identify metabolites with high associations unique to cancer in general and specific for BC. In this review, we study the association of non-targeted and targeted metabolites pathway with patients with BC and healthy controls in numerous places and numerous publications as mentioned before. A high chance of identifying biomarkers from metabolites by conducting more studies was found.

Metabolomics face a number of challenges. i) Data analysis: Metabolomics results contain vast and complex data to analyse, which is one of the challenges. Computational tools and expertise such as websites and programs are needed to analyse the data. ii) Standardization: Metabolomic analysis involves multiple steps, including data analysis, sample preparation, and data acquisition. These steps need to be optimised to give us protocols to produce the same results accurately. iii) Analytical variability: Metabolomics is a susceptible technique, and slight variations in sample preparation or data acquisition can lead to significant differences in results. This variability can confer difficulty in reproducing results between laboratories and in developing robust and reliable biomarkers. Despite these challenges, metabolomics has the potential to revolutionise cancer research and improve patient outcomes. According to the valuable data released from the original work, it will help in accurate diagnosis and early detection of the BC. The future plan of this article aim to produce an exact phenotype for BC detection tool.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the Fundamental Research Grant Scheme (grant no. 203/PPSP/6171345) of the Ministry of Higher Education.

Availability of data and materials

Not applicable.

Authors' contributions

MMBY, MM, WNBWA, RAR, WFWAR and TADAATD conceptualized the study. OMA, SSBM, NARBMR, NFABBH and LHY prepared the original draft. OMA, MMBY, MM, WNBWA, RAR, WFWAR, SSBM, NARBMR, NFABBH, LHY and TADAATD wrote, reviewed and edited the manuscript. All authors revised the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Jobard E, Dossus L, Baglietto L, Fornili M, Lécuyer L, Mancini FR, Gunter MJ, Trédan O, Boutron-Ruault MC, Elena-Herrmann B, et al: Investigation of circulating metabolites associated with breast cancer risk by untargeted metabolomics: A case-control study nested within the French E3N cohort. Br J Cancer. 124:1734–1743. 2021.PubMed/NCBI View Article : Google Scholar

2 

Wang Q and Xu R: MetabolitePredict: A de novo human metabolomics prediction system and its applications in rheumatoid arthritis. J Biomed Inform. 71:222–228. 2017.PubMed/NCBI View Article : Google Scholar

3 

Silva CL, Olival A, Perestrelo R, Silva P, Tomás H and Câmara JS: Untargeted urinary1H NMR-based metabolomic pattern as a potential platform in breast cancer detection. Metabolites. 9(269)2019.PubMed/NCBI View Article : Google Scholar

4 

Morad HM, Abou-Elzahab MM, Aref S and El-Sokkary AMA: Diagnostic value of 1H NMR-based metabolomics in acute lymphoblastic leukemia, acute myeloid leukemia, and breast cancer. ACS Omega. 7:8128–8140. 2022.PubMed/NCBI View Article : Google Scholar

5 

Patel A: Benign vs malignant tumors. JAMA Oncol. 6(1488)2020.PubMed/NCBI View Article : Google Scholar

6 

Britt KL, Cuzick J and Phillips KA: Key steps for effective breast cancer prevention. Nat Rev Cancer. 20:417–436. 2020.PubMed/NCBI View Article : Google Scholar

7 

Ferlay J, Colombet M, Soerjomataram I, Parkin DM, Piñeros M, Znaor A and Bray F: Cancer statistics for the year 2020: An overview. Int J Cancer. 149:778–789. 2021.PubMed/NCBI View Article : Google Scholar

8 

Anastasiadi Z, Lianos GD, Ignatiadou E, Harissis HV and Mitsis M: Breast cancer in young women: An overview. Updates Surg. 69:313–317. 2017.PubMed/NCBI View Article : Google Scholar

9 

Nicolini A, Ferrari P and Duffy MJ: Prognostic and predictive biomarkers in breast cancer: Past, present and future. Semin Cancer Biol. 52:56–73. 2018.PubMed/NCBI View Article : Google Scholar

10 

Li J, Guan X, Fan Z, Ching LM, Li Y, Wang X, Cao WM and Liu DX: Non-invasive biomarkers for early detection of breast cancer. Cancers (Basel). 12(2767)2020.PubMed/NCBI View Article : Google Scholar

11 

Barzaman K, Karami J, Zarei Z, Hosseinzadeh A, Kazemi MH, Moradi-Kalbolandi S, Safari E and Farahmand L: Breast cancer: Biology, biomarkers, and treatments. Int Immunopharmacol. 84(106535)2020.PubMed/NCBI View Article : Google Scholar

12 

Barzaman K, Karami J, Zarei Z, Hosseinzadeh A, Kazemi MH, Moradi-Kalbolandi S, Safari E and Farahmand L: Breast cancer: Biology, biomarkers, and treatments. Int Immunopharmacol. 84(106535)2020.PubMed/NCBI View Article : Google Scholar

13 

Waks AG and Winer EP: Breast cancer treatment: A review. JAMA. 321:288–300. 2019.PubMed/NCBI View Article : Google Scholar

14 

Sharma GN, Dave R, Sanadya J, Sharma P and Sharma KK: Various types and management of breast cancer: An overview. J Adv Pharm Technol Res. 1:109–126. 2010.PubMed/NCBI

15 

Akram M, Iqbal M, Daniyal M and Khan AU: Awareness and current knowledge of breast cancer. Biol Res. 50(33)2017.PubMed/NCBI View Article : Google Scholar

16 

Feng Y, Spezia M, Huang S, Yuan C, Zeng Z, Zhang L, Ji X, Liu W, Huang B, Luo W, et al: Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis. 5:77–106. 2018.PubMed/NCBI View Article : Google Scholar

17 

Thirumurugan D, Cholarajan A, Raja SSS and Vijayakumar R: An introductory chapter: Secondary metabolites. In: Vijayakumar R, Raja SSS (edis). Secondary Metabolites-Sources and Applications. Crotia: InTech-Open Science, pp138, 2018.

18 

Chen H and Wang L: Sugar strategies for biomass biochemical conversion. Technologies for Biochemical Conversion of Biomass. Metallurgical Industry Press, pp137-164, 2017.

19 

Abdel-Aziz SM, Abo Elsoud MM and Anise AAH: Microbial biosynthesis: A repertory of vital natural products. Food Biosynthesis. Elsevier Inc., 25-54, 2017.

20 

Tiwari R and Rana CS: Plant secondary metabolites: A review. Int J Eng Res Gen Sci. 3:661–670. 2015.

21 

Chandran H, Meena M, Barupal T and Sharma K: Plant tissue culture as a perpetual source for production of industrially important bioactive compounds. Biotechnol Rep (Amst). 26(e00450)2020.PubMed/NCBI View Article : Google Scholar

22 

Abdullah MA, Bahamid AAA, Alshajrawi OMS, Nazir MS and Tahir Z: Integrated biomaterials engineering of oil palm fibres and microalgae for bioenergy, environmental remediation, and conversion into value-added-products. IOP Conf Ser Earth Environ Sci. 448(012091)2020.

23 

Jones DP, Park Y and Ziegler TR: Nutritional metabolomics: Progress in addressing complexity in diet and health. Annu Rev Nutr. 32:183–202. 2012.PubMed/NCBI View Article : Google Scholar

24 

Hanna VS and Hafez EAA: Synopsis of arachidonic acid metabolism: A review. J Adv Res. 11:23–32. 2018.PubMed/NCBI View Article : Google Scholar

25 

Pezzuto F, Buonaguro L, Buonaguro FM and Tornesello ML: The role of circulating free DNA and MicroRNA in non-invasive diagnosis of HBV- and HCV-related hepatocellular carcinoma. Int J Mol Sci. 19(1007)2018.PubMed/NCBI View Article : Google Scholar

26 

Lu T and Li J: Clinical applications of urinary cell-free DNA in cancer: Current insights and promising future. Am J Cancer Res. 7:2318–2332. 2017.PubMed/NCBI

27 

Nam H, Chung BC, Kim Y, Lee KY and Lee D: Combining tissue transcriptomics and urine metabolomics for breast cancer biomarker identification. Bioinformatics. 25:3151–3157. 2009.PubMed/NCBI View Article : Google Scholar

28 

Henneges C, Bullinger D, Fux R, Friese N, Seeger H, Neubauer H, Laufer S, Gleiter CH, Schwab M, Zell A and Kammerer B: Prediction of breast cancer by profiling of urinary RNA metabolites using Support Vector Machine-based feature selection. BMC Cancer. 9(104)2009.PubMed/NCBI View Article : Google Scholar

29 

Dinges SS, Hohm A, Vandergrift LA, Nowak J, Habbel P, Kaltashov IA and Cheng LL: Cancer metabolomic markers in urine: Evidence, techniques and recommendations. Nat Rev Urol. 16:339–362. 2019.PubMed/NCBI View Article : Google Scholar

30 

Frickenschmidt A, Frohlich H, Bullinger D, Zell A, Laufer S, Gleiter CH, Liebich H and Kammerer B: Metabonomics in cancer diagnosis: Mass spectrometry-based profiling of urinary nucleosides from breast cancer patients. Biomarkers. 13:435–449. 2008.PubMed/NCBI View Article : Google Scholar

31 

Chen Y, Zhang R, Song Y, He J, Sun J, Bai J, An Z, Dong L, Zhan Q and Abliz Z: RRLC-MS/MS-based metabonomics combined with in-depth analysis of metabolic correlation network: Finding potential biomarkers for breast cancer. Analyst. 134:2003–2011. 2009.PubMed/NCBI View Article : Google Scholar

32 

Cho SH, Jung BH, Lee SH, Lee WY, Kong G and Chung BC: Direct determination of nucleosides in the urine of patients with breast cancer using column-switching liquid chromatography-tandem mass spectrometry. Biomed Chromatogr. 20:1229–1236. 2006.PubMed/NCBI View Article : Google Scholar

33 

Lécuyer L, Victor Bala A, Deschasaux M, Bouchemal N, Nawfal Triba M, Vasson MP, Rossary A, Demidem A, Galan P, Hercberg S, et al: NMR metabolomic signatures reveal predictive plasma metabolites associated with long-term risk of developing breast cancer. Int J Epidemiol. 47:484–494. 2018.PubMed/NCBI View Article : Google Scholar

34 

Lecuyer L, Dalle C, Lyan B, Demidem A, Rossary A, Vasson MP, Petera M, Lagree M, Ferreira T, Centeno D, et al: Plasma metabolomic signatures associated with long-term breast cancer risk in the SU.VI.MAX prospective cohort. Cancer Epidemiol Biomarkers Prev. 28:1300–1307. 2019.PubMed/NCBI View Article : Google Scholar

35 

An R, Yu H, Wang Y, Lu J, Gao Y, Xie X and Zhang J: Integrative analysis of plasma metabolomics and proteomics reveals the metabolic landscape of breast cancer. Cancer Metab. 10(13)2022.PubMed/NCBI View Article : Google Scholar

36 

Gasparri ML, Casorelli A, Bardhi E, Besharat AR, Savone D, Ruscito I, Farooqi AA, Papadia A, Mueller MD, Ferretti E and Benedetti Panici P: Beyond circulating microRNA biomarkers: Urinary microRNAs in ovarian and breast cancer. Tumour Biol. 39(1010428317695525)2017.PubMed/NCBI View Article : Google Scholar

37 

Ilyas MN, Ab A, Al-Hatamleh MAI, Al-Shajrawi OM, Ariff TM and Simbak N: Rising trends of obesity in Malaysia; role of inflammation and inflammatory markers in obesity related insulin resistance: A nuclear factor kappa B (Nfkb) perspective. Exp Clin Endocrinol Diabetes. 109:S135–S148. 2017.

38 

Rudnicka E, Suchta K, Grymowicz M, Calik-Ksepka A, Smolarczyk K, Duszewska AM, Smolarczyk R and Meczekalski B: Chronic low grade inflammation in pathogenesis of PCOS. Int J Mol Sci. 22(3789)2021.PubMed/NCBI View Article : Google Scholar

39 

Slupsky CM, Steed H, Wells TH, Dabbs K, Schepansky A, Capstick V, Faught W and Sawyer MB: Urine metabolite analysis offers potential early diagnosis of ovarian and breast cancers. Clin Cancer Res. 16:5835–5841. 2010.PubMed/NCBI View Article : Google Scholar

40 

Bax C, Lotesoriere BJ, Sironi S and Capelli L: Review and comparison of cancer biomarker trends in urine as a basis for new diagnostic pathways. Cancers (Basel). 11(1244)2019.PubMed/NCBI View Article : Google Scholar

41 

Cala M, Aldana J, Sánchez J, Guio J and Meesters RJW: Urinary metabolite and lipid alterations in Colombian Hispanic women with breast cancer: A pilot study. J Pharm Biomed Anal. 152:234–241. 2018.PubMed/NCBI View Article : Google Scholar

42 

Pasikanti KK, Esuvaranathan K, Hong Y, Ho PC, Mahendran R, Raman Nee Mani L, Chiong E and Chan EC: Urinary metabotyping of bladder cancer using two-dimensional gas chromatography time-of-flight mass spectrometry. J Proteome Res. 12:3865–3873. 2013.PubMed/NCBI View Article : Google Scholar

43 

Silva C, Perestrelo R, Silva P, Tomás H and Câmara JS: Breast cancer metabolomics: From analytical platforms to multivariate data analysis. A review. Metabolites. 9(102)2019.PubMed/NCBI View Article : Google Scholar

44 

Putluri N, Shojaie A, Vasu VT, Vareed SK, Nalluri S, Putluri V, Thangjam GS, Panzitt K, Tallman CT, Butler C, et al: Metabolomic profiling reveals potential markers and bioprocesses altered in bladder cancer progression. Cancer Res. 71:7376–7386. 2011.PubMed/NCBI View Article : Google Scholar

45 

Yu L, Jiang C, Huang S, Gong X, Wang S and Shen P: Analysis of urinary metabolites for breast cancer patients receiving chemotherapy by CE-MS coupled with on-line concentration. Clin Biochem. 46:1065–1073. 2013.PubMed/NCBI View Article : Google Scholar

46 

Woo HM, Kim KM, Choi MH, Jung BH, Lee J, Kong G, Nam SJ, Kim S, Bai SW and Chung BC: Mass spectrometry based metabolomic approaches in urinary biomarker study of women's cancers. Clin Chim Acta. 400:63–69. 2009.PubMed/NCBI View Article : Google Scholar

47 

Men Y, Li L, Zhang F, Kong X, Zhang W, Hao C and Wang G: Evaluation of heavy metals and metabolites in the urine of patients with breast cancer. Oncol Lett. 19:1331–1337. 2020.PubMed/NCBI View Article : Google Scholar

48 

Asiago VM, Alvarado LZ, Shanaiah N, Gowda GAN, Owusu-Sarfo K, Ballas RA and Raftery D: Early detection of recurrent breast cancer using metabolite profiling. Cancer Res. 70:8309–8318. 2010.PubMed/NCBI View Article : Google Scholar

49 

Oakman C, Tenori L, Claudino WM, Cappadona S, Nepi S, Battaglia A, Bernini P, Zafarana E, Saccenti E, Fornier M, et al: Identification of a serum-detectable metabolomic fingerprint potentially correlated with the presence of micrometastatic disease in early breast cancer patients at varying risks of disease relapse by traditional prognostic methods. Ann Oncol. 22:1295–1301. 2011.PubMed/NCBI View Article : Google Scholar

50 

Tenori L, Oakman C, Claudino WM, Bernini P, Cappadona S, Nepi S, Biganzoli L, Arbushites MC, Luchinat C, Bertini I and Di Leo A: Exploration of serum metabolomic profiles and outcomes in women with metastatic breast cancer: A pilot study. Mol Oncol. 6:437–444. 2012.PubMed/NCBI View Article : Google Scholar

51 

Wei S, Liu L, Zhang J, Bowers J, Gowda GAN, Seeger H, Fehm T, Neubauer HJ, Vogel U, Clare SE and Raftery D: Metabolomics approach for predicting response to neoadjuvant chemotherapy for breast cancer. Mol Oncol. 7:297–307. 2013.PubMed/NCBI View Article : Google Scholar

52 

Jobard E, Pontoizeau C, Blaise BJ, Bachelot T, Elena-Herrmann B and Trédan O: A serum nuclear magnetic resonance-based metabolomic signature of advanced metastatic human breast cancer. Cancer Lett. 343:33–41. 2014.PubMed/NCBI View Article : Google Scholar

53 

Tenori L, Oakman C, Morris PG, Gralka E, Turner N, Cappadona S, Fornier M, Hudis C, Norton L, Luchinat C and Di Leo A: Serum metabolomic profiles evaluated after surgery may identify patients with oestrogen receptor negative early breast cancer at increased risk of disease recurrence. Results from a retrospective study. Mol Oncol. 9:128–139. 2015.PubMed/NCBI View Article : Google Scholar

54 

Kim Y, Koo I, Jung BH, Chung BC and Lee D: Multivariate classification of urine metabolome profiles for breast cancer diagnosis. BMC Bioinformatics. 11 (Suppl 2)(S4)2010.PubMed/NCBI View Article : Google Scholar

55 

Bathen TF, Geurts B, Sitter B, Fjøsne HE, Lundgren S, Buydens LM, Gribbestad IS, Postma G and Giskeødegård GF: Feasibility of MR metabolomics for immediate analysis of resection margins during breast cancer surgery. PLoS One. 8(e61578)2013.PubMed/NCBI View Article : Google Scholar

56 

Borgan E, Sitter B, Lingjærde OC, Johnsen H, Lundgren S, Bathen TF, Sørlie T, Børresen-Dale AL and Gribbestad IS: Merging transcriptomics and metabolomics-advances in breast cancer profiling. BMC Cancer. 10(628)2010.PubMed/NCBI View Article : Google Scholar

57 

Debik J, Euceda LR, Lundgren S, Gythfeldt HDVL, Garred Ø, Borgen E, Engebraaten O, Bathen TF and Giskeødegård GF: Assessing treatment response and prognosis by serum and tissue metabolomics in breast cancer patients. J Proteome Res. 18:3649–3660. 2019.PubMed/NCBI View Article : Google Scholar

58 

Haukaas TH, Euceda LR, Giskeødegård GF, Lamichhane S, Krohn M, Jernström S, Aure MR, Lingjærde OC, Schlichting E, Garred Ø, et al: Metabolic clusters of breast cancer in relation to gene- and protein expression subtypes. Cancer Metab. 4(12)2016.PubMed/NCBI View Article : Google Scholar

59 

Chae EY, Shin HJ, Kim S, Baek HM, Yoon D, Kim S, Shim YE, Kim HH, Cha JH, Choi WJ, et al: The role of high-resolution magic angle spinning 1H nuclear magnetic resonance spectroscopy for predicting the invasive component in patients with ductal carcinoma in situ diagnosed on preoperative biopsy. PLoS One. 11(e0161038)2016.PubMed/NCBI View Article : Google Scholar

60 

Euceda LR, Haukaas TH, Giskeødegård GF, Vettukattil R, Engel J, Silwal-Pandit L, Lundgren S, Borgen E, Garred Y, Garred G, et al: Evaluation of metabolomic changes during neoadjuvant chemotherapy combined with bevacizumab in breast cancer using MR spectroscopy. Metabolomics. 13(37)2017.

61 

Cala MP, Aldana J, Medina J, Sánchez J, Guio J, Wist J and Meesters RJW: Multiplatform plasma metabolic and lipid fingerprinting of breast cancer: A pilot control-case study in Colombian Hispanic women. PLoS One. 13(e0190958)2018.PubMed/NCBI View Article : Google Scholar

62 

Lécuyer L, Victor Bala A, Deschasaux M, Bouchemal N, Nawfal Triba M, Vasson MP, Rossary A, Demidem A, Galan P, Hercberg S, et al: NMR metabolomic signatures reveal predictive plasma metabolites associated with long-term risk of developing breast cancer. Int J Epidemiol. 47:484–494. 2018.PubMed/NCBI View Article : Google Scholar

63 

Suman S, Sharma RK, Kumar V, Sinha N and Shukla Y: Metabolic fingerprinting in breast cancer stages through 1H NMR spectroscopy-based metabolomic analysis of plasma. J Pharm Biomed Anal. 160:38–45. 2018.PubMed/NCBI View Article : Google Scholar

64 

Louis E, Bervoets L, Reekmans G, De Jonge E, Mesotten L, Thomeer M and Adriaensens P: Phenotyping human blood plasma by 1H-NMR: A robust protocol based on metabolite spiking and its evaluation in breast cancer. Metabolomics. 11:225–236. 2015.

65 

Vignoli A, Muraro E, Miolo G, Tenori L, Turano P, Di Gregorio E, Steffan A, Luchinat C and Corona G: Effect of estrogen receptor status on circulatory immune and metabolomics profiles of HER2-positive breast cancer patients enrolled for neoadjuvant targeted chemotherapy. Cancers (Basel). 12(314)2020.PubMed/NCBI View Article : Google Scholar

66 

McCartney A, Vignoli A, Tenori L, Fornier M, Rossi L, Risi E, Luchinat C, Biganzoli L and Di Leo A: Metabolomic analysis of serum may refine 21-gene expression assay risk recurrence stratification. NPJ Breast Cancer. 5(26)2019.PubMed/NCBI View Article : Google Scholar

67 

Jiang L, Lee SC and Ng TC: Pharmacometabonomics analysis reveals serum formate and acetate potentially associated with varying response to gemcitabine-carboplatin chemotherapy in metastatic breast cancer patients. J Proteome Res. 17:1248–1257. 2018.PubMed/NCBI View Article : Google Scholar

68 

Wojtowicz W, Wróbel A, Pyziak K, Tarkowski R, Balcerzak A, Bębenek M and Młynarz P: Evaluation of MDA-MB-468 cell culture media analysis in predicting triple-negative breast cancer patient sera metabolic profiles. Metabolites. 10(173)2020.PubMed/NCBI View Article : Google Scholar

69 

Men Y, Li L, Zhang F, Kong X, Zhang W, Hao C, et al: Evaluation of heavy metals and metabolites in the urine of patients with breast cancer. Oncol Lett. 19:1331–1337. 2020.PubMed/NCBI View Article : Google Scholar

70 

Li N, Deng Y, Zhou L, Tian T, Yang S, Wu Y, Zheng Y, Zhai Z, Hao Q, Song D, et al: Global burden of breast cancer and attributable risk factors in 195 countries and territories, from 1990 to 2017: Results from the Global Burden of Disease Study 2017. J Hematol Oncol. 12(140)2019.PubMed/NCBI View Article : Google Scholar

71 

Slupsky CM, Steed H, Wells TH, Dabbs K, Schepansky A, Capstick V, Faught W and Sawyer MB: Urine metabolite analysis offers potential early diagnosis of ovarian and breast cancers. Clin Cancer Res. 16:5835–5841. 2010.PubMed/NCBI View Article : Google Scholar

72 

Cairns RA, Harris IS and Mak TW: Regulation of cancer cell metabolism. Nat Rev Cancer. 11:85–95. 2011.PubMed/NCBI View Article : Google Scholar

73 

Lau AN and Vander Heiden MG: Metabolism in the tumor microenvironment. Annu Rev Cancer Biol. 4:17–40. 2020.

74 

Phan LM, Yeung SCJ and Lee MH: Cancer metabolic reprogramming: Importance, main features, and potentials for precise targeted anti-cancer therapies. Cancer Biol Med. 11:1–19. 2014.PubMed/NCBI View Article : Google Scholar

75 

Sullivan LB, Gui DY and Van Der Heiden MG: Altered metabolite levels in cancer: Implications for tumour biology and cancer therapy. Nat Rev Cancer. 16:680–693. 2016.PubMed/NCBI View Article : Google Scholar

76 

Gu I, Gregory E, Atwood C, Lee SO and Song YH: Exploring the role of metabolites in cancer and the associated nerve crosstalk. Nutrients. 14(1722)2022.PubMed/NCBI View Article : Google Scholar

77 

Potter M, Newport E and Morten KJ: The Warburg effect: 80 Years on. Biochem Soc Trans. 44:1499–1505. 2016.PubMed/NCBI View Article : Google Scholar

78 

Nalbantoglu S and Karadag A: Metabolomics bridging proteomics along metabolites/oncometabolites and protein modifications: Paving the way toward integrative multiomics. J Pharm Biomed Anal. 199(114031)2021.PubMed/NCBI View Article : Google Scholar

79 

Kinnaird A, Zhao S, Wellen KE and Michelakis ED: Metabolic control of epigenetics in cancer. Nat Rev Cancer. 16:694–707. 2016.PubMed/NCBI View Article : Google Scholar

80 

Al-Shajrawi OM, Basit E and Baig AA: HIF1 (rs11549465) and NFKB1 (rs28362491) variants association with obesity in Malaysia. Meta Gene. 25(100753)2020.

81 

Choi YK and Park KG: Targeting glutamine metabolism for cancer treatment. Biomol Ther (Seoul). 26:19–28. 2018.PubMed/NCBI View Article : Google Scholar

82 

Butler M, van der Meer LT and van Leeuwen FN: Amino acid depletion therapies: Starving cancer cells to death. Trends Endocrinol Metab. 32:367–381. 2021.PubMed/NCBI View Article : Google Scholar

83 

Wang ZH, Peng WB, Zhang P, Yang XP and Zhou Q: Lactate in the tumour microenvironment: From immune modulation to therapy. EBioMedicine. 73(103627)2021.PubMed/NCBI View Article : Google Scholar

84 

Doldo E, Costanza G, Agostinelli S, Tarquini C, Ferlosio A, Arcuri G, Passeri D, Scioli MG and Orlandi A: Vitamin A, cancer treatment and prevention: The new role of cellular retinol binding proteins. Biomed Res Int. 2015(624627)2015.PubMed/NCBI View Article : Google Scholar

85 

Zeng LH, Wang QM, Feng LY, Ke YD, Xu QZ, Wei AY, Zhang C and Ying RB: High-dose vitamin C suppresses the invasion and metastasis of breast cancer cells via inhibiting epithelial-mesenchymal transition. Onco Targets Ther. 12:7405–7413. 2019.PubMed/NCBI View Article : Google Scholar

86 

Sailo BL, Banik K, Padmavathi G, Javadi M, Bordoloi D and Kunnumakkara AB: Tocotrienols: The promising analogues of vitamin E for cancer therapeutics. Pharmacol Res. 130:259–272. 2018.PubMed/NCBI View Article : Google Scholar

87 

Miyazawa S, Moriya S, Kokuba H, Hino H, Takano N and Miyazawa K: Vitamin K2 induces non-apoptotic cell death along with autophagosome formation in breast cancer cell lines. Breast Cancer. 27:225–235. 2020.PubMed/NCBI View Article : Google Scholar

88 

Qiu F, Chen YR, Liu X, Chu CY, Shen LJ, Xu J, Gaur S, Forman HJ, Zhang H, Zheng S, et al: Arginine starvation impairs mitochondrial respiratory function in ASS1-deficient breast cancer cells. Sci Signal. 7(ra31)2014.PubMed/NCBI View Article : Google Scholar

89 

Cheng CT, Qi Y, Wang YC, Chi KK, Chung Y, Ouyang C, Chen YR, Oh ME, Sheng X, Tang Y, et al: Arginine starvation kills tumor cells through aspartate exhaustion and mitochondrial dysfunction. Commun Biol. 1(178)2018.PubMed/NCBI View Article : Google Scholar

90 

Ji JX, Cochrane DR, Tessier-Cloutier B, Chen SY, Ho G, Pathak KV, Alcazar IN, 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. 26:4402–4413. 2020.PubMed/NCBI View Article : Google Scholar

91 

Krall AS, Xu S, Graeber TG, Braas D and Christofk HR: Asparagine promotes cancer cell proliferation through use as an amino acid exchange factor. Nat Commun. 7(11457)2016.PubMed/NCBI View Article : Google Scholar

92 

Hoang G, Udupa S and Le A: Application of metabolomics technologies toward cancer prognosis and therapy. Int Rev Cell Mol Biol. 347:191–223. 2019.PubMed/NCBI View Article : Google Scholar

93 

Elgogary A, Xu Q, Poore B, Alt J, Zimmermann SC, Zhao L, Fu J, Chen B, Xia S, Liu Y, et al: Combination therapy with BPTES nanoparticles and metformin targets the metabolic heterogeneity of pancreatic cancer. Proc Natl Acad Sci USA. 113:E5328–E5336. 2016.PubMed/NCBI View Article : Google Scholar

94 

Lane AN, Fan TWM and Higashi RM: Isotopomer-based metabolomic analysis by NMR and mass spectrometry. Methods Cell Biol. 84:541–588. 2008.PubMed/NCBI View Article : Google Scholar

95 

Le A, Lane AN, Hamaker M, Bose S, Gouw A, Barbi J, Tsukamoto T, Rojas CJ, Slusher BS, Zhang H, et al: Glucose-independent glutamine metabolism via TCA cycling for proliferation and survival in B cells. Cell Metab. 15:110–121. 2012.PubMed/NCBI View Article : Google Scholar

96 

Beger R: A review of applications of metabolomics in cancer. Metabolites. 3:552–574. 2013.PubMed/NCBI View Article : Google Scholar

97 

Zhang A, Sun H, Wang P, Han Y and Wang X: Modern analytical techniques in metabolomics analysis. Analyst. 137:293–300. 2012.PubMed/NCBI View Article : Google Scholar

98 

Lyon DE, Starkweather A, Yao Y, Garrett T, Kelly DL, Menzies V, Dereziński P, Datta S, Kumar S and Jackson-Cook C: Pilot study of metabolomics and psychoneurological symptoms in women with early stage breast cancer. Biol Res Nurs. 20:227–236. 2018.PubMed/NCBI View Article : Google Scholar

99 

Silva CL, Perestrelo R, Silva P, Tomás H and Câmara JS: Volatile metabolomic signature of human breast cancer cell lines. Sci Rep. 7(43969)2017.PubMed/NCBI View Article : Google Scholar

100 

Cífková E, Lísa M, Hrstka R, Vrána D, Gatěk J, Melichar B and Holčapek M: Correlation of lipidomic composition of cell lines and tissues of breast cancer patients using hydrophilic interaction liquid chromatography/electrospray ionization mass spectrometry and multivariate data analysis. Rapid Commun Mass Spectrom. 31:253–263. 2017.PubMed/NCBI View Article : Google Scholar

101 

Cavaco C, Pereira JAM, Taunk K, Taware R, Rapole S, Nagarajaram H and Câmara JS: Screening of salivary volatiles for putative breast cancer discrimination: An exploratory study involving geographically distant populations. Anal Bioanal Chem. 410:445–468. 2018.PubMed/NCBI View Article : Google Scholar

102 

Porto-Figueira P, Pereira JAM and Câmara JS: Exploring the potential of needle trap microextraction combined with chromatographic and statistical data to discriminate different types of cancer based on urinary volatomic biosignature. Anal Chim Acta. 1023:53–63. 2018.PubMed/NCBI View Article : Google Scholar

103 

Budczies J, Brockmöller SF, Müller BM, Barupal DK, Richter-Ehrenstein C, Kleine-Tebbe A, Griffin JL, Orešič M, Dietel M, Denkert C and Fiehn O: Comparative metabolomics of estrogen receptor positive and estrogen receptor negative breast cancer: Alterations in glutamine and beta-alanine metabolism. J Proteomics. 94:279–288. 2013.PubMed/NCBI View Article : Google Scholar

104 

Dougan MM, Li Y, Chu LW, Haile RW, Whittemore AS, Han SS, Moore SC, Sampson JN, Andrulis IL, John EM and Hsing AW: Metabolomic profiles in breast cancer: A pilot case-control study in the breast cancer family registry. BMC Cancer. 18(532)2018.PubMed/NCBI View Article : Google Scholar

105 

Sas KM, Karnovsky A, Michailidis G and Pennathur S: Metabolomics and diabetes: Analytical and computational approaches. Diabetes. 64:718–732. 2015.PubMed/NCBI View Article : Google Scholar

106 

Ahad T and Nissar J: Fingerprinting in determining the adultration of food. J Pharmacogn Phytochem. 6:1543–1553. 2017.

107 

Claudino WM, Goncalves PH, di Leo A, Philip PA and Sarkar FH: Metabolomics in cancer: A bench-to-bedside intersection. Crit Rev Oncol Hematol. 84:1–7. 2012.PubMed/NCBI View Article : Google Scholar

108 

De Castro F, Benedetti M, Del Coco L and Fanizzi FP: NMR-based metabolomics in metal-based drug research. Molecules. 24(2240)2019.PubMed/NCBI View Article : Google Scholar

109 

Cacciola F, Farnetti S, Dugo P, Marriott PJ and Mondello L: Comprehensive two-dimensional liquid chromatography for polyphenol analysis in foodstuffs. J Sep Sci. 40:7–24. 2017.PubMed/NCBI View Article : Google Scholar

110 

Emwas AHM: The strengths and weaknesses of NMR spectroscopy and mass spectrometry with particular focus on metabolomics research. Methods Mol Biol. 1277:161–193. 2015.PubMed/NCBI View Article : Google Scholar

111 

Beltran A, Suarez M, Rodríguez MA, Vinaixa M, Samino S, Arola L, Correig X and Yanes O: Assessment of compatibility between extraction methods for NMR- and LC/MS-based metabolomics. Anal Chem. 84:5838–5844. 2012.PubMed/NCBI View Article : Google Scholar

112 

Emwas AH, Roy R, McKay RT, Tenori L, Saccenti E, Gowda GAN, Raftery D, Alahmari F, Jaremko L, Jaremko M and Wishart DS: Nmr spectroscopy for metabolomics research. Metabolites. 9(123)2019.PubMed/NCBI View Article : Google Scholar

113 

Dunn WB, Broadhurst DI, Atherton HJ, Goodacre R and Griffin JL: Systems level studies of mammalian metabolomes: The roles of mass spectrometry and nuclear magnetic resonance spectroscopy. Chem Soc Rev. 40:387–426. 2011.PubMed/NCBI View Article : Google Scholar

114 

Qiu S, Cai Y, Yao H, Lin C, Xie Y, Tang S and Zhang A: Small molecule metabolites: Discovery of biomarkers and therapeutic targets. Signal Transduct Target Ther. 8(132)2023.PubMed/NCBI View Article : Google Scholar

115 

Burke HB: Predicting clinical outcomes using molecular biomarkers. Biomark Cancer. 8:89–99. 2016.PubMed/NCBI View Article : Google Scholar

116 

Carlomagno N, Incollingo P, Tammaro V, Peluso G, Rupealta N, Chiacchio G, Sandoval Sotelo ML, Minieri G, Pisani A, Riccio E, et al: Diagnostic, predictive, prognostic, and therapeutic molecular biomarkers in third millennium: A breakthrough in gastric cancer. Biomed Res Int. 2017(7869802)2017.PubMed/NCBI View Article : Google Scholar

117 

Mayeux R: Biomarkers: Potential uses and limitations. NeuroRx. 1:182–188. 2004.PubMed/NCBI View Article : Google Scholar

118 

Duffy MJ, Harbeck N, Nap M, Molina R, Nicolini A, Senkus E and Cardoso F: Clinical use of biomarkers in breast cancer: Updated guidelines from the European Group on Tumor Markers (EGTM). Eur J Cancer. 75:284–298. 2017.PubMed/NCBI View Article : Google Scholar

119 

Brennan M and Lim B: The actual role of receptors as cancer markers, biochemical and clinical aspects: Receptors in breast cancer. Adv Exp Med Biol. 867:327–337. 2015.PubMed/NCBI View Article : Google Scholar

120 

Li Z, Wei H, Li S, Wu P and Mao X: The role of progesterone receptors in breast cancer. Drug Des Devel Ther. 16:305–314. 2022.PubMed/NCBI View Article : Google Scholar

121 

Brisken C, Hess K and Jeitziner R: Progesterone and overlooked endocrine pathways in breast cancer pathogenesis. Endocrinology. 156:3442–3450. 2015.PubMed/NCBI View Article : Google Scholar

122 

Trabert B, Sherman ME, Kannan N and Stanczyk FZ: Progesterone and breast cancer. Endocr Rev. 41:320–344. 2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2025
Volume 22 Issue 4

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Alshajrawi OM, Tengku Din T, Marzuki S, Maulidiani M, Mohd Rusli N, Badrol Hisham N, Hui Ying L, Yahya M, Wan Azman W, Ramli RA, Ramli RA, et al: Exploring the complex relationship between metabolomics and breast cancer early detection (Review). Mol Clin Oncol 22: 35, 2025.
APA
Alshajrawi, O.M., Tengku Din, T., Marzuki, S., Maulidiani, M., Mohd Rusli, N., Badrol Hisham, N. ... Wan Abdul Rahman, W. (2025). Exploring the complex relationship between metabolomics and breast cancer early detection (Review). Molecular and Clinical Oncology, 22, 35. https://doi.org/10.3892/mco.2025.2830
MLA
Alshajrawi, O. M., Tengku Din, T., Marzuki, S., Maulidiani, M., Mohd Rusli, N., Badrol Hisham, N., Hui Ying, L., Yahya, M., Wan Azman, W., Ramli, R. A., Wan Abdul Rahman, W."Exploring the complex relationship between metabolomics and breast cancer early detection (Review)". Molecular and Clinical Oncology 22.4 (2025): 35.
Chicago
Alshajrawi, O. M., Tengku Din, T., Marzuki, S., Maulidiani, M., Mohd Rusli, N., Badrol Hisham, N., Hui Ying, L., Yahya, M., Wan Azman, W., Ramli, R. A., Wan Abdul Rahman, W."Exploring the complex relationship between metabolomics and breast cancer early detection (Review)". Molecular and Clinical Oncology 22, no. 4 (2025): 35. https://doi.org/10.3892/mco.2025.2830