The application of MS-based lipidomics provides a valid integrative approach to studying pharmacometabolomic changes because it gives information about whole lipid metabolism pathways that can be closely correlated to the other metabolic networks in the understanding of all mechanisms mediating statin effects
[42][43]. In a recent study, the potential molecular mechanisms underlying the association between metabolic improvement and microbiota composition following simvastatin treatment were investigated to explain the gut microbiome involvement in the statin response variability
[44]. A metabolomic profiling using an ultra-high-performance liquid chromatography (UHPLC) system coupled with a hybrid triple quadrupole TOF (Q-TOF) mass spectrometer was performed to study the interactions of endogenous serum metabolites with the gut microbiota following simvastatin treatment in high-lipid diet-induced hyperlipidemic rats. Differential endogenous metabolites were identified that affected the metabolism of amino acids (phenylalanine and tyrosine), unsaturated fatty acids (linoleic acid and 9-hydroxyoctadecadienoic acid), and the functions of gut microbial metabolism (
m-coumaric acid and 3-(2-hydroxyphenyl) propionic acid)
[44]. These data suggested that simvastatin therapy strongly modulates the serum metabolic profile in hyperlipidemic rats, and, since these metabolic pathways are involved in gut flora interactions, they could be potential therapeutic targets for the improvement of simvastatin hypolipidemic efficacy. Indeed, hyperlipidemia is a metabolic syndrome that is commonly linked to cardiovascular diseases. Phenylalanine is a nutrient precursor for gut microbiota-generated metabolites, which are known to be associated with cardiovascular diseases and adverse cardiovascular events, while tyrosine promotes lipid metabolism, therefore representing a potential biomarker for hyperlipidemia
[44]. Both phenylalanine and tyrosine showed increased levels following statin treatment. In the same way, levels of both linoleic acid and 9-hydroxyoctadecadienoic acid were significantly increased after simvastatin therapy, which confirms their beneficial effects against cardiovascular risk, including hyperlipidemia and hypertension
[44]. The concentration of the metabolites of the gut microflora,
m-coumaric acid and a derivative of phenylpropionic acid, were also higher in hyperlipidemic rats after statin treatment, showing their antilipogenic and cholesterol-lowering properties
[44].
3.1.3. Adverse Effects of Statins
Statins are highly effective and safe for most people, but they can cause minor or severe side effects that should never be neglected. Statin-related myotoxicity, for example, can range from mild muscle pain up to rhabdomyolysis, which is a serious and fatal disorder that sometimes occurs in patients following pharmacological treatment [45]. Statin-associated rhabdomyolysis risk has been reported as dose-dependent and concentration-dependent [46].
Metabolomics provides an accurate signature of all metabolite changes in biological fluids, cells, and tissues that can be a source for biomarker discovery. A metabolomic analysis of skeletal muscle and plasma using LC-MS and GC-MS was performed on a rat model treated with two myotoxicants, cerivastatin and tetramethyl-p-phenylenediamine, to induce a skeletal injury and identify candidate biomarkers for skeletal muscle toxicity [47]. They observed in skeletal muscle a significant increase in 2-hydroxyglutarate in cerivastatin-treated rats and hexanoylcarnitine in both types of treated rats. These increases were also measured in plasma samples at different times after dosing, demonstrating the possibility to use plasma 2-hydroxyglutarate and hexanoylcarnitine as valid and easily detectable biomarkers for the early detection of skeletal muscle toxicity in rats, with better sensitivity than the conventional markers creatine kinase and aspartate aminotransferase whose utility in clinics is limited due to their low diagnostic power [48]. Moreover, this study confirmed the importance and benefit of metabolomics for biomarker discovery in toxicological studies.
Among the potential statin-related adverse events, there is also an increased incidence of type II diabetes mellitus that can lead to premature discontinuation of treatment. Therefore, it is important to evaluate a correlation between statin-induced metabolic changes and statin-induced hyperglycemia and insulin resistance, to identify pre-drug treatment metabolites predictive of increased diabetic risk [49]. In this regard, a pharmacometabolomic study was performed by GC-TOF-MS on plasma pre- and post-treatment with simvastatin for 6 weeks from patients enrolled for the CAP study [49] to measure changes in intermediary metabolism and the associated high plasma glucose levels as a potentially adverse response to simvastatin. Some patients developed hyperglycemia and pre-diabetes, as well as a dysfunction of beta cells and insulin resistance in more than 50% of patients following statin therapy. An initial metabolic profile of simvastatin-induced insulin resistance was identified, including ethanolamine, hydroxylamine, hydroxycarbamate, and isoleucine, which can be predictive biomarkers of individuals at risk of developing a statin-induced new onset pre-type II diabetes mellitus [49]. In particular, the metabolite ethanolamine was identified as the most likely to predict simvastatin-induced diabetic risk, indicating that decarboxylation and oxidation were significantly associated with statin-induced hyperglycemia and insulin resistance. Pharmacometabolomics allows having a baseline metabolic signature before starting the drug therapy that can be then used to find predictive biomarkers able to stratify patients and to identify subjects who are at higher risk of adverse side effects, enabling personalized selection of the most appropriate medication for each patient and personalized monitoring of their prognosis.
3.1.4. Beneficial Effects of Statins
Although the possible side effects of therapy with statins are unpleasant, it is important not to forget the considerable benefits of taking them for the treatment of several pathologies. Metabonomics is used not only for clinical diagnosis, but also for evaluating the clinical course of a disease, prognosis, and treatment effect of drugs, such as statins
[50].
Many years ago, Ooga et al. performed
[51] a metabolic analysis in Watanabe heritable hyperlipidemic (WHHL) rabbits as a model of hypercholesterolemia to obtain a determination of all metabolite concentrations and a characterization of the metabolic imbalance of their pathological condition. Numerous metabolites were measured in plasma and several tissues from WHHL and healthy control rabbits using CE-TOF-MS and LC-TOF-MS systems. Several significant metabolic differences between the healthy and the pathological conditions were observed, and the metabolomic features observed in the pathological rabbit model including the modulation of glutathione and phosphatidylcholine metabolism showing advanced oxidative stress in several tissues, especially in the liver
[51].
Shifting the attention to human subjects with hypercholesterolemia, comprehensive cross-sectional profiling of lipids and metabolites was performed by Christensen et al. in children with and without familial hypercholesterolemia (FH) aiming to characterize the alterations associated with elevated LDL-C in FH patients
[52]. Elevated plasma cholesterol is the most important risk factor for atherosclerosis and cholesterol-lowering treatment with statins is required to stop or slow down atherosclerotic development in FH children. Plasma metabolites were measured by high-throughput NMR spectroscopy to compare the differences between statin-treated and non-statin-treated FH children, and healthy children
[52]. It is important to investigate hypercholesterolemia-associated metabolic aberrations in HF children to better understand the disease, and thereby improve the treatment of hypercholesterolemia in children and, more in general terms, the treatment of atherosclerotic processes. The scholars observed increased levels of atherogenic ApoB-containing lipoproteins and lipid fractions in both statin-treated and non-statin-treated FH children compared to healthy children. In addition, FH children showed alterations in HDL subfractions, and in particular, their small HDL particles were characterized by a higher content of cholesteryl esters, and lower levels of free cholesterol and phospholipids
[52].
Another common application of metabonomics is the study of the development and progression of diseases following a specific pharmacological treatment.
3.2. PCSK9 Inhibitors
PCSK9 inhibitors are pharmacological agents used to reduce blood LDL-C levels and improve cardiovascular outcomes both in primary and secondary prevention
[53]. They are human monoclonal antibodies that bind PCSK9 protein with high affinity to lower LDL-C concentrations by blocking the degradation of cholesterol receptors available on the hepatocyte cell surface, which are responsible for removing LDL-C from blood
[54]. PCSK9 inhibitors seem to show a more effective lipid-lowering profile than statins
[55], even if the efficacy and safety among PCSK9 inhibitors and statins are still a subject of intensive study.
A Mendelian randomization study by Ference et al. also compared the effects of lower LDL-C levels mediated by variants located in
HMG-CoA reductase (
HMGCR), the gene encoding the target of statins, or in
PCSK9 on the risk of cardiovascular events and the risk of diabetes
[56]. The results showed that variants in
PCSK9 had a nearly identical effect as statin therapy on the risk of cardiovascular diseases and diabetes per unit decrease in plasma LDL-C level. Of note, the clinical benefit of
PCSK9 and
HMGCR variants increased when presented together.
Based on evidence from the two major clinical trials on PCSK9 inhibitors, the FOURIER
[57] and the ODYSSEY
[58] outcome trials that used evolocumab and alirocumab, respectively, as fully humanized monoclonal antibodies against PCSK9, a recent paper by Gallego-Colon et al. underlines that the 2019 European Society of Cardiology/European Atherosclerosis Society guidelines for the management of dyslipidemias establish the use of PCSK9 inhibitors to very high-risk atherosclerotic cardiovascular disease patients who are unresponsive to a maximum tolerated dose of statins and ezetimibe
[59]. Therefore, the discovery of PCSK9 inhibitors has defined a new era of lipid-lowering therapies for patients with atherosclerotic cardiovascular disease which can change future clinical practice.
An untargeted metabolomics approach was also performed to obtain a global view of metabolic and lipidomic pathways and characterize metabolites and lipids that were modified in plasma from patients with FH who received treatment with PCSK9 inhibitors
[60]. Familial hypercholesterolemia causes extremely high circulating LDL-C levels, which are due to mutations of different genes involved in LDL-C metabolism, such as
PCSK9. After 12 weeks of treatment with evolocumab, the scholars observed a significant reduction of LDL-C levels compared to baseline, together with increments in creatine, indole, and indoleacrylic acid concentrations. Instead, a significant decrease in choline and phosphatidylcholine levels, as well as a reduction in platelet-activating factor 16, were reported. The study highlighted for the first time a reduction in inflammation and platelet activation metabolites in FH patients after therapy with PCSK9 inhibitors
[60]. Moreover, due to the small sample size, further studies are required to clarify the underlying mechanisms and the impact on cardiovascular events, confirming data in a larger number of participants with targeted analysis.
3.3. Fibrates
Fibrates are activators of peroxisome proliferator-activated receptor alpha (PPARα), used to prevent and treat hyperlipidemia often in combination with statins, thanks to their ability to increase fatty acid β-oxidation, fatty acid transport, and HDL metabolism, leading to a global reduction of triglyceride and cholesterol levels
[37][61].
Patterson et al. identified pantothenic acid and acylcarnitines as specific potential indicators of PPARα activation of fatty acid β-oxidation induced by fibrates using a metabolomic approach
[62]. They treated healthy volunteers with fenofibrate (200 mg/day) for 14 days and analyzed urinary metabolites at time 0, after 2 days, and after 14 days, by LC-MS, using an ultra-performance liquid chromatography (UPLC) system coupled to a high-resolution mass spectrometer (Q-TOF). They evidenced a dramatic decrease in urinary pantothenic acid (>5 fold) and acylcarnitines (>20 folds), and to confirm that these molecules could be biomarkers of PPARα activation, they treated wild-type and Ppara-null mice with 0.1% fenofibrate for 7 days. Of note, only wild-type mice exhibited a reduction of both urinary pantothenic acid (40 folds) and acylcarnitines (88 folds), suggesting that the effect is strongly associated with the activation of PPARα and transcends species
[62].
Further, a combined transcriptomic and metabolomic approach has been applied to compare in a mouse model 2 weeks of fenofibrate treatment with respect to fish oil treatment
[63]. Fish oil is indeed rich in eicosapentaenoic acid and docosahexaenoic acid, fatty acids that act through PPARα activation and suppress the activity of the prolipogenic transcription factor SREBP-1. Fish oil specifically decreased the levels of various phospholipid species, while fenofibrate specifically increased the levels of Krebs cycle intermediates (i.e., fumaric acid, isocitric acid, malic acid, succinic acid and α-ketoglutaric acid) and most amino acids. These data correlate well with the induction of genes involved in the Krebs cycle and in the urea cycle or in the metabolism of amino groups. Comparing both plasma metabolome and hepatic transcriptome, it emerged that despite being similarly potent toward modulating plasma lipids, fish oil caused only modest changes in gene expression likely in comparison to fenofibrate, reflecting the activation of multiple mechanistic pathways with fish oil, typical of nutritional interventions
[63].
Combination Therapy of Statins and Fibrates
Fibrates are frequently used in combination with statins, working in synergy to reduce plasma lipids, even if this type of treatment is associated with a higher incidence of fatal side effects, such as acute tubular necrosis and rhabdomyolysis. Several hypotheses have been formulated including pharmacokinetic interference, displacement of statins from their binding sites, synergistic action on skeletal muscle, or inhibition of statin glucuronidation by fibrates.
3.4. Nutraceutical and Dietary Habits
In the past decades, an increasing number of studies have suggested that nutraceuticals and dietary habits may be also effective for CVD prevention
[64][65][66], with significant effects on reducing CVD risk and population mortality
[67].
In particular, natural micronutrients and non-nutrient components in these foods, such as polyphenols, have been shown to modulate cholesterol metabolism
[67]. Sommella et al. focused their attention on
Malus pumila Miller cv. Annurca, an apple native to southern Italy, containing high levels of procyanidin B2, a dimeric procyanidin, with favorable biochemical effects against metabolic disorders and atherosclerosis
[67]. They demonstrated that 800 mg/day of Annurca apple polyphenolic extract (AAE) substantially reduced both LDL-C (37.6%) and increased HDL-C (49.3%), similarly to statin treatment
[68], and applied an untargeted metabolomic approach to depicting the molecular mechanism activated by this nutraceutical treatment
[67]. They used deuterium labeling for 72 h coupled with GC-MS and Fourier transform-ion cyclotron resonance (FT-ICR) mass spectrometry to highlight primary metabolic pathways influenced by AAE in in vitro cultured human hepatocytes, HuH7 cells. Their results suggested that AAE acts differently from statins, promoting mitochondrial activity, reprogramming fatty acid metabolism, and inhibiting lipogenesis and cholesterogenesis. AAE diverts acetyl-CoA to the Krebs cycle to produce adenosine triphosphate (ATP) and energy for the cell, instead of becoming HMG-CoA. Glutamine levels are also reduced by AAE suggesting that glutamine can be indeed one of the sources of increased mitochondrial activity. Furthermore, AAE stimulates glycolysis ultimately increasing mitochondrial respiration. Thus, inhibition of lipogenesis and cholesterogenesis could be ascribed to a modulation of the entire metabolic process connected with the use of citrate.
In addition, the beneficial effects of probiotics to improve lipid profiles have been demonstrated in animal models and humans. Ding et al. studied the effects of
Lactobacillus plantarum LP3, from traditional fermented yak milk, on the plasma lipid profile, gut microbiota, and cecum metabolome, by LC-MS, in rats treated with a high-fat diet
[69]. Together with a significant reduction of TC, TG, and LDL-C, they evidenced adjustments in the biosynthesis of fatty acids, steroids, and bile acids, and the metabolism of linoleic acid, linolenic acid, and arachidonic acid were the main metabolic pathways in obese rats. The ability of
Lactobacillus plantarum LP3 to reduce the ratio of Firmicutes to Bacteroidetes in obese rats could explain the reduction in metabolites associated with the biosynthesis of fatty acids
[69].
Dietary plant-derived polyphenols are another class of molecules with protective effects against cardiovascular diseases
[70]. Zhou et al. used a metabolomic approach based on GC-MS analysis of extracts from liver tissues to evaluate the synergistic protective effects of quercetin and resveratrol in mice that were fed a high-fat diet
[71]. The integration of metabolomic and transcriptomic results clearly showed the enhancement of glycolysis, fatty acid oxidation, and gluconeogenesis. The metabolites that were reduced due to the high-fat diet resulted in being restored by quercetin or resveratrol treatment, such as 4-aminobutyric acid, ornithine, histidine, and lysine
[71].
4. Conclusions
It is increasingly evident that metabolomic approaches in pharmacology could be useful not only in the understanding of drug safety, toxicity, and metabolism, but also in the prediction of drug response and in the identification of biological mechanisms, even if some limitations should be acknowledged. Indeed, there is still a lack of standardized protocols for both sample preparation (i.e., collection, storage, and processing) and data acquisition, which are very important for a clinical application of these approaches. Data have been obtained in different compartments, both in clinical settings or animal models, at different times, thus making it difficult to compare them and have a comprehensive view of the metabolomic effects. Another important issue that should be taken into consideration is the influence of the environment (i.e., smoking, food, and physical activity) on the metabolic phenotype, thus requiring a large number of samples to obtain reproducible results, as well as very accurate experimental design. It would be important to also perform longitudinal studies increasing the compliance of patients with the introduction of remote sampling or less invasive collection procedures.
Metabolomics combined with multi-omics strategies and advanced bioinformatics tools could definitely improve the drug repurposing which has gained importance in recent years for identifying novel therapeutic indications for already registered drugs. Since lipid-lowering drugs have pleiotropic effects beyond their known mechanism of action, the discovery of repurposed drugs has implications for precision medicine to treat individual patients providing a decrease in the cost of a new drug development and benefits for the treatment of cardiovascular diseases.