Next Article in Journal
Imaging and Molecular Mechanisms of Alzheimer’s Disease: A Review
Next Article in Special Issue
Relationship of Circulating Irisin with Body Composition, Physical Activity, and Cardiovascular and Metabolic Disorders in the Pediatric Population
Previous Article in Journal
Inflammation-Accelerated Senescence and the Cardiovascular System: Mechanisms and Perspectives
Previous Article in Special Issue
The Functional Role of Zinc Finger E Box-Binding Homeobox 2 (Zeb2) in Promoting Cardiac Fibroblast Activation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fish, Fish Oils and Cardioprotection: Promise or Fish Tale?

1
Division of Cardiology, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
2
Cape Fear Valley Hospital, Fayetteville, NC 28304, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(12), 3703; https://doi.org/10.3390/ijms19123703
Submission received: 8 November 2018 / Revised: 19 November 2018 / Accepted: 20 November 2018 / Published: 22 November 2018
(This article belongs to the Special Issue Nutrition and Cardiovascular Health)

Abstract

:
Fish and commercially available fish oil preparations are rich sources of long-chain omega-3 polyunsaturated fatty acids. Eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are the most important fatty acids in fish oil. Following dietary intake, these fatty acids get incorporated into the cell membrane phospholipids throughout the body, especially in the heart and brain. They play an important role in early brain development during infancy, and have also been shown to be of benefit in dementia, depression, and other neuropsychiatric disorders. Early epidemiologic studies show an inverse relationship between fish consumption and the risk of coronary heart disease. This led to the identification of the cardioprotective role of these marine-derived fatty acids. Many experimental studies and some clinical trials have documented the benefits of fish oil supplementation in decreasing the incidence and progression of atherosclerosis, myocardial infarction, heart failure, arrhythmias, and stroke. Possible mechanisms include reduction in triglycerides, alteration in membrane fluidity, modulation of cardiac ion channels, and anti-inflammatory, anti-thrombotic, and anti-arrhythmic effects. Fish oil supplements are generally safe, and the risk of toxicity with methylmercury, an environmental toxin found in fish, is minimal. Current guidelines recommend the consumption of either one to two servings of oily fish per week or daily fish oil supplements (around 1 g of omega-3 polyunsaturated fatty acids per day) in adults. However, recent large-scale studies have failed to demonstrate any benefit of fish oil supplements on cardiovascular outcomes and mortality. Here, we review the different trials that evaluated the role of fish oil in cardiovascular diseases.

1. Introduction

Cardiovascular disease (CVD) is a leading cause of death in the United States [1]. However, despite extensive advances in our knowledge of nutritional options in the prevention and therapy of CVD, the benefits of dietary fish and fish oil supplementation on CVD remains debatable. Fish oil is a rich source of long-chain omega-3 (ω-3) polyunsaturated fatty acids (PUFAs)—eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA). The possible cardioprotective role of fish consumption was first identified in the early studies of Greenland’s Inuit population who were found to have a low incidence of myocardial infarction (MI) compared to their Danish counterparts. The benefit was attributed to the high fish consumption by the Inuits [2,3]. This was followed by nearly four decades of research, including animal studies, epidemiological studies, randomized controlled trials (RCTs), meta-analyses of epidemiological cohort studies, and trial meta-analyses. Many of these studies demonstrated the cardioprotective effects of fish consumption and fish oil supplementation [4,5,6,7,8,9,10]. The rising amount of evidence led to recommendations regarding consumption of seafood/fish and/or their dietary supplementation with ω-3 PUFA from the Food and Drug Administration (FDA) [11]. In 2012, a prescription fish oil formulation was approved by the US FDA. Fish oil continues to be the most popular natural supplement in the US, used by nearly 18.8 million adults [12]. However, several recent large-scale studies have failed to demonstrate any significant benefits of fish oil supplements on cardiovascular outcomes and mortality [13,14,15,16,17]. In this review, we discuss the structure and metabolism of marine-derived ω-3 PUFAs, their proposed benefits and molecular mechanisms of action, and the evidence regarding their role in CVD. We conclude by providing possible reasons for the conflicting evidence and recommendations regarding the dietary intake of fish and fish oil supplements.

2. Structure and Metabolism of PUFAs

Fatty acids are long-chain hydrocarbons with a carboxylic acid group at one end (alpha terminal) and methyl group at the other end (omega terminal). They can be classified based on the number of double bonds in their side chains—saturated fatty acids (no double bond), monounsaturated fatty acids or MUFAs (single double bond), and polyunsaturated fatty acids or PUFAs (two or more double bonds). PUFAs can be classified further by the length of the carbon chain and the position of the first double bond from the methyl terminal into omega-6 (ω-6 or n-6) or omega-3 (ω-3 or n-3). For example, linoleic acid (LA) or 9,12-octadecadienoic acid (C18:2) has 18 carbon atoms with 2 double bonds. Since the first double bond from the methyl terminal is at the sixth position, it is an ω-6 PUFA. Similarly, alpha-linolenic acid (ALA) is 9,12,15-octadecatrienoic acid (C18:3), meaning it has 18 carbon atoms with 3 double bonds. However, in this case, the first double bond is at the third position from the methyl terminal, and hence it is an ω-3 PUFA. Both LA and ALA are considered essential fatty acids since they cannot be synthesized by humans and must be ingested via their diet.
The essential fatty acids LA and ALA are then metabolized to other fatty acids through desaturase and elongase enzymes. LA (ω-6) is metabolized to arachidonic acid (5,8,11,14-eicosatetraenoic acid, C20:4, ω-6). Similarly, ALA (ω-3) is converted to EPA (5,8,11,14,17-eicosapentaenoic acid, C20:5, ω-3) and DHA (4,7,10,13,16,19-docosahexaenoic acid, C22:6, ω-3). Thus, EPA and DHA are traditionally considered non-essential since, technically speaking, they can be synthesized from ALA. However, this pathway is slow and inefficient [18]. Therefore, for all practical purposes, the dietary intake of EPA and DHA is “essential” and crucial to obtain health benefits.
After absorption from the intestine as chylomicrons, fatty acids are transported to the liver and other tissues. PUFAs are subsequently incorporated into the phospholipid bilayer of plasma membranes, and affect membrane fluidity and signaling. ω-6 and ω-3 PUFAs have opposite effects in the body. Diets rich in ω-6 are precursors of eicosanoids associated with inflammation, vasoconstriction, and platelet aggregation [19]. Acute self-limited inflammation is a protective response to infection and injury. However, excessive inappropriate inflammation has been linked to atherosclerosis and cancer. On the other hand, ω-3 PUFAs are precursors of anti-inflammatory molecules and provide benefits against chronic inflammatory conditions, like diabetes, ischemic heart disease, and cancer [20]. These molecular mechanisms are discussed in further detail in a later section of this review.
The structure and metabolism of major PUFAs is depicted in Figure 1.

3. Dietary Sources of Major PUFAs

Most vegetable oils and crop seeds, like corn, sunflower, soybean, and canola oils, are a rich source of ω-6 LA with lesser amounts of ω-3 ALA. On the other hand, flax, walnuts, and chia seeds are a rich source of ALA, as are some green leafy vegetables.
Fatty fish and other seafood are the most important dietary sources of EPA and DHA. Wild (marine) fish feed on phyto- and zoo-planktons, and are therefore a richer source of EPA and DHA than cultivated (farmed) fish which feed on cereals and vegetable oils [20]. Cod liver and algal oil have been proposed to be non-traditional marine sources of EPA and DHA. Finally, marine-derived ω-3 fortified food products, like cereals, pastas, dairy products, eggs, meat, salad dressings, and oils are now available. They are a potential option for vegetarians and those who dislike seafood [18]. The principal dietary sources of PUFAs are shown in Table 1 [21,22].

4. Proposed Cardioprotective Benefits of ω-3 PUFAs and Their Molecular Mechanisms of Action

Fish and fish oil consumption has been shown to decrease cardiovascular events and mortality in many secondary prevention trials that included high-risk patients with recent myocardial infarction or heart failure. Several molecular mechanisms for this cardioprotective effect of ω-3 PUFAs have been proposed.

4.1. Anti-Inflammatory Effect

ω-3 PUFAs have pleiotropic anti-inflammatory effects. They readily replace arachidonic acid in cell membranes. This results in decreased production of ω-6 arachidonic acid-derived inflammatory mediators, like prostaglandin (PG) E2, thromboxane (TX) A2, and leukotrienes (LTs) A4, B4, C4, D4, and E4 [23,24]. ω-3 PUFAs generate less potent inflammatory substrates, like 3-series PGs and TXs and 5-series LTs [24]. 3-series PGs have less potent biological effects (compared to 2-series PGs) and TX A3 lacks pro-platelet aggregatory properties. 5-series LTs are relatively less effective as pro-inflammatory molecules. EPA and DHA are metabolized to anti-inflammatory mediators, like resolvins, protectins, and the G protein-coupled receptor 120 [24,25]. Additionally, cell membranes with higher ω-3: ω-6 PUFA ratio have higher fluidity. Marine-derived ω-3 PUFAs have been shown to reduce the circulating levels of pro-inflammatory cytokines like interleukin (IL)-1, IL-6, and the tumor necrosis factor (TNF)-α [26]. ω-3 PUFAs also regulate intracellular signaling pathways to inactivate nuclear transcriptional factors. They decrease expression of inflammatory genes via the downregulation of nuclear factor (NF)-κB. The inhibition of NF-κB is mediated by the activation of peroxisome proliferator-activated receptors (PPAR) [27].

4.2. Improved Endothelial Function

Endothelial dysfunction from loss of endothelial-derived nitric oxide (NO) synthesis results in predisposition to accelerated atherosclerosis and adverse vascular events. Marine-derived ω-3 PUFAs cause translocation and activation of endothelial NO synthase (eNOS) into the cytosol, resulting in vasodilation and improved endothelial function [25,28]. Recently, EPA was shown to prevent saturated fatty acid-induced vascular endothelial dysfunction through regulation of long-chain acyl-coA synthetase expression [29]. Endothelial function is additionally improved by reduced expression of endothelial vascular cell adhesion molecules, resulting in attenuated leukocyte adhesion to endothelium [30,31].

4.3. Atherosclerotic Plaque Stabilization

EPA and DHA inhibit the proliferation and migration of smooth muscle cells (SMCs), a central step in atherosclerotic plaque formation and progression. Vasa vasorum is a network of microvessels extending up to the plaque base, which is vital for plaque progression. ω-3 PUFAs interfere with the neovascularization of vasa vasorum, thereby suppressing plaque development [25].
Apart from reducing plaque progression, ω-3 PUFAs also contribute to plaque stability. Plaque vulnerability predisposes people to plaque erosion or rupture, which causes acute coronary syndrome. High tissue levels of EPA and DHA decrease macrophage infiltration and the release of matrix metalloproteinases (MMPs), resulting in greater plaque stability [23]. The addition of EPA to statin therapy has been shown to reduce the lipid core in coronary plaques [32]. Plaques in patients receiving fish oil are more likely to be fibrous cap atheromas, with fewer macrophages and lesser inflammation, and are therefore more likely to be stable [33].

4.4. Effect on Lipid Metabolism

ω-3 PUFAs modulate the activity of genes that control lipid homeostasis. Large doses of fish oil interfere with the synthesis of very low-density lipoprotein (VLDL) via inhibition of sterol receptor element-binding protein-1c. This results in a marked lowering of serum TG levels [23]. Although ω-3 fatty acids do not affect the serum levels of total cholesterol and low-density lipoprotein (LDL), fish oil has been shown to reduce remnant lipoproteins and post-prandial lipemia after fatty meals [30,31]. Remnant lipoprotein levels and post-prandial lipemia are involved in the pathogenesis of sudden cardiac death (SCD) [23]. Marine-derived ω-3 fatty acids also cause a favorable change in high-density lipoprotein (HDL) by increasing the large, cholesterol-rich HDL2 fraction and lowering the small, TG-rich HDL3 fraction [30].

4.5. Anti-Thrombotic Effect

EPA inhibits the synthesis of platelet TXA2 which causes platelet aggregation and vasoconstriction. Both EPA and DHA antagonize the TXA2 and PGH2 receptors in human platelets [34]. There are reports that ω-3 PUFA reduce fibrinogen levels and increase tissue plasminogen-activator concentrations [30,31,35].

4.6. Anti-Arrhythmic Effect

ω-3 PUFAs modulate the activity of multiple ion channels and stabilize the cardiomyocyte membrane, thereby preventing tachyarrhythmias and SCD [5]. EPA and DHA inhibit the voltage-gated sodium channels in cardiac myocytes, increase the voltage threshold for depolarization, and prolong the refractory period. They also modulate certain calcium channels, decrease free cytosolic calcium, and reduce membrane excitability further [24]. Part of the anti-arrhythmic action of these fatty acids is also due to their autonomic effects, like increased vagal tone [36]. In addition, low serum levels of EPA and DHA have been found to increase the risk of cardiogenic syncope in patients with Brugada syndrome [37].

4.7. Cardiac Remodeling

The OMEGA-REMODEL trial reported the beneficial effects of high-dose ω-3 PUFA therapy on cardiac remodeling in patients with MI. Cardiac magnetic resonance confirmed reduced ventricular remodeling and myocardial fibrosis after PUFA supplementation [38]. Similar findings of the preferential effects of ω-3 PUFAs on cardiac remodeling and heart failure were also seen in the Gruppo Italiano per lo Studio della Streptochinasi nell’Infarto Miocardico-Heart Failure (GISSI-HF) trial [10].
Cardiac myofibroblasts, activated by inflammatory signals or pressure overload, cause cardiac remodeling. Recently, a novel EPA metabolite called 18-hydroxy eicosapentaenoic acid (18-HEPE) has been identified, which prevents cardiac remodeling under pressure overload [39]. Higher dietary intake of EPA ethyl ester increases the plasma concentration of 18-HEPE, and may have beneficial long-term effects by preventing cardiac fibrosis [24].
Moreover, EPA and DHA improve cardiac mitochondrial function by increasing the efficiency of adenosine triphosphate (ATP) production [40].

4.8. Improved Exercise Tolerance

Decreased exercise capacity is a known risk factor for CVD. Recently, ω-3 PUFAs have been shown to improve exercise tolerance. This may be due to favorable effects on erythrocyte rheology and skeletal muscle function [25,41]. Improved exercise capacity may contribute to a lower risk of adverse cardiac events.

4.9. Improved Cognitive Function

Poor cognitive function is a risk factor for cardiovascular events [42]. Serum EPA levels have been shown to be independently associated with cognitive function in CVD patients [43]. Thus, improved dietary ω-3 PUFA intake might conceivably improve cognition and decrease the risk of cardiovascular events [25].
Figure 2 summarizes the various molecular mechanisms by which ω-3 PUFAs, like EPA and DHA, have been proposed to exert their cardioprotective effects.

5. Evidence from Trials and Meta-Analyses

The Diet and Reinfarction Trial (DART) was the first RCT to assess the benefits of dietary fish and fish oil in the secondary prevention of MI [4]. A total of 2033 patients with recent MI were followed for two years. Consumption of at least two servings (200–400 g) of fish per week was associated with a reduction in all-cause mortality. Similar beneficial effects were also noticed in participants who took fish oil capsules instead of fish.
The GISSI-Prevenzione trial included 11,324 patients with previous MI (within three months) [6]. The intervention group received one fish oil capsule per day, in addition to standard care. The intervention group was noted to have a 41% reduction in all-cause mortality and 53% reduction in SCD as early as four months into the study. The difference in all-cause mortality remained significant after a follow-up of 3.5 years, and was proposed to be primarily due to the anti-arrhythmic effects of EPA and DHA.
The DART-2 was a secondary prevention study conducted in men with stable angina (and not previous MI, unlike the first DART study) [44]. Patients who were advised to consume two portions of oily fish every week or take three fish oil capsules daily had a higher risk of cardiac mortality and SCD. Subgroup analysis revealed that this difference was driven by the fish oil capsule group. The study was largely criticized for its poor design and lack of blinding.
The Japan EPA Lipid Intervention Study (JELIS), a combined primary (14,981 patients) and secondary intervention (3664 patients) trial, included a total of 18,645 patients [9]. The EPA (1.8 g daily) plus statin group showed a reduction in major coronary events compared to the statin-alone group. Subgroup analysis revealed that the significant reduction in coronary events was mainly seen in patients with a history of coronary artery disease, and there was no benefit of EPA in primary prevention. However, the results of this study may have been diluted, as the consumption of fish in the Japanese population is high at baseline.
The GISSI-HF trial was designed to study the effects of daily EPA and DHA supplementation in patients with HF [10]. The ω-3 PUFA group had a reduction in all-cause mortality and cardiovascular hospitalizations compared to the control group.
The Alpha Omega trial was a secondary prevention study using 4837 patients with prior MI [14]. Low-dose EPA and DHA in margarine were given daily to the intervention group, while the control group received only plain margarine. There was no difference in cardiovascular events between the two groups. The lack of benefit from EPA and DHA in the Alpha Omega study [14], compared to the GISSI Prevenzione [6] and JELIS [9] trials (all secondary prevention trials in prior MI patients), may have been, in part, due to the lower treatment dose of the intervention arm.
The OMEGA study included German patients with MI in the two weeks prior to enrolment [13]. The treatment group received EPA and DHA daily, versus the control group, which received olive oil. After a follow-up of 1 year, no significant difference was found in the rates of adverse cardiovascular events, SCD, and all-cause mortality between the two groups. However, the study was concluded to be underpowered due to the lower-than-expected event rates and overestimation of the effect of ω-3 PUFAs.
SU.FOL.OM3 was a randomized trial evaluating the effects of B-vitamin and ω-3 PUFA supplementation in 2501 French patients with a recent acute coronary or cerebral ischemic event [15]. ω-3 PUFA intake did not significantly affect the major cardiovascular event rate. Like the OMEGA study, the event rate was lower than anticipated and so the trial was underpowered.
The Outcome Reduction with an Initial Glargine Intervention (ORIGIN) trial was the first study to investigate the effects of ω-3 PUFA supplementation on cardiovascular events in patients with pre-diabetes and diabetes [16]. Analysis of 12,536 patients over 6.2 years did not reveal any benefits of ω-3 PUFAs in reducing cardiovascular death, compared to the olive oil given to the control group. These results were in contrast to the findings of the GISSI Prevenzione [6] and GISSI-HF [10] trials. Inter-trial differences in patient baseline characteristics and concomitant therapies could be a possible explanation.
The Risk and Prevention study investigated the efficacy of ω-3 PUFAs in Italian patients with high risk of CVD but without previous MI [17]. After a follow-up of five years, there was no difference in the primary endpoint of cardiovascular death and hospitalization between the ω-3 PUFA treatment group and olive oil control group. However, subgroup analysis revealed that the ω-3 PUFA group had fewer heart failure hospitalizations. Additionally, women in the treatment group had lower rates of primary endpoint than the control group. The results of this study may not be generalizable due to the Mediterranean dietary habits in the Italian population.
The PREDIMED trial in 7447 patients reported that a Mediterranean diet, with 50 g of extra-virgin olive oil daily, significantly reduced cardiovascular events and death at a follow-up period of 4.8 years [45]. It remains unknown whether the benefit was due to the Mediterranean diet alone, olive oil alone, or whether it was a combined effect. The OMEGA [13], ORIGIN [16], and Risk and Prevention [17] trials did not detect a significant difference between the overall outcomes in the ω-3 PUFA group and olive oil control group. However, the results of the PREDIMED trial [45] indicate that olive oil itself may have some cardioprotective benefits and is likely not an ideal control.
The Age-Related Eye Disease Study 2 (AREDS2) trial included 4203 patients at high risk of CVD and either intermediate or advanced macular degeneration [46]. Patients were randomly grouped to the ω-3 PUFA group (650 mg EPA plus 350 mg DHA), macular xanthophyll group (10 mg lutein plus 2 mg zeaxanthin), combination therapy, or matching placebos. Long-chain ω-3 PUFAs or macular xanthophylls did not reduce the risk of CVD events.
Rizos et al. performed a meta-analysis of 20 RCTs including 68,680 patients to study the role of ω-3 PUFA supplementation on major cardiovascular outcomes [47]. Overall, ω-3 PUFAs were not associated with cardiovascular benefits.
Studies have also evaluated the role of ω-3 PUFAs in the secondary prevention of atrial fibrillation [48,49]. The FORWARD trial included 586 participants with previous atrial fibrillation who were randomized to receive either ω-3 PUFA for 1 g per day, or a placebo for one year [48]. PUFA supplementation did not reduce recurrent atrial fibrillation. Mariani et al. performed a meta-analysis of 16 trials covering 4677 patients and concluded that ω-3 PUFAs have no effect in preventing recurrent or post-operative atrial fibrillation [49].
The results of the ASCEND trial were recently published [50]. It was a randomized, placebo-controlled, blinded trial in 15,480 patients followed for 7.4 years. The study aimed to assess the efficacy and safety of taking 100 mg of aspirin daily in preventing cardiovascular events and cancer in diabetic patients without known CVD. This study also investigated whether daily ω-3 PUFA supplementation decreased cardiovascular events in this population. Aspirin use was noted to prevent cardiovascular events in patients, but also caused major bleeding events. Compared to the placebo group that received olive oil capsules, 1 g of ω-3 PUFA supplementation daily failed to decrease the risk of serious vascular events in diabetics without known CVD.
The Reduction of Cardiovascular Events with EPA-Intervention Trial (REDUCE-IT) was recently concluded [51]. The study involved 8179 high-risk patients with hypertriglyceridemia on statin therapy who were randomized to receive either 4 g of ethyl EPA daily, or a placebo. After a median follow-up of 4.9 years, there was an approximately 25% reduction in the risk of major adverse cardiovascular events in the treatment group.
The Vitamin D and Omega-3 Trial (VITAL) results were recently announced [52]. The study included 25,871 participants with an objective to assess the effect of daily vitamin D3 (2000 IU) and fish oil supplement (1 g) in the primary prevention of cancer and CVD. After a follow-up of over five years, ω-3 PUFA supplementation did not result in a lower incidence of cardiovascular events or cancer, compared to the placebo.
The STRENGTH study is an ongoing RCT that will enroll approximately 13,000 patients with hypertriglyceridemia, low HDL, and a high risk for CVD [53]. Patients are being randomized to receive either statin with corn oil or statin with prescription ω-3 carboxylic acids. The study is anticipated to be completed in 2019.
Results of various published trials and meta-analyses discussed above are presented in Table 2.

6. Available ω-3 PUFA Formulations

ω-3 PUFA ethyl esters was the first formulation approved by the FDA in the year 2004. It was marketed under the trade name Omacor® by Reliant Pharmaceuticals and approved for use in patients with serum triglyceride levels greater than 500 mg/dL. It was later renamed to Lovaza® (GlaxoSmithKline). Ethyl EPA or icosapent ethyl, marketed under the name Vascepa® by Amarin Pharmaceuticals, was approved in 2012. It differed from the earlier formulation in that it contained only ethyl esters of EPA without any DHA [54].
In 2014, ω-3 carboxylic acids, marketed under the name Epanova® by AstraZeneca, was approved for hypertriglyceridemia greater than 500 mg/dL. This formulation consists of free fatty acids instead of the prodrug, and therefore does not require pancreatic lipase for conversion to active form. Thus, it can be taken independent of meals with good bioavailability [54]. The ECLIPSE [55] and ECLIPSE II [56] studies compared the pharmacokinetics of ethyl esters and carboxylic acids. Both studies reported much higher bioavailability with the carboxylic acid formulations.
Despite newer options, ethyl esters continue to be the most commonly prescribed due to its generic options.
Differences between various ω-3 prescription products are shown in Table 3 [22].

7. Side-Effects and Safety Concerns

Fish oil supplements are generally well tolerated. The most common side effects are gastrointestinal, like nausea, eructation, and diarrhea. Certain large fish (like king mackerel, shark, swordfish) have a higher chance of being contaminated with methyl mercury and therefore should be avoided by pregnant/breastfeeding women and children. There have been some concerns about an increased risk of minor bleeding with ω-3 fatty acid supplementation, especially in patients taking aspirin and statins. However, no major bleeding events have been reported in trials to date.

8. Conclusions

Data from early epidemiologic and observational studies have shown there to be cardioprotective benefits from fish and fish oil consumption. However, most primary prevention trials and recent secondary prevention trials have failed to replicate similar results. Several possibilities could explain the difference in results—the high efficacy of modern-day pharmacotherapy (like statins) and revascularization that attenuates the benefit of ω-3 fatty acids, a lower dose of EPA and DHA supplementation in trials than what was needed, insufficient length of follow-up to see benefits, an improved diet with a higher consumption of fish and other seafood which may account for the decreased magnitude of benefits from fish oil capsules over time, improper study design with use of olive oil as a control (olive oil itself has cardioprotective properties making it an unideal control), and fewer-than-anticipated events leading to underpowered studies. Some trials with a large sample size and strong study design are ongoing, and may shed useful light on the subject.
Based on the current evidence, individuals are advised to consume a healthy diet with two servings of fatty fish every week. Such a food-based approach also supplies several other beneficial nutrients apart from ω-3 PUFAs. For those who cannot consume fish, fish oil supplements containing EPA and DHA have a good safety profile and may be reasonable options, especially in patients with pre-existing CVD, heart failure, and hypertriglyceridemia.

Author Contributions

All authors contributed to literature review and writing of this manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bauer, U.E.; Briss, P.A.; Goodman, R.A.; Bowman, B.A. Prevention of chronic disease in the 21st century: Elimination of the leading preventable causes of premature death and disability in the USA. Lancet 2014, 384, 45–52. [Google Scholar] [CrossRef]
  2. Bang, H.O.; Dyerberg, J.; Nielsen, A. Plasma lipid and lipoprotein pattern in Greenlandic West-coast Eskimos. Lancet 1971, 297, 1143–1146. [Google Scholar] [CrossRef]
  3. Bang, H.O.; Dyerberg, J.; Sinclair, H.M. The composition of the Eskimo food in north western Greenland. Am. J. Clin. Nutr. 1980, 33, 2657–2661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Burr, M.L.; Gilbert, J.F.; Holliday, R.A.; Elwood, P.C.; Fehily, A.M.; Rogers, S.; Sweetnam, P.M.; Deadman, N.M. Effects of changes in fat, fish, and fibre intakes on death and myocardial reinfarction: Diet and reinfarction trial (DART). Lancet 1989, 334, 757–761. [Google Scholar] [CrossRef]
  5. Yang, B.; Saldeen, T.G.; Nichols, W.W.; Mehta, J.L. Dietary fish oil supplementation attenuates myocardial dysfunction and injury caused by global ischemia and reperfusion in isolated rat hearts. J. Nutr. 1993, 123, 2067–2074. [Google Scholar] [PubMed]
  6. GISSI-Prevenzione Investigators. Dietary supplementation with n-3 polyunsaturated fatty acids and vitamin E after myocardial infarction: Results of the GISSI-Prevenzione trial. Lancet 1999, 354, 447–455. [Google Scholar] [CrossRef]
  7. Whelton, S.P.; He, J.; Whelton, P.K.; Muntner, P. Meta-analysis of observational studies on fish intake and coronary heart disease. Am. J. Cardiol. 2004, 93, 1119–1123. [Google Scholar] [CrossRef] [PubMed]
  8. He, K.; Song, Y.; Daviglus, M.L.; Liu, K.; Van Horn, L.; Dyer, A.R.; Greenland, P. Accumulated evidence on fish consumption and coronary heart disease mortality: A Meta-analysis of cohort studies. Circulation 2004, 109, 2705–2711. [Google Scholar] [CrossRef] [PubMed]
  9. Yokoyama, M.; Origasa, H.; Matsuzaki, M.; Matsuzawa, Y.; Saito, Y.; Ishikawa, Y.; Oikawa, S.; Sasaki, J.; Hishida, H.; Itakura, H.; et al. Effects of eicosapentaenoic acid on major coronary events in hypercholesterolaemic patients (JELIS): A randomised open-label, blinded endpoint analysis. Lancet 2007, 369, 1090–1098. [Google Scholar] [CrossRef]
  10. Tavazzi, L.; Maggioni, A.P.; Marchioli, R.; Barlera, S.; Franzosi, M.G.; Latini, R.; Lucci, D.; Nicolosi, G.L.; Porcu, M.; Tognoni, G. Effect of n-3 polyunsaturated fatty acids in patients with chronic heart failure (the GISSI-HF trial): A randomised, double-blind, placebo-controlled trial. Lancet 2008, 372, 1223–1230. [Google Scholar] [PubMed]
  11. Kris-Etherton, P.M.; Harris, W.S.; Appel, L.J. Fish consumption, fish oil, omega-3 fatty acids, and cardiovascular disease. Circulation 2002, 106, 2747–2757. [Google Scholar] [CrossRef] [PubMed]
  12. Clarke, T.C.; Black, L.I.; Stussman, B.J.; Barnes, P.M.; Nahin, R.L. Trends in the Use of Complementary Health Approaches among Adults: United States, 2002–2012; National Health Statistics Reports No. 79; National Center for Health Statistics: Hyattsville, MD, USA, 2015; pp. 1–16.
  13. Rauch, B.; Schiele, R.; Schneider, S.; Diller, F.; Victor, N.; Gohlke, H.; Gottwik, M.; Steinbeck, G.; Del Castillo, U.; Sack, R.; et al. OMEGA, a randomized, placebo-controlled trial to test the effect of highly purified omega-3 fatty acids on top of modern guideline-adjusted therapy after myocardial infarction. Circulation 2010, 122, 2152–2159. [Google Scholar] [CrossRef] [PubMed]
  14. Kromhout, D.; Giltay, E.J.; Geleijnse, J.M. n–3 Fatty acids and cardiovascular events after myocardial infarction. N. Engl. J. Med. 2010, 363, 2015–2026. [Google Scholar] [CrossRef] [PubMed]
  15. Galan, P.; Kesse-Guyot, E.; Czernichow, S.; Briancon, S.; Blacher, J.; Hercberg, S. Effects of B vitamins and omega 3 fatty acids on cardiovascular diseases: A randomised placebo controlled trial. BMJ 2010, 341, c6273. [Google Scholar] [CrossRef] [PubMed]
  16. ORIGIN Trial Investigators. n–3 Fatty acids and cardiovascular outcomes in patients with dysglycemia. N. Engl. J. Med. 2012, 367, 309–318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Risk and Prevention Study Collaborative Group. N–3 fatty acids in patients with multiple cardiovascular risk factors. N. Engl. J. Med. 2013, 368, 1800–1808. [Google Scholar] [CrossRef] [PubMed]
  18. Bowen, K.J.; Harris, W.S.; Kris-Etherton, P.M. Omega-3 fatty acids and cardiovascular disease: Are there benefits? Curr. Treat. Opt. Cardiovasc. Med. 2016, 18, 69. [Google Scholar] [CrossRef] [PubMed]
  19. Dennis, E.A.; Norris, P.C. Eicosanoid storm in infection and inflammation. Nat. Rev. Immunol. 2015, 15, 511. [Google Scholar] [CrossRef] [PubMed]
  20. Saini, R.K.; Keum, Y.S. Omega-3 and omega-6 polyunsaturated fatty acids: Dietary sources, metabolism, and significance—A review. Life Sci. 2018, 203, 255–267. [Google Scholar] [CrossRef] [PubMed]
  21. Dietary Guidelines Advisory Committee. Scientific Report of the 2015 Dietary Guidelines Advisory Committee; US Department of Health and Human Services: Washington, DC, USA, 2015.
  22. Burke, M.F.; Burke, F.M.; Soffer, D.E. Review of cardiometabolic effects of prescription omega-3 fatty acids. Curr. Atheroscler. Rep. 2017, 19, 60. [Google Scholar] [CrossRef] [PubMed]
  23. Kromhout, D.; Yasuda, S.; Geleijnse, J.M.; Shimokawa, H. Fish oil and omega-3 fatty acids in cardiovascular disease: Do they really work? Eur. Heart J. 2011, 33, 436–443. [Google Scholar] [CrossRef] [PubMed]
  24. Endo, J.; Arita, M. Cardioprotective mechanism of omega-3 polyunsaturated fatty acids. J. Cardiol. 2016, 67, 22–27. [Google Scholar] [CrossRef] [PubMed]
  25. Yagi, S.; Fukuda, D.; Aihara, K.I.; Akaike, M.; Shimabukuro, M.; Sata, M. N-3 polyunsaturated fatty acids: Promising nutrients for preventing cardiovascular disease. J. Atheroscler. Thromb. 2017, 24, 999–1010. [Google Scholar] [CrossRef] [PubMed]
  26. Calder, P.C. The role of marine omega-3 (n-fatty acids in inflammatory processes, atherosclerosis and plaque stability. Mol. Nutr. Food Res. 2012, 56, 1073–1080. [Google Scholar] [CrossRef] [PubMed]
  27. De Winther, M.P.; Kanters, E.; Kraal, G.; Hofker, M.H. Nuclear factor κB signaling in atherogenesis. Arter. Thromb. Vasc. Biol. 2005, 25, 904–914. [Google Scholar] [CrossRef] [PubMed]
  28. Omura, M.; Kobayashi, S.; Mizukami, Y.; Mogami, K.; Todoroki-Ikeda, N.; Miyake, T.; Matsuzaki, M. Eicosapentaenoic acid (EPA) induces Ca2+-independent activation and translocation of endothelial nitric oxide synthase and endothelium-dependent vasorelaxation. FEBS Lett. 2001, 487, 361–366. [Google Scholar] [CrossRef]
  29. Ishida, T.; Naoe, S.; Nakakuki, M.; Kawano, H.; Imada, K. Eicosapentaenoic acid prevents saturated fatty acid-induced vascular endothelial dysfunction: Involvement of long-chain acyl-CoA synthetase. J. Atheroscler. Thromb. 2015, 22, 1172–1185. [Google Scholar] [CrossRef] [PubMed]
  30. Jain, A.P.; Aggarwal, K.K.; Zhang, P.Y. Omega-3 fatty acids and cardiovascular disease. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 441–445. [Google Scholar] [PubMed]
  31. Haglund, O.; Mehta, J.L.; Saldeen, T. Effects of fish oil on some parameters of fibrinolysis and lipoprotein(a) in healthy subjects. Am. J. Cardiol. 1994, 74, 189–192. [Google Scholar] [CrossRef]
  32. Niki, T.; Wakatsuki, T.; Yamaguchi, K.; Taketani, Y.; Oeduka, H.; Kusunose, K.; Ise, T.; Iwase, T.; Yamada, H.; Soeki, T.; et al. Effects of the addition of eicosapentaenoic acid to strong statin therapy on inflammatory cytokines and coronary plaque components assessed by integrated backscatter intravascular ultrasound. Circ. J. 2016, 80, 450–460. [Google Scholar] [CrossRef] [PubMed]
  33. Thies, F.; Garry, J.M.; Yaqoob, P.; Rerkasem, K.; Williams, J.; Shearman, C.P.; Gallagher, P.J.; Calder, P.C.; Grimble, R.F. Association of n-3 polyunsaturated fatty acids with stability of atherosclerotic plaques: A randomised controlled trial. Lancet 2003, 361, 477–485. [Google Scholar] [CrossRef]
  34. Swann, P.G.; Venton, D.L.; Le Breton, G.C. Eicosapentaenoic acid and docosahexaenoic acid are antagonists at the thromboxane A2/prostaglandin H2 receptor in human platelets. FEBS Lett. 1989, 243, 244–246. [Google Scholar] [CrossRef]
  35. Atakisi, O.; Atakisi, E.; Ozcan, A.; Karapehlivan, M.; Kart, A. Protective effect of omega-3 fatty acids on diethylnitrosamine toxicity in rats. Eur. Rev. Med. Pharmacol. Sci. 2013, 17, 467–471. [Google Scholar] [PubMed]
  36. Christensen, J.H.; Schmidt, E.B. Autonomic nervous system, heart rate variability and n-3 fatty acids. J. Cardiovasc. Med. 2007, 8, 19–22. [Google Scholar] [CrossRef] [PubMed]
  37. Yagi, S.; Soeki, T.; Aihara, K.I.; Fukuda, D.; Ise, T.; Kadota, M.; Bando, S.; Matsuura, T.; Tobiume, T.; Yamaguchi, K.; et al. Low serum levels of eicosapentaenoic acid and docosahexaenoic acid are risk factors for cardiogenic syncope in patients with Brugada syndrome. Int. Heart J. 2017, 58, 720–723. [Google Scholar] [CrossRef] [PubMed]
  38. Heydari, B.; Abdullah, S.; Pottala, J.V.; Shah, R.; Abbasi, S.; Mandry, D.; Francis, S.A.; Lumish, H.; Ghoshhajra, B.B.; Hoffmann, U.; et al. Effect of Omega-3 Acid Ethyl Esters on Left Ventricular Remodeling After Acute Myocardial Infarction Clinical Perspective: The OMEGA-REMODEL Randomized Clinical Trial. Circulation 2016, 134, 378–391. [Google Scholar] [CrossRef] [PubMed]
  39. Endo, J.; Sano, M.; Isobe, Y.; Fukuda, K.; Kang, J.X.; Arai, H.; Arita, M. 18-HEPE, an n-3 fatty acid metabolite released by macrophages, prevents pressure overload–induced maladaptive cardiac remodeling. J. Exp. Med. 2014, 211, 1673–1687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Duda, M.K.; O’shea, K.M.; Stanley, W.C. ω-3 polyunsaturated fatty acid supplementation for the treatment of heart failure: Mechanisms and clinical potential. Cardiovasc. Res. 2009, 84, 33–41. [Google Scholar] [CrossRef] [PubMed]
  41. Da Boit, M.; Hunter, A.M.; Gray, S.R. Fit with good fat? The role of n-3 polyunsaturated fatty acids on exercise performance. Metabolism 2017, 66, 45–54. [Google Scholar] [CrossRef] [PubMed]
  42. Yagi, S.; Akaike, M.; Ise, T.; Ueda, Y.; Iwase, T.; Sata, M. Renin–angiotensin–aldosterone system has a pivotal role in cognitive impairment. Hypertens. Res. 2013, 36, 753. [Google Scholar] [CrossRef] [PubMed]
  43. Yagi, S.; Hara, T.; Ueno, R.; Aihara, K.I.; Fukuda, D.; Takashima, A.; Hotchi, J.; Ise, T.; Yamaguchi, K.; Tobiume, T.; et al. Serum concentration of eicosapentaenoic acid is associated with cognitive function in patients with coronary artery disease. Nutr. J. 2014, 13, 112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Burr, M.L.; Ashfield-Watt, P.A.; Dunstan, F.D.; Fehily, A.M.; Breay, P.; Ashton, T.; Zotos, P.C.; Haboubi, N.A.; Elwood, P.C. Lack of benefit of dietary advice to men with angina: Results of a controlled trial. Eur. J. Clin. Nutr. 2003, 57, 193. [Google Scholar] [CrossRef] [PubMed]
  45. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Primary prevention of cardiovascular disease with a Mediterranean diet. N. Engl. J. Med. 2013, 368, 1279–1290. [Google Scholar] [CrossRef] [PubMed]
  46. Bonds, D.E.; Harrington, M.; Worrall, B.B.; Bertoni, A.G.; Eaton, C.B.; Hsia, J.; Robinson, J.; Clemons, T.E.; Fine, L.J.; Chew, E.Y. Effect of long-chain ω-3 fatty acids and lutein+ zeaxanthin supplements on cardiovascular outcomes: Results of the Age-Related Eye Disease Study 2 (AREDSrandomized clinical trial. JAMA Intern. Med. 2014, 174, 763–771. [Google Scholar] [CrossRef] [PubMed]
  47. Rizos, E.C.; Ntzani, E.E.; Bika, E.; Kostapanos, M.S.; Elisaf, M.S. Association between omega-3 fatty acid supplementation and risk of major cardiovascular disease events: A systematic review and meta-analysis. JAMA 2012, 308, 1024–1033. [Google Scholar] [CrossRef] [PubMed]
  48. Macchia, A.; Grancelli, H.; Varini, S.; Nul, D.; Laffaye, N.; Mariani, J.; Ferrante, D.; Badra, R.; Figal, J.; Ramos, S.; et al. Omega-3 fatty acids for the prevention of recurrent symptomatic atrial fibrillation: Results of the FORWARD (Randomized Trial to Assess Efficacy of PUFA for the Maintenance of Sinus Rhythm in Persistent Atrial Fibrillation) trial. J. Am. Coll. Cardiol. 2013, 61, 463–468. [Google Scholar] [CrossRef] [PubMed]
  49. Mariani, J.; Doval, H.C.; Nul, D.; Varini, S.; Grancelli, H.; Ferrante, D.; Tognoni, G.; Macchia, A. N-3 polyunsaturated fatty acids to prevent atrial fibrillation: Updated systematic review and meta-analysis of randomized controlled trials. J. Am. Heart Assoc. 2013, 2, e005033. [Google Scholar] [CrossRef] [PubMed]
  50. The ASCEND Study Collaborative Group. Effects of n-3 fatty acid supplements in diabetes mellitus. N. Engl. J. Med. 2018, 379, 1540–1550. [Google Scholar] [CrossRef] [PubMed]
  51. A Study of AMR101 to Evaluate Its Ability to Reduce Cardiovascular Events in High Risk Patients with Hypertriglyceridemia and on Statin. 2016. Available online: https://clinicaltrials.gov/ct2/show/NCT01492361 (accessed on 13 October 2018).
  52. Manson, J.E.; Cook, N.R.; Lee, I.M.; Christen, W.; Bassuk, S.S.; Mora, S.; Gibson, H.; Albert, C.M.; Gordon, D.; Copeland, T.; et al. Marine n− 3 fatty acids and prevention of cardiovascular disease and cancer. N. Engl. J. Med. 2018. [Google Scholar] [CrossRef] [PubMed]
  53. US National Institutes of Health. Outcomes Study to Assess STatin Residual Risk Reduction With EpaNova in HiGh CV Risk PatienTs With Hypertriglyceridemia (STRENGTH); US National Institutes of Health: Bethesda, MD, USA, 2015.
  54. Benes, L.B.; Bassi, N.S.; Kalot, M.A.; Davidson, M.H. Evolution of Omega-3 Fatty Acid Therapy and Current and Future Role in the Management of Dyslipidemia. Cardiol. Clin. 2018, 36, 277–285. [Google Scholar] [CrossRef] [PubMed]
  55. Davidson, M.H.; Johnson, J.; Rooney, M.W.; Kyle, M.L.; Kling, D.F. A novel omega-3 free fatty acid formulation has dramatically improved bioavailability during a low-fat diet compared with omega-3-acid ethyl esters: The ECLIPSE (Epanova® compared to Lovaza® in a pharmacokinetic single-dose evaluation) study. J. Clin. Lipidol. 2012, 6, 573–584. [Google Scholar] [CrossRef] [PubMed]
  56. Offman, E.; Marenco, T.; Ferber, S.; Johnson, J.; Kling, D.; Curcio, D.; Davidson, M. Steady-state bioavailability of prescription omega-3 on a low-fat diet is significantly improved with a free fatty acid formulation compared with an ethyl ester formulation: The ECLIPSE II study. Vasc. Health Risk Manag. 2013, 9, 563. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structure and metabolism of major polyunsaturated fatty acids (PUFAs).
Figure 1. Structure and metabolism of major polyunsaturated fatty acids (PUFAs).
Ijms 19 03703 g001
Figure 2. Pleiotropic cardioprotective effects of ω-3 PUFAs.
Figure 2. Pleiotropic cardioprotective effects of ω-3 PUFAs.
Ijms 19 03703 g002
Table 1. Dietary sources of major PUFAs.
Table 1. Dietary sources of major PUFAs.
PUFADietary Source
Linoleic acid (ω-6)Corn, safflower, soybean, sunflower oils
Alpha-linolenic acid (plant-derived ω-3)Flaxseed oil, canola (rapeseed) oil, walnuts, seeds of chia, perilla, green leafy vegetables
Eicosapentaenoic and Docosahexaenoic acids (marine-derived ω-3)Fish, fish oil, other seafood, beef, lamb, ω-3 fortified foods
Table 2. Trials and meta-analyses of ω-3 PUFAs in cardiovascular disease (CVD).
Table 2. Trials and meta-analyses of ω-3 PUFAs in cardiovascular disease (CVD).
StudyStudy DesignNumber of PatientsInterventionFollow-UpOutcome
DART [4]
1989
Secondary prevention RCT2033200–400 g fish per week2 years29% reduction in mortality
GISSI Prevenzione [6]
1999
Secondary prevention RCT11,324882 mg EPA and DHA daily3.5 years15–20% reduction in mortality and CV events
DART-2 [44]
2003
Secondary prevention RCT31142 fish servings per week or 3 fish oil capsules daily3-9 yearsHigher cardiac mortality and SCD
JELIS [9]
2007
Primary and secondary prevention RCT18,6451.8 g EPA daily4.6 years19% reduction in coronary events in CAD patients, no benefit in primary prevention
GISSI-HF [10]
2008
Secondary prevention RCT6975840 mg EPA and DHA daily3.9 years9% reduction in mortality and 8% reduction in hospitalizations
Alpha Omega [14]
2010
Secondary prevention RCT4837226 mg EPA and 150 mg DHA daily3.4 yearsNo benefit
OMEGA [13]
2010
Secondary prevention RCT3851460 mg EPA and 380 mg DHA daily
(vs olive oil control)
1 yearNo benefit
SU.FOL.OM3 [15]
2010
Secondary prevention RCT2501600 mg EPA and DHA daily4.7 yearsNo benefit
ORIGIN [16]
2012
Secondary prevention RCT12,536465 mg EPA and 375 mg DHA daily
(vs olive oil control)
6.2 yearsNo benefit
Rizos et al. [47]
2012
Meta-analysis of 20 RCTs68,6801000 mg EPA and DHA daily
(median)
-No benefit
Risk and Prevention [17]
2013
Primary prevention RCT12,513850 mg of EPA and DHA daily
(vs olive oil control)
5 yearsNo benefit
AREDS2 [46]
2014
Primary prevention RCT4203650 mg EPA plus 350 mg DHA daily4.8 yearsNo benefit
ASCEND [50]
2018
Primary prevention RCT15,4801 g ω-3 PUFA daily
(vs olive oil control)
7.4 yearsNo benefit
REDUCE-IT [51]
2018
Primary prevention RCT81794 g ethyl EPA daily4.9 years25% reduction in major CV events
VITAL [52]
2018
Primary prevention RCT25,8711 g ω-3 PUFA daily5.3 yearsNo benefit
Table 3. ω-3 PUFA formulations.
Table 3. ω-3 PUFA formulations.
Ethyl Esters of EPA and DHAEthyl Esters of EPA OnlyFree Fatty Acids of EPA and DHA
Brand nameLovaza® (GlaxoSmithKline)Vascepa® (Amarin Pharmaceuticals)Epanova® (AstraZeneca)
Approval date200420122014
EPA/DHA (g per capsule)EPA 0.465 g
DHA0.375 g
EPA 1 gEPA 0.550 g
DHA 0.200 g
Dosing2 g (2 capsules) twice daily or 4 g (4 capsules)once daily WITH MEALS2 g (2 capsules) twice daily WITH MEALS2 g (2 capsules)or 4 g (4 capsules) once daily WITH OR WITHOUT MEALS

Share and Cite

MDPI and ACS Style

Goel, A.; Pothineni, N.V.; Singhal, M.; Paydak, H.; Saldeen, T.; Mehta, J.L. Fish, Fish Oils and Cardioprotection: Promise or Fish Tale? Int. J. Mol. Sci. 2018, 19, 3703. https://doi.org/10.3390/ijms19123703

AMA Style

Goel A, Pothineni NV, Singhal M, Paydak H, Saldeen T, Mehta JL. Fish, Fish Oils and Cardioprotection: Promise or Fish Tale? International Journal of Molecular Sciences. 2018; 19(12):3703. https://doi.org/10.3390/ijms19123703

Chicago/Turabian Style

Goel, Akshay, Naga Venkata Pothineni, Mayank Singhal, Hakan Paydak, Tom Saldeen, and Jawahar L. Mehta. 2018. "Fish, Fish Oils and Cardioprotection: Promise or Fish Tale?" International Journal of Molecular Sciences 19, no. 12: 3703. https://doi.org/10.3390/ijms19123703

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

Article Metrics

Back to TopTop