Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Transplant. Mar 24, 2016; 6(1): 125-134
Published online Mar 24, 2016. doi: 10.5500/wjt.v6.i1.125
Post-transplant dyslipidemia: Mechanisms, diagnosis and management
Arnav Agarwal, G V Ramesh Prasad, Division of Nephrology, Department of Medicine, University of Toronto, Toronto, ON M5C 2T2, Canada
Arnav Agarwal, G V Ramesh Prasad, Renal Transplant Program, St. Michael’s Hospital, Toronto, ON M5C 2T2, Canada
Author contributions: Agarwal A and Prasad GVR conducted the research and wrote the paper.
Conflict-of-interest statement: No potential conflict of interest. No financial support.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: G V Ramesh Prasad, MBBS, MSc, MA, FRCPC, FACP, FASN, Division of Nephrology, Department of Medicine, University of Toronto, 61 Queen Street East, 9th Floor, Toronto, ON M5C 2T2, Canada. prasadr@smh.ca
Telephone: +1-416-8673722 Fax: +1-416-8673709
Received: July 29, 2015
Peer-review started: July 31, 2015
First decision: October 27, 2015
Revised: November 26, 2015
Accepted: February 16, 2016
Article in press: February 17, 2016
Published online: March 24, 2016

Abstract

Post-transplant dyslipidemia is highly prevalent and presents unique management challenges to the clinician. The two major outcomes to consider with post-transplant therapies for dyslipidemia are preserving or improving allograft function, and reducing cardiovascular risk. Although there are other cardiovascular risk factors such as graft dysfunction, hypertension, and diabetes, attention to dyslipidemia is warranted because interventions for dyslipidemia have an impact on reducing cardiac events in clinical trials specific to the transplant population. Dyslipidemia is not synonymous with hyperlipidemia. Numerous mechanisms exist for the occurrence of post-transplant dyslipidemia, including those mediated by immunosuppressive drug therapy. Statin therapy has received the most attention in all solid organ transplant recipient populations, although the effect of proper dietary advice and adjuvant pharmacological and non-pharmacological agents should not be dismissed. At all stages of treatment appropriate monitoring strategies for side effects should be implemented so that the benefits from these therapies can be achieved. Clinicians have a choice when there is a conflict between various transplant society and lipid society guidelines for therapy and targets.

Key Words: Cholesterol, Dyslipidemia, Triglycerides, Statins, Immunosuppression

Core tip: Post-transplant dyslipidemia is highly prevalent in all solid organ transplant recipient populations. Guidelines for therapy are derived mostly from general population experiences, although the mechanisms for dyslipidemia due to immunosuppression are distinct and known. Statin therapy has understandably received the most attention in transplant populations but the potential efficacy of other therapeutic strategies should not be ignored.



INTRODUCTION

The great success of solid organ transplantation (SOT) over the past 50 years is demonstrated by the fact that both excellent short-term allograft survival and adequate long-term allograft function without the development of overwhelming comorbidity are routinely expected. Immunosuppressive medication regimens have advanced to the point that acute rejection has declined significantly, and even chronic forms of rejection are being delayed and their effects mitigated. As a result, increased clinician attention is being focused on the general well-being of transplant recipients, apart from allograft health per se, towards which cardiovascular (CV) health is an important component. In turn, each of the traditional CV disease (CVD) risk factors has received a share of the thrust on management strategies in transplant populations[1], including dyslipidemia[2]. However, most interventions are typically mapped to transplant recipients on the basis of evidence garnered from the general population. While the mechanisms for post-transplant dyslipidemia have largely been worked out, it is still not sufficiently known whether there is value to measuring isolated cholesterol subfractions, designing interventions for specific subfractions, or altering immunosuppressive medication regimens towards the goal of improving lipid profiles and CV health.

This review article provides a comprehensive overview of dyslipidemia in SOT recipients, based on the currently available literature. The prevalence and types of post-transplant dyslipidemia are first described, followed by the factors associated with lipid abnormalities, mechanisms of dyslipidemia after transplantation, the consequences of dyslipidemia, and finally its clinical diagnosis, monitoring, and treatment.

PREVALENCE AND TYPES OF DYSLIPIDEMIA

At one time, the prevalence of hyperlipidemia, which is the most common form of dyslipidemia, was estimated to be as high as 80% in kidney transplant recipients (KTR)[3]. Reports of the high prevalence of hyperlipidemia go back as far as 1973[4]. In the azathioprine-corticosteroid era of post-transplant immunosuppression, the prevalence rate was estimated at 50%-78%[5-7]. Hypertriglyceridemia was just as common as hypercholesterolemia. However, with the introduction of cyclosporine, hypercholesterolemia has become the predominant abnormality[8], particularly low density lipoprotein (LDL) cholesterol elevation[9]. An early prevalence estimate of hyperlipidemia of over 50% has been reported in heart transplant recipients (HTR)[10]. Lung transplantation has been associated with a prevalence of hypercholesterolemia and hypertriglyceridemia of 32% and 41% respectively[11]. Estimates of dyslipidemia in liver transplant recipients (LTR) include 43%[12] and 31%-51%[13]. The point prevalence of hyperlipidemia is unlikely to vary over time post-transplant. In KTR, hyperlipidemia is persistent if untreated. It is also possible that the prevalence is higher with time, due to inadequate surveillance in long-term patients. Cumulative factors such as advancing age, immunosuppression, weight gain, and the development of diabetes may all contribute to developing hyperlipidemia over time. Hyperlipidemia has also been documented in children after kidney transplantation[14].

Dyslipidemia is not synonymous with hyperlipidemia, so it is conceivable that dyslipidemia may still be present despite normal lipid levels. Increased levels of very low-density lipoprotein (VLDL) cholesterol and decreased high-density lipoprotein (HDL) cholesterol levels despite normal “total” cholesterol levels are well-described[15]. A low HDL has been noted in lung transplant recipients[16] but not necessarily in HTR[17]. In particular, a low level of the HDL2 sub-fraction has been reported after kidney transplantation[18]. There is also a higher amount of oxidized LDL cholesterol[19,20]. The lipid profile of LTR has also recently been elaborated. Compared to controls with no chronic medical disease, LTR had higher apolipoprotein B, small dense LDL cholesterol, and VLDL cholesterol concentrations[21]. VLDL cholesterol concentration was also related to cyclosporine levels[21]. Despite the initial excitement surrounding HDL sub-fractions and oxidized LDL cholesterol[18-20], measurement of these lipid forms has yet to reach clinical practice almost thirty years after their description. The prevalence of small dense LDL cholesterol has been estimated at 26%-33% in KTR[22]. Elevations in serum apolipoprotein B and lipoprotein (a)[23], as well as decreased apolipoprotein A-I[24], and decreased ratios of apolipoproteins C-II to C-III[25,26] also generated significant interest, but at the present time none of these are routinely measured in a clinical setting. More recently, “non-HDL cholesterol”, which is simply the total cholesterol minus HDL cholesterol level, has received attention in transplant patients[27]. However, the importance of this particular measure has not yet been placed in full context.

FACTORS ASSOCIATED WITH LIPID ABNORMALITIES

Given the variety of lipid abnormalities seen, it is useful to divide factors contributing to dyslipidemia into those that contribute primarily to hypercholesterolemia and those that contribute primarily to hypertriglyceridemia, notwithstanding their qualitative impact that cannot be routinely assessed in the clinic. These risk factors are summarized in Table 1 (partially adapted from[8]).

Table 1 Factors associated with lipid abnormalities after transplantation.
HypercholesterolemiaHypertriglyceridemia
Genetic predispositionGenetic predisposition
AgeExcessive dietary intake of carbohydrates, cholesterol, and saturated fat
Excessive dietary intake of cholesterol and saturated fatsObesity
ObesityProteinuria
ProteinuriaRenal insufficiency
Anti-hypertensive agents, e.g., diuretics, beta-blockersCorticosteroids
CorticosteroidsMammalian target-of-rapamycin inhibitors (sirolimus)
Calcineurin-inhibitors (cyclosporine, possibly tacrolimus)
Mammalian target-of-rapamycin inhibitors (sirolimus, everolimus)

Hypercholesterolemia is considered more prevalent based on the available literature, although the literature is dominated by North American and Western European publications. Genetic predisposition may be based on the prevalence of various polymorphisms of the lipoprotein system. For example, the GA genotype of the apo A-1 promoter region has been associated with a greater rise in LDL cholesterol after heart transplantation[28]. Conversely, some genes such as the TP-binding cassette subfamily B member 1 (ABCB1) lose their association with LDL cholesterol after heart transplantation[29]. Advanced age is another non-modifiable risk factor. However, modifiable risk factors such as a diet high in saturated fat may be just as important as a contributor to hypercholesterolemia. Obesity, proteinuria either as a result of native or transplant kidney disease, or the use of thiazide diuretics or beta-blockers for hypertension and heart disease may also contribute. Corticosteroids, cyclosporine, and sirolimus may all cause elevations in cholesterol levels[8]. Although tacrolimus is generally believed to cause less elevation in LDL cholesterol than cyclosporine, this may not always be the case, particularly in LTR in whom lipid levels may correlate with tacrolimus levels[30]. The association with sirolimus is particularly strong. LDL cholesterol levels were higher in the sirolimus arm of the Symphony study[31].

In the case of post-transplant hypertriglyceridemia, as with hypercholesterolemia, genetic predisposition plays an important role. The apolipoprotein E 2/2 and 2/3 genotypes are associated with elevated triglycerides after kidney transplantation[32]. The apo A-1 promoter region[28] also correlates with elevated triglycerides. The development of hypertriglyceridemia in response to sirolimus has been subject to genetic analysis, with positive associations demonstrated with the ABCB1 1236 TT homozygote and the interleukin-10 1082AA homozygote in the case of KTR[33]. Age, however, seems to be less important as a risk factor for hypertriglyceridemia. A diet rich in simple sugars predisposes to hypertriglyceridemia, and although obesity and proteinuria are also associated with hypertriglyceridemia, poor renal function per se appears to be an additional risk factor[8]. Sirolimus is more strongly associated with hypertriglyceridemia than hypercholesterolemia, with even a lower drug exposure leading to this abnormality[31], although the contribution of other immunosuppressive drugs is less clear. More common is the association of hypertriglyceridemia with other metabolic syndrome components[1].

MECHANISMS OF POST-TRANSPLANT DYSLIPIDEMIA

Immunosuppressive agents contribute significantly and specifically to lipid abnormalities after SOT.

Corticosteroids induce insulin resistance. The resultant hyperinsulinemia leads to increased hepatic uptake of free fatty acids (FFA)[34]. FFA constitutes the main substrate for VLDL cholesterol synthesis. FFA synthetase and acetyl-CoA carboxylase are also increased by steroids[35] and so hepatic synthesis of VLDL is increased. Insulin resistance also leads to a reduction in lipoprotein lipase, which leads to reduced triglyceride clearance[36]. There is an increased conversion of VLDL to LDL cholesterol, leading to a rise in LDL cholesterol levels. Yet another contributory mechanism is down-regulation of LDL receptor expression[37]. Finally, corticosteroids increase the activity of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA), which is the rate-limiting step in the cholesterol biosynthetic pathway[37].

Cyclosporine interferes with the binding of LDL cholesterol to the LDL receptor. As a result, there is a decline in LDL clearance, leading to a rise in LDL cholesterol levels. In this respect, there may be an additive effect of cyclosporine with corticosteroids. Cyclosporine also interferes with bile acid synthesis[38] by interfering with the enzyme 26 hydroxylase[15]. Decreased bile acid synthesis in turn leads to LDL receptor down-regulation, further reducing the clearance of cholesterol. Cyclosporine, by virtue of being highly lipophilic, is transported within the core of LDL cholesterol particles. In the process, it may change the molecular configuration of LDL[39] and alter the normal feedback regulation of cholesterol synthesis[8]. Glucose intolerance may even potentiate the effect of cyclosporine on lipid levels. The effects of tacrolimus on lipid metabolism are generally similar to those of cyclosporine, so it remains unclear why tacrolimus is associated with less hyperlipidemia.

Sirolimus provides a fascinating instance of a strong connection between pharmacotherapy and dyslipidemia on the one hand, yet ongoing debate about its cardiovascular effects both harmful and protective on the other. Sirolimus may inhibit lipoprotein lipase[40] and decrease lipolysis. There may also be hepatic over-production of lipoprotein in general[41]. Other effects include a decrease in apolipoprotein B100 catabolism[42]. Finally, sirolimus alters insulin signaling, increases the activity of tissue lipase, and increases the secretion of VLDL cholesterol[40]. Sirolimus is almost never used as monotherapy for transplant-related immunosuppression and so likely acts in a synergistic manner with other immunosuppressive agents in promoting dyslipidemia. Sirolimus is also used as an anti-proliferative agent in endovascular stents, but the amount of exposure is unlikely to promote lipid abnormalities in that instance.

CONSEQUENCES OF DYSLIPIDEMIA POST-TRANSPLANTATION

SOT recipients, especially KTR, are at high risk for the development of post-transplant CVD. The link between dyslipidemia and CVD may not be as strong as, for instance, diabetes[1], but there is no reason to believe that the association does not hold in transplant populations as it does in the general population. The underlying assumptions, however, are not so straightforward. Atherosclerosis is accelerated after transplantation[8], and this can be linked at least retrospectively to cardiovascular events[43]. The association of elevations in cholesterol to cardiovascular events may be stronger with cholesterol than with triglycerides, and likewise, more associated with ischemic heart disease than other forms of CVD such as cerebrovascular disease or peripheral vascular disease[44]. It has been estimated that an increase in LDL cholesterol concentration by 2 mmol/L doubles the risk for major adverse cardiac events (MACE), comparable to an age increase by 23 years[45]. A low level of HDL cholesterol has been associated with a threefold increase in post-transplant MACE[46] and also an increase in all-cause mortality[46]. Non-HDL cholesterol has been found to be as powerful a predictor of MACE as diabetes in KTR[47].

Despite some correlative success between various lipid level abnormalities and MACE, consistent demonstration of the association remains quite difficult, since a large proportion of MACE is explained by unmeasured risk factors outside of the traditional Framingham risk factors, including dyslipidemia[48]. Moreover, hyperlipidemia has not been found to be an independent risk factor for MACE in non-Caucasian populations in whom non-traditional risk factors may be more important[49]. The Assessment of Lescol in Renal Transplantation (ALERT) study database[50] has formed the basis for significant understanding of the link of dyslipidemia to human pathology, but all links remain associative. Data from another large database, the Patient Outcomes in Renal Transplantation study, however, indicate that dyslipidemia adds little predictive value to more transplant-specific and graft-related variables in predicting acute myocardial infarction (MI), coronary artery revascularization or sudden death[51]. Nonetheless, hypertriglyceridemia in particular has been associated with the progression of coronary artery calcification (CAC) in KTR[52], although it must be understood that CAC is only a surrogate marker for CVD and is itself controversial in that respect at best. Information regarding dyslipidemia and CVD risk in SOT outside of kidney transplantation is limited. In HTR, hypercholesterolemia has been associated with non-fatal MACE in a retrospective analysis[53]. Although LTR display a higher CVD risk and CVD is the leading cause of non-graft related deaths[54], demonstration of dyslipidemia as a CVD risk factor lags behind other risk factors such as diabetes and hypertension[54]. While other studies in liver transplantation have also either not addressed or failed to demonstrate a relationship of dyslipidemia to CVD[55], a link with CVD has been found with metabolic syndrome and hypertriglyceridemia[56].

Dyslipidemia, or at least one aspect of it (hypertriglyceridemia and low HDL cholesterol), is one among five components constituting the metabolic syndrome. Therefore, it is helpful to understand the contribution of dyslipidemia to post-transplant morbidity relative to its sister CVD risk factors such as hypertension, microalbuminuria, obesity and dysglycemia. As one example, in a cohort study of 1182 stable KTR with close to 7500 patient-years of follow-up, dyslipidemia did not attain statistical significance as a stand-alone CVD risk factor, but provided additive value to dysglycemia and microalbuminuria in predicting MACE ahead of hypertension and obesity[1]. Interventions for dyslipidemia have an impact on reducing cardiac deaths and non-fatal MI in clinical trials specific to the transplant population[2]. Therefore, attention to dyslipidemia is indeed warranted.

In contrast to other populations, SOT permits the assessment of the relationship of dyslipidemia to the performance of the allograft itself. It is possible, at least theoretically, that an allograft is predisposed differently to metabolic injury compared to a native organ due to its intersection with injury from the actions of the immune system. Hyperlipidemia is a paradigmatic contributor to chronic kidney allograft injury as a “non-immune” risk factor[57]. Atherosclerosis is believed to be an integral part of the rejection process, by virtue of the accumulation of oxidized LDL cholesterol in the kidney interstitium leading to fibrosis[58]. However, this may be a bidirectional relationship, with lipid abnormalities perpetuated by allograft dysfunction. Hypercholesterolemia has been associated with kidney allograft loss in the context of prior acute rejection[59]. Hypercholesterolemia itself may predispose to acute rejection, by altering cyclosporine pharmacokinetics and increased binding with less tissue release[60]. At a clinical level, overall there has been little progress in understanding beyond earlier studies that demonstrate associations between early post-kidney transplant lipid levels and subsequent graft function or death-censored graft loss[61,62]. A demonstrable effect of lipid levels on graft function may be blunted by more aggressive lipid lowering in transplant recipients for cardiovascular protection with the advent of other potent medical therapies, as well as due to data on safety and efficacy of lipid-lowering therapies from studies such as ALERT. Effective immunosuppressive therapy, and other graft-related variables such as donor organ quality may also be too overpowering to allow for demonstrating any effects of lipid profiles on graft function.

DIAGNOSIS AND MONITORING

The diagnosis of dyslipidemia in SOT recipients typically starts with a lipid profile obtained after 8 to 12 h of fasting. Although non-fasting lipid level measurement has been occasionally recommended for the general population, transplant recipients should be considered a high-risk group for CVD and should therefore be subject to fasting measurements. Normal “cut-offs” for hyperlipidemia are typically the same as those used for the general population[15], in the absence of any evidence to the contrary. Measurements of lipid parameters beyond total, HDL and LDL cholesterol, or triglycerides are rarely performed outside of research studies. All recipients require at least one such fasting lipid profile, with the first profile obtained at some point during the first year. An initial evaluation as soon as three months post-transplant has been recommended[8]. A Canadian commentary on the 2009 KDIGO Clinical Practice Guideline[63] advises initial measurement 2-3 mo post-transplant, 2-3 mo after a change in treatment, and annually thereafter[63]. Annual monitoring is corroborated by older European guidelines[64]. More recently, the need for repeat lipid level measurement in many forms of chronic kidney disease has been questioned[65], mostly on the basis of lack of evidence for utility and the absence of clinical trial data. A useful approach might be to gauge the transplant recipient’s overall cardiac risk profile, and reserve lipid monitoring to those at a perceived higher CV risk, understanding that chronic graft dysfunction may itself be a high-risk equivalent.

TREATMENT

All transplant recipients require consultation with a dietician on a regular, if infrequent basis. A diet low in total fat, saturated fatty acids, and cholesterol can be prescribed as an initial measure, particularly in KTR who by definition have chronic kidney disease (CKD). Hypertriglyceridemia may be controlled with the help of a diet low in simple sugars and alcohol. The American Heart Association Step I diet can be considered as a starting point for those with an elevated LDL cholesterol level. Limiting dietary cholesterol intake to under 300 mg/d and caloric intake from fat to under 30% of the total caloric intake may be helpful. A further Step II approach would be to limit these further to under 200 mg/d and 10% respectively. However, evidence of the efficacy of such diets in transplant recipients is lacking. Balance of the saturated to polyunsaturated fat intake should be sought. Losing excess body weight is important, and control of total caloric intake is likely to have the biggest impact[3]. Improved glycemic control will also help to improve hyperlipidemia. Adherence to prescribed diets can be highly variable, and so culture-specific dietary interventions may be needed to improve adherence. Incorporation of soy protein into the diet[15] has not been tested in SOT recipients. The success of dietary intervention alone at improving dyslipidemia has been estimated at under 20% in KTR[66].

Non-conventional pharmacological therapies have received some attention, particularly in KTR. There may be attempts by SOT recipients to reduce their lipid levels through herbal supplements. Obviously, this can be quite dangerous in the context of immunosuppressive medication. For example, red yeast rice (Monascus purpureus) is a remedy designed to lower cholesterol levels. Red yeast rice contains varieties of mevinic acid, a naturally occurring statin, that has been associated with rhabdomyolysis[67]. Since statin concentrations show batch variability and production is unregulated, herbal remedies should be discouraged. Fish oil is rich in omega-3 polyunsaturated fatty acids and can lower serum triglycerides[68] by reducing its hepatic synthesis. Fish oil may even have a beneficial effect on graft function[69], although further studies are clearly needed before this therapy can be endorsed. Finally, the use of antioxidants particularly antioxidant vitamins has also been considered based on the rationale that oxidized LDL cholesterol is particularly atherogenic. However, antioxidants are not considered efficacious at preventing CVD in the general population[70]. The administration of homocysteine-lowering therapies is also not recommended[68].

HMG-CoA reductase inhibitors, or statins, are widely used in KTR, LTR and HTR. They are potent reducers of LDL cholesterol levels, and are generally considered safe as long as patients are appropriately monitored. Some statins may have modest beneficial effects in lowering serum triglycerides and raising HDL cholesterol levels[15]. There are also claims that statins have pleiotropic effects, involving a favorable modulation of endothelial function that translates into improved CV health[71]. Since CKD may be a high-risk equivalent for CVD, this paradigm seems appealing. Perhaps the most commonly used statin is atorvastatin, despite the fact that the single prospective randomized trial of statins vs placebo in KTR, the ALERT study, utilized a different but older statin, namely fluvastatin[2]. This large trial was successful in demonstrating benefit for secondary CVD endpoints, but not the primary composite endpoint. Since a greater reduction in LDL cholesterol is believed to translate into greater cardiovascular advantage, atorvastatin or another more potent statin such as rosuvastatin may be preferred by clinicians. Atorvastatin and rosuvastatin are not as dependent on time of day for administration as the other statins[15]. Maximum doses used are generally less than those for the general population, although the rationale for this practice in SOT recipients is based more on the known interaction of calcineurin-inhibitors through the CYP3A4 isoenzyme system[72] than clinical evidence. Transplant recipients are also prescribed multiple other medications that can interact through this busy enzyme system, and so regular monitoring for the major statin-induced side effects, namely myositis or rhabdomyolysis, as well as hepatitis, is warranted. Simvastatin has recently been singled out as an offender with regards to rhabdomyolysis[15]. However, statins remain appealing agents to use, being once-daily drugs and especially since they have also been shown to improve patient survival[73]. Detailed guidelines on the use of specific statins in KTR are available[15]. The recommended target for LDL cholesterol is a level under 2.0 mmol/L[63] although this may be based more on extrapolation from the general population. A non-HDL cholesterol target of under 3.36 mmol/L in adults and 4.14 mmol/L in adolescents is a recommendation that serves as a surrogate for forms of cholesterol besides LDL cholesterol[63]. It might be easier to initiate statin therapy early after the transplant, when other medications are being adjusted and patients are more receptive to new suggestions for optimizing their overall health. As more time elapses post-transplant, longer-term risks such as CVD may become less appreciated and the introduction of new medications may be perceived as an unnecessary risk or potential threat to allograft health.

Statins are also used in other SOT recipients besides KTR. Statins are generally considered safe in LTR with no severe complications[74], although pravastatin in particular has been recommended[75]. Statins also reduce accelerated graft atherosclerosis and mortality in HTR, especially pravastatin and simvastatin[76], although atorvastatin has also been studied[77]. The benefit of statins has also been extended to pediatric and adolescent HTR[78]. Although the literature with other solid organs is not as expansive as that for KTR, there is no reason to believe that safety and efficacy concerns are substantially different among them.

If a maximal dose of statin proves to be insufficient at bringing the LDL cholesterol level to target, then consideration can be given to adding a second agent. Ezetimibe inhibits cholesterol absorption at the level of the intestinal brush border. Ezetimibe is generally safe in KTR[79] although consultation at this point with a lipid metabolism specialist could be considered, particularly when increased transaminase levels have previously been noted with statin therapy. There are no time-of-day restrictions with ezetimibe. Ezetimibe can be considered for use in LTR[75,80] and in HTR[81] in whom it has also been tested as monotherapy[82]. Ezetimibe also increases HDL cholesterol levels in some HTR[83].

Fibrates reduce hepatic VLDL cholesterol synthesis and increase lipoprotein lipase activity, decreasing triglyceride levels and increasing HDL cholesterol levels to some extent. LDL cholesterol levels may also decline, but not to the same extent as triglycerides. Among fibrates, fenofibrate is generally preferred over gemfibrozil due to less myotoxicity when added to a statin, as a result of less drug interaction. A concern regarding fibrate use is the potential for decline in kidney function in the presence of existing renal insufficiency[84]. The use of fibrates should be avoided in advanced CKD since fibrates are metabolized by the kidneys[15]. Their efficacy at preventing cardiac events in other population groups such as type 2 diabetes has also been seriously questioned[85] and they are rarely, if ever used in combination with statins. Fibrates are believed to be generally well tolerated in LTR[86]. Severe hypertriglyceridemia however may require plasma exchange in order to manage the associated pancreatitis[87].

Niacin and bile acid sequestrants have both been explored for use in SOT recipients. Niacin could be considered as an option for montherapy to reduce LDL cholesterol levels in those intolerant to statins[15]. Niacin has been studied favorably in combination with simvastatin in the general population at preventing coronary disease[70], although this has also been questioned[88]. If used, a gradual dose escalation is required, and liver enzyme monitoring is warranted. Bile acid sequestrants are not popular in transplant recipients due to their gastrointestinal side effects including nausea and bloating, which patients are often already prone to as a result of immunosuppressive drug therapy. They can also interfere with the absorption of immunosuppressive drugs and should be separately administered from them by at least two hours.

Table 2 provides one suggested summary approach to post-transplant hyperlipidemia that can be tailored to individual clinic circumstances. However, relevant national society guidelines should preferably be followed. Clinicians have a choice when there is a conflict between various transplant society and lipid society guidelines for therapy and targets. There are few, if any clinical trials where modification of immunosuppressive therapy has been pursued with the intention of addressing dyslipidemia or reducing CVD risk and similarly, large database reviews are not sufficiently informative in this respect.

Table 2 A suggested approach to managing post-transplant dyslipidemia.
Initial post-transplant periodManage acute graft-related concerns
Optimize immunosuppressive medication to graft function
2-3 mo post-transplantMeasure 8-12 h fasting lipid profile
If LDL cholesterol and/or triglyceride level above target1Dietician consult
2-3 mo post-dietary interventionMeasure 8-12 h fasting lipid profile
If LDL cholesterol and/or triglyceride level still above target1Initiate statin therapy, e.g., atorvastatin 10 mg/d or rosuvastatin 5 mg/d
Assess for potential drug interactions
Monitor creatine kinase and liver transaminase levels
2-3 mo post-statin initiationMeasure 8-12 h fasting lipid profile
If LDL cholesterol and/or triglyceride level still above target1Repeat all of the above until targets are achieved. Increase statin dose as tolerated to a maximum acceptable dose with each measurement not at target. If targets are not achieved then consider adding a supplemental agent, e.g., ezetimibe 10 mg/d
If LDL cholesterol and/or triglyceride level still above target1Consider consultation with lipid specialist
LDL and triglyceride target levels achievedAnnual monitoring of lipid levels. Consider more frequent monitoring for side effects
At all times post-transplantGauge overall cardiovascular risk
CONCLUSION

Post-transplant dyslipidemia is highly prevalent and presents unique management challenges to the clinician. There are two major outcomes when considering post-transplant therapies: preserving or improving allograft function and reducing cardiovascular risk. Attention to dyslipidemia is warranted because interventions for dyslipidemia have an impact on reducing cardiac events in clinical trials specific to the transplant population. Dyslipidemia is not synonymous with hyperlipidemia. Numerous mechanisms exist for the occurrence of post-transplant dyslipidemia, including those mediated by immunosuppressive drug therapy. Statin therapy has received the most attention in all SOT recipient populations, although the effect of proper dietary advice and adjuvant pharmacological or non-pharmacological agents should not be dismissed. At all stages of treatment appropriate monitoring for side effects should be implemented so that the benefits from these therapies can be achieved.

Footnotes

P- Reviewer: Vlachopanos G S- Editor: Gong XM L- Editor: A E- Editor: Wang CH

References
1.  Prasad GV, Huang M, Silver SA, Al-Lawati AI, Rapi L, Nash MM, Zaltzman JS. Metabolic syndrome definitions and components in predicting major adverse cardiovascular events after kidney transplantation. Transpl Int. 2015;28:79-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
2.  Holdaas H, Fellström B, Jardine AG, Holme I, Nyberg G, Fauchald P, Grönhagen-Riska C, Madsen S, Neumayer HH, Cole E. Effect of fluvastatin on cardiac outcomes in renal transplant recipients: a multicentre, randomised, placebo-controlled trial. Lancet. 2003;361:2024-2031.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 680]  [Cited by in F6Publishing: 627]  [Article Influence: 29.9]  [Reference Citation Analysis (0)]
3.  Gonyea JE, Anderson CF. Weight change and serum lipoproteins in recipients of renal allografts. Mayo Clin Proc. 1992;67:653-657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 44]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
4.  Ghosh P, Evans DB, Tomlinson SA, Calne RY. Plasma lipids following renal transplantation. Transplantation. 1973;15:521-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 55]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
5.  Cattran DC, Steiner G, Wilson DR, Fenton SA. Hyperlipidemia after renal transplantation: natural history and pathophysiology. Ann Intern Med. 1979;91:554-559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 86]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
6.  Nicholls AJ, Cumming AM, Catto GR, Edward N, Engeset J. Lipid relation ships in dialysis and renal transplant patients. Q J Med. 1981;50:149-160.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Kasiske BL, Umen AJ. Persistent hyperlipidemia in renal transplant patients. Medicine (Baltimore). 1987;66:309-316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 119]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
8.  Hricik DE. Hyperlipidemia in renal transplant recipients. Graft. 2000;3:177-184.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Raine AE, Carter R, Mann JI, Chapman JR, Morris PJ. Increased plasma LDL cholesterol after renal transplantation associated with cyclosporine immunosuppression. Transplant Proc. 1987;19:1820-1821.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Becker DM, Markakis M, Sension M, Vitalis S, Baughman K, Swank R, Kwiterovich PO, Pearson TA, Achuff SC, Baumgartner WA. Prevalence of hyperlipidemia in heart transplant recipients. Transplantation. 1987;44:323-325.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
11.  Nash EF, Stephenson A, Helm EJ, Durie PR, Tullis E, Singer LG, Chaparro C. Impact of lung transplantation on serum lipids in adults with cystic fibrosis. J Heart Lung Transplant. 2011;30:188-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
12.  Dopazo C, Bilbao I, Castells LL, Sapisochin G, Moreiras C, Campos-Varela I, Echeverri J, Caralt M, Lázaro JL, Charco R. Analysis of adult 20-year survivors after liver transplantation. Hepatol Int. 2015;9:461-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
13.  Parekh J, Corley DA, Feng S. Diabetes, hypertension and hyperlipidemia: prevalence over time and impact on long-term survival after liver transplantation. Am J Transplant. 2012;12:2181-2187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 66]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
14.  Van Gool S, Van Damme-Lombaerts R, Cobbaert C, Proesmans W, Eggermont E. Lipid and lipoprotein abnormalities in children on hemodialysis and after renal transplantation. Transplant Proc. 1991;23:1375-1377.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Riella LV, Gabardi S, Chandraker A. Dyslipidemia and its therapeutic challenges in renal transplantation. Am J Transplant. 2012;12:1975-1982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 62]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
16.  Reed RM, Hashmi S, Eberlein M, Iacono A, Netzer G, DeFilippis A, Girgis RE, Toth PP, Scharf S, Jones S. Impact of lung transplantation on serum lipids in COPD. Respir Med. 2011;105:1961-1968.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
17.  Singh N, Jacobs F, Rader DJ, Vanhaecke J, Van Cleemput J, De Geest B. Impaired cholesterol efflux capacity and vasculoprotective function of high-density lipoprotein in heart transplant recipients. J Heart Lung Transplant. 2014;33:499-506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
18.  Ettinger WH, Bender WL, Goldberg AP, Hazzard WR. Lipoprotein lipid abnormalities in healthy renal transplant recipients: persistence of low HDL2 cholesterol. Nephron. 1987;47:17-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
19.  Ghanem H, van den Dorpel MA, Weimar W, Man in ‘T Veld AJ, El-Kannishy MH, Jansen H. Increased low density lipoprotein oxidation in stable kidney transplant recipients. Kidney Int. 1996;49:488-493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 65]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
20.  Sutherland WH, Walker RJ, Ball MJ, Stapley SA, Robertson MC. Oxidation of low density lipoproteins from patients with renal failure or renal transplants. Kidney Int. 1995;48:227-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 70]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
21.  Chhatrala R, Siddiqui MB, Stravitz RT, Driscoll C, Sanyal A, Sargeant C, Luketic V, Sharma A, Sterling R, Matherly S. Evolution of serum atherogenic risk in liver transplant recipients: Role of lipoproteins and metabolic and inflammatory markers. Liver Transpl. 2015;21:623-630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 36]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
22.  Badiou S, Garrigue V, Dupuy AM, Chong G, Cristol JP, Mourad G. Small dense low-density lipoprotein in renal transplant recipients: a potential target for prevention of cardiovascular complications? Transplant Proc. 2006;38:2314-2316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
23.  Hilbrands LB, Demacker PN, Hoitsma AJ, Stalenhoef AF, Koene RA. The effects of cyclosporine and prednisone on serum lipid and (apo)lipoprotein levels in renal transplant recipients. J Am Soc Nephrol. 1995;5:2073-2081.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Gokal R, Mann JI, Moore RA, Morris PJ. Hyperlipidaemia following renal transplantation. A study of the prevalence, ‘natural history’ and dietary treatment. Q J Med. 1979;48:507-517.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Cassader M, Ruiu G, Gambino R, Alemanno N, Triolo G, Pagano G. Lipoprotein-apolipoprotein changes in renal transplant recipients: a 2-year follow-up. Metabolism. 1991;40:922-925.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
26.  Averna MR, Barbagallo CM, Sparacino V, Caputo F, Mancino C, Calabrese S, Notarbartolo A. Follow-up of lipid and apoprotein levels in renal transplant recipients. Nephron. 1991;58:255-256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
27.  Spinelli GA, Felipe CR, Park SI, Mandia-Sampaio EL, Tedesco-Silva H, Medina-Pestana JO. Lipid profile changes during the first year after kidney transplantation: risk factors and influence of the immunosuppressive drug regimen. Transplant Proc. 2011;43:3730-3737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 46]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
28.  González-Amieva A, López-Miranda J, Marín C, Pérez-Martinez P, Gómez P, Paz-Rojas E, Arizón JM, Jiménez-Perepérez JA, Concha M, Pérez-Jiménez F. The apo A-I gene promoter region polymorphism determines the severity of hyperlipidemia after heart transplantation. Clin Transplant. 2003;17:56-62.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Taegtmeyer AB, Breen JB, Smith J, Rogers P, Kullak-Ublick GA, Yacoub MH, Banner NR, Barton PJ. Effect of ABCB1 genotype on pre- and post-cardiac transplantation plasma lipid concentrations. J Cardiovasc Transl Res. 2011;4:304-312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
30.  Li HY, Li B, Wei YG, Yan LN, Wen TF, Zhao JC, Xu MQ, Wang WT, Ma YK, Yang JY. Higher tacrolimus blood concentration is related to hyperlipidemia in living donor liver transplantation recipients. Dig Dis Sci. 2012;57:204-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
31.  Claes K, Meier-Kriesche HU, Schold JD, Vanrenterghem Y, Halloran PF, Ekberg H. Effect of different immunosuppressive regimens on the evolution of distinct metabolic parameters: evidence from the Symphony study. Nephrol Dial Transplant. 2012;27:850-857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
32.  Li HF, Han CF, Wang YX, Lu YS, Zou HQ, Xu QQ. Effect of apolipoprotein E gene polymorphism on serum lipid level before and after renal transplantation. Transplant Proc. 2010;42:2513-2517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
33.  Sam WJ, Chamberlain CE, Lee SJ, Goldstein JA, Hale DA, Mannon RB, Kirk AD, Hon YY. Associations of ABCB1 and IL-10 genetic polymorphisms with sirolimus-induced dyslipidemia in renal transplant recipients. Transplantation. 2012;94:971-977.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
34.  Pagano G, Bruno A, Cavallo-Perin P, Cesco L, Imbimbo B. Glucose intolerance after short-term administration of corticosteroids in healthy subjects. Prednisone, deflazacort, and betamethasone. Arch Intern Med. 1989;149:1098-1101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
35.  Hays AP, Hill RB. Enzymes of lipid synthesis in the liver of the cortisone-treated rat. Biochim Biophys Acta. 1965;98:646-648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
36.  Ibels SL, Reardon MF, Nestel PJ. Plasma post-heparin lipolytic activity and triglyceride clearance in uremic and hemodialysis patients and renal allograft recipients. J Lab Clin Med. 1976;87:648-658.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Kobashigawa JA, Kasiske BL. Hyperlipidemia in solid organ transplantation. Transplantation. 1997;63:331-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 257]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
38.  Princen HM, Meijer P, Wolthers BG, Vonk RJ, Kuipers F. Cyclosporin A blocks bile acid synthesis in cultured hepatocytes by specific inhibition of chenodeoxycholic acid synthesis. Biochem J. 1991;275:501-505.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 85]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
39.  de Groen PC. Cyclosporine, low-density lipoprotein, and cholesterol. Mayo Clin Proc. 1988;63:1012-1021.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 111]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
40.  Morrisett JD, Abdel-Fattah G, Hoogeveen R, Mitchell E, Ballantyne CM, Pownall HJ, Opekun AR, Jaffe JS, Oppermann S, Kahan BD. Effects of sirolimus on plasma lipids, lipoprotein levels, and fatty acid metabolism in renal transplant patients. J Lipid Res. 2002;43:1170-1180.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Massy ZA, De Bandt JP, Morelon E, Thevenin M, Lacour B, Kreis H. Hyperlipidaemia and post-heparin lipase activities in renal transplant recipients treated with sirolimus or cyclosporin A. Nephrol Dial Transplant. 2000;15:928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
42.  Kasiske BL, de Mattos A, Flechner SM, Gallon L, Meier-Kriesche HU, Weir MR, Wilkinson A. Mammalian target of rapamycin inhibitor dyslipidemia in kidney transplant recipients. Am J Transplant. 2008;8:1384-1392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 116]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
43.  Drüeke TB, Abdulmassih Z, Lacour B, Bader C, Chevalier A, Kreis H. Atherosclerosis and lipid disorders after renal transplantation. Kidney Int Suppl. 1991;31:S24-S28.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Aakhus S, Dahl K, Widerøe TE. Cardiovascular morbidity and risk factors in renal transplant patients. Nephrol Dial Transplant. 1999;14:648-654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 175]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
45.  Soveri I, Holdaas H, Jardine A, Gimpelewicz C, Staffler B, Fellström B. Renal transplant dysfunction--importance quantified in comparison with traditional risk factors for cardiovascular disease and mortality. Nephrol Dial Transplant. 2006;21:2282-2289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 45]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
46.  Barn K, Laftavi M, Pierce D, Ying C, Boden WE, Pankewycz O. Low levels of high-density lipoprotein cholesterol: an independent risk factor for late adverse cardiovascular events in renal transplant recipients. Transpl Int. 2010;23:574-579.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
47.  Holme I, Fellstrom B, Jardine A, Holdaas H. Comparison of predictive ability of lipoprotein components to that of traditional risk factors of coronary events in renal transplant recipients. Atherosclerosis. 2010;208:234-239.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
48.  Silver SA, Huang M, Nash MM, Prasad GV. Framingham risk score and novel cardiovascular risk factors underpredict major adverse cardiac events in kidney transplant recipients. Transplantation. 2011;92:183-189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
49.  Prasad GV, Vangala SK, Silver SA, Wong SC, Huang M, Rapi L, Nash MM, Zaltzman JS. South Asian ethnicity as a risk factor for major adverse cardiovascular events after renal transplantation. Clin J Am Soc Nephrol. 2011;6:204-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
50.  Holdaas H, Fellström B, Holme I, Nyberg G, Fauchald P, Jardine A, Grönhagen-Riska C, Madsen S, Neumayer HH, Cole E. Effects of fluvastatin on cardiac events in renal transplant patients: ALERT (Assessment of Lescol in Renal Transplantation) study design and baseline data. J Cardiovasc Risk. 2001;8:63-71.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Israni AK, Snyder JJ, Skeans MA, Peng Y, Maclean JR, Weinhandl ED, Kasiske BL. Predicting coronary heart disease after kidney transplantation: Patient Outcomes in Renal Transplantation (PORT) Study. Am J Transplant. 2010;10:338-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 138]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
52.  Seyahi N, Cebi D, Altiparmak MR, Akman C, Ataman R, Pekmezci S, Serdengecti K. Progression of coronary artery calcification in renal transplant recipients. Nephrol Dial Transplant. 2012;27:2101-2107.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 40]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
53.  Kobashigawa JA, Starling RC, Mehra MR, Kormos RL, Bhat G, Barr ML, Sigouin CS, Kolesar J, Fitzsimmons W. Multicenter retrospective analysis of cardiovascular risk factors affecting long-term outcome of de novo cardiac transplant recipients. J Heart Lung Transplant. 2006;25:1063-1069.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Albeldawi M, Aggarwal A, Madhwal S, Cywinski J, Lopez R, Eghtesad B, Zein NN. Cumulative risk of cardiovascular events after orthotopic liver transplantation. Liver Transpl. 2012;18:370-375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 135]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
55.  Fouad TR, Abdel-Razek WM, Burak KW, Bain VG, Lee SS. Prediction of cardiac complications after liver transplantation. Transplantation. 2009;87:763-770.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 113]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
56.  Laish I, Braun M, Mor E, Sulkes J, Harif Y, Ben Ari Z. Metabolic syndrome in liver transplant recipients: prevalence, risk factors, and association with cardiovascular events. Liver Transpl. 2011;17:15-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 174]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
57.  Halloran PF, Melk A, Barth C. Rethinking chronic allograft nephropathy: the concept of accelerated senescence. J Am Soc Nephrol. 1999;10:167-181.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Bosmans JL, Holvoet P, Dauwe SE, Ysebaert DK, Chapelle T, Jürgens A, Kovacic V, Van Marck EA, De Broe ME, Verpooten GA. Oxidative modification of low-density lipoproteins and the outcome of renal allografts at 1 1/2 years. Kidney Int. 2001;59:2346-2356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
59.  Wissing KM, Abramowicz D, Broeders N, Vereerstraeten P. Hypercholesterolemia is associated with increased kidney graft loss caused by chronic rejection in male patients with previous acute rejection. Transplantation. 2000;70:464-472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 97]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
60.  Batiuk TD, Pazderka F, Enns J, De Castro L, Halloran PF. Cyclosporine inhibition of leukocyte calcineurin is much less in whole blood than in culture medium. Transplantation. 1996;61:158-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
61.  Carvalho MF, Soares V. Hyperlipidemia as a risk factor of renal allograft function impairment. Clin Transplant. 2001;15:48-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
62.  Roodnat JI, Mulder PG, Zietse R, Rischen-Vos J, van Riemsdijk IC, IJzermans JN, Weimar W. Cholesterol as an independent predictor of outcome after renal transplantation. Transplantation. 2000;69:1704-1710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 70]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
63.  Knoll GA, Blydt-Hansen TD, Campbell P, Cantarovich M, Cole E, Fairhead T, Gill JS, Gourishankar S, Hebert D, Hodsman A. Canadian Society of Transplantation and Canadian Society of Nephrology commentary on the 2009 KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Kidney Dis. 2010;56:219-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 40]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
64.  EBPG Expert Group on Renal Transplantation. European best practice guidelines for renal transplantation. Section IV: Long-term management of the transplant recipient. IV.5.3. Cardiovascular risks. Hyperlipidaemia. Nephrol Dial Transplant. 2002;17 Suppl 4:26-28.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Wanner C, Tonelli M; Kidney Disease: Improving Global Outcomes Lipid Guideline Development Work Group Members. KDIGO Clinical Practice Guideline for Lipid Management in CKD: summary of recommendation statements and clinical approach to the patient. Kidney Int. 2014;85:1303-1309.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 328]  [Cited by in F6Publishing: 350]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
66.  Moore RA, Callahan MF, Cody M, Adams PL, Litchford M, Buckner K, Galloway J. The effect of the American Heart Association step one diet on hyperlipidemia following renal transplantation. Transplantation. 1990;49:60-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 43]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
67.  Prasad GV, Wong T, Meliton G, Bhaloo S. Rhabdomyolysis due to red yeast rice (Monascus purpureus) in a renal transplant recipient. Transplantation. 2002;74:1200-1201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 86]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
68.  Arnadottir M, Berg AL. Treatment of hyperlipidemia in renal transplant recipients. Transplantation. 1997;63:339-345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
69.  van der Heide JJ, Bilo HJ, Donker JM, Wilmink JM, Tegzess AM. Effect of dietary fish oil on renal function and rejection in cyclosporine-treated recipients of renal transplants. N Engl J Med. 1993;329:769-773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 152]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
70.  Brown BG, Zhao XQ, Chait A, Fisher LD, Cheung MC, Morse JS, Dowdy AA, Marino EK, Bolson EL, Alaupovic P. Simvastatin and niacin, antioxidant vitamins, or the combination for the prevention of coronary disease. N Engl J Med. 2001;345:1583-1592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1505]  [Cited by in F6Publishing: 1575]  [Article Influence: 68.5]  [Reference Citation Analysis (0)]
71.  Blum A, Shamburek R. The pleiotropic effects of statins on endothelial function, vascular inflammation, immunomodulation and thrombogenesis. Atherosclerosis. 2009;203:325-330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
72.  Amundsen R, Åsberg A, Ohm IK, Christensen H. Cyclosporine A- and tacrolimus-mediated inhibition of CYP3A4 and CYP3A5 in vitro. Drug Metab Dispos. 2012;40:655-661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 67]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
73.  Cosio FG, Pesavento TE, Pelletier RP, Henry M, Ferguson RM, Kim S, Lemeshow S. Patient survival after renal transplantation III: the effects of statins. Am J Kidney Dis. 2002;40:638-643.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Martin JE, Cavanaugh TM, Trumbull L, Bass M, Weber F, Aranda-Michel J, Hanaway M, Rudich S. Incidence of adverse events with HMG-CoA reductase inhibitors in liver transplant patients. Clin Transplant. 2008;22:113-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
75.  Mells G, Neuberger J. Reducing the risks of cardiovascular disease in liver allograft recipients. Transplantation. 2007;83:1141-1150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 63]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
76.  Bilchick KC, Henrikson CA, Skojec D, Kasper EK, Blumenthal RS. Treatment of hyperlipidemia in cardiac transplant recipients. Am Heart J. 2004;148:200-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 47]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
77.  Kirklin JK, Benza RL, Rayburn BK, McGiffin DC. Strategies for minimizing hyperlipidemia after cardiac transplantation. Am J Cardiovasc Drugs. 2002;2:377-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
78.  Penson MG, Fricker FJ, Thompson JR, Harker K, Williams BJ, Kahler DA, Schowengerdt KO. Safety and efficacy of pravastatin therapy for the prevention of hyperlipidemia in pediatric and adolescent cardiac transplant recipients. J Heart Lung Transplant. 2001;20:611-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
79.  Kohnle M, Pietruck F, Kribben A, Philipp T, Heemann U, Witzke O. Ezetimibe for the treatment of uncontrolled hypercholesterolemia in patients with high-dose statin therapy after renal transplantation. Am J Transplant. 2006;6:205-208.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Almutairi F, Peterson TC, Molinari M, Walsh MJ, Alwayn I, Peltekian KM. Safety and effectiveness of ezetimibe in liver transplant recipients with hypercholesterolemia. Liver Transpl. 2009;15:504-508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 48]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
81.  Crespo-Leiro MG, Paniagua MJ, Marzoa R, Grille Z, Naya C, Flores X, Rodriguez JA, Mosquera V, Franco R, Castro-Beiras A. The efficacy and safety of ezetimibe for treatment of dyslipidemia after heart transplantation. Transplant Proc. 2008;40:3060-3062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
82.  Makkar KM, Sanoski CA, Goldberg LR, Spinler SA. An observational study of ezetimibe in cardiac transplant recipients receiving calcineurin inhibitors. Ann Pharmacother. 2013;47:1457-1462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
83.  Le VV, Racine N, Pelletier GB, Carrier M, Cossette M, White M. Impact of ezetimibe on cholesterol subfractions in dyslipidemic cardiac transplant recipients receiving statin therapy. Clin Transplant. 2009;23:249-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 61]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
84.  Lipscombe J, Lewis GF, Cattran D, Bargman JM. Deterioration in renal function associated with fibrate therapy. Clin Nephrol. 2001;55:39-44.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Ginsberg HN, Elam MB, Lovato LC, Crouse JR, Leiter LA, Linz P, Friedewald WT, Buse JB, Gerstein HC, Probstfield J. Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med. 2010;362:1563-1574.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1929]  [Cited by in F6Publishing: 1818]  [Article Influence: 129.9]  [Reference Citation Analysis (0)]
86.  Singh S, Watt KD. Long-term medical management of the liver transplant recipient: what the primary care physician needs to know. Mayo Clin Proc. 2012;87:779-790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
87.  Kella DK, Shoukat S, Sperling L. Plasma exchange for severe hypertriglyceridemia-induced pancreatitis in an orthotopic heart transplant recipient. J Clin Lipidol. 2012;6:474-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
88.  Boden WE, Probstfield JL, Anderson T, Chaitman BR, Desvignes-Nickens P, Koprowicz K, McBride R, Teo K, Weintraub W. Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med. 2011;365:2255-2267.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2112]  [Cited by in F6Publishing: 2042]  [Article Influence: 157.1]  [Reference Citation Analysis (0)]