mTOR inhibitors and dyslipidemia in transplant recipients: A cause for concern?

10
mTOR inhibitors and dyslipidemia in transplant recipients: A cause for concern? Hallvard Holdaas a, , Luciano Potena b , Faouzi Saliba c a Section of Nephrology, Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway b Heart Failure and Heart Transplant Program, Academic Hospital S. Orsola-Malpighi, Alma-Mater University of Bologna, Bologna, Italy c AP-HP Hôpital Paul Brousse, Centre Hépato-Biliaire, Villejuif, France abstract Post-transplant dyslipidemia is exacerbated by mammalian target of rapamycin (mTOR) inhibitors. Early clinical trials of mTOR inhibitors used xed dosing with no concomitant reduction in calcineurin inhibitor (CNI) expo- sure, leading to concerns when consistent and marked dyslipidemia was observed. With use of modern concentration-controlled mTOR inhibitor regimens within CNI-free or reduced-exposure CNI regimens, however, the dyslipidemic effect persists but is less pronounced. Typically, total cholesterol levels are at the upper end of normal, or indicate borderline risk, in kidney and liver transplant recipients, and are lower in heart transplant pa- tients under near-universal statin therapy. Of note, it is possible that mTOR inhibitors may offer a cardioprotective effect. Experimental evidence for delayed progression of atherosclerosis is consistent with evi- dence from heart transplantation that coronary artery intimal thickening and the incidence of cardiac allograft vasculopathy are reduced with everolimus versus cyclosporine therapy. Preliminary data also indicate that mTOR inhibitors may improve arterial stiffness, a predictor of cardiovascular events, and may reduce ventricular remodeling and decrease left ventricular mass through an anti-brotic effect. Post-transplant dyslipidemia under mTOR inhibitor therapy should be monitored and managed closely, but unless unresponsive to therapy should not be regarded as a barrier to its use. © 2014 Elsevier Inc. All rights reserved. 1. Introduction Cardiovascular disease is the leading cause of death following kidney transplantation [1]. The risk of fatal or non-fatal cardiovascular events in kidney transplant patients is estimated to be 50-fold higher than in the general population [2], with cardiovascular events occurring in up to 5% of kidney transplant recipients each year [24]. Following liver trans- plantation, the risk of cardiovascular events is also increased compared to controls, though to a lesser extent than after kidney transplantation [5,6]. In heart transplant patients, as might be expected, cardiovascular disease is the most common cause of death, mainly related to cardiac allograft vasculopathy (CAV) [7,8]. The nature of chronic kidney disease means that transplant patients are generally a high-risk group. In the last 20 years, for example, the proportion of patients wait-listed for kidney transplantation in the US aged 50 years or older has more than doubled [9]. In addition, these contributory factors are frequently compounded by an extended period of chronic kidney disease prior to transplantation [10]. Poor renal func- tion after transplantation, moreover, can lead to progressive anemia, an upward inuence on blood pressure and chronic inammation. Finally, all organ transplant patients are exposed to the adverse effects of main- tenance immunosuppression [11]. The inuence of immunosuppres- sion on cardiovascular risk is generally indirect, via increased risk of hypertension, diabetes, nephrotoxicity and dyslipidemia. In kidney [12], liver [5] and heart [13] transplantation, a signicant association has been shown between increased total cholesterol and cardiovascular events and represents one of the major modiable risk factors. Indeed, an elevated cholesterol level confers a greater increase in risk for ischemic heart disease following kidney transplantation com- pared to non-transplanted controls [12]. 2. Dyslipidemia after organ transplantation Dyslipidemia is a routine nding in solid organ transplantation, affecting more than half of all kidney transplant patients during the rst year post-transplant [11]. Typically, patients show an initial in- crease in total cholesterol and triglyceridemia during the rst three months after transplantation which then tends to decline to a plateau around 69 months (Fig. 1) [14,15], at least partly in response to use of lipid-lowering therapy [14,15]. In liver transplant patients, a persis- tent increase in serum lipid levels has also been documented [16], while in heart transplant recipients an increase in plasma levels of total cholesterol, low-density lipoprotein (LDL cholesterol) and triglyc- erides is typical within the rst few months post-transplant [17]. Prior to near-universal use of statin therapy in heart transplant recipients, Transplantation Reviews xxx (2014) xxxxxx Corresponding author at: Section of Nephrology, Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Postbox 4950 Nydalen, N-0424 Oslo, Norway. Tel.: +47 23 07 00 00; fax: +47 23073865. E-mail address: [email protected] (H. Holdaas). http://dx.doi.org/10.1016/j.trre.2014.08.003 0955-470X/© 2014 Elsevier Inc. All rights reserved. Contents lists available at ScienceDirect Transplantation Reviews journal homepage: www.elsevier.com/locate/trre Please cite this article as: Holdaas H, et al, mTOR inhibitors and dyslipidemia in transplant recipients: A cause for concern?, Transplant Rev (2014), http://dx.doi.org/10.1016/j.trre.2014.08.003

Transcript of mTOR inhibitors and dyslipidemia in transplant recipients: A cause for concern?

Transplantation Reviews xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Transplantation Reviews

j ourna l homepage: www.e lsev ie r .com/ locate / t r re

mTOR inhibitors and dyslipidemia in transplant recipients: A causefor concern?

Hallvard Holdaas a,⁎, Luciano Potena b, Faouzi Saliba c

a Section of Nephrology, Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norwayb Heart Failure and Heart Transplant Program, Academic Hospital S. Orsola-Malpighi, Alma-Mater University of Bologna, Bologna, Italyc AP-HP Hôpital Paul Brousse, Centre Hépato-Biliaire, Villejuif, France

a b s t r a c t

Post-transplant dyslipidemia is exacerbated bymammalian target of rapamycin (mTOR) inhibitors. Early clinicaltrials of mTOR inhibitors used fixed dosing with no concomitant reduction in calcineurin inhibitor (CNI) expo-sure, leading to concerns when consistent and marked dyslipidemia was observed. With use of modernconcentration-controlledmTOR inhibitor regimenswithin CNI-free or reduced-exposure CNI regimens, however,the dyslipidemic effect persists but is less pronounced. Typically, total cholesterol levels are at the upper end ofnormal, or indicate borderline risk, in kidney and liver transplant recipients, and are lower in heart transplant pa-tients under near-universal statin therapy. Of note, it is possible that mTOR inhibitors may offer acardioprotective effect. Experimental evidence for delayed progression of atherosclerosis is consistent with evi-dence from heart transplantation that coronary artery intimal thickening and the incidence of cardiac allograftvasculopathy are reduced with everolimus versus cyclosporine therapy. Preliminary data also indicate thatmTOR inhibitors may improve arterial stiffness, a predictor of cardiovascular events, and may reduce ventricularremodeling and decrease left ventricularmass through an anti-fibrotic effect. Post-transplant dyslipidemia undermTOR inhibitor therapy should be monitored and managed closely, but unless unresponsive to therapy shouldnot be regarded as a barrier to its use.

© 2014 Elsevier Inc. All rights reserved.

1. Introduction

Cardiovascular disease is the leading cause of death following kidneytransplantation [1]. The risk of fatal or non-fatal cardiovascular events inkidney transplant patients is estimated to be 50-fold higher than in thegeneral population [2], with cardiovascular events occurring in up to 5%of kidney transplant recipients each year [2–4]. Following liver trans-plantation, the risk of cardiovascular events is also increased comparedto controls, though to a lesser extent than after kidney transplantation[5,6]. In heart transplant patients, as might be expected, cardiovasculardisease is the most common cause of death, mainly related to cardiacallograft vasculopathy (CAV) [7,8].

The nature of chronic kidney disease means that transplant patientsare generally a high-risk group. In the last 20 years, for example, theproportion of patients wait-listed for kidney transplantation in the USaged 50 years or older has more than doubled [9]. In addition, thesecontributory factors are frequently compounded by an extended periodof chronic kidney disease prior to transplantation [10]. Poor renal func-tion after transplantation, moreover, can lead to progressive anemia, anupward influence on blood pressure and chronic inflammation. Finally,

⁎ Corresponding author at: Section of Nephrology, Department of Transplant Medicine,Oslo University Hospital, Rikshospitalet, Postbox 4950 Nydalen, N-0424 Oslo, Norway.Tel.: +47 23 07 00 00; fax: +47 23073865.

E-mail address: [email protected] (H. Holdaas).

http://dx.doi.org/10.1016/j.trre.2014.08.0030955-470X/© 2014 Elsevier Inc. All rights reserved.

Please cite this article as: HoldaasH, et al, mTOR inhibitors and dyslipidemihttp://dx.doi.org/10.1016/j.trre.2014.08.003

all organ transplant patients are exposed to the adverse effects of main-tenance immunosuppression [11]. The influence of immunosuppres-sion on cardiovascular risk is generally indirect, via increased risk ofhypertension, diabetes, nephrotoxicity and dyslipidemia.

In kidney [12], liver [5] and heart [13] transplantation, a significantassociation has been shown between increased total cholesterol andcardiovascular events and represents one of the major modifiable riskfactors. Indeed, an elevated cholesterol level confers a greater increasein risk for ischemic heart disease following kidney transplantation com-pared to non-transplanted controls [12].

2. Dyslipidemia after organ transplantation

Dyslipidemia is a routine finding in solid organ transplantation,affecting more than half of all kidney transplant patients during thefirst year post-transplant [11]. Typically, patients show an initial in-crease in total cholesterol and triglyceridemia during the first threemonths after transplantation which then tends to decline to a plateauaround 6–9 months (Fig. 1) [14,15], at least partly in response to useof lipid-lowering therapy [14,15]. In liver transplant patients, a persis-tent increase in serum lipid levels has also been documented [16],while in heart transplant recipients an increase in plasma levels oftotal cholesterol, low-density lipoprotein (LDL cholesterol) and triglyc-erides is typical within the first few months post-transplant [17]. Priorto near-universal use of statin therapy in heart transplant recipients,

a in transplant recipients: A cause for concern?, Transplant Rev (2014),

T1

Fig. 1.Mean total cholesterol level and use of lipid-lowering therapy during the first yearafter kidney transplantation inpatients randomized to standard-exposure tacrolimuswithsirolimus (target C0 8 ng/mL), tacrolimus with MMF or standard-exposure CsA withsirolimus (target C0 8 ng/mL), all with concomitant steroids [14].

2 H. Holdaas et al. / Transplantation Reviews xxx (2014) xxx–xxx

a clear correlationwas demonstrated between higher lipid levels andincreased risk of CAV [18].

Several factors influence the risk of dyslipidemia, including acuterejection, graft dysfunction, proteinuria and the immunosuppressive reg-imen [19]. Post-transplant dyslipidemia can be caused or exacerbated bycorticosteroids or calcineurin inhibitors (CNIs), but the most notablechanges in lipid status have been observed following the introduction ofmammalian target of rapamycin (mTOR) inhibitors [20]. When consider-ing the lipogenic effect of mTOR inhibitors, two distinct issues need to beassessed: first, any adverse effect on lipid status, and second, a potentialcardioprotective effect which may offset increased lipid levels.

3. Etiology of mTOR inhibitor lipogenic effect

The lipogenic effect of mTOR inhibitors, as for CNI therapy, arisesfrom several mechanisms [21] which are not fully elucidated. ThemTOR signaling pathway plays a role in regulating the uptake of lipidsinto adipose tissue and their breakdown by lipoprotein lipase [22]. Dis-ruption of the mTOR pathway appears to inhibit uptake of lipids intoadipocytes, resulting in 20–30% reduction in lipid storage [21]. mTORinhibitors also promote lipolysis, increasing basal lipolysis by approxi-mately 20%, and enhance the expression of lipogenic genes in adiposetissue [21,23]. The antilipolytic effect of insulin is impaired in thepresence of sirolimus, and mTOR inhibitors may delay the peripheralclearance of fatty acids [24] resulting in reduced incorporation of fattyacids into very low density lipoprotein (VLDL) [25]. Levels of free fattyacids have been shown to rise in mice after administration of sirolimusdue to augmented lipolysis and suppressed adipocyte storage [25].

4. Dyslipidemic effect of mTOR inhibitors: early clinical evidence

The early clinical trials of mTOR inhibitors used fixed dosing, oftenwith a substantial loading dose, and did not reduce concomitant expo-sure to CNIs. Results from these studies raised concerns about thelipogenic effect of such regimens. A pooled analysis [26] assessed datafrom two large controlled studies of 1295 de novo kidney transplantpatients receiving sirolimus 2 mg/day or 5 mg/day with standard-exposure cyclosporine (CsA) and steroids versus azathioprine orplacebo [27,28]. Compared to controls, mean cholesterol and triglyceridelevels were 17 mg/dL and 59 mg/dL higher in the sirolimus 2 g/daycohort, and 30 mg/dL and 103 mg/dL higher, respectively, in thesirolimus 5 mg/day cohort at one year post-transplant. Levels peakedaround months 2–3 and responded to lipid-lowering therapy [26].

Please cite this article as: Holdaas H, et al,mTOR inhibitors and dyslipidemihttp://dx.doi.org/10.1016/j.trre.2014.08.003

Similar observations were made in a pooled analysis of two randomizedtrials in which sirolimus dose was concentration-controlled but withhigh trough concentrations (30 ng/mL tomonth 2 and 15 ng/mL thereaf-ter), although again the effectwasmanageablewith lipid-lowering agents[29]. Ameta-analysis of trials performedup to 2005 confirmed thatmTORinhibitorswere associatedwith an increased risk of hypercholesterolemiacompared to antimetabolite therapy [30]. A wider systematic review ofstudies published up to 2008, which compared sirolimus or everolimusversus various non-mTOR inhibitor regimens, also concluded that therewas an increased prevalence of lipid-lowering treatment and higherlevels of cholesterol and triglycerides among kidney transplant patientsreceiving an mTOR inhibitor [20].

However, as evidence accumulated it became apparent that theeffect of mTOR inhibitors on lipid status is dose-dependent. For bothsirolimus [27,31] and everolimus [32], a lower fixed dosewas associatedwith smaller changes in lipid profile thanhigher doses, even in the pres-ence of standard-exposure CNI therapy. Modern mTOR regimens arenow routinely concentration-controlled, with no loading dose foreverolimus and smaller loading doses (if any) for sirolimus, and targettrough concentrations have declined over the last decade. A maximumtrough concentration target of 8 ng/mL for everolimus, and usually10 ng/mL for sirolimus, is generally considered suitable in kidney,liver and heart transplantation [33,34], most frequently administeredin combination with reduced-dose CNI when used as de novo immuno-suppression, or in a CNI-free regimen when introduced in maintenancepatients. This raises the question of whether the dyslipidemic effect ofmTOR inhibitors is still a cause for concern when using modern, lessintensive regimens with widespread administration of statin therapy.

5. Clinical experience using modern mTOR inhibitor regimens

No trials have been undertaken specifically to compare the lipogeniceffect of mTOR inhibitors versus other immunosuppressants. Interpre-tation of data from comparative clinical trials is complicated by thefact that the use and dose of lipid-lowering drugs are generally notspecified in the study protocol, and changes in their administrationover time are not usually recorded. Also, trials of mTOR inhibitors typi-cally exclude patients with very high lipid levels, and discontinuationdue to lipid abnormalities can potentially skew results in favor of themTOR inhibitor treatment group.With these caveats in mind, however,examination of lipid parameters in studies which have used contempo-rary, concentration-controlled regimens can be informative. TheEVERHEART study, which is currently ongoing, includes a standardizedhypolipidemic protocol to specifically study hypercholesterolemia withor without everolimus in the first fewweeks after heart transplantation(NCT01017029).

5.1. Kidney transplantation

Randomized trials of de novo kidney transplant patients comparingeverolimus or sirolimus versus mycophenolate mofetil (MMF) usingcontemporary mTOR exposure levels, and which reported lipid levels,are summarized in Table 1 [14,35,36]. Where the mTOR inhibitor wasadministered with standard-exposure CNI, use of lipid-loweringtherapy was higher in the mTOR inhibitor arm, with mixed resultsconcerning an effect on total cholesterol, LDL-cholesterol and triglycer-ides [14,36]. Between-group differences were less marked but persistedin a recent large randomized trial in which everolimus with low-doseCsA was compared to MMF with standard-exposure CsA, an approachwhich is now considered more appropriate. More studies have com-pared de novo mTOR inhibitor therapy in a CNI-free regimen versus aCNI-based regimen in [15,37–41]. Here, lipid-modifying agents wereagain used more frequently in patients receiving an mTOR inhibitor,and total cholesterol was typically higher versus CNI-treated patients,although differences for LDL-cholesterol and triglycerides were lessconsistent (Table 1). Of note, in these studies the increase in LDL was

a in transplant recipients: A cause for concern?, Transplant Rev (2014),

T2

T3

F2

3H. Holdaas et al. / Transplantation Reviews xxx (2014) xxx–xxx

accompanied by a concomitant increase inHDL, leading to a stable or re-duced ratio of LDL to HDL such that the estimated lipid-related risk ofcardiovascular events was unchanged or lowered [42].

Several randomized trials in kidney transplant patients have com-pared CNI continuation versus switch to everolimus [43–45] orsirolimus [46–48] more than three months post-transplant, by whichtime lipid changes have typically peaked. In most cases, patients werefollowed for a minimum of one year post-conversion. In all but onereport from an interim analysis [44], mean total cholesterol was signif-icantly higher in the mTOR inhibitor group at last follow-up, with LDL-cholesterol and triglyceride levels also higher than in controls wheredata were provided. Holdaas et al included a third treatment group intheir randomized trial of 394 patients more than six months after kid-ney transplantation, in which patients started everolimus therapywith reduced CNI exposure [43]. As with CNI elimination, total choles-terol and triglycerides were higher after the introduction of everolimus.

For studies in which kidney transplant patients were converted tomTOR inhibitor therapy, mean total cholesterol level at last follow-upwas within the borderline high range (200–239 mg/dL) [43,45–48],LDL-cholesterol was either normal or borderline high (130–159 mg/dL)and triglycerides were borderline high (150–199 mg/dL) or high(≥200 mg/dL). Most trials did not report changes in lipid levels overtime. In the CONVERT trial, the increases in total cholesterol, LDL-cholesterol and triglycerides peaked at two months after conversionto sirolimus then declined [46]. However, more sirolimus-treated pa-tients were treated with lipid-lowering therapy (77.7% versus 54.6% inthe standard CNI group at month 24, p b 0.001).

5.2. Liver transplantation

In liver transplantation, three randomized trials have evaluated earlyconversion from CNI therapy to either sirolimus [49] or everolimus[50,51] (i.e. starting by one month post-transplant). Use of lipid-lowering therapy was far lower than in the studies of kidney transplantpatients (typically b20%). Each of the studies showed either total orLDL-cholesterol to be higher in the mTOR inhibitor group (Table 2).The largest trial in this setting, in which everolimus with reduced tacro-limus was administered from month 1 after liver transplantation,showed the combination regimen to be associated with significantlyhigher levels of triglycerides, total cholesterol and LDL-cholesterol atone year post-transplant compared to a standard CNI-based regimen,but also with higher HDL-cholesterol (Table 2) [52]. The use of lipid-lowering therapy in the everolimus/tacrolimus group was similar tothat of the tacrolimus-only cohort (23.3% versus 17.8%, p = 0.944).

Few trials have assessed conversion to anmTOR inhibitor regimen inmaintenance liver transplant patients and have only rarely reportedlipid levels. However the available data have consistently indicated ahigher rate of hypercholesterolemia as an adverse event following con-version from CNI therapy [53–56].Watson et al described a significantlygreater increase in total cholesterol at month 3 after conversion fromCNI to sirolimus therapy versus controls among a small group of 30maintenance liver transplant patients [57]. The difference becamenon-significant at month 12 but more sirolimus-treated patientsreceived statin therapy than in the CNI continuation group [57].

5.3. Heart transplantation

The situation in heart transplantation is somewhat different, sincealmost all patients receive statins or other lipid-lowering drugs. Theonly randomized trial to compare a concentration-controlled mTORinhibitor (everolimus) with early CNI discontinuation versus mainte-nance CsA therapy found no pronounced differences in lipid status atone year [58], while two comparative studies of everolimus versusMMF each showed significantly higher total cholesterol and triglyceridelevels [59,60] in the mTOR inhibitor group (Table 3). Use of lipid-

Please cite this article as: HoldaasH, et al, mTOR inhibitors and dyslipidemihttp://dx.doi.org/10.1016/j.trre.2014.08.003

lowering therapy was slightly higher (5–8% more patients) in thepatients receiving mTOR inhibitor in the latter two studies.

In a randomized trial of heart (n = 190) or lung (n = 92) mainte-nance transplant patients, Gullestad et al observed a significantly great-er increase in total cholesterol, LDL-cholesterol and triglycerides afterconversion to everolimus with reduced CNI exposure versus patientswho remained on standard CNI therapy [61], differences that continuedto two years after randomization [62]. From randomization to two yearspost-transplant, themean change in total cholesterolwas 0.5 mmol/L inthe everolimus group versus 0.1 mmol/L in the control arm (p b 0.001).The changes in LDL-cholesterol (0.3 versus 0.0 mmol/L, p= 0.010) andtriglycerides (0.3 versus 0.0 mmol/L, p= 0.002) were also significantlydifferent [62]. Conversely, Potena et al reported that as a consequence ofstatin dosing, heart recipients randomized to everolimus and very lowCNI showed no difference in lipid levels at three years after randomiza-tion compared to patients receiving MMF and low CNI [63].

5.4. General

Across all three organ types, the mean level of total cholesterol atone year post-transplant was in the range 173–225 mg/dL in patientsreceiving an mTOR inhibitor with various concomitant medications.These values straddle the upper range of ‘normal’ and ‘borderline risk’.For LDL-cholesterol, the mean levels were in the range 104–127 mg/dL,just below ‘borderline risk’. Mean triglyceride levels were in the range89–228 mg/dL, again varying between the upper range of normal andlower values of borderline risk. Presumably due to the high rate oflipid-lowering medication, values were consistently lowest in theheart transplant populations.

6. Possible cardioprotective effects of mTOR inhibitors

Coronary stents coatedwith everolimus or sirolimus arewidely usedin revascularization procedures to reduce restenosis and narrowing ofthe lumen [64,65]. The use of such stents was stimulated by data fromanimal models showing that mTOR inhibitors inhibit transplant athero-sclerosis [66,67]. At the experimental level, mTOR inhibitors have beenfound to delay the progression of atherosclerosis by inhibiting the accu-mulation of lipids, resulting in smaller atherosclerotic plaques[25,67–69]. In mice with cholesterol-induced atherosclerosis, plaquesizes showed a dose-dependent reduction in the presence of everolimus[70]. Moreover, animal models also suggest that mTOR inhibition stabi-lizes plaques, reducing the risk of rupture by promoting macrophageclearance and limiting the inflammatory response within smooth mus-cle cells in the artery walls [71,72].

Clinically, the antiatherogenic effect within graft arteries of everoli-mus has been demonstrated in the recent A2310 study of 721 de novoheart transplant patient randomized to everolimus with low-dose CsAor toMMFwith standard-exposure CsA [59]. In a substudy, 189 patientswere evaluated by intravascular ultrasound at baseline and atmonth 12post-transplant [59,73]. By month 12, themean increase in maximal in-timal thickness of coronary arteries was significantly smaller in theeverolimus group versusMMF (Fig. 2). The incidence of cardiac allograftvasculopathy (defined as an increase of 0.5 mm ormore in themaximalintimal thickness) was also significantly lower with everolimus at oneyear post-transplant (Fig. 2). These results, which confirmed earlydata from a large randomized trial of everolimus versus azathioprine[74], showed that the benefit observed with everolimus was indepen-dent of patients' lipid levels [73]. A direct influence on arterial thicken-ing would be consistent with evidence from kidney transplantationsuggesting that mTOR inhibition in a CNI-free regimen is associatedwith inhibition of fibrosis progression in hepatitis C-positive patientsreceiving a kidney transplant [75,76]. While intravascular ultrasound dataare of course only an indirect assessment of risk, a study in heart trans-plant patients has shown that thosewith an increase inmaximal intimalthickness increases of 0.5 mm or more in the first year after transplant

a in transplant recipients: A cause for concern?, Transplant Rev (2014),

Table 1Randomized studies of concentration-controll mTOR inhibitor therapy versus MPA or CNI therapy in de novo kidney transplant recipients.

Study Follow-up(months)

reatmentroup

n Immunosuppressiona Lipid-modifyingtherapy

P value Total-C P value LDL-C P value MeanHDL-C

P value Triglycerides P value

mTOR vs MPAMendez et al 2005 [35] 6 RL 185 SRL 4–12 ng/mL

TACSteroids

42% 0.003 29.3%N240 mg/dL

0.0003 - - - - 55.6%N200 mg/dL

0.004

MF 176 MMFTACSteroids

22% 12.2%N240 mg/dL

- - 38.0%N200 mg/dL

Ciancio et al 2004 [14] 12 RL/TAC 50 SRL 8 ng/mLTACSteroids

54% P b 0.001 vsTAC/MMF

202 mg/dL P = 0.61 - - - 163 mg/dL 0.19

MF/TAC 50 TACMMFSteroids

16% 193 mg/dL 151 mg/dL

RL/CsA 50 SRL 8 ng/mLCsASteroids

80% P b 0.001 vsTAC/MMF

201 mg/dL 187 mg/dL

Tedesco-Silva et al 2010 [36] 12 VR 274 EVR 3–8 g/mLLow-dose CsASteroids

64.6% - 15.7%N350 mg/dL

- - - 7.7%N270 mg/dL

- 4.4%N750 mg/dL

-

VR 278 EVR 6–12 g/mLLow-dose CsASteroids

72.3% 16.5%N350 mg/dL

- 5.0%N270 mg/dL

6.1%N750 mg/dL

MF 273 MMFCsASteroids

57.5% 6.3%N350 mg/dL

- 6.5%N270 mg/dL

2.6%N750 mg/dL

mTOR vs CNIFlechner et al 2013 [37] 12 RL 152 SRLb

MMFSteroids

61.2% - - - - - - -

RL/TAC 152 SRLb

TAC to week 13Steroids

58.6% - - - - - -

MF/TAC 139 MMFTACSteroids

50.4% - P b 0.05 vsSRL groups

- P b 0.05 vsSRL groups

- - - P b 0.05 vsSRL groups

4H.H

oldaasetal./Transplantation

Reviewsxxx

(2014)xxx–xxx

Pleasecite

thisarticle

as:Holdaas

H,etal,m

TORinhibitors

anddyslipidem

iain

transplantrecipients:Acause

forconcern?,TransplantRev(2014),

http://dx.doi.org/10.1016/j.trre.2014.08.003

ed

Tg

S

M

S

M

S

E

E

M

S

S

M

Table 1 (continued)

Study Follow-up(months)

Treatmentgroup

n Immunosuppressiona Lipid-modifyingtherapy

P value Total-C P value LDL-C P value MeanHDL-C

P value Triglycerides P value

Ekberg et al 2010 [38]Claes et al 2012 [15]

12 SRL 380 SRL 4-8 ng/mLMMFSteroids

39% 0.007 acrossgroups

- - 119 mg/dL P b 0.05 55 mg/dL n.s. 206 mg/dL b0.05 vs TACand MMF/lowCsA

MMF/CsA 384 Standard CsAMMFSteroids

34% - 112 mg/dL n.s. 55 mg/dL 185 mg/dL

MMF/CsA 408 Low-dose CsAMMFSteroids

32% - 117 mg/dL P b 0.05 55 mg/dL 174 mg/dL

MMF/TAC 403 Low-dose TACMMFSteroids

26% - 107 mg/dL Reference 53 mg/dL 165 mg/dL

Buchler et al 2007 [39] 12 SRL 71 SRL 10–15 ng/mLSteroids to month 5MMF

70.5% 0.03 5.7 mmol/L 0.03 3.3 mmol/L 0.12 1.4 mmol/L - 2.3 mmol/L 0.18

CsA 74 CsASteroids to month 5MMF

51.0% 5.1 mmol/L 3.0 mmol/L 1.4 mmol//L 1.7 mmol/L

Larson et al 2006 [40] 12 SRL 81 SRL 8–12 ng/mLMMFSteroids

- - 219 mg/dL 0.02 - - - -

TAC 84 TACMMFSteroids

- 200 mg/dL - - -

Flechner et al 2002 [41] 12 SRL 31 SRL 5–12 ng/mLMMFSteroids

64.5% - 225 mg/dL 0.72 125 mg/dL 0.61 52 mg/dL 0.79 228 mg/dL 0.38

CsA 30 CsAMMFSteroids

53.3% 220 mg/dL 143 mg/dL 52 mg/dL 205 mg/dL

C, cholesterol; CNI, calcineurin inhibitor; CsA, cyclosporine; EVR, everolimus;HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol;MMF,mycophenolatemofetil;MPA,mycophenolic acid;mTOR,mammalian targetof rapamycin; SRL, sirolimus; TAC, tacrolimus.Lipid values are shown as means unless stated otherwise.

a Maintenance immunosuppression.b Original protocol: 10–15 mg loading dose, 10–15 ng/mL toweek 13, 8–12 ng/mL duringweeks 14–26, 5–12 ng/mL duringweeks 27–104. Revised protocol: 15 mg loading dose × 2, 10–15 ng/mL toweek 26, 8–15 ng/mL duringweeks 27–104.

5H.H

oldaasetal./Transplantation

Reviewsxxx

(2014)xxx–xxx

Pleasecite

thisarticle

as:HoldaasH

,etal,mTO

Rinhibitors

anddyslipidem

iain

transplantrecipients:Acause

forconcern?,TransplantRev(2014),

http://dx.doi.org/10.1016/j.trre.2014.08.003

Table 2Randomized studies of early conve ion to concentration-controlled mTOR inhibitor therapy versus CNI therapy in liver transplant recipients.

Study Fo w-up(m ths)

Treatmentgroup

n Immunosuppressiona Lipid-modifyingtherapy

P value Total-C P value LDL-C P value Mean HDL-C P value Triglycerides P value

Teperman et al 2013 [49] 12 SRL 148 SRL 5–10 ng/mL from week 4–12MMF±Steroids

13.5% - - - 120 mg/dL 0.001 47 mg/dL 0.55 210 mg/dL 0.07

CsA/TAC 145 CsA/TACMMF±Steroids

4.8% - 101 mg/dL 46 mg/dL 169 mg/dL

De Simone et al 2012 [52] 12 EVRLow-doseTAC

245 EVR 3–8 ng/mL from week 4Low-dose TACSteroids

23.3% 0.944 209 mg/dL b0.001 121 mg/dL b0.001 51 mg/dL b0.001 197 mg/dL b0.001

TAC 243 TACSteroids

17.8% 175 mg/dL 101 mg/dL 47 mg/dL 141 mg/dL

Fischer et al 2012 [50] 12 EVR 101 EVR 5–12 ng/mLCsA to week 8±Steroids

- - 22.8%b ≤0.05 - - 5.9%c n.s.

CsA 102 CsA±Steroids

- 10.8%b - - 2.9%c

Masetti et al 2010 [51] 12 EVR 52 EVR 6–10 ng/mL from day 10CsA to day 30Steroids

9.6% - ~200 mg/dL 0.01 ~120 mg/dL 0.11 ~50 mg/dL 0.03 ~170 mg/dL 0.68

CsA 26 CsASteroids

7.7% ~160 mg/dL ~80 mg/dL ~32 mg/dL ~200 mg/dL

C, cholesterol; CsA, cyclosporine; E , everolimus; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; MMF, mycophenolate mofetil; mTOR, mammalian target of rapamycin; n.s., not significant; SRL, sirolimus;TAC, tacrolimus.Lipid values are shown as means u ess stated otherwise.

a Maintenance immunosuppres n.b Incidence of hypercholesterole ia reported as an adverse event by the investigator.c Incidence of hypertriglyceride a reported as an adverse event by the investigator.

6H.H

oldaasetal./Transplantation

Reviewsxxx

(2014)xxx–xxx

Pleasecite

thisarticle

as:Holdaas

H,etal,m

TORinhibitors

anddyslipidem

iain

transplantrecipients:Acause

forconcern?,TransplantRev(2014),

http://dx.doi.org/10.1016/j.trre.2014.08.003

rs

lloon

VR

nlsiommi

Table 3Randomized studies of concentration-controlled mTOR inhibitor therapy versus MPA or CNI therapy in de novo heart transplant recipients.

Study Follow-up(months)

Treatment group n Immunosuppression Lipid-modifyingtherapy

P value Total-C P value LDL-C P value Mean HDL-C P value Triglycerides P value

mTOR vs MPAEisen et al 2013a [59] 24 EVR 282 EVR 3–8 ng/mL

Low-dose CsA from month 2Steroids

94.6% - 5.0 mmol/L b0.001 2.7 mmol/L 0.054 1.4 mmol/L b0.001 2.3 mmol/L 0.065

MMF 271 MMFStandard CsASteroids

94.0% 4.6 mmol/L 2.6 mmol/L 1.3 mmol/L 2.0 mmol/L

Lehmkuhl et al 2009 [60] 12 EVR 92 EVR 3-8 ng/mLLow-dose CsASteroids

93.4% 5.5 mmol/L 0.002 - - - - 2.3 mmol/L 0.001

MMF 84 MMFStandard CsASteroids

85.5% 5.0 mmol/L - - 1.7 mmol/L

mTOR vs CNIAndreassen et al 2013 [58] 12 EVR 56 EVR

Low-dose CsA to week 7MMFSteroids

91% - 5.3 mmol/L n.s. 2.9 mmol/L n.s. 1.6 mmol/L n.s. - -

CsA 59 CsAMMFSteroids

86% 5.1 mmol/L 2.8 mmol/L 1.6 mmol/L -

C, cholesterol; CsA, cyclosporine; EVR, everolimus; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; MMF, mycophenolate mofetil; MPA, mycophenolic acid; mTOR, mammalian target of rapamycin; n.s., notsignificant; TAC, tacrolimus.Lipid values are shown as means unless stated otherwise.

a Randomization to a third treatment group (everolimus 6–12 ng/mL) was terminated early due to higher mortality.

7H.H

oldaasetal./Transplantation

Reviewsxxx

(2014)xxx–xxx

Pleasecite

thisarticle

as:HoldaasH

,etal,mTO

Rinhibitors

anddyslipidem

iain

transplantrecipients:Acause

forconcern?,TransplantRev(2014),

http://dx.doi.org/10.1016/j.trre.2014.08.003

Fig. 2. (A) Mean (SD) change in maximum intimal thickness of coronary arteries frombaseline to month 12. (B) Proportion of patients with cardiac allograft vasculopathy atmonth 12, defined as ≥0.5 mm increase in maximum intimal thickness from baseline tomonth 12. Data from de novo heart transplant patients randomized to everolimus withreduced-exposure CsA or MMF with standard-exposure CsA, both with steroids, from asubstudy of the A2310 study inwhich intravascular ultrasoundwas performed at baselineand month 12 post-transplant [59].

8 H. Holdaas et al. / Transplantation Reviews xxx (2014) xxx–xxx

experienced a higher rate of death or graft loss, with more majoradverse cardiac events by five years post-transplant [77]. It seems feasi-ble that reduced intimal thickening under mTOR inhibitor therapy mayhelp to counteract the lipogenic effect.

A cardioprotective effect of mTOR inhibitors is also suggested bydata concerning pulsewave velocity, awell-establishedmarker for arte-rial stiffness. Arterial stiffness increases with age, but is exacerbated byatherosclerosis and is known to increase the risk of cardiovascularevents following kidney transplantation [78,79]. A prospective trial of27 maintenance kidney transplant patients found that after conversionfrom CsA to everolimus, pulse wave velocity stabilized over thefollowing ninemonths, but increased in the CsA group, reflecting ongo-ing atherosclerotic processes [80]. Similarly, a subpopulation analysisfrom the randomized CONCEPT trial reported that a CNI-freesirolimus-based regimen reduced aortic stiffness as measured by pulsewave velocity [81].

Lastly, preliminary data from analyses of kidney transplant patientsat one center [82] and heart transplant patients at another center[83,84] have indicated that conversion from CNI therapy to everolimusor sirolimus is associated with reduced ventricular remodeling anddecreased left ventricular mass, with improved diastolic function [84]. Thismay arise from a direct effect on the myocardium [84]. Progressivefibrosis, a key characteristic of endomyocardial remodeling after hearttransplantation, was shown to be significantly attenuated by everoli-mus therapy versus MMF therapy in a prospective study of 132 denovo heart transplant patients [85]. There was a significant differencein the extent of fibrosis on cardiac biopsy as early as one month post-

Please cite this article as: Holdaas H, et al,mTOR inhibitors and dyslipidemihttp://dx.doi.org/10.1016/j.trre.2014.08.003

transplant, with less scarring in the everolimus-treated patients up tothe end of the three-year study.

Overall, the evidence for cardioprotective properties of mTORinhibitors following transplantation is growing and merits fur-ther investigation.

7. Cardiac outcomes

The scale and duration of controlled trials make it difficult to com-pare the rate of cardiac events reliably between different immunosup-pressive regimens. To date, no trial of mTOR inhibitor therapy has hada cardiovascular primary endpoint. During three-year follow-up ofpatients in the SYMPHONY study, the largest trial of immunosuppressionin kidney transplantation to date, data on fatal and non-fatal cardiovas-cular events were recorded [86]. At three years, the incidence of any car-diovascular event was low in all groups with no significant differencebetween the low-dose sirolimus arm and the CNI-based treatmentgroups. Less detailed three-year [87] and five-year [88] follow-up datafrom studies of sirolimus with or without low-dose CNI versus standardCNI regimens have also not indicated any difference in cardiovasculardeaths within kidney transplant populations. In heart transplantation, apivotal trial randomized 634 patients to de novo treatment with everoli-mus at a fixed dose of 1.5 mg/day or 3.0 mg/day (switched toconcentration-controlled therapy after one year) or to azathioprine, allwith standard CsA and steroids [74]. At four years post-transplant, non-fatal major adverse cardiac events (excluding events during month 1)had occurred in 7.9% of everolimus-treated patients versus 13.6% ofazathioprine-treated patients (p = 0.0331) [89], but this difference hasnot been confirmed by other studies. More recently, two-year mortalitywas similar in a study in de novo heart transplant patients randomizedto everolimus (target C0 3–8 ng/mL) with reduced-CsA or MMF withstandard CsA [59]. Recruitment to a third group (everolimus targetingC0 6–12 ng/mL) was terminated early due to a higher rate of infectiousdeaths, but there were no apparent differences in the rate ofcardiovascular-related deaths between any of the three arms.

8. Conclusions

Dyslipidemia is a recognized class effect of mTOR inhibitors whichappears to be more pronounced than that seen with mycophenolicacid (MPA) or CNIs following solid organ transplantation. Interpretationof available studies is complicated by relatively small numbers and fre-quent differences in the use of lipid-lowering therapy between studytreatment groups. Also, longer-term data to two or even five yearspost-transplant would be desirable but have not been widely reported.Nevertheless, the absolute differences in lipid profile between mTORinhibitor-based and other regimens are less marked than during earlystudies and a dyslipidemic effect may be at least partly offset by theantiatherogenic action of mTOR inhibitor therapy. More generally, itshould be borne in mind that use of an mTOR inhibitor permits reduc-tion or elimination of CNI therapy, which could potentially offer advan-tages in terms of reducing CNI-related hypertension and nephrotoxicity[90]. A strong association has been demonstrated between renal func-tion and cardiovascular death in kidney and heart transplant recipients,independent of conventional risk factors for cardiovascular disease[10,91]. Improved renal function has been convincingly demonstratedin a series of trials using mTOR inhibitors with CNI therapy reductionor withdrawal either de novo or in maintenance organ transplantpatients versus standard CNI-based regimens [92–95] although protein-uria, which has an associated lipogenic effect, is a potential concernin maintenance patients with poor renal function [63,96].

Lipid status in kidney transplant patients should be managed as fornon-transplant patientswith chronic kidney disease [11]. Statin therapyis effective in lowering cholesterol levels and appears to reduce the riskof cardiovascular events following kidney transplantation [12,97–99].There does not appear to be a clinically relevant pharmacokinetic

a in transplant recipients: A cause for concern?, Transplant Rev (2014),

9H. Holdaas et al. / Transplantation Reviews xxx (2014) xxx–xxx

interaction between everolimus and statin agents during coadministra-tion [100]. No formal guidelines exist for management of hypercho-lesterolemia in liver transplant patients, but it would seem reasonable tomonitor andmanage as for other high-risk individuals [101]. Use of statintherapy is almost universal following heart transplantation and maylargely negate the effect of elevated total cholesterol on cardiovascularrisk [13].

In conclusion, dyslipidemia in organ transplant patients receivingmTOR inhibitor therapy should be monitored and managed closely,but unless unresponsive to therapy should not be regarded as a barrierto use of this class of immunosuppressant. It would seem reasonable toavoid mTOR inhibitors in patients with significant dyslipidemia beforeor after transplantation [11]. Cardiovascular risk is not higher in thepresence of mTOR inhibitor therapy and increased lipid levels may beat least partly balanced by a direct cardioprotective effect and, potential-ly, reduction in other CNI-related risk factors.

Acknowledgments

A draft of this manuscript was prepared by a freelance writer withfunding from Novartis Pharma AG. The authors were fully responsiblefor all content and editorial decisions and received no financial supportor other form of compensation related to the development of this man-uscript. The opinions expressed in the manuscript are those of theauthors andNovartis Pharmaceuticals had no influence on the contents.

H Holdaas has served as a consultant to Bristol-Myers Squibb,Novartis, AstraZeneca, Astellas and Schering-Plough, and has receivedlecture fees from Novartis and AstraZeneca. L Potena has received lec-ture fees from Novartis and has served as a consultant for Diaxhonitand Biotest. F Saliba has received speaker honorarium and/or grantresearch from Novartis, Astellas, Gilead, Roche, Merck Sharip &Dohme, Pfizer, Gambro and vital therapies.

References

[1] Lentine KL, Costa SP, Weir MP, et al. Cardiac disease evaluation and managementamong kidney and liver transplantation candidates: a scientific statement fromthe American Heart Association and the American College of Cardiology Founda-tion. Circulation 2012;126:617–63.

[2] Ojo AO. Cardiovascular complications after renal transplantation and their preven-tion. Transplantation 2006;82:603–11.

[3] Lentine KL, Brennan DC, Schnitzler MA. Incidence and predictors of myocardial in-farction after kidney transplantation. J Am Soc Nephrol 2005;16:496–506.

[4] Kasiske BL, Maclean JR, Snyder JJ. Acute myocardial infarction and kidney trans-plantation. J Am Soc Nephrol 2006;17:900–7.

[5] Guckelberger O, Mutzke F, Glanemann, et al. Validation of cardiovascular riskscores in a liver transplant population. Liver Transpl 2006;12:394–401.

[6] Madhwal S, Atreja A, Albeldawi M, Lopez R, Post A, Costa MA. Is liver transplanta-tion a risk factor for cardiovascular disease? A meta-analysis of observational stud-ies. Liver Transpl 2012;18:1140–6.

[7] Colvin-Adams M, Smith JM, Heubner BM, et al. OPTN/SRTR 2011 Annual Data Re-port: heart. Am J Transplant 2013;13:119–48.

[8] Stehlik J, Hosenpud JD, Edwards LB, Hertz MI, Mehra MR. International Society forHeart and Lung Transplantation. ISHLT International Registry for Heart and LungTransplantation—into the fourth decade, from strength to strength. J Heart LungTransplant 2013;32:941–50.

[9] Organ Procurement and Transplant Network Database. Available at http://optn.transplant.hrsa.gov/. [Accessed 2 October 2013].

[10] Fellstrom B, Jardine AG, Soveri I, et al. Renal dysfunction as a risk factor for mortal-ity and cardiovascular disease in renal transplantation: experience from the Assess-ment of Lescol in Renal Transplantation trial. Transplantation 2005;79:1160–3.

[11] Kidney Disease Improving Global Outcomes (KDIGO) clinical practice guideline forlipid management in chronic kidney disease. Kidney Int Suppl 2013;3:263–5.

[12] Holdaas H, Fellström B, Jardine AG, et al. Effect of fluvastatin on cardiac outcomes inrenal transplant recipients: a multicentre, randomised, placebo-controlled trial.Lancet 2003;361:2024–31.

[13] Arora S, Aukrust P, Andreassen A, et al. The prognostic importance of modifiablerisk factors after heart transplantation. Am Heart J 2009;158:431–6.

[14] Ciancio G, Burke GW, Gaynor JJ, et al. A randomized, long-term trial of tacrolimus/sirolimus versus tacrolimus/mycophenolate mofetil versus cyclosporine (Neoral)/sirolimus in renal transplantation. II. Survival, function and protocol complianceat one year. Transplantation 2004;77:252–8.

[15] Claes K, Meier-Kriesche H-U, Schold JD, Vanrenterghem Y, Halloran PF, Ekberg H.Effect of different immunosuppressive regimens on the evolution of distinct

Please cite this article as: HoldaasH, et al, mTOR inhibitors and dyslipidemihttp://dx.doi.org/10.1016/j.trre.2014.08.003

metabolic parameters: evidence from the Symphony study. Nephrol Dial Trans-plant 2012;27:850–7.

[16] Muñoz SJ, Elgenaidi H. Cardiovascular risk factors after liver transplantation. LiverTranspl 2005;11:S52–6.

[17] Kirklin JK, Benza RL, Rayburn BK, McGiffin DC. Strategies for minimizing hyperlip-idemia after cardiac transplantation. Am J Cardiovasc Drugs 2002;2:377–87.

[18] Escobar A, Ventura HO, Stapleton DD, et al. Cardiac allograft vasculopathy assessedby intravascular ultrasonography and nonimmunologic risk factors. Am J Cardiol1994;74:1042–6.

[19] Gaston RS, Kasiske BL, Fieberg AM, et al. Use of cardioprotectivemedications in kid-ney transplant recipients. Am J Transplant 2009;9:1811–5.

[20] Kasiske BL, de Mattos A, Flechner SM, et al. Mammalian target of rapamycin inhib-itor dyslipidemia in kidney transplant recipients. Am J Transplant 2008;8:1384–92.

[21] PereiraMJ, Palming J, Rizell M, et al. The immunosuppressive agents rapamycin, cy-closporine A and tacrolimus increase lipolysis, inhibit lipid storage and alter ex-pression of genes involved in lipid metabolism in human adipose tissue. Mol CellEndocrinol 2013;365:260–9.

[22] Blanchard PG, Festuccia WT, Houde YP, et al. Major involvement of mTOR in thePPAR -induced stimulation of adipose tissue lipid uptake and fat accretion. J LipidRes 2012;53:1117–25.

[23] Ma KL, Ruan XZ, Powis SH, Chen Y, Moorhead JF, Varghese Z. Sirolimus modifiescholesterol homeostasis in hepatic cells: a potential molecular mechanism forsirolimus-associated dyslipidemia. Transplantation 2007;84:1029–36.

[24] Brown NF, Stefanovic-Racic M, Sipula IJ, Perdomo G. The mammalian target ofrapamycin regulates lipid metabolism in primary cultures of rat hepatocytes. Me-tabolism 2007;56:1500–7.

[25] Morrisett JD, Abdel-Fattah G, Hoogeveen R, et al. Effects of sirolimus on plasmalipids, lipoprotein levels, and fatty acid metabolism in renal transplant patients. JLipid Res 2002;43:1170–80.

[26] Blum CB. Effects of sirolimus on lipids in renal allograft recipients: an analysis usingthe Framingham risk model. Am J Transplant 2002;2:551–9.

[27] Kahan BD, for The Rapamune US Study Group. Efficacy of sirolimus compared withazathioprine for reduction of acute renal allograft rejection: a randomizedmulticentre study. Lancet 2000;356:194–202.

[28] MacDonald AS, for the Rapamune Global Study Group. A worldwide, phase III, ran-domized, controlled, safety and efficacy study of a sirolimus/cyclosporine regimenfor prevention of acute rejection in recipients of primary mismatched renal allo-grafts. Transplantation 2001;71:271–80.

[29] Legendre C, Campistol JM, Squifflet JP, Burke JT. Cardiovascular risk factors ofsirolimus compared with cyclosporine: early experience from two randomized tri-als in renal transplantation. Transplant Proc 2003;35:S151–3.

[30] Webster AC, Lee VW, Chapman JR, Craig JC. Target of rapamycin inhibitors(sirolimus and everolimus) for primary immunosuppression of kidney transplantrecipients: a systematic review and meta-analysis of randomized trials. Transplan-tation 2006;81:1234–48.

[31] Vitko S, Wlodarczyk Z, Kyllönen L, et al. Tacrolimus combined with two differentdosages of sirolimus in kidney transplantation: results of a multicenter study. AmJ Transplant 2006;6:531–8.

[32] Vitko S, Tedesco H, Eris J, et al. Everolimus with optimized cyclosporine dosing inrenal transplant recipients: 6-month safety and efficacy results of two randomizedstudies. Am J Transplant 2004;4:626–35.

[33] Gurk-Turner C, Manitpisitkul W, Cooper M. A comprehensive review of everolimusclinical reports: a new mammalian target of rapamycin inhibitor. Transplantation2012;94:659–68.

[34] Zuckermann A, Wang SS, Epailly E, et al. Everolimus immunosuppression in denovo heart transplant recipients: what does the evidence tell us now? TransplantRev 2013;27:76–84.

[35] Mendez R, Gonwa T, Yang HC, et al. A prospective, randomized trial of tacrolimus incombination with sirolimus or mycophenolate mofetil in kidney transplantation:results at 1 year. Transplantation 2005;80:303–9.

[36] Tedesco Silva Jr H, Cibrik D, Johnston T, et al. Everolimus plus reduced-exposureCsA versus mycophenolic acid plus standard-exposure CsA in renal-transplant re-cipients. Am J Transplant 2010;10:1401–13.

[37] Flechner SM, Gurkan A, Hartmann A, et al. A randomized, open-label study ofsirolimus versus cyclosporine in primary de novo renal allograft recipients. Trans-plantation 2013;95:1233–41.

[38] Ekberg H, Bernasconi C, Nöldeke J, et al. Cyclosporine, tacrolimus and sirolimus re-tain their distinct toxicity profiles despite low doses in the Symphony study.Nephrol Dial Transplant 2010;25:2004–10.

[39] Büchler M, Caillard S, Barbier S, et al. Sirolimus versus cyclosporine in kidney recip-ients receiving thymoglobulin, mycophenolate mofetil and a 6-month course ofsteroids. Am J Transplant 2007;7:2522–31.

[40] Larson TS, Dean PG, Stegall MD, et al. Complete avoidance of calcineurin inhibitorsin renal transplantation: a randomized trial comparing sirolimus and tacrolimus.Am J Transplant 2006;6:514–22.

[41] Flechner SM, Goldfarb D, Modlin C, et al. Kidney transplantation without calcine-urin inhibitor drugs: a prospective, randomized trial of sirolimus versus cyclospor-ine. Transplantation 2002;74:1070–6.

[42] Fernandez ML, Webb D. The LDL to HDL cholesterol ratio as a valuable tool to eval-uate coronary heart disease risk. J Am Coll Nutr 2008;27:1–5.

[43] Holdaas H, Rostaing L, Serón D, et al. Conversion of long-term kidney transplant re-cipients from calcineurin inhibitor therapy to everolimus: a randomized, multicen-ter, 24-month study. Transplantation 2011;92:410–8.

[44] Bemelman FJ, de Maar EF, Press RR, et al. Minimization of maintenance immuno-suppression early after renal transplantation: an interim analysis. Transplantation2009;88:421–8.

a in transplant recipients: A cause for concern?, Transplant Rev (2014),

10 H. Holdaas et al. / Transplantation Reviews xxx (2014) xxx–xxx

[45] Budde K, Becker T, Arns A, et al. Everolimus-based, calcineurin-inhibitor-free regi-men in recipients of de-novo kidney transplants: an open-label, randomised, con-trolled trial. Lancet 2011;377:837–47.

[46] Schena FP, Pascoe MD, Alberu J, et al. Conversion from calcineurin inhibitors tosirolimus maintenance therapy in renal allograft recipients: 24-month efficacyand safety results from the CONVERT trial. Transplantation 2009;87:233–42.

[47] Lebranchu Y, Thierry A, Toupance O, et al. Efficacy on renal function of early con-version from cyclosporine to sirolimus 3 months after renal transplantation:CONCEPT study. Am J Transplant 2009;9:1115–23.

[48] Morales JM, Grinyó JM, Campistol JM, et al. Improved renal function, with similarproteinuria, after two years of early tacrolimus withdrawal from a regimen ofsirolimus plus tacrolimus. Transplantation 2008;86:620–2.

[49] Teperman L, Moonka D, Sebastian A, et al. Calcineurin inhibitor-free mycopheno-late mofetil/sirolimus maintenance in liver transplantation: the randomizedspare-the-nephron trial. Liver Transpl 2013;19:675–89.

[50] Fischer L, Klempnauer J, Beckebaum S, et al. A randomized, controlled study to as-sess the conversion from calcineurin-inhibitors to everolimus after liver transplan-tation – PROTECT. Am J Transplant 2012;12:1855–65.

[51] Masetti M, Montalti R, Rompianesi G, et al. Early withdrawal of calcineurin inhibi-tors and everolimus monotherapy in de novo liver transplant recipients preservesrenal function. Am J Transplant 2010;10:2252–62.

[52] De Simone P, Nevens F, De Carlis L, et al. Everolimus with reduced tacrolimus im-proves renal function in de novo liver transplant recipients: a randomized con-trolled trial. Am J Transplant 2012;12:3008–20.

[53] AbdelmalekMF, Humar A, Stickel F, et al. Sirolimus conversion regimen versus con-tinued calcineurin inhibitors in liver allograft recipients: a randomized trial. Am JTransplant 2012;12:694–705.

[54] De Simone P, Metselaar HJ, Fischer L, et al. Conversion from a calcineurin inhibitorto everolimus therapy in maintenance liver transplant recipients: a prospective,randomized, multicenter trial. Liver Transpl 2009;15:1262–9.

[55] Shenoy S, Hardinger KL, Crippin J, et al. Sirolimus conversion in liver transplant re-cipients with renal dysfunction: a prospective, randomized, single-center trial.Transplantation 2007;83:1389–92.

[56] Saliba F, Dharancy S, Lohro R, et al. Conversion to everolimus in maintenance livertransplant patients: a multicenter, retrospective analysis. Liver Transpl 2011;17:905–13.

[57] Watson CJ, Gimson AE, Alexander GJ, et al. A randomized controlled trial of lateconversion from calcineurin inhibitor (CNI)-based to sirolimus-based immunosup-pression in liver transplant recipients with impaired renal function. Liver Transpl2007;13:1694–702.

[58] Andreassen AK, Andersson B, Gustafsson F, Eiskjær H, Rådegran G, Gude E. Evero-limus initiation and early withdrawal of calcineurin inhibitor therapy in de novoheart transplant recipients – a multicenter, randomized Scandinavian trial. Am JTransplant 2014;14:1828–38 Epublication ahead.

[59] Eisen H, Kobashigawa J, Starling RC, et al. Everolimus versus mycophenolate mofe-til in heart transplantation: a randomized, multicenter trial. Am J Transplant 2013;13:1203–16.

[60] Lehmkuhl HB, Arizon J, Viganò M, et al. Everolimus with reduced cyclosporine ver-sus MMF with standard cyclosporine in de novo heart transplant recipients. Trans-plantation 2009;88:115–22.

[61] Gullestad L, Iversen M, Mortensen SA, et al. Everolimus with reduced calcineurininhibitor in thoracic transplant recipients with renal dysfunction: a multicenter,randomized trial. Transplantation 2010;89:864–72.

[62] Gullestad L,Mortensen SA, EiskjaerH, et al. Two-year outcomes in thoracic transplantrecipients after conversion to everolimuswith reduced calcineurin inhibitor within amulticenter, open-label, randomized trial. Transplantation 2010;90:1581–9.

[63] Potena L, Prestinenzi P, Bianchi IG, et al. Cyclosporine lowering with everolimusversus mycophenolate mofetil in heart transplant recipients: long-term follow-up of the SHIRAKISS randomized, prospective study. J Heart Lung Transplant2012;31:565–70.

[64] Morice MC, Serruys PW, Sousa JE, et al. A randomized comparison of a sirolimus-eluting stent with a standard stent for coronary revascularization. N Engl J Med2002;346:1773–80.

[65] Costa RA, Lansky AJ, Mintz GS, et al. Angiographic results of the first human expe-rience with everolimus-eluting stents for the treatment of coronary lesions (theFUTURE I trial). Am J Cardiol 2005;95:113–6.

[66] ViklickýO, ZouH,Müller V, Lacha J, SzabóA,HeemannU. SDZ-RADpreventsmanifes-tation of chronic rejection in rat renal allografts. Transplantation 2000;69:497–502.

[67] Ma KL, Ruan XZ, Powis SH, Moorhead JF, Varghese Z. Anti-atherosclerotic effects ofsirolimus on human vascular smooth muscle cells. Am J Physiol Heart Circ Physiol2007;292:H2721–8.

[68] Baetta R, Granata A, Canavesi M, et al. Everolimus inhibits monocyte/macrophagemigration in vitro and their accumulation in carotid lesions of cholesterol-fed rab-bits. J Pharmacol Exp Ther 2009;328:419–25.

[69] Goueffic Y, Potter-Perigo S, Chan CK, et al. Sirolimus blocks the accumulation ofhyaluronan (HA) by arterial smooth muscle cells and reduces monocyte adhesionto the ECM. Atherosclerosis 2007;195:23–30.

[70] Mueller MA, Beutner F, Teupser D, Ceglarek U, Thiery J. Prevention of atherosclero-sis by the mTOR inhibitor everolimus in LDLR _/_ mice despite severe hypercholes-terolemia. Atherosclerosis 2008;198:39–48.

[71] ChenWQ, Zhong L, Zhang L, et al. Oral rapamycin attenuates inflammation and en-hances stability of atherosclerotic plaques in rabbits independent of serum lipidlevels. Br J Pharmacol 2009;156:941–51.

Please cite this article as: Holdaas H, et al,mTOR inhibitors and dyslipidemihttp://dx.doi.org/10.1016/j.trre.2014.08.003

[72] Verheye S,MartinetW, KockxMM, et al. Selective clearance ofmacrophages in ath-erosclerotic plaques by autophagy. J Am Coll Cardiol 2007;49:706–15.

[73] Kobashigawa JA, Pauly DF, Starling RC, et al. Cardiac allograft vasculopathy by intra-vascular ultrasound in heart transplant patients: substudy from the everolimus ormycophenolate mofetil randomized, multicenter trial. JACC Heart Fail 2013;1:389–99.

[74] Eisen HJ, Tuzcu EM, Dorent R, et al. Everolimus for the prevention of allograft rejec-tion and vasculopathy in cardiac-transplant recipients. N Engl J Med 2003;349:847–58.

[75] McKenna GJ, Trotter JF, Klintmalm E, et al. Limiting hepatitis C virus progression inliver transplant recipients using sirolimus-based immunosuppression. Am J Trans-plant 2011;11:2379–87.

[76] Flechner SM, Kurian SM, Solez K, et al. De novo kidney transplantation without useof calcineurin inhibitors preserves renal structure and function at two years. Am JTransplant 2004;4:1776–85.

[77] Kobashigawa JA, Tobis JM, Starling RC, et al. Multicenter intravascular ultrasoundvalidation study among heart transplant recipients: outcomes after five years. JAm Coll Cardiol 2005;45:1532–7.

[78] Verbeke F, Maréchal C, Van Laecke S, et al. Aortic stiffness and central wavereflections predict outcome in renal transplant recipients. Hypertension 2011;58:833–8.

[79] Bahous SA, Blacher J, Safar ME. Aortic stiffness, kidney disease, and renal transplan-tation. Curr Hypertens Rep 2009;11:98–103.

[80] Seckinger J, Sommerer C, Hinkel UP, Hoffmann O, Zeier M, Schwenger V. Switch ofimmunosuppression from cyclosporine A to everolimus: impact on pulse wave ve-locity in stable de-novo renal allograft recipients. J Hypertens 2008;26:2213–9.

[81] Joannidès R, Monteil C, de Ligny BH, et al. Immunosuppressant regimen based onsirolimus decreases aortic stiffness in renal transplant recipients in comparison tocyclosporine. Am J Transplant 2011;11:2414–22.

[82] Paoletti E, Ratto E, Bellino D, Marsano L, Cassottana P, Cannella G. Effect of earlyconversion from CNI to sirolimus on outcomes in kidney transplant recipientswith allograft dysfunction. J Nephrol 2012;25:709–18.

[83] Raichlin E, Chandrasekaran K, Kremers WK, et al. Sirolimus as primary immuno-suppressant reduces left ventricular mass and improves diastolic function of thecardiac allograft. Transplantation 2008;86:1395–400.

[84] Kushwaha SS, Raichlin E, Sheinin Y, et al. Sirolimus affects cardiomyocytes to re-duce left ventricular mass in heart transplant recipients. Eur Heart J 2008;29:2742–50.

[85] Hiemann NE, Wellnhofer E, Lehmkuhl HB, Knosalla C, Hetzer R, Meyer R. Everoli-mus prevents endomyocardial remodeling after heart transplantation. Transplan-tation 2011;92:1165–72.

[86] Ekberg H, Bernasconi C, Tedesco-Silva H, et al. Calcineurin inhibitor minimizationin the Symphony study: observational results 3 years after transplantation. Am JTransplant 2009;9:1876–85.

[87] Ciancio G, Burke GW, Gaynor JJ, et al. A randomized long-term trial of tacrolimus/sirolimus versus tacrolimus/mycophenolate versus cyclosporine/sirolimus in renaltransplantation: three-year analysis. Transplantation 2006;81:845–52.

[88] Lebranchu Y, Snanoudj R, Toupance O, et al. Five-year results of a randomized trialcomparing de novo sirolimus and cyclosporine in renal transplantation: theSPIESSER Study. Am J Transplant 2012;12:1801–10.

[89] Eisen H, Yang X. The impact of proliferation signal inhibitors on the healthcare bur-den of major adverse cardiac events following heart transplantation. Transplanta-tion 2006;82:S13–8.

[90] Zeier M, Van Der Giet M. Calcineurin inhibitor sparing regimens using m-target ofrapamycin inhibitors: an opportunity to improve cardiovascular risk following kid-ney transplantation? Transpl Int 2011;24:30–42.

[91] Arora S, Andreassen A, Simonsen S, et al. Prognostic importance of renal function1 year after heart transplantation for all-cause and cardiac mortality and develop-ment of allograft vasculopathy. Transplantation 2007;84:149–54.

[92] Holdaas H, Midtvedt K, Åsberg A. A drug safety evaluation of everolimus in kidneytransplantation. Expert Opin Drug Saf 2012;11:1013–22.

[93] Kawahara T, Asthana S, Kneteman NM. m-TOR inhibitors: what role in liver trans-plantation? J Hepatol 2011;55:1441–51.

[94] KochM. Everolimus in liver and lung transplantation. Drugs Today (Barc) 2009;45:11–20.

[95] Manito N, Delgado JF, Crespo-Leiro MG, et al. Clinical recommendations for the useof everolimus in heart transplantation. Transplant Rev (Orlando) 2010;24:129–42.

[96] Diekmann F, Andrés A, Oppenheimer F. mTOR inhibitor-associated proteinuria inkidney transplant recipients. Transplant Rev (Orlando) 2012;26:27–9.

[97] Navaneethan SD, Perkovic V, Johnson DW, Nigwekar SU, Craig JC, Strippoli GF.HMG CoA reductase inhibitors (statins) for kidney transplant recipients. CochraneDatabase Syst Rev 2009;2:CD005019.

[98] Palmer SC, Craig JC, Navaneethan SD, Tonelli M, Pellegrini F, Strippoli GF. Benefitsand harms of statin therapy for persons with chronic kidney disease: a systematicreview and meta-analysis. Ann Intern Med 2012;157:263–75.

[99] Holdaas H, Fellström B, Cole E, et al. Long-term cardiac outcomes in renal trans-plant recipients receiving fluvastatin: the ALERT extension study. Am J Transplant2005;5:2929–36.

[100] Kovarik JM, Hartmann S, Hubert M, et al. Pharmacokinetic and pharmacodynamicassessments of HMG-CoA reductase inhibitors when coadministered with everoli-mus. J Clin Pharmacol 2002;42:222–8.

[101] Mells G, Neuberger J. Reducing the risks of cardiovascular disease in liver allograftrecipients. Transplantation 2007;83:1141–50.

a in transplant recipients: A cause for concern?, Transplant Rev (2014),