Metabolic and cardiovascular complications in the liver transplant recipient

Laura De Luca, Rachel Westbrook, Emmanuel A. Tsochatzis

Royal Free Hospital, London, UK
Sheila Sherlock Liver Unit and UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK

Correspondence to: Emmanuel A. Tsochatzis, The Royal Free Sheila Sherlock Liver Centre and UCL Institute for Liver and Digestive Health, The Royal Free Hospital, London, UK, Tel.: +44 2077 940500 (ext.: 31142), e-mail: e.tsochatzis@ucl.ac.uk
Received 15 September 2014; accepted 29 October 2014
© 2015 Hellenic Society of Gastroenterology

Abstract

Metabolic syndrome (MS) is an established risk factor for atherosclerosis and cardiovascular disease that affects 20-30% of the adult population in the western world, correlating with increased incidence of cardiovascular disease. Survival following liver transplantation (LT) has been steadily improving over the last 2 decades, with graft loss becoming a relatively rare cause of morbidity and mortality post LT. The improvement in short-term survival following LT has resulted in an increased incidence of metabolic and cardiovascular complications, which affect the mid- and long term survival. Patients following LT typically gain weight and might develop diabetes, hypertension and dyslipidemia as a consequence of their immunosuppressive therapy and their lifestyle. In this paper we review the prevalence of metabolic and cardiovascular complications following LT, their impact on post-transplant morbidity and mortality and their optimal management.

Keywords iver transplantation, metabolic syndrome, cardiovascular disease, epidemiology, risk factors, morbidity, mortality, NAFLD, NASH, hypertension, obesity, dyslipidemia, diabetes mellitus

Ann Gastroenterol 2015; 28 (2): 160-172


Introduction

Metabolic syndrome (MS) comprises several metabolic disorders including visceral obesity, diabetes mellitus (DM), dyslipidemia and hypertension (Table 1). It is an established risk factor for atherosclerosis and cardiovascular (CV) disease (CVD) [1-4] and is associated with nonalcoholic fatty liver disease (NAFLD), gallstones, obstructive sleep apnea, gout, depression, musculoskeletal diseases, and polycystic ovary syndrome [3].

Table 1 Criteria for the clinical diagnosis of metabolic syndrome [65]

thumblarge

MS has 3 potential etiologies, namely insulin resistance [5-7], obesity and disorders of adipose tissue [8], and a constellation of independent factors that mediate its specific components (e.g., molecules of hepatic, vascular, and immunologic origin) [9,10]. Additional factors such as aging, pro-inflammatory states and hormonal changes have also been implicated as contributors.

In the Western world dramatic changes in lifestyle and diet are fueling a persistent and sustained increase in the prevalence of MS. It is currently estimated to affect 20-30% of the adult population [11]. Its increasing prevalence correlates with an increasing incidence of CVD; the risk of atherosclerotic CVD accompanying MS is approximately double compared with the absence of the syndrome [3]. MS confers a risk independent of traditional CVD risk factors (relative risk [RR] 1.54) indicating that the associated CVD risk cannot be explained entirely by its individual components [12]. In a recent meta-analysis of 43 studies with 172,573 individuals, MS was shown to convey a RR for CVD events and death of 1.78 [2].

NAFLD is the hepatic manifestation of the MS and the leading cause of abnormal liver function tests in the Western world [13]. NAFLD occurs in subjects of all ages, even in patients with a normal body weight [14]. In MS, fatty liver infiltration causes a spectrum of disease ranging from simple steatosis, to nonalcoholic steatohepatitis (NASH), fibrosis and ultimately cirrhosis. Once cirrhosis is established, steatosis often disappears and many patients with NASH are labeled as having cryptogenic cirrhosis [15].

Liver transplantation (LT) is the most effective treatment for patients with decompensated chronic liver disease and significantly improves both quality of life and survival. In the pre-LT era, patients with advanced liver disease would die within months [16], whereas transplanted patients now have 1- and 5-year survival of 90% and 80% respectively [17,18]. Post-LT survival has been steadily improving over the last 2 decades. This is likely due to a combination of greater surgical expertise reducing technical complications, better selection of patients reducing peri-operative deaths, and perhaps most impressive improvement in the efficacy and tolerability of post-transplant immunosuppressive therapy (IS) reducing graft loss from acute and chronic rejection [19]. These advances have resulted in graft loss becoming a relatively rare cause of morbidity and mortality 1 year after LT.

Non-liver causes of death following LT are increasing with malignancy (22%), CVD (11%), infection (9%), and renal failure (6%) becoming leading causes of mortality one year post LT (Fig. 1) [20]. MS is a common thread of risk for each of these making the prevalence, etiology, prevention, and management of post-transplant MS (PTMS) of increasing interest and importance to the transplant community.

thumblarge

Figure 1 Etiology of death in liver transplant recipients 1 year post transplant [20]

LT does not have an impact on MS and patients transplanted for NAFLD with an underlying MS remain at risk of its sequalae [21,22]. Patients transplanted for etiologies other than NASH typically gain weight and often develop DM, hypertension and dyslipidemia as a consequence of IS therapy and a resultant MS. Therefore, all transplanted patients have an increased risk of non-liver-related morbidity and mortality owing to MS.

With the increasing prevalence of MS and its sequalae having an impact on morbidity and mortality post LT, physicians need to be aware of the syndrome and its management. In this paper, we review the prevalence of metabolic and CV complications following LT, their impact on post-transplant morbidity and mortality, and their optimal management.

Post-LT development of obesity

The World Health Organization (WHO) defines obesity as a body mass index (BMI) >30 kg/m2 and morbid obesity as a BMI >35 kg/m2. The increasing average BMI of the general population is reflected in patients being assessed for LT, as 15-30% of patients listed for LT have a BMI >30 kg/m2 [23]. Moreover, cirrhosis secondary to NAFLD is rapidly becoming a leading indication for LT. In patients with cirrhosis and fluid retention, weight and subsequently BMI can be inaccurate; the estimation of dry weight by subtracting the amount of ascites from the total body weight is a more reliable estimate.

Obese patients listed for LT should be advised and helped reduce their BMI, as this is associated with worse peri- and post-operative outcomes, including prolonged intensive therapy unit (ITU) and hospital stay, prolonged wound healing, and higher infection rates [24].

In the analysis of the UNOS database (1988-1996) by Nair et al, 7% of LT recipients were severely or morbidly obese (BMI ≥35 kg/m2); this was associated with an increased prevalence of adverse CV events leading to a 5-year mortality significantly higher in the severely obese (51%) and morbidly obese (58%) groups compared with the non-obese group (44%) [25]. This effect is more striking in the presence of DM [26].

Following LT, weight gain is the norm and is due to multiple factors such as the reversal of cirrhosis and its associated catabolic state, increased appetite due to the absence of chronic disease, and the effect of steroids. With few exceptions, patients that are overweight or obese prior to transplant will remain so after, while one-third of patients of normal weight pre-LT become obese post-transplant [23,28]. Obesity rates post-LT are 20-68% and are highest in those patients who are transplanted over the age of 50, those who are obese prior to LT and those on high-dose steroids.

In the 1st post-LT year, weight gain is frequently overlooked due to the focus of physicians on issues such as graft function, infection risks and renal function. The potential impact of post-LT weight gain includes increased risk of DM, and MS and its associated complications, such as CVD, renal disease, and de novo NASH in the allograft. In light of the above, patient education regarding weight control via diet and exercise should begin during the LT assessment process and reinforced throughout the post-LT period along with rapid tapering of corticosteroids in obese patients, and consideration to pharmacotherapy where the above interventions are unsuccessful.

Post-transplant development of DM

New-onset DM (NODM) is increasingly recognized as a complication of organ transplantation. NODM results in increased susceptibility to infectious and CV complications, may lead to diminished long-term graft survival, and has a major impact on the quality of life and survival [29-32].

The development of NODM is multi-factorial, however studies have shown an increased incidence in patients transplanted for HCV (OR 5.8, 95%CI 1.9-17.9), presence of DM, pre LT (OR 24.4, 95%CI 8.2-73.2), male gender (OR 3.57, 95%CI 1.2-10) [33-36].

The choice and dose of immunosuppressive medications is the major modifiable risk factor for NODM post LT. Corticosteroids have a well-known diabetogenic effect. The main mechanism underlying this effect is development of insulin resistance along with increased gluconeogenesis [37]. Although steroids are required in high doses for the first few weeks after LT, they should be rapidly tapered and discontinued unless otherwise needed to prevent disease recurrence or rejection. Calcineurin inhibitors (CNI) are also associated with NODM post-LT, as they directly damage pancreatic islet cells. The risk of NODM is significantly higher with tacrolimus (TAC) than cyclosporine A (CSA). Individualizing the immunosuppression regimen in the light of a patient’s risk profile would seem a prudent, as opposed to a “one size fits all” strategy. There is strong evidence to support that CNI minimization improves long-term outcomes with no adverse effects to graft survival [38-40].

In a meta-analysis including 3043 transplant recipients [41], NODM was reported in 13.4% of patients after solid organ transplantation, with a higher incidence in patients receiving immunosuppressive regimen of TAC than CSA (16.6% vs. 9.8%). This trend was consistent across patients that received renal, liver, heart and lung transplants. Sixteen studies with 1106 patients have reported the incidence of NODM in patients undergoing LT. The mean incidence of NODM across the 14 TAC-based studies was 18.2%, compared with 7.7% across the 12 CSA-based studies, with no evident impact of concomitant therapy in the incidence of NODM in either treatment group. The average NODM rates in the 7 prospective randomized trials included in the meta-analysis (338 LT recipients) were 15.9% for TAC-treated patients and 4.9% for CSA-treated patients.

There is convincing evidence from the non-LT population that tight glycemic control significantly reduces morbidity and mortality in patients with either type 1 or type 2 DM [42,43]. Although this approach has not been specifically tested in the LT population, it is reasonable to assume that similar benefits would be derived from effective management of glucose levels [44]. Little information exists on the use of anti-diabetic compounds in patients who undergo transplantation, and no comparative trials have been conducted [45-48].

In view of the current absence of precise recommendations, clinical judgement should be used when selecting anti-diabetic therapy, based on the medical history, severity of glucose deregulation, and properties of the anti-diabetic agents [47].

Post-transplant development of systemic hypertension

Arterial hypertension is an established risk factor for CV-related morbidity and mortality in the general population [49]. Although it affects a minority of patients prior to LT, its prevalence increases to 70% post LT [50]. Immunosuppressive medication is largely responsible for the development of hypertension post LT, with CNI and corticosteroids being the most strongly implicated. The primary mechanism of CNI induced hypertension is through widespread arterial vasoconstriction that results in increased systemic vascular resistance. The effect of vasoconstriction in the kidney is to promote sodium reabsorption and volume expansion.

Several reports have suggested that the incidence of hypertension in patients treated with TAC is lower than in patients treated with CSA within the first 1-2 years after kidney transplantation [51-53], up to 3 years after heart transplantation [54], and during the first year after LT [55-57]. Canzanello et al demonstrated that at 24 months post-LT, the prevalence of hypertension in the CSA and TAC groups were 82% and 64%, respectively. For those patients who were hypertensive by 24 months, the onset of hypertension was significantly delayed in the TAC group compared with the CSA group: 40% versus 71% and 73% versus 93% at 1 and 12 months, respectively. Within the TAC group, hypertensive patients had significantly lower glomerular filtration rates compared with normotensive patients, 74±12 versus 47±6 mL/min respectively. These results indicate that, compared with CSA, the onset of hypertension after LT is delayed and less prevalent with TAC [58].

The general principles for treating non-LT patients with essential hypertension apply to transplant recipients as well, however the choice of antihypertensive drugs in patients treated with CNI must be undertaken with particular attention to drug interactions.

Dihydropyridine calcium channel blocking drugs, such as amlodipine or felodipine, are a good first line choice since part of the mechanism of hypertension is due to renal arteriolar vasoconstriction. They act most potently on vascular smooth muscle to reduce systemic vascular resistance, have minimal interactions with CNIs and limited side effects. They appear to improve renal blood flow, although this has not been apparent during long-term therapy in LT recipients [59]. β-Blockers are widely used and may facilitate the return of high cardiac outputs toward normal levels. Angiotensin-converting enzyme (ACE) inhibitors and angiotensin-receptor blockers, which block the renin-angiotensin system, are of limited value when used as monotherapy for hypertensive patients early after LT as plasma renin activity is low during this period. Furthermore, ACE inhibitors and angiotensin-2 blockers may magnify the collateral effects of the CNI treatment such as hyperkalemia and metabolic acidosis. They can be used effectively during later periods after transplantation when the activation of the renin-angiotensin system becomes more evident. The use of diuretic agents in post-transplantation hypertension is debatable. The tendency of diuretics to increase serum creatinine levels, primarily by augmenting volume contraction in the face of CNI-mediated vasoconstriction in the kidney, has led to concerns regarding the stability of renal function.

Post-transplant development of dyslipidemia

Dyslipidemia occurs in all solid organ transplantations, but prevalence rates vary with the organ, being greatest for heart transplant recipients (60-80%) and least for liver recipients (45-69%) [60,61].

Pre-transplant dyslipidemia usually persists post-transplant and requires continuation of medical treatment. Patients with end-stage liver disease (ESLD) often have low serum cholesterol levels because of impaired hepatic synthesis and esterification [60,62,63], however patients with cholestatic liver disease may have increased serum cholesterol if the liver synthetic function is reasonably preserved. Serum triglycerides may be elevated both in obstructive jaundice and, less often, in parenchymal liver disease. Using the MS definition criteria (Table 1), the majority of ESLD patients have dyslipidemia based on low HDL-C levels due to the primary liver failure.

The recently published 2013 ACC/AHA Guideline changes the treatment paradigm for dyslipidemia. The new guideline no longer targets LDL-C levels but focuses on treating cholesterol to reduce atherosclerotic CVD (ASCVD) risk. Strong randomized control trial evidence supports a net benefit from statin therapy in individuals with clinical ASCVD (secondary prevention), and in three groups of individuals without clinical ASCVD (primary prevention), namely primary elevation of LDL-C >190 mg/dL; DM aged 40-75 years with LDL-C 70-189 mg/dL and without clinical ASCVD; without clinical ASCVD or DM with LDL-C 70-189 mg/dL and estimated 10-year ASCVD risk >7.5%. Statin therapy may be also considered in those who do not meet these criteria if other indications of increased ASCVD risk are present [64].

In post-transplant studies, the definition of hyperlipidemia varies widely and few use the NCEP-ATP III definitions [65]. Various individual patient risk factors along with the choice of immunosuppressive agents can influence the prevalence of post-transplant dyslipidemia. With regard to immunosuppression, CSA compared to TAC is associated with more frequent hyperlipidemia (14% versus 5%) [55,66-69] and hypertriglyceridemia (49% vs. 17%) [55]; in multiple series, changing from CSA to TAC improved hyperlipidemia [70-72]. The reason for this effect with CSA could be related to inhibition of bile salt synthesis [73]. Long-term corticosteroid use can also contribute to hyperlipidemia [74,75]. Steroid-free or sparing regimens were associated with improved lipid levels [76,77], including less hypertriglyceridemia in one [78] but not all series [79]. Sirolimus, an immunosuppressive agent used either in conjunction or instead of a CNI, is associated with high rates of dyslipidemia (up to 55%) [80]. This might result from changes in insulin signaling pathways resulting in excess triglyceride production and secretion [81]. Rates of dyslipidemia were lower when sirolimus was combined with TAC compared to CSA [82]. There is similar evidence of a synergistic hyperlipidemic effect between CSA and everolimus [83].

The observation that TAC is less likely to cause hypercholesterolemia than CSA has led several groups to propose conversion of liver transplant recipients to TAC-based immunosuppressive therapy from CSA-based therapy in the setting of persistent hypercholesterolemia, with some evidence of efficacy [84].

Post-transplant dyslipidemia is generally resistant to dietary interventions. For hypercholesterolemia post-LT a good first-line approach is statin treatment. Statins have been extensively used in solid organ transplant recipients for decades and are safe, efficacious and well tolerated [85]. Pravastatin is the most studied and used statin in post-transplant patients. It is not metabolized via the P450 enzyme system so has no interaction with IS therapy; atorvastatin, simvastatin, lovastatin, cerivastatin and fluvastatin however have also been frequently used in transplanted patients with little documented ill effect.

Hypertriglyceridemia with normal cholesterol levels is also common post LT. Hypertriglyceridemia responds to fish oil (ω-3) and this could be the preferred regimen since very few side effects and drug interactions can be expected [86]. Fish oil does not significantly affect CSA (a highly lipophilic agent) levels [87], and is not known to affect TAC metabolism. It also has anti-inflammatory and anti-proliferative properties, and improves hepatic steatosis [88].

Alternative agents include the fibric acid derivatives (gemfibrozil, clofibrate, fenofibrate), which are generally well tolerated. Fibrates are highly protein bound and are metabolized by cytochrome P450, with some evidence of a mild effect of increasing CNI serum levels [87].

Post-transplant development of NAFLD

The development of de novo NAFLD post LT is due to the previously discussed high incidence of its risk factors post LT including obesity, hyperlipidemia, DM, hypertension, and MS. A single-center retrospective report of 68 transplant recipients without prior fatty liver described de novo NAFLD in 18% and de novo NASH in 9% post LT. The regression model indicated that the use of ACE inhibitors was associated with a reduced risk of post-LT NAFLD (OR 0.09, 95%CI 0.010-0.92), while >10% increase in BMI was associated with a higher risk of developing NAFLD (OR 19.38, 95%CI 3.50-107.40) [89]. Although fatty liver and steatohepatitis are widely accepted to re-occur after LT, Contos et al demonstrated that none of the 30 long-term survivors (up to 5 years) developed graft dysfunction or graft loss caused by recurrence of NAFLD, and the morbidity and mortality is usually due to non-liver related causes, in particular CVD [90]. Further studies to define the long-term liver (>10 years) outcomes of such patients and methods to prevent recurrent fatty liver disease are needed.

Predictors of post-transplant MS

Higher age at transplantation, an increase in BMI post-LT, pre-LT DM, history of smoking, the immunosuppressant regimen used (CSA), and the indication for LT (hepatitis C, alcohol, or cryptogenic cirrhosis) are the risk factors most consistently associated with PTMS [23,91,92]. Anastácio et al assessed the prevalence and the predictors of PTMS in a cross-sectional study of 184 patients. MS was present in 39-50% post-LT depending on the criteria used. Predictors of PTMS were older age, shorter time since transplantation, alcohol abuse etiology, excessive weight at LT, physical activity reduction after LT and low intake of calcium, potassium, fiber and folic acid [93]. It could not be assessed if these factors were present before the onset of liver impairment or whether they were a consequence of it. Laryea et al [92] also found significantly higher pre-LT BMI, triglycerides, and abnormal HDL-C among patients with MS compared to those who did not develop PTMS.

The choice of immunosuppression has been implicated in the development of various components of MS as discussed above. However, given the superiority of TAC over CSA in terms of both graft and patient survival, no changes in the primary immunosuppressive regimen can be justified on this basis [94]. The predictors and risk factors of PTMS and for the individual components of the syndrome identified from the published data are shown in Table 2.

Table 2 Studies that evaluate independent predictors and risk factors of post-transplant metabolic syndrome and its individual components

thumblarge

Post-transplant renal dysfunction

Both acute and chronic kidney disease occur frequently in liver transplant recipients. In its purest form, hepatorenal syndrome represents a functional form of renal dysfunction, theoretically reversible [95]. However, most patients with renal dysfunction at the time of transplant do not recover normal function after LT [96]. A significant proportion (5-50%) of recipients develop acute renal failure in the immediate postoperative period, with more frequent occurrence in those who have renal impairment at the time of LT [97-100].

In a retrospective study of 202 patients, pre-transplant renal impairment was an independent predictor of post-transplant cardiac events (HR 2.19) and reduced cardiac event-free survival (HR 2.27) [101]. In a study of 798 LT recipients, CV system (CVS)-related deaths >1 year post-LT were significantly associated with renal insufficiency pre- or post-LT (HR 3.59) [20]. About 10-20% of long-term survivors after LT develop permanent renal dysfunction or failure, attributed to a number of factors, including the use of CNIs [103,104]. In a retrospective study of 54 post-liver transplant patients, low eGFR predicted patients with high Framingham CV risk score, suggesting that liver recipients with low eGFR should undergo close management of CV risk factors [105]. DM, coronary artery disease, and primary graft non-function predicted the development of severe renal failure in a retrospective study of 172 patients [106]. A meta-analysis of randomized trials directly comparing TAC trough concentrations showed that “reduced” TAC trough concentrations (<10 ng/mL) within the first month after LT were associated with less severe renal impairment at 1 year (RR 0.51), with no significant influence on acute rejection (RR 0.92) compared to “conventional” TAC trough levels (>10 ng/mL) [38]. In a study that evaluated early TAC exposure (<15 days) in relation to long-term outcomes in 493 consecutive LT patients, mean TAC of 7-10 ng/mL were associated with reduced risk of graft loss (RR 0.46) compared to the recommended levels of 10-15 ng/mL. Lower TAC levels did not significantly influence chronic rejection or chronic renal impairment [39]. The relevance of renal failure among liver transplant recipients and the impact that this frequent complication has on subsequent management necessitates further studies on modifiable risk factors such as the dose and choice of immunosuppression.

Post-transplant development of CV events

CV risk of LT recipients differs substantially from kidney and heart recipients. The primary difference is related to hemodynamic and metabolic changes associated with chronic liver disease, which lead to peripheral vasodilatation, low arterial blood pressure, and reduced serum cholesterol levels [109,110]. A number of post-mortem studies in cirrhotic patients have shown little pathologic evidence of atherosclerosis and a decreased incidence of vascular diseases [111,112].

Nevertheless, CVD is one of the leading causes of morbidity and mortality in LT patients. It is the 3rd most common late (after 1 year) cause of death, accounting for 12–16% of deaths (primary or major contributing causes) in the USA [20], and the 6th cause of overall mortality in the UK [113].

It is estimated that 27% of patients considered for LT have unknown underlying coronary artery disease [114,115]. The purpose of cardiac evaluation pre-LT is to assess perioperative risk and to exclude concomitant cardiopulmonary disorders that would preclude a good long-term outcome. Severe cardiac disease with unacceptable perioperative risk is a contraindication to LT.

Patients routinely undergo pre-transplant CVS assessment for risk stratification which includes: full history and examination, witnessed climb of 2 flights of stairs with pre- and post-O2 saturations, 12 lead ECG, and transthoracic echo with assessment of left ventricular, right ventricular and valvular function (with an estimation of systolic pulmonary artery pressure). Further investigation (right-side heart angiography, dobutamine stress echo, cardiac CT angiography) are based on medical history and findings from the initial screening tests [116]. Coronary revascularization (percutaneous coronary intervention [PCI] or coronary artery bypass grafting [CABG]) should be considered in LT candidates with obstructive coronary heart disease (CHD) if the extent contraindicates LT [117].

It is unclear to what extent pre-transplant cardiac abnormalities affect long-term outcome after LT. In a case-control study of 42 LT recipients with known pre-LT CHD and 42 recipients without CHD, mortality rates were higher in the CHD than in the control group at 1 and 3 years although lower than previously reported in other series (at 3 years 26% vs. 50%) [118]. The overall improvement in the CVS outcomes from historical series is possibly due to better risk stratification strategy and better management of the identified CHD (PCI, CABG). In a study of 1221 LT recipients divided in 3 sub-periods (reflecting the change in the risk stratification policy) the rate of cardiac catheterization during the pre-transplant assessment increased during the 3 time periods, as did the rate of PCI. All cause mortality decreased over time, as did the myocardial infarction (MI) rate [119].

LT recipients have a high prevalence of risk factors for CVD, exceeding that of the general population and thus have a higher predicted risk of developing CHD. Despite this, in a retrospective study of 181 consecutive adult liver transplant recipients, there were no deaths from CHD or stroke during the 54 months study period. The Framingham coronary risk equations provide an estimate of the 10-year risk of developing CHD. The 10-year probability for CHD (using the Framingham risk score) is higher in LT recipients (11%) than the general population (7%). Neal et al showed that the incidence ratios for MI and stroke were not significantly different in LT recipients compared with a matched non-transplant population [120]. Johnston et al assessed 110 consecutive liver recipients (median follow-up 3.9 years) for CV risk factors and reported a relative risk of ischemic cardiac events of 3.07 and a RR for CV deaths of 2.56 in allograft recipients compared to an age-matched population without transplants [121]. Vanwagner et al in a retrospective cohort study of 242 patients with >12 months follow up compared the incidence of CV events between patients transplanted for NASH and alcohol-induced cirrhosis. They demonstrated that patients with NASH have an increased risk of post-transplant CV events when compared to patients transplanted for alcohol-induced liver disease, even after controlling for traditional preoperative cardiac risk factors (26% vs. 8%; OR 4.12, 95%CI 1.91-8.90) [122]. In a cohort of 252 transplant recipients, MS was diagnosed in 5.4% of patients before and in 51.9% after transplantation. They split the cohort in 2 groups (with and without the PTMS): there was no difference between the groups in mortality or causes of death. The prevalence of major vascular events in the whole cohort increased from 2.3% before transplantation to 10.3% after but the difference was no statistically significant. The cumulative incidence of CV morbidity was statistically higher in patients with PTMS [123]. In 775 adult LT recipients, PTMS was significantly more prevalent in patients with CV events versus patients with no CV events (61.4% vs. 34.1%). The 1- and 3-year overall cumulative risks of CV events were 4.5% and 10.1% respectively [124]. These studies are summarized in Table 3.

Table 3 Studies that evaluate the post-transplant development of cardiovascular events

thumblarge

Therefore, it is important to promptly identify and treat all the parameters of the MS that contribute to the development of CVD. The correct management and timely treatment may reduce significantly the risk of major CV events.

Predictors of CV events post LT

Albeldawi et al determined the cumulative risk of CV events after LT and analyzed the predictive risk factors. Independent predictors of CV events were older age at transplantation (OR 1.2, 95%CI 1.1-1.3), male sex (OR 2.0 95%CI=1.2-3.3) NODM (OR 2.0, 95%CI 1.3-3.3) post-transplant hypertension (OR 1.8, 95%CI=1.1-3.0), and use of mycophenolate mofetil (OR 2.0, 95%CI 1.3-3.2). They showed that patients with post-transplant hypertension and DM, i.e. with potentially modifiable risk factors, are approximately twice as likely to experience a CV event [124]. Regarding the etiology of the ESLD, compared with all other etiologies, patients undergoing transplantation for NASH had a significantly higher risk of a CV event 1 and 3 years after transplantation (15.3% and 19.3), whereas patients undergoing transplantation for primary biliary cirrhosis or primary sclerosing cholangitis had a significantly lower risk of a CV event 1 and 3 years after transplantation (0% and 4.5%) [124].

In the paper of Dec et al, pre-existing cardiac disease and older age at transplantation were the only independent predictors of major complications. Major cardiac events were significantly associated with a lower 5-year survival rate (event: 32% vs. event free: 52%). The frequency of major intraoperative (21% vs. 2%) and post operative (57% vs. 17%) cardiac complications was significantly higher for recipients with known heart disease compared to those without pre-existing heart disease [125]. Again the importance of recognizing and treating the risk factors is crucial to avoid CV complications [126]. It may therefore be important to identify and stratify high-risk patients for CVD prior to LT and offer targeted post-LT interventions.

Concluding remarks

At present, there are no established guidelines in the treatment and prevention of CV profile and metabolic complications in liver transplant recipients. We reviewed the prevalence and incidence of PTMS and its individual components in this population and the rate of CVS events affecting morbidity and mortality. We presented an overall view on the relationship between these and other modifiable factors such as immunosuppression and the current evidence for treating the individual components of the PTMS in this population. We further highlighted the need of high-quality studies to confirm these relationships and the need for practice guidelines to deal with this growing problem.

MS is a cluster of modifiable factors where early interventions can potentially prevent more deleterious consequences. The LT recipients should be considered as a high CV risk population independently of the cause of the liver disease, and the British Transplant Society recommendations on NASH-transplanted patients should be followed; these suggest an intensive control of glucose serum levels, early steroid withdrawal and low doses of CNI in the post-transplant period [127].

In conclusion, liver transplant recipients and especially those with PTMS are at high risk for CVS events. However, the literature is limited and lacks high-quality studies. Future prospective studies are necessary to accurately document the prevalence and incidence of these complications and determine whether aggressive risk reduction strategies can attenuate the increased CVS risk seen in this population.

References

1. Third report of the National Cholesterol Education Program (NCEP) expert panel on detection, evaluation, and treatment of high blood cholesterol in adults (adult treatment panel III) final reportCirculation 2002; 106: 3143-3421.

2. Gami AS, Witt BJ, Howard DE, Metabolic syndrome and risk of incident cardiovascular events and death: a systematic review and meta-analysis of longitudinal studiesJ Am Coll Cardiol 2007; 49: 403-414.

3. Grundy SM, Cleeman JI, Daniels SR, Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific StatementCirculation 2005; 112: 2735-2752.

4. Reaven GM, Banting lecture 1988. Role of insulin resistance in human diseaseDiabetes 1988; 37: 1595-1607.

5. Abbasi F, Brown BW, JrLamendola C, McLaughlin T, Reaven GM, Relationship between obesity, insulin resistance, and coronary heart disease riskJ Am Coll Cardiol 2002; 40: 937-943.

6. Bogardus C, Lillioja S, Mott DM, Hollenbeck C, Reaven G, Relationship between degree of obesity and in vivo insulin action in manAm J Physiol 1985; 248: E286-291.

7. Ferrannini E, Haffner SM, Mitchell BD, Stern MP, Hyperinsulinaemia: the key feature of a cardiovascular and metabolic syndromeDiabetologia 1991; 34: 416-422.

8. Tsochatzis E, Papatheodoridis GV, Archimandritis AJ, The evolving role of leptin and adiponectin in chronic liver diseasesAm J Gastroenterol 2006; 101: 2629-2640.

9. Tsochatzis E, Papatheodoridis GV, Hadziyannis E, Serum adipokine levels in chronic liver diseases: association of resistin levels with fibrosis severityScand J Gastroenterol 2008; 43: 1128-1136.

10. Tsochatzis EA, Papatheodoridis GV, Archimandritis AJ, Adipokines in nonalcoholic steatohepatitis: from pathogenesis to implications in diagnosis and therapyMediators Inflamm 2009 2009; 831670

11. Grundy SM, Metabolic syndrome pandemicArterioscler Thromb Vasc Biol 2008; 28: 629-636.

12. Despres JP, Lemieux I, Abdominal obesity and metabolic syndromeNature 2006; 444: 881-887.

13. Ruhl CE, Everhart JE, Determinants of the association of overweight with elevated serum alanine aminotransferase activity in the United StatesGastroenterology 2003; 124: 71-79.

14. Bacon BR, Farahvash MJ, Janney CG, Neuschwander-Tetri BA, Nonalcoholic steatohepatitis: an expanded clinical entityGastroenterology 1994; 107: 1103-1109.

15. Caldwell SH, Oelsner DH, Iezzoni JC, Hespenheide EE, Battle EH, Driscoll CJ, Cryptogenic cirrhosis: clinical characterization and risk factors for underlying diseaseHepatology 1999; 29: 664-669.

16. Tsochatzis EA, Bosch J, Burroughs AK, Liver cirrhosisLancet 2014; 383: 1749-1761.

17. Belle SH, Porayko MK, Hoofnagle JH, Lake JR, Zetterman RK, Changes in quality of life after liver transplantation among adults. National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) Liver Transplantation Database (LTD)Liver Transpl Surg 1997; 3: 93-104.

18. Germani G, Theocharidou E, Adam R, Liver transplantation for acute liver failure in Europe: outcomes over 20 years from the ELTR databaseJ Hepatol 2012; 57: 288-296.

19. Adam R, McMaster P, O’Grady JG, Evolution of liver transplantation in Europe: report of the European Liver Transplant RegistryLiver Transpl 2003; 9: 1231-1243.

20. Watt KD, Pedersen RA, Kremers WK, Heimbach JK, Charlton MR, Evolution of causes and risk factors for mortality post-liver transplant: results of the NIDDK long-term follow-up studyAm J Transplant 2010; 10: 1420-1427.

21. Madhwal S, Atreja A, Albeldawi M, Lopez R, Post A, Costa MA, Is liver transplantation a risk factor for cardiovascular disease? A meta-analysis of observational studiesLiver Transpl 2012; 18: 1140-1146.

22. Pagadala M, Dasarathy S, Eghtesad B, McCullough AJ, Posttransplant metabolic syndrome: an epidemic waiting to happenLiver Transpl 2009; 15: 1662-1670.

23. Bianchi G, Marchesini G, Marzocchi R, Pinna AD, Zoli M, Metabolic syndrome in liver transplantation: relation to etiology and immunosuppressionLiver Transpl 2008; 14: 1648-1654.

24. Choban PS, Flancbaum L, The impact of obesity on surgical outcomes: a reviewJ Am Coll Surg 1997; 185: 593-603.

25. Nair S, Verma S, Thuluvath PJ, Obesity and its effect on survival in patients undergoing orthotopic liver transplantation in the United StatesHepatology 2002; 35: 105-109.

26. Dare AJ, Plank LD, Phillips AR, Additive effect of pretransplant obesity, diabetes, and cardiovascular risk factors on outcomes after liver transplantationLiver Transpl 2014; 20: 281-290.

27. Everhart JE, Lombardero M, Lake JR, Wiesner RH, Zetterman RK, Hoofnagle JH, Weight change and obesity after liver transplantation: incidence and risk factorsLiver Transpl Surg 1998; 4: 285-296.

28. Miles AM, Sumrani N, Horowitz R, Diabetes mellitus after renal transplantation: as deleterious as non-transplant-associated diabetes?Transplantation 1998; 65: 380-384.

29. Roth D, Milgrom M, Esquenazi V, Fuller L, Burke G, Miller J, Posttransplant hyperglycemia. Increased incidence in cyclosporine-treated renal allograft recipientsTransplantation 1989; 47: 278-281.

30. Sumrani NB, Delaney V, Ding ZK, Diabetes mellitus after renal transplantation in the cyclosporine era-an analysis of risk factorsTransplantation 1991; 51: 343-347.

31. Weir MR, Fink JC, Risk for posttransplant diabetes mellitus with current immunosuppressive medicationsAm J Kidney Dis 1999; 34: 1-13.

32. Baid S, Cosimi AB, Farrell ML, Posttransplant diabetes mellitus in liver transplant recipients: risk factors, temporal relationship with hepatitis C virus allograft hepatitis, and impact on mortalityTransplantation 2001; 72: 1066-1072.

33. Bigam DL, Pennington JJ, Carpentier A, Hepatitis C-related cirrhosis: a predictor of diabetes after liver transplantationHepatology 2000; 32: 87-90.

34. Trail KC, McCashland TM, Larsen JL, Morbidity in patients with posttransplant diabetes mellitus following orthotopic liver transplantationLiver Transpl Surg 1996; 2: 276-283.

35. Tueche SG, Diabetes mellitus after liver transplant new etiologic clues and cornerstones for understandingTransplant Proc 2003; 35: 1466-1468.

36. Chan JC, Cockram CS, Critchley JA, Drug-induced disorders of glucose metabolism. Mechanisms and managementDrug Saf 1996; 15: 135-157.

37. Rodriguez-Peralvarez M, Germani G, Darius T, Lerut J, Tsochatzis E, Burroughs AK, Tacrolimus trough levels, rejection and renal impairment in liver transplantation: a systematic review and meta-analysisAm J Transplant 2012; 12: 2797-2814.

38. Rodriguez-Peralvarez M, Germani G, Papastergiou V, Early tacrolimus exposure after liver transplantation: relationship with moderate/severe acute rejection and long-term outcomeJ Hepatol 2013; 58: 262-270.

39. Rodriguez-Peralvarez M, Tsochatzis E, Naveas MC, Reduced exposure to calcineurin inhibitors early after liver transplantation prevents recurrence of hepatocellular carcinomaJ Hepatol 2013; 59: 1193-1199.

40. Heisel O, Heisel R, Balshaw R, Keown P, New onset diabetes mellitus in patients receiving calcineurin inhibitors: a systematic review and meta-analysisAm J Transplant 2004; 4: 583-595.

41. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research GroupN Engl J Med 1993; 329: 977-986.

42. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) GroupLancet 1998; 352: 837-853.

43. Shields PL, Tang H, Neuberger JM, Gunson BK, McMaster P, Pirenne J, Poor outcome in patients with diabetes mellitus undergoing liver transplantationTransplantation 1999; 68: 530-535.

44. Benhamou PY, Penfornis A, Natural history, prognosis, and management of transplantation-induced diabetes mellitusDiabetes Metab 2002; 28: 166-175.

45. Davidson J, Wilkinson A, Dantal J, New-onset diabetes after transplantation: 2003 International consensus guidelines. Proceedings of an international expert panel meeting. Barcelona, Spain, 19 February 2003Transplantation 2003; 75: Ss3-24.

46. Marchetti P, Strategies for risk reduction and management of posttransplant diabetes mellitusTransplant Proc 2001; 33: 27s-31s.

47. Montori VM, Basu A, Erwin PJ, Velosa JA, Gabriel SE, Kudva YC, Posttransplantation diabetes: a systematic review of the literatureDiabetes Care 2002; 25: 583-592.

48. Midtvedt K, Neumayer HH, Management strategies for posttransplant hypertensionTransplantation 2000; 70: Ss64-69.

49. Watt KD, Charlton MR, Metabolic syndrome and liver transplantation: a review and guide to managementJ Hepatol 2010; 53: 199-206.

50. Ellis D, Shapiro R, Jordan ML, Comparison of FK-506 and cyclosporine regimens in pediatric renal transplantationPediatr Nephrol 1994; 8: 193-200.

51. Kokado Y, Takahara S, Kameoka H, Okuyama A, Hypertension in renal transplant recipients and its effect on long-term renal allograft survivalTransplant Proc 1996; 28: 1600-1602.

52. Pirsch JD, Miller J, Deierhoi MH, Vincenti F, Filo RS, A comparison of tacrolimus (FK506) and cyclosporine for immunosuppression after cadaveric renal transplantation. FK506 Kidney Transplant Study GroupTransplantation 1997; 63: 977-983.

53. Swenson JM, Fricker FJ, Armitage JM, Immunosuppression switch in pediatric heart transplant recipients: cyclosporine to FK 506J Am Coll Cardiol 1995; 25: 1183-1188.

54. Canzanello VJ, Schwartz L, Taler SJ, Evolution of cardiovascular risk after liver transplantation: a comparison of cyclosporine A and tacrolimus (FK506)Liver Transpl Surg 1997; 3: 1-9.

55. Clark W, Tacrolimus: immunosuppression following liver and kidney transplantJ Clin Pharm Ther 1996; 21: 135-141.

56. Todo S, Fung JJ, Tzakis A, One hundred ten consecutive primary orthotopic liver transplants under FK 506 in adultsTransplant Proc 1991; 23: 1397-1402.

57. Canzanello VJ, Textor SC, Taler SJ, Late hypertension after liver transplantation: a comparison of cyclosporine and tacrolimus (FK 506)Liver Transpl Surg 1998; 4: 328-334.

58. Textor SC, Schwartz L, Wilson DJ, Systemic and renal effects of nifedipine in cyclosporine-associated hypertensionHypertension 1994; 23: I220-224.

59. Gisbert C, Prieto M, Berenguer M, Hyperlipidemia in liver transplant recipients: prevalence and risk factorsLiver Transpl Surg 1997; 3: 416-422.

60. Kobashigawa JA, Kasiske BL, Hyperlipidemia in solid organ transplantationTransplantation 1997; 63: 331-338.

61. Imagawa DK, Dawson S, 3rdHolt CD, Hyperlipidemia after liver transplantation: natural history and treatment with the hydroxy-methylglutaryl-coenzyme A reductase inhibitor pravastatinTransplantation 1996; 62: 934-942.

62. Munoz SJ, Deems RO, Moritz MJ, Martin P, Jarrell BE, Maddrey WC, Hyperlipidemia and obesity after orthotopic liver transplantationTransplant Proc 1991; 23: 1480-1483.

63. Stone NJ, Robinson JG, Lichtenstein AH, 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice GuidelinesJ Am Coll Cardiol 2014; 63: 2889-2934.

64. Executive summary of the third report of the National Cholesterol Education Program (NCEP) expert panel on detection, evaluation, and treatment of high blood cholesterol In adults (adult treatment panel III)Jama 2001; 285: 2486-2497.

65. Charco R, Cantarell C, Vargas V, Serum cholesterol changes in long-term survivors of liver transplantation: a comparison between cyclosporine and tacrolimus therapyLiver Transpl Surg 1999; 5: 204-208.

66. Guckelberger O, Bechstein WO, Neuhaus R, Cardiovascular risk factors in long-term follow-up after orthotopic liver transplantationClin Transplant 1997; 11: 60-65.

67. Rabkin JM, Corless CL, Rosen HR, Olyaei AJ, Immunosuppression impact on long-term cardiovascular complications after liver transplantationAm J Surg 2002; 183: 595-599.

68. Stegall MD, Wachs ME, Everson G, Prednisone withdrawal 14 days after liver transplantation with mycophenolate: a prospective trial of cyclosporine and tacrolimusTransplantation 1997; 64: 1755-1760.

69. Manzarbeitia C, Reich DJ, Rothstein KD, Braitman LE, Levin S, Munoz SJ, Tacrolimus conversion improves hyperlipidemic states in stable liver transplant recipientsLiver Transpl 2001; 7: 93-99.

70. Neal DA, Gimson AE, Gibbs P, Alexander GJ, Beneficial effects of converting liver transplant recipients from cyclosporine to tacrolimus on blood pressure, serum lipids, and weightLiver Transpl 2001; 7: 533-539.

71. Pratschke J, Neuhaus R, Tullius SG, Treatment of cyclosporine-related adverse effects by conversion to tacrolimus after liver transplantationTransplantation 1997; 64: 938-940.

72. Hulzebos CV, Bijleveld CM, Stellaard F, Cyclosporine A-induced reduction of bile salt synthesis associated with increased plasma lipids in children after liver transplantationLiver Transpl 2004; 10: 872-880.

73. Fernandez-Miranda C, de la Calle A, Morales JM, Lipoprotein abnormalities in long-term stable liver and renal transplanted patients. A comparative studyClin Transplant 1998; 12: 136-141.

74. Stegall MD, Everson GT, Schroter G, Prednisone withdrawal late after adult liver transplantation reduces diabetes, hypertension, and hypercholesterolemia without causing graft lossHepatology 1997; 25: 173-177.

75. Boillot O, Mayer DA, Boudjema K, Corticosteroid-free immunosuppression with tacrolimus following induction with daclizumab: a large randomized clinical studyLiver Transpl 2005; 11: 61-67.

76. Segev DL, Sozio SM, Shin EJ, Steroid avoidance in liver transplantation: meta-analysis and meta-regression of randomized trialsLiver Transpl 2008; 14: 512-525.

77. Moench C, Barreiros AP, Schuchmann M, Tacrolimus monotherapy without steroids after liver transplantation-a prospective randomized double-blinded placebo-controlled trialAm J Transplant 2007; 7: 1616-1623.

78. Klintmalm GB, Washburn WK, Rudich SM, Corticosteroid-free immunosuppression with daclizumab in HCV(+) liver transplant recipients: 1-year interim results of the HCV-3 studyLiver Transpl 2007; 13: 1521-1531.

79. Neff GW, Montalbano M, Tzakis AG, Ten years of sirolimus therapy in orthotopic liver transplant recipientsTransplant Proc 2003; 35: 209s-216s.

80. Morrisett JD, Abdel-Fattah G, Hoogeveen R, Effects of sirolimus on plasma lipids, lipoprotein levels, and fatty acid metabolism in renal transplant patientsJ Lipid Res 2002; 43: 1170-1180.

81. Trotter JF, Wachs ME, Trouillot TE, Dyslipidemia during sirolimus therapy in liver transplant recipients occurs with concomitant cyclosporine but not tacrolimusLiver Transpl 2001; 7: 401-408.

82. Deters M, Klabunde T, Meyer H, Resch K, Kaever V, Effects of curcumin on cyclosporine-induced cholestasis and hypercholesterolemia and on cyclosporine metabolism in the ratPlanta Med 2003; 69: 337-343.

83. Roy A, Kneteman N, Lilly L, Tacrolimus as intervention in the treatment of hyperlipidemia after liver transplantTransplantation 2006; 82: 494-500.

84. Martin JE, Cavanaugh TM, Trumbull L, Incidence of adverse events with HMG-CoA reductase inhibitors in liver transplant patientsClin Transplant 2008; 22: 113-119.

85. McKenney JM, Sica D, Role of prescription omega-3 fatty acids in the treatment of hypertriglyceridemiaPharmacotherapy 2007; 27: 715-728.

86. Asberg A, Interactions between cyclosporin and lipid-lowering drugs: implications for organ transplant recipientsDrugs 2003; 63: 367-378.

87. Lee S, Gura KM, Puder M, Omega-3 fatty acids and liver diseaseHepatology 2007; 45: 841-845.

88. Seo S, Maganti K, Khehra M, De novo nonalcoholic fatty liver disease after liver transplantationLiver Transpl 2007; 13: 844-847.

89. Contos MJ, Cales W, Sterling RK, Development of nonalcoholic fatty liver disease after orthotopic liver transplantation for cryptogenic cirrhosisLiver Transpl 2001; 7: 363-373.

90. Francioso S, Angelico F, Baiocchi L, High prevalence of metabolic syndrome and long-term survival after liver transplantationJ Hepatol 2008; 48: S82-S82.

91. Laryea M, Watt KD, Molinari M, Metabolic syndrome in liver transplant recipients: prevalence and association with major vascular eventsLiver Transpl 2007; 13: 1109-1114.

92. Anastacio LR, Ferreira LG, Ribeiro H, de S, Liboredo JC, Lima AS, Correia MI, Metabolic syndrome after liver transplantation: prevalence and predictive factorsNutrition 2011; 27: 931-937.

93. O’Grady JG, Burroughs A, Hardy P, Elbourne D, Truesdale A, Tacrolimus versus microemulsified ciclosporin in liver transplantation: the TMC randomised controlled trialLancet 2002; 360: 1119-1125.

94. Moreau R, Lebrec D, Acute renal failure in patients with cirrhosis: perspectives in the age of MELDHepatology 2003; 37: 233-243.

95. Ruiz R, Barri YM, Jennings LW, Hepatorenal syndrome: a proposal for kidney after liver transplantation (KALT)Liver Transpl 2007; 13: 838-843.

96. Bilbao I, Charco R, Balsells J, Risk factors for acute renal failure requiring dialysis after liver transplantationClin Transplant 1998; 12: 123-129.

97. Brown RS, JrLombardero M, Lake JR, Outcome of patients with renal insufficiency undergoing liver or liver-kidney transplantationTransplantation 1996; 62: 1788-1793.

98. Gonwa TA, Klintmalm GB, Levy M, Jennings LS, Goldstein RM, Husberg BS, Impact of pretransplant renal function on survival after liver transplantationTransplantation 1995; 59: 361-365.

99. Jeyarajah DR, Gonwa TA, McBride M, Hepatorenal syndrome: combined liver kidney transplants versus isolated liver transplantTransplantation 1997; 64: 1760-1765.

100. Josefsson A, Fu M, Bjornsson E, Castedal M, Kalaitzakis E, Pre-transplant renal impairment predicts posttransplant cardiac events in patients with liver cirrhosisTransplantation 2014; 98: 107-114.

101. Davis CL, Gonwa TA, Wilkinson AH, Pathophysiology of renal disease associated with liver disorders: implications for liver transplantation. Part ILiver Transpl 2002; 8: 91-109.

102. Davis CL, Gonwa TA, Wilkinson AH, Identification of patients best suited for combined liver-kidney transplantation: part IILiver Transpl 2002; 8: 193-211.

103. Mansell H, Worobetz LJ, Sylwestrowicz T, Shoker AS, A retrospective study of the Framingham cardiovascular risk scores in a liver transplant populationTransplant Proc 2013; 45: 308-314.

104. Pawarode A, Fine DM, Thuluvath PJ, Independent risk factors and natural history of renal dysfunction in liver transplant recipientsLiver Transpl 2003; 9: 741-747.

105. Cicognani C, Malavolti M, Morselli-Labate AM, Zamboni L, Sama C, Barbara L, Serum lipid and lipoprotein patterns in patients with liver cirrhosis and chronic active hepatitisArch Intern Med 1997; 157: 792-796.

106. Textor SC, Wiesner R, Wilson DJ, Systemic and renal hemodynamic differences between FK506 and cyclosporine in liver transplant recipientsTransplantation 1993; 55: 1332-1339.

107. Grant WC, Wasserman F, Rodensky PL, Thomson RV, The incidence of myocardial infarction in portal cirrhosisAnn Intern Med 1959; 51: 774-779.

108. Vanecek R, Atherosclerosis and cirrhosis of the liverBull World Health Organ 1976; 53: 567-570.

109. Gelson W, Hoare M, Dawwas MF, Vowler S, Gibbs P, Alexander G, The pattern of late mortality in liver transplant recipients in the United KingdomTransplantation 2011; 91: 1240-1244.

110. Carey WD, Dumot JA, Pimentel RR, The prevalence of coronary artery disease in liver transplant candidates over age 50Transplantation 1995; 59: 859-864.

111. Tiukinhoy-Laing SD, Rossi JS, Bayram M, Cardiac hemodynamic and coronary angiographic characteristics of patients being evaluated for liver transplantationAm J Cardiol 2006; 98: 178-181.

112. Martin P, DiMartini A, Feng S, Brown R, JrFallon M, Evaluation for liver transplantation in adults: 2013 practice guideline by the American Association for the Study of Liver Diseases and the American Society of TransplantationHepatology 2014; 59: 1144-1165.

113. Raval Z, Harinstein ME, Skaro AI, Cardiovascular risk assessment of the liver transplant candidateJ Am Coll Cardiol 2011; 58: 223-231.

114. Diedrich DA, Findlay JY, Harrison BA, Rosen CB, Influence of coronary artery disease on outcomes after liver transplantationTransplant Proc 2008; 40: 3554-3557.

115. Maddur H, Bourdillon PD, Liangpunsakul S, Role of cardiac catheterization and percutaneous coronary intervention in the preoperative assessment and management of patients before orthotopic liver transplantationLiver Transpl 2014; 20: 664-672.

116. Neal DA, Tom BD, Luan J, Is there disparity between risk and incidence of cardiovascular disease after liver transplant?Transplantation 2004; 77: 93-99.

117. Johnston SD, Morris JK, Cramb R, Gunson BK, Neuberger J, Cardiovascular morbidity and mortality after orthotopic liver transplantationTransplantation 2002; 73: 901-906.

118. Vanwagner LB, Bhave M, Te HS, Feinglass J, Alvarez L, Rinella ME, Patients transplanted for nonalcoholic steatohepatitis are at increased risk for postoperative cardiovascular eventsHepatology 2012; 56: 1741-1750.

119. 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 eventsLiver Transpl 2011; 17: 15-22.

120. Albeldawi M, Aggarwal A, Madhwal S, Cumulative risk of cardiovascular events after orthotopic liver transplantationLiver Transpl 2012; 18: 370-375.

121. Dec GW, Kondo N, Farrell ML, Dienstag J, Cosimi AB, Semigran MJ, Cardiovascular complications following liver transplantationClin Transplant 1995; 9: 463-471.

122. Tsochatzis EA, Bosch J, Burroughs AK, New therapeutic paradigm for patients with cirrhosisHepatology 2012; 56: 1983-1992.

123. Newsome PN, Allison ME, Andrews PA, Guidelines for liver transplantation for patients with non-alcoholic steatohepatitisGut 2012; 61: 484-500.

124. Lim LG, Cheng CL, Wee A, Prevalence and clinical associations of posttransplant fatty liver diseaseLiver Int 2007; 27: 76-80.

Notes

Conflict of Interest: None