Next Article in Journal
From Genes to Treatment: Literature Review and Perspectives on Acid Sphingomyelinase Deficiency in Children
Previous Article in Journal
Demonstrating an Academic Core Facility for Automated Medical Image Processing and Analysis: Workflow Design and Practical Applications
Previous Article in Special Issue
Severe Hyporesponsiveness to Erythropoiesis-Stimulating Agents in Patients on Chronic Hemodialysis—Reconsidering the Relationship with Thrombo-Inflammation and Oxidative Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

From Risk Assessment to Management: Cardiovascular Complications in Pre- and Post-Kidney Transplant Recipients: A Narrative Review

1
Nephrology Department, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France
2
Inserm UMR_S 1109 Immuno-Rhumatology Laboratory, Translational Medicine Federation of Strasbourg (FMTS), FHU Target, Faculté de Médecine, Université de Strasbourg, 67000 Strasbourg, France
3
INI-CRCT (Cardiovascular and Renal Trialists), F-CRIN Network, 67000 Strasbourg, France
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Diagnostics 2025, 15(7), 802; https://doi.org/10.3390/diagnostics15070802
Submission received: 14 February 2025 / Revised: 17 March 2025 / Accepted: 21 March 2025 / Published: 21 March 2025
(This article belongs to the Special Issue Advances in Nephrology)

Abstract

:
Kidney transplantation remains the best treatment for chronic kidney failure, offering better outcomes and quality of life compared with dialysis. Cardiovascular disease (CVD) is a major cause of morbidity and mortality in kidney transplant recipients and is associated with decreased patient survival and worse graft outcomes. Post-transplant CVD results from a complex interaction between traditional cardiovascular risk factors, such as hypertension and diabetes, and risk factors specific to kidney transplant recipients including chronic kidney disease, immunosuppressive drugs, or vascular access. An accurate assessment of cardiovascular risk is now needed to optimize the management of cardiovascular comorbidities through the detection of risk factors and the screening of hidden pretransplant coronary artery disease. Promising new strategies are emerging, such as GLP-1 receptor agonists and SGLT2 inhibitors, with a high potential to mitigate cardiovascular complications, although further research is needed to determine their role in kidney transplant recipients. Despite this progress, a significant gap remains in understanding the optimal management of post-transplant CVD, especially coronary artery disease, stroke, and peripheral artery disease. Addressing these challenges is essential to improve the short- and long-term outcomes in kidney transplant recipients. This narrative review aims to provide a comprehensive overview of cardiovascular risk assessment and post-transplant CVD management.

1. Introduction

Kidney transplantation is associated with a long-term reduction in cardiovascular risk compared with wait-listed patients, but cardiovascular disease (CVD) remains a prominent cause of morbidity and mortality in kidney transplant recipients. Post-transplant CVD results from complex interactions between (1) traditional cardiovascular risk factors (i.e., diabetes, hypertension, dyslipidemia and tobacco), (2) specific factors linked with chronic kidney disease and transplantation such as mineral bone disease or graft dysfunction, and (3) previously known or hidden cardiovascular comorbidities. The spectrum of post-transplant CVD covers coronary artery disease, stroke, and peripheral artery disease, with some peculiar complications such as early post-transplant coronary artery disease, undetected despite pretransplant screening, and accelerated arteriosclerosis occurring in the transplant. We aimed to conduct a narrative review on the risk assessment and management of cardiovascular disease in the post-kidney transplantation period.
In this narrative review, we tried to deliver an overview of post-transplant cardiovascular risk and disease, starting with risk assessment. We first briefly describe the epidemiology of cardiovascular events before studying the field of traditional and specific cardiovascular risk factors. We also treat the specific and crucial screening of coronary artery disease before transplantation. In the second part, we were interested in the management of the previously described risk factors, with a comprehensive overview of cardiorenal medical therapies, immunosuppression, and vascular access management. Finally, in the third part, we focus on the management of the main cardiovascular diseases occurring after transplantation, especially coronary artery disease, cerebrovascular disease, atrial fibrillation, and peripheral artery disease.
The selection of the references did not follow a systematic review approach, but we previously defined a set of general criteria to identify studies relevant to the topic after a search in the PubMed database. We prioritized studies including observational data or clinical trials related to cardiovascular risk factors and diseases in the particular population of kidney transplant recipients. The authors selected them based on their general evaluation of relevance, methodology, and sample size as well as their contribution to understanding the field of post-transplant cardiovascular disease. We tried to capture a broad range of perspectives including both well-established studies and emerging research that provide new insights into the field.

2. Assessment of Cardiovascular Risk in Kidney Transplant Recipients

2.1. Epidemiology of Cardiovascular Events

2.1.1. Cardiovascular Morbidity and Mortality

CVD is the leading cause of morbidity and mortality in chronic kidney disease [1,2], particularly among patients undergoing dialysis [3]. Kidney transplantation is the optimal treatment for kidney failure, as it is associated with improved survival compared with waitlisted patients [4]. Notably, much of this survival benefit is due to a reduction in cardiovascular mortality compared with dialysis [5,6]. Historically, CVD has been the primary cause of death among kidney transplant recipients, accounting for over 30% of deaths in the 1990s [7,8]. Today, CVD remains the leading cause of death in this population [9,10]; however, its incidence has significantly decreased [11,12], likely due to advancements in primary and secondary prevention strategies as well as improvements in treatment and despite the older median age at the time of waitlisting and transplantation. Notable disparities exist between continents, with CVD-related deaths accounting for approximately 20% in Europe compared with 25–30% in the United States [9] and in Australia and New Zealand [10]. These discrepancies are mainly attributable to inherent differential cardiovascular risk between high-income countries, with a CVD-related mortality consistently higher in the United States [13]. Furthermore, a high rate of sudden deaths, potentially attributable to cardiovascular causes, could lead to an underestimation of these figures [14].
CVD after kidney transplantation encompasses various conditions including coronary artery disease (CAD), heart failure, cardiac arrhythmias, and stroke. In the FAVORIT study, prevalent CVD was reported in 20% of patients five years post-transplantation [15]. According to a recent UK study, the cumulative incidence of CVD in naive recipients was 5.4% at five years and 14.3% at ten years [16], while in the U.S., it was reported as 8.3% at five years [17]. Compared with the general population, the relative risk of cardiovascular death in recipients aged 40 to 50 was 46 [18]. Additionally, CVD places a significant burden on national healthcare systems, with a growing number of cases and a median cost for post-transplant CVD-related hospitalizations [19].
The primary cause of CVD-related morbidity and mortality in kidney transplant recipients is CAD. Numerous studies have highlighted the incidence of myocardial infarction, ranging from 11.1% at three years post-transplant [20] to 14% after five years [15]. However, these data primarily pertain to U.S. recipients. A post hoc analysis of the multinational PORT study reported a cumulative incidence of CAD of 3.1% at one year and 7.6% at five years. These discrepancies are largely attributed to a higher incidence of CAD in North American patients compared with European recipients (approximately 6% at five years) [21]. Among waitlisted patients undergoing transplantation, the prevalence of CAD is reported to be as high as 20% [22], although the impact of CAD screening on post-transplant mortality remains unclear [23].
Heart failure with reduced ejection fraction (HFrEF) prevalence is not well-defined in waitlisted patients [24], but baseline systolic dysfunction appears to be associated with increased mortality [25]. While left ventricular ejection fraction and other sonographic parameters often improve post-transplantation [26], Lentine et al. reported an HFrEF incidence of 18.3% after transplantation, which was associated with reduced patient survival. Unfortunately, there are no data on the incidence of heart failure with the preserved ejection fraction (HFpEF) in kidney transplant recipients. Atrial fibrillation (AF), the most common cardiac rhythm disorder, is detected in 6% of kidney transplant recipients at the baseline and is associated with increased mortality [27]. The cumulative incidence of AF after transplantation is approximately 7.3% [28]. Pulmonary hypertension is another frequent cardiovascular complication in kidney transplant recipients [29], with a prevalence of around 20% according to small retrospective cohorts [30,31]. One prominent cause may be high cardiac output secondary to arteriovenous fistulas [32]. Valvular calcifications are observed in approximately 30% of recipients after one year of follow-up [33]. Although kidney transplantation reduces hospitalizations for valvular heart disease compared with waitlisted patients, its effect on disease progression remains unclear [34]. In a recent French cohort, recipients with prosthetic heart valves had decreased overall survival, with only mechanical valves being independently associated with mortality compared with bioprosthetic valves. Transcatheter aortic valve implantation appears to be a safe option for recipients with high-grade aortic stenosis [35]. Ventricular arrhythmias are detected in up to 30% of recipients [36] and may contribute to the high rate of sudden death observed in this population [14].
Accelerated arteriosclerosis should also be considered as a part of post-transplant cardiovascular disease. Arteriosclerosis in kidney transplants is defined as a vascular fibrous intimal thickening, according to the Banff classification [37], progressing after kidney transplantation and distinct from baseline donor lesions [38]. The pathophysiology of transplant arteriosclerosis remains unclear but probably results from two main mechanisms: hypertension and other cardiovascular risk factors can indeed induce these types of lesions [39], but the most recent hypothesis incriminates the alloimmune response related to donor-specific antibodies. In this study by Loupy et al. [40], risk factors for arteriosclerosis were donor parameters, inducing baseline lesions, cold ischemia time, recipient hypertension requiring medical therapy, and circulating donor-specific antibodies. In the same study, transplant arteriosclerosis was associated with graft failure and cardiovascular events, especially in the presence of donor-specific antibodies.

2.1.2. Early Cardiovascular Events

The postoperative period carries a high risk of cardiovascular events, particularly CAD events and myocardial infarctions (MIs). Notably, Kasiske et al. demonstrated that the risk of MI was significantly higher in the postoperative period compared with waitlisted patients, with hazard ratios of 3.57 in deceased donor recipients and 2.81 in living donor recipients during the first three months post-transplant. Subsequently, the risk decreased, and transplantation became protective against MI [41]. In the study by Lentine et al. [20], the cumulative incidence of post-transplant MI was 4.3% at six months. CVD remains the leading cause of death in the initial months following kidney transplantation, with a mortality rate of 1.18 per 100 patient-years, as reported in the Australian study by Ying [10].
In an observational study by De Lima et al., 8.4% of recipients experienced a CAD-related event within the first year post-transplantation [42]. Similarly, Cheng et al., using Medicare data, reported incidences of 2.9% for MI and 2.6% for cardiac death [23]. An analysis of the U.S. Renal Data System found an MI incidence of 4.7% within the first month after transplantation [43]. Interestingly, Medicare data also showed a decline in the incidence of these outcomes over time. In Europe, the data are more limited, but the incidence of early coronary events appears to be lower. The PORT study reported a reduced incidence of such events [21], and a UK study using National Health Service data found the incidence of major adverse cardiovascular events (MACE) to be 0.9% within 90 days post-transplantation [44].
Despite these findings, each study indicated that pretransplant CAD screening, whether invasive or non-invasive, did not adequately predict or prevent early post-transplant coronary events. While risk factors for coronary events are well-defined [21], new strategies must be developed to prevent these early complications, which significantly contribute to the morbidity and mortality of newly transplanted recipients.

2.2. Cardiovascular Risk Factors

CVD in kidney transplant recipients is characterized by distinctive risk factors and atypical mechanisms. Both pre- and post-transplant factors contribute significantly to the development of heart failure, CAD, arrhythmias, and ultimately, cardiovascular mortality in this population. Traditional cardiovascular risk factors, such as diabetes, hypertension, dyslipidemia, and smoking, remain important but are exacerbated by transplant-specific factors. These include prolonged dialysis duration, impaired graft function, systemic inflammation from rejection episodes, post-transplant diabetes, and the effects of immunosuppressive therapies such as calcineurin inhibitors (e.g., tacrolimus-related diabetes risk and cyclosporine-induced hypertension) and corticosteroids (Figure 1).

2.2.1. Traditional Risk Factors

Age remains a prominent risk factor, as in the general population [45]. Despite its high prevalence in transplant recipients, hypertension remains a significant contributor to CVD. A post hoc analysis of the FAVORIT trial by Carpenter et al. described a linear relationship between systolic blood pressure and the occurrence of CVD and death, with no association observed for diastolic blood pressure [46]. Several studies also demonstrated that high blood pressure increased the risk of graft loss [47,48].
Sex differences in cardiovascular risk are less well-studied. Liu et al. used a propensity score-matched cohort to show that women had a lower risk of MACE among kidney transplant recipients aged 50 or older with a waiting time of less than six years [49]. The PORT study added that men were at higher risk of CAD [21].
Regarding dyslipidemia, Roodnat et al. found that elevated serum cholesterol negatively impacted graft and patient survival [50]. The extension of the ALERT study formally demonstrated that reducing LDL cholesterol with fluvastatin reduced long-term cardiac death or nonfatal MI by 29% [51,52].
Diabetes mellitus is a major risk factor in kidney transplant recipients. Post-transplant diabetes mellitus (PTDM) was reported to occur in 9.1% of recipients in the cyclosporine era and was associated with poorer kidney outcomes [53]. Tacrolimus use significantly increased PTDM risk [54], which in turn mediated a higher risk of cardiovascular events [55]. However, pretransplant diabetes [56] and impaired glucose tolerance [57] also play a critical role. In a Canadian cohort of 5000 recipients, pretransplant diabetes was associated with a higher risk of MACE than both euglycemic and PTDM patients [58]. Additionally, diabetic kidney disease independently increased the risk of cardiovascular death compared with other histologic subtypes in the USRDS analysis.
Tobacco use is a significant risk factor for cardiovascular events, with a dose-dependent relationship between pack-years at transplantation and cardiovascular outcomes [59]. Incident smoking post-transplantation is also associated with poorer graft outcomes [60].

2.2.2. Non-Classical Risk Factors

In addition to traditional risk factors, the PORT study identified several atypical risk factors specific to transplant recipients [21]. These include obesity, prolonged dialysis duration, and delayed graft function, which are associated with CAD events in the first year post-transplantation. Acute rejection, a panel reactive antibody > 10%, and graft dysfunction were linked to a higher risk of CAD within three years of follow-up.
Obesity exacerbates other risk factors such as diabetes and hypertension, with each five-unit increase in body mass index (BMI) raising the cardiovascular risk by 26% [61]. An increase in the waist-to-hip ratio post-transplantation is also associated with cardiovascular events [62], as is metabolic syndrome [63].
Dialysis duration is a critical factor, reflecting cumulative cardiovascular impairment, mineral bone disease, and calcium-phosphorus dysregulation. Each additional year of pretransplant dialysis increases the cardiovascular death risk by 14% [64]. A Swedish study of 889 recipients suggested that parathyroidectomy might reduce cardiovascular events incidence compared with medical therapy [65].
Delayed graft function (DGF) also impacts cardiovascular outcomes. Tapiawala et al. reported an increased rate of cardiovascular death among patients with delayed graft function [66].
High-flow arteriovenous fistula is associated with worse echocardiographic parameters [67], and fistula by itself is associated with heart failure after transplantation with a hazard ratio of 2.14 [68].
Acute rejection has been identified as an independent cardiovascular risk factor, even in the absence of graft dysfunction [21,69]. Similarly, proteinuria is associated with cardiovascular events [70], though its incidence remains unchanged despite the use of renin-angiotensin-aldosterone system (RAAS) blockers [71].
Kidney function directly influences cardiovascular outcomes. A post hoc analysis of the FAVORIT trial by Weiner et al. showed that each 5 mL/min/1.73 m2 decline in glomerular filtration rate (GFR) increased the MACE risk by 15%, with risks becoming more pronounced below a GFR of 45 mL/min/1.73 m2 [72].
Immunosuppressive therapy significantly contributes to cardiovascular risk. Calcineurin inhibitors are associated with dyslipidemia, hypertension, and PTDM [73], while corticosteroids contribute to bone mineral disease and metabolic syndrome. In contrast, belatacept was associated with lower PTDM rates [74], reduced proteinuria, and improved survival compared with calcineurin inhibitors [75]. mTOR inhibitors have not been proven to be effective in mitigating cardiovascular complications [73].
Other factors, including systemic inflammation [76] and left ventricular hypertrophy, also increase the cardiovascular risk. Left ventricular hypertrophy, often driven by anemia and hypertension, persists in some recipients despite improvement after transplantation [77,78].

2.3. Strategies for Screening for Coronary Artery Disease in Potential Kidney Transplant Recipient (PKTR)

2.3.1. Pretransplant Coronary Artery Disease Screening: Why?

The goal of pretransplant cardiac evaluation is to identify high-risk patients who may benefit from specific management strategies prior to transplantation, thereby reducing the risk of MACE and early cardiovascular death post-transplant. This evaluation also helps to pinpoint patients for whom transplantation may be deemed too risky or contraindicated. Screening for undiagnosed CAD is particularly important due to its high prevalence in CKD patients [79,80] and its role as the leading cause of post-transplant mortality [12].
CKD patients often present with atypical or absent symptoms of CAD [81] such as less frequent chest pain compared with the general population [82,83]. This discrepancy is partially attributed to the high prevalence of diabetes and uremic neuropathy in this population. However, the benefits of screening must be weighed against the significant risks associated with invasive procedures like coronary angiography, which can lead to complications including hematoma at the puncture site, arterial injury, stroke, MI, and sudden cardiac arrest.
Recently, the European Society of Cardiology (ESC) 2024 guidelines for managing chronic coronary syndromes in the general population [84] introduced a stratification model categorizing patients into three risk groups, each benefiting from tailored screening approaches. However, CKD patients exhibit unique characteristics, such as a higher prevalence of CAD and elevated pre- and post-transplant cardiovascular risks, which diminish the applicability of these standard recommendations to the KTR population. Additionally, the performance of common screening tools is often suboptimal in CKD patients.
Unlike other surgical procedures, kidney transplantation carries the additional risk of graft dysfunction, particularly in the context of organ shortage. Consequently, the primary focus of pretransplant CAD screening should be on identifying patients who genuinely require optimization to reduce perioperative and long-term cardiovascular risks. At the same time, it is crucial to minimize unnecessary risks, delays, and costs for patients unlikely to benefit from intervention.

2.3.2. Performance of the Main Available Tests in the CKD Population

Coronary CT angiography (CCTA) has proven diagnostic value in the general population but is less reliable in ESRD patients due to extensive coronary calcifications. A recent meta-analysis reported a sensitivity of 96% and specificity of 66% for detecting significant coronary stenosis [85], but also highlighted a high risk of false positives [86]. The development of computed tomographic fractional flow reserve (FFR-CT), a physiological simulation technique using routine CTA data to model coronary flow, pressure, and resistance, may improve diagnostic accuracy. This method helps assess the functional significance of coronary lesions, especially in cases where calcifications compromise CCTA interpretation [87]. FFR-CT has been significantly associated with post-transplant MACE. While a normal CCTA can reassure against macrovascular disease in patients with lower CAD likelihood, it does not address microvascular conditions. Conversely, a positive result may necessitate additional ischemia testing, often leading to delays and increased costs.
Coronary artery calcium scoring (CACS) uses adjusted thresholds for ESRD and dialysis-dependent ESRD populations. While helpful, its diagnostic performance is limited when used alone, with a sensitivity of 67% and specificity of 77% [86]. Combined with clinical data (e.g., fewer than three risk factors) and a CACS cutoff of <400, it offers a strong negative predictive value for CAD and correlates with the risk of MACE [88]. Dahl et al. [89] found that high-risk plaques and CACS were associated with MACE, but not with mortality, although calcified and low-attenuation plaque burdens were independently associated with both outcomes. Combining CACS with CCTA can improve the diagnostic accuracy. However, clinicians remain hesitant to use CCTA routinely, given concerns about radiation exposure and the potential toxicity of iodine contrast agents [90].
Stress cardiovascular magnetic resonance (CMR) is an effective tool for detecting myocardial ischemia but must be used cautiously in patients with ESRD due to potential risks associated with gadolinium-based contrast agents (GBCAs) [91]. While newer macrocyclic GBCAs may offer a safer alternative, their widespread use and validation in this population remain limited. Emerging techniques such as native T1 mapping and arterial spin labelling are currently under investigation and may provide contrast-free alternatives [92]. Dobutamine stress CMR, a non-contrast approach, has demonstrated high diagnostic accuracy for detecting CAD in potential kidney transplant recipients (PKTRs) [93,94]. However, large-scale studies are still needed to establish its role within broader screening strategies, particularly given the considerations around its cost and accessibility.
Single photon emission computed tomography (SPECT) provides functional assessment by evaluating myocardial perfusion and viability. However, diagnostic performance in CKD patients is limited due to factors such as triple-vessel disease, left ventricular hypertrophy (LVH), fibrosis, endothelial dysfunction, and volume overload [95]. Sensitivity and specificity for detecting stenosis >70% have been reported at 67% and 77%, respectively [96,97]. Despite these limitations, SPECT remains commonly used due to its accessibility, low risk, and strong predictive value for MACE [98,99].
Stress echocardiography (SE) offers advantages like accessibility, affordability, and the absence of radiation exposure. It provides a comprehensive evaluation of myocardial kinetics, blood flow, and valve function. However, diagnostic performance can be affected by concentric remodeling and eccentric hypertrophy, which may lead to false negatives [100]. SE shows a sensitivity of 76% and specificity of 88% for detecting CAD with stenosis >70% [96]. Compared with SPECT, SE demonstrates superior diagnostic and prognostic value and is recommended as a first-line test by the AHA 2022 statement. However, test selection should consider factors such as patient echogenicity, exercise capacity, and conditions like arrhythmia or hypo-/hypertension. Notably, SE and SPECT are comparable to coronary angiography in predicting all-cause mortality and MACE in kidney transplant recipients [101,102].
Positron emission tomography (PET-CT) is emerging as the most promising non-invasive diagnostic tool in the general population [103], offering lower radiation exposure and shorter test duration compared with SPECT. PET-CT not only evaluates CAD, but also assesses coronary flow reserve and myocardial blood flow (MBF), making it the gold standard for the non-invasive assessment of coronary microvascular dysfunction [104]. A recent single-center study compared PET-CT and SPECT in 393 pretransplant patients, finding PET-CT significantly associated with post-transplant MACE (hazard ratio [HR] 3.02 vs. HR 1.39 for SPECT) [105]. Although PET-CT’s diagnostic performance in the PKTR population has not been specifically studied, its promising results in the general population suggest similar utility. However, PET-CT’s high cost and limited availability warrant further research to define its role in pretransplant evaluation.
To enhance discrimination, imaging can be combined with biomarkers like high-sensitivity cardiac troponin T (hs-cTnT), which shows promise even in ESRD patients [106]. A recent study by Szramowska et al. evaluated 100 patients undergoing pre-kidney transplant screening with kinetic abnormalities, positive scintigraphy, or SE [107]. Testing for hs-cTnT and coronary angiography revealed significant stenosis in 50% of patients. An hs-cTnT cutoff value of 0.069 ng/mL demonstrated 61.4% sensitivity and 82.2% specificity for diagnosing obstructive CAD. Incorporating an age threshold of 52 years, the study proposed an algorithm where patients under 52 with hs-cTnT < 0.069 ng/mL could forgo further CAD evaluation, while those older or with hs-cTnT > 0.069 ng/mL would be referred for coronary angiography. Although this strategy may reduce unnecessary coronary angiographies, the study’s small, single-center design requires further validation.
Finally, commonly used risk scores, such as the Revised Cardiac Risk Index (RCRI) for 30-day perioperative cardiovascular risk [108] and the Framingham risk score for post-transplant MACE, show limited predictive value in CKD patients as they do not account for transplant-specific risk factors [109].

2.3.3. Which Screening Strategy to Choose?

Screening strategies for asymptomatic patients on the kidney transplant waiting list have evolved significantly over the decades. According to the latest AHA 2022 statement [90], KTR presenting with symptoms or clinical signs of angina, heart failure, significant ischemia on ECG, left ventricular ejection fraction (LVEF) < 40%, kinetic abnormalities, arrhythmias, or valvular disease are classified as high risk. These patients should be referred directly for specialized cardiac evaluation, with coronary angiography considered as a first-line investigation.
For asymptomatic patients, new data now allow for the better identification of individuals who would benefit from revascularization. All candidates should undergo a minimum evaluation including a comprehensive risk factor assessment, symptom inquiry, ECG, and echocardiography. Notably, the KDIGO 2020 Guidelines [110] recommend echocardiography only for asymptomatic patients who have been on dialysis for more than two years and have risk factors for pulmonary hypertension. Despite these guidelines, echocardiography remains a widely used screening tool due to its affordability, accessibility, and absence of associated risks.
Following this initial evaluation, patients with no significant abnormalities on ECG or echocardiography—defined as no valvular disease, LVEF >40%, and no kinetic abnormalities—can be categorized as low risk. Low-risk criteria include being under 60 years of age, no diabetes, no history of cerebrovascular or peripheral artery disease, and less than five years on dialysis. These patients generally do not require further evaluation.
Patients who do not meet the low-risk criteria are classified as intermediate risk and should proceed with additional screening. Patients with a history of known CAD are treated as higher risk compared with those without CAD, and their management depends on prior coronary angiogram findings, LVEF, and clinical signs. Based on this assessment, a strategy involving either optimized medical treatment or non-invasive ischemia testing (+/− coronary angiography) can be indicated (Figure 2).
The AHA 2022 statement was evaluated in a retrospective observational study [111], which demonstrated that potential kidney transplant candidates (PKTCs) who were not recommended for further screening had a low revascularization rate and fewer adverse outcomes. However, the 2022 algorithm directed a higher percentage of patients for cardiac referral or screening compared with the AHA 2012 algorithm (73% vs. 53%).
Regarding ischemia screening, the AHA 2022 statement does not specify a gold standard test. However, they appear to favor stress echocardiography, likely due to its cost-effectiveness and potentially superior specificity compared with other imaging modalities.

3. Management of Post-Transplant Cardiovascular Risk

3.1. Treatment of Traditional Risk Factors in Kidney Transplant Recipients

Managing traditional risk factors in KTR is crucial and differs significantly from approaches used in chronic kidney disease (CKD) and dialysis patients (Table 1).
Hypertension: Calcium channel blockers are considered first-line treatment for hypertension in KTRs, as they have been shown to reduce the risk of graft loss (relative risk [RR] 0.75) compared with angiotensin-converting enzyme inhibitors (ACEis) [112]. However, ACEis should be prioritized in cases with significant proteinuria, given their renoprotective effects. Angiotensin receptor blockers (ARBs), beta-blockers, and diuretics are also valuable options depending on the patient’s clinical profile. The American Heart Association (AHA) recommends a target blood pressure of less than 130/80 mmHg for KTRs, as established in 2018 [113].
Dyslipidemia: Dyslipidemia should be addressed in all KTRs following the KDIGO 2013 Guidelines on lipid management [114]. In the ALERT study, fluvastatin therapy reduced the LDL cholesterol levels by 32% and achieved a significant 21% reduction in long-term MACE occurrence [51,52]. Ezetimibe can be used as a second-line therapy in combination with statins [115]. For hypertriglyceridemia, drug treatment is not currently recommended as per the KDIGO Guidelines. LDL cholesterol targets for KTRs are generally aligned with those for the general population and depend on cardiovascular risk. According to the ESC 2019 Guidelines, KTRs fall into high or very high-risk categories. Most recipients should aim for an LDL target of <0.7 g/L, while those at very high risk (e.g., GFR < 30 mL/min/1.73 m2, history of CVD, or diabetes with organ damage) should target <0.55 g/L [116]. Clinicians should note that cyclosporin is related to an increased systemic exposure to statins through interaction with cytochrome P450 3A4, p-glycoprotein, and organic anion transport polypeptide. However, the impact of tacrolimus on statin metabolism is less clear [117,118]. The American Heart Association still recommends reducing statins to the minimum available dosage in patients treated with calcineurin inhibitors, except for pravastatin and fluvastatin [119].
Diabetes: For pretransplant diabetes, insulin therapy is often necessary, particularly in the perioperative period [120]. Given their proven benefits in reducing cardiovascular risk [121] and slowing CKD progression [122], SGLT2 inhibitors should be considered first-line therapy for diabetes management in KTRs [123,124]. GLP-1 receptor agonists are another first-line option, supported by recent studies demonstrating their cardiovascular benefits [125,126] and tolerability in KTRs [127]. Additional options include metformin (with caution in patients at risk of lactic acidosis) and DPP-4 inhibitors. The KDIGO 2009 Guidelines recommend an HbA1c target of 7–7.5% in KTRs and advocate for aspirin use in secondary prevention or in primary prevention for recipients at high cardiovascular risk [128]. Furthermore, strategies such as early corticosteroid discontinuation [129] and switching to belatacept [74] may improve diabetes management and outcomes.
Smoking: Smoking cessation is a critical concern in KTRs. Smoking increases the risk of graft failure, cardiovascular events, and malignancies. Smoking cessation should be prioritized, and patients should be encouraged to quit during every clinical visit. First-line therapies include structured cessation programs and nicotine replacement therapies [130,131]. Second-line options include varenicline, which requires a dose adjustment based on GFR and bupropion, which should be used cautiously due to seizure risks.
Table 1. Management of traditional cardiovascular risk factors among kidney transplant recipients.
Table 1. Management of traditional cardiovascular risk factors among kidney transplant recipients.
Modality of ScreeningTiming of ScreeningTreatmentTargetGuidelines
HypertensionHome blood pressure self-monitoring and measurement at office visit.Blood pressure measurement at every visit
  • First-line: Calcium channel blockers, particularly after transplantation.
  • Consider ARBs or ACE inhibitors if proteinuria is present.
  • Alternative options: Thiazide diuretics, loop diuretics, beta-blockers.
  • BP < 130/80 mmHg
  • For older recipients, consider < 140/90 mmHg.
AHA 2017 [113]
DyslipidemiaMeasurement of
LDL-C, HDL-C, and triglycerides
Annually
  • All kidney transplant recipients should be treated with statins (per KDIGO 2013 Lipid Guidelines). Start with a low dose due to interactions with CNIs.
  • Add ezetimibe to enhance LDL-C lowering if needed.
  • For hypertriglyceridemia: Lifestyle changes recommended.
  • Most patients are at high or very high cardiovascular (CV) risk.
  • LDL-C target < 0.7 g/L for patients with GFR < 30 mL/min/1.73 m2, history of CVD, or diabetes with organ damage.
  • LDL-C target < 0.7 g/L in other transplant recipients.
KDIGO 2013 [114]
2019 ESC/EAS Guidelines [116]
DiabetesFasting plasma glucose, HbA1c, or oral glucose tolerance testingWeekly during the first month, then at months 3, 6, 9, and 12, followed by annual assessments or when increasing the CNI dose.
  • Early initiation of insulin.
  • Consider first-line therapies: SGLT2 inhibitors or GLP-1 receptor agonists.
  • Other options: Metformin (if eGFR > 30 mL/min), DPP-4 inhibitors.
  • Aspirin for secondary prevention or primary prevention in patients at high CV risk.
  • Consider early corticosteroid discontinuation or a switch to belatacept to prevent post-transplant diabetes.
HbA1c 7–7.5%KDIGO 2009 [128]
SmokingDiscuss tobacco use at each visit and assess whether the patient is willing to quit or needs assistance.At every visit
  • Enroll patients in a smoking cessation program.
  • Consider smoking cessation therapies:
  • First-line: Nicotine replacement therapy.
  • Second-line: Varenicline (dose-adjusted for GFR) or bupropion (note seizure risk when used with cyclosporine).
Smoking cessationKDIGO 2009 [128]

3.2. Treatment of Non-Classical Risk Factors Management in Kidney Transplant Recipients

Several non-classical risk factors, including obesity, PTDM, proteinuria, and bone mineral disease, play a significant role in the cardiovascular outcomes of KTRs and are amenable to treatment.
Obesity: Although obesity is not a contraindication to kidney transplantation, it is associated with worse outcomes. Additionally, new transplant recipients are prone to post-transplant weight gain, primarily due to corticosteroid use, which further exacerbates cardiovascular risk [132]. Addressing obesity and weight gain involves lifestyle modifications, and bariatric surgery may be considered when necessary [133]. However, bariatric surgery has limitations, including the risk of enteric hyperoxaluria and nephrolithiasis, necessitating the development of alternative strategies to mitigate these risks. Emerging therapies, particularly GLP-1 receptor agonists like semaglutide, have shown promise in reducing cardiovascular risk and promoting weight loss in obese patients without diabetes [134], but evidence in KTRs remains limited.
Post-transplant diabetes mellitus (PTDM): Managing PTDM involves the early initiation of insulin therapy [120], standard diabetes treatments such as GLP-1 agonists and SGLT2 inhibitors, and optimizing immunosuppressive regimens. Switching to belatacept [74] or discontinuing corticosteroids [129] may also be beneficial in selected cases to improve glycemic control and reduce cardiovascular risk.
Proteinuria: Proteinuria in KTRs can be managed with RAAS blockers, although these drugs have not demonstrated significant benefits on graft or patient survival [71,135]. SGLT2 inhibitors offer potential benefits but lack robust data in the transplant population [122].
Bone mineral disease: Hyperparathyroidism is a leading contributor to bone mineral disease post-transplantation. Parathyroidectomy remains the gold standard treatment and has been shown to reduce cardiovascular events [65]. Medical treatments, such as cinacalcet, can lower the parathyroid hormone levels, but their effects on cardiovascular risk are unclear [74].
Graft dysfunction and acute rejection: Both graft dysfunction and acute rejection are significant risk factors for cardiovascular complications. Their management should adhere to standard practices including appropriate immunosuppressive regimens and vigilant monitoring [136,137,138].

3.3. The Role of Lifestyle Modifications

According to the KDIGO 2009 Guidelines, kidney transplant recipients should follow a “healthy lifestyle, with exercise, proper diet, and weight reduction as needed” [128]. These recommendations are obviously adapted from those concerning the general population, according to the 2019 ACC/AHA Guidelines, lifestyle factors can indeed influence and reduce cardiovascular risk in the general population [139]. Concerning nutrition and diet, patients should prioritize vegetables such as fruits, legumes, nuts, and whole grains as well as fish intake. Saturated fat should be replaced by mono- or polyunsaturated fats. The amount of cholesterol and salt consumption should be reduced. A high potassium intake could also be encouraged in the general population but should be carefully evaluated in kidney transplant recipients. Finally, as part of a healthy diet, the intake of trans fats, processed meats, refined carbohydrates, and sweetened beverages should be minimized. Patients should also be encouraged, if able, to routinely perform at least 150 min of moderate-intensity or 75 min of high-intensity physical activity per week. Lifestyle changes also imply weight loss, tobacco weaning, and alcohol avoidance. They participate in a global reduction in cardiovascular disease in the general population, mainly through the reduction in blood pressure and LDL-cholesterol levels, and the improvement in glycemic control. They also participate in reducing the BMI and are the baseline treatment for obesity [139]. The 2019 KDIGO Guidelines also state that there is no reason to believe that a healthy lifestyle and a proper diet do not prevent cardiovascular complications as in the general population [128].
Several randomized control trials have analyzed the impact of physical activity in the field of kidney transplantation, but are globally underpowered to show a real difference in the improvement in cardiovascular risk factors. Most of these are centered on the improvement in physical capacity and kidney function. A meta-analysis by Zhang et al. concerning exercise intervention in kidney transplant recipients and including 16 randomized control trials revealed a positive impact of exercise on renal function and physical performance, with a lower creatinine and improved VO2 peak and 6-min walk test [140]. However, a randomized control trial of 96 patients did not show significant improvement in cardiovascular risk, evaluated according to the Framingham equations, in the exercise arm in the first year after transplantation [141].
Concerning the impact of dietary changes, they were associated in several clinical trials to better kidney function and overall survival. In a randomized clinical trial including 632 kidney transplant recipients, the Mediterranean diet was associated with better graft function [142]. Another trial including 632 recipients determined that the DASH diet was associated with better graft function and lower all-cause mortality [143]. However, a recent systematic review regarding the dietary interventions for body weight management did not demonstrate any effect due to a lack of significant studies in chronic kidney disease [144] and kidney transplant recipients [145]. Unfortunately, the impact of dietary changes on specific cardiovascular outcomes are rarely depicted in the literature. In summary, if some specific diet styles are associated with better outcomes, the impact of dietary interventions remains to be proven.
The proper impact of physical activity and dietary changes remains unclear on specific cardiovascular risk factors and outcomes in kidney transplant recipients, mostly due to unadapted designs in clinical studies. However, due to their large beneficial impact in the general population, and based on the available literature, we can conclude that lifestyle changes are formally indicated in all kidney transplant recipients, regardless of their cardiovascular risk.

3.4. RAAS Inhibitors in Kidney Transplant Recipients

RAAS inhibitors, including ACEis and ARBs, are widely recognized for their role in reducing proteinuria, controlling hypertension, and slowing the progression of CKD. In KTRs, the use of RAAS inhibitors is particularly compelling due to the high prevalence of hypertension and proteinuria, both of which are associated with poor graft outcomes and increased cardiovascular morbidity. However, the utility of RAAS inhibitors in improving long-term graft survival and overall patient outcomes remains debated due to conflicting evidence from clinical trials and observational studies.
Proteinuria is a strong predictor of graft loss in KTRs. RAAS inhibitors effectively reduce proteinuria by decreasing intraglomerular pressure through efferent arteriolar vasodilation. This mechanism not only reduces protein excretion, but also mitigates glomerular damage. For example, Brenner et al. demonstrated in an RCT involving 1513 patients with diabetic nephropathy that losartan reduced proteinuria by 35% and cardiovascular events by 16%, highlighting its renoprotective and cardioprotective effect [146]. Similarly, in an RCT by Midtvedt et al., lisinopril effectively reduced proteinuria in hypertensive KTRs, though nifedipine demonstrated a more pronounced improvement in GFR, emphasizing the nuanced effects of different antihypertensive strategies [147].
Observational studies have also explored the impact of RAAS inhibitors on the long-term outcomes of KTRs. Heinze et al. reported a significant 35% reduction in graft loss risk (HR 0.65) and a 39% reduction in mortality (HR 0.61) in KTRs treated with RAAS inhibitors [148]. These findings suggest a beneficial effect on both graft longevity and patient survival, likely mediated by antiproteinuric and antihypertensive mechanisms. However, contrasting results were presented by Opelz et al. in the Collaborative Transplant Study (CTS), which analyzed 17,209 KTRs and found no significant improvements in graft survival (HR 1.05) or patient survival (HR 1.01) among those receiving ACEis or ARBs. Subgroup analyses of high-risk patients, such as those with diabetic nephropathy or hypertension, also failed to demonstrate meaningful benefits, raising questions about the generalizability of positive findings [149].
Further complicating the narrative are the results from more recent investigations, such as Knoll et al., which evaluated ramipril in a multicenter RCT of 213 KTRs with proteinuria. The study found no significant differences in key outcomes, including doubling of serum creatinine, ESRD, or mortality, compared with the placebo. Additionally, adverse effects such as hyperkalemia were more common in the ramipril group, underscoring the potential risks of RAAS blockade [150]. Similarly, Cheungpasitporn et al., in a meta-analysis of 20,024 KTRs, concluded that RAAS inhibitors offered no significant reduction in allograft loss or mortality, emphasizing the heterogeneity of the study designs and patient populations [151].
Despite these challenges, the potential cardioprotective and anti-inflammatory effects of RAS inhibitors remain of interest. Kovarik et al. demonstrated that ACEis suppressed systemic angiotensin II levels and promoted the alternative RAAS pathway, generating anti-inflammatory peptides like angiotensin-(1–7). These effects could theoretically mitigate chronic allograft nephropathy and cardiovascular complications. However, the phenomenon of “ACE escape”, where intrarenal angiotensin II synthesis persists despite systemic RAAS blockade, raises questions about the long-term efficacy of these agents [152].
In conclusion, while RAAS inhibitors effectively reduce proteinuria and may offer cardiovascular protection (Table 2), their role in improving graft and patient survival in kidney transplant recipients remains inconclusive. The conflicting evidence underscores the need for individualized treatment approaches, the careful monitoring of potential side effects, and the identification of patient subgroups most likely to benefit. Future research, particularly well-designed, large-scale RCTs, is essential to clarify the long-term benefits and risks of RAAS inhibitors in this unique population. For now, their use should be guided by patient-specific factors, with an emphasis on balancing the potential benefits with the known risks.
Table 2. Summary of key studies on RAS inhibitors in kidney transplant recipients.
Table 2. Summary of key studies on RAS inhibitors in kidney transplant recipients.
Study (Year)Population (N)DesignKey FindingsReference
Midtvedt et al. (2001)72RCTNifedipine improved GFR by 20%; lisinopril reduced proteinuria.[147]
Heinze et al. (2006)1513Observational cohortRAS inhibitors reduced graft loss risk (HR 0.65) and mortality (HR 0.61).[148]
Opelz et al. (2006)17,209Observational cohortNo significant improvement in graft (HR 1.05) or patient survival (HR 1.01).[149]
Knoll et al. (2016)213RCTNo significant effect on ESRD, mortality, or creatinine doubling in KTRs.[150]
Cheungpasitporn et al. (2016)20,024Meta-analysisNo reduction in allograft loss or mortality; heterogeneous study designs.[151]
Kovarik et al. (2019)48Cross-sectionalACEis promoted anti-inflammatory RAS pathways but showed intrarenal escape.[152]
RAS: renin-angiotensin system, ACEis: angiotensin-converting enzyme inhibitors, ARBs: angiotensin receptor blockers, RCT: randomized controlled trial, KTRs: kidney transplant recipients, GFR: glomerular filtration rate, ESRD: end-stage renal disease.

3.5. Emerging Therapies to Manage Cardiovascular Risk

Emerging therapies for cardiorenal protection in kidney transplant recipients, including SGLT2 inhibitors, finerenone, and semaglutide, represent promising strategies to mitigate the high cardiovascular and renal risks in this population. SGLT2 inhibitors, such as empagliflozin and dapagliflozin, have shown significant benefits in improving glycemic control, reducing albuminuria, and preserving allograft function. They achieve this by reducing glomerular hyperfiltration and intraglomerular pressure while exerting anti-inflammatory and antifibrotic effects [153,154].
A pivotal study leveraging the TriNetX database has provided compelling real-world evidence regarding the efficacy of SGLT2 inhibitors in diabetic kidney transplant recipients (KTRs). This study analyzed a cohort of 50,120 diabetic KTRs, and after propensity score matching, compared the outcomes between 1970 SGLT2 inhibitor users and an equal number of matched non-users with a median follow-up of 3.4 years. SGLT2 inhibitor users exhibited a remarkable reduction in all-cause mortality (adjusted hazard ratio [aHR]: 0.32, p < 0.001), MACE (aHR: 0.48, p < 0.001), and major adverse kidney events (MAKE; aHR: 0.52, p < 0.001) [123]. Further subgroup analyses revealed that these benefits extended across various demographic and clinical profiles including age, sex, baseline kidney function, and comorbidity burden. Importantly, the study also confirmed the safety profile of SGLT2 inhibitors in this population, with no significant increase in genitourinary infections or acute kidney injury. The observed reduction in albuminuria and attenuation of diabetic nephropathy progression underscores the nephroprotective potential of these agents [123]. However, these results should be interpreted with caution, since only 44% of the participants in this study were treated using RAAS inhibitors. All randomized controlled trials performed in non-transplanted CKD patients [121,122] compared the RAAS blockers-SGLT2 inhibitors’ association with RAAS blockers alone, and a similar design should be applied to the transplant population. It is rather reassuring that no significant difference in the effect size of the SGLT2 inhibitors was described in this study between the subgroups with or without RAAS inhibitor treatment.
These findings strongly advocate for the integration of SGLT2 inhibitors into the therapeutic regimen for diabetic KTRs. With their demonstrated ability to significantly reduce mortality and adverse cardiac and kidney events, these agents hold transformative potential for improving the post-transplant outcomes. Future studies, including randomized controlled trials, are essential to further elucidate the long-term safety and efficacy of these agents in diverse KTR populations.
Finerenone, a nonsteroidal mineralocorticoid receptor antagonist, offers additional nephroprotection by mitigating inflammation and fibrosis. Preclinical and early clinical data suggest that it may prevent calcineurin inhibitor nephrotoxicity and improve cardiovascular outcomes in KTRs [155]. Meanwhile, semaglutide, a GLP-1 receptor agonist, has demonstrated weight reduction and glycemic control benefits in KTRs with diabetes or metabolic syndrome, with no significant adverse impact on graft function in small studies. Its cardioprotective and nephroprotective effects make it a valuable addition to the therapeutic arsenal [156].
Together, these therapies hold the potential to reshape the management of KTRs, improving graft longevity and patient survival while addressing the dual challenges of cardiovascular and renal disease. However, further randomized controlled trials are needed to validate their long-term safety and efficacy in this unique population [157].

3.6. Challenges of Immunosuppression from a Cardiovascular Perspective

3.6.1. T-Cells and Their Cardiovascular Impact

While rejection is clearly associated with an increased cardiovascular risk [158], the role of T-cell immunosuppression in this context remains a matter of debate in the literature. T lymphocytes are central to the pathogenesis of hypertension, vascular remodeling, and atherosclerosis [159,160]. The impact of specific T-cell subtypes is an area warranting further investigation. For instance, low CD4 counts, as observed in HIV patients, have been linked to elevated cardiovascular risk [161]. Among the T-cell subtypes, CD4 Th1 T-cells and CD8 T-cells appear to promote atherosclerosis, whereas Th2 T-cells exert protective effects [162]. Similarly, regulatory T-cells (T-Reg) not only provide protection against atherosclerosis, but may also mitigate vascular lesions following cardiac or kidney injury [159,160,162,163,164].
The potential cardiovascular impact of T-cell-depleting induction therapies in transplant recipients has been highlighted, particularly in a small retrospective study [165]. These therapies are thought to shift T-cell subsets, reducing the CD4 T-cell counts while promoting the expansion of late-stage differentiated CD8 T-cells, which may contribute to cardiovascular risk [166]. However, larger-scale observational data, such as a cohort of 162,998 KTRs from the United States Renal Data System (USRDS), provide a more reassuring perspective, suggesting no significant cardiovascular harm associated with T-cell depletion [167].
Despite these findings, the cardiovascular effects of T-cell modulation remain an unresolved issue. Further research is essential to clarify the mechanisms through which T-cell subsets influence cardiovascular outcomes in transplant recipients and guide clinical strategies for balancing immunosuppression with cardiovascular risk management.

3.6.2. Off-Targets Adverse Effects of Maintenance Therapy: A Personalized Strategy

Maintenance therapy in solid organ transplantation typically relies on a regimen centered on calcineurin inhibitors (CNIs) combined with mycophenolate mofetil (MMF) and corticosteroids [128]. The cardio-renal off-target effects of these immunosuppressive therapies are summarized in Table 3.

3.6.3. Addressing CNI Toxicity

While CNIs are highly effective in preventing rejection, they are associated with significant off-target effects, particularly metabolic complications such as hypertension, diabetes, and dyslipidemia [168]. Despite their experimental mitochondrial protective effects on ischemia-reperfusion injury [169], their cardiotoxicity and endothelial toxicity are increasingly recognized. These toxicities contribute to accelerated atherosclerosis, cardiac remodeling, and chronic cardiovascular-kidney disorder (CCKD), especially in kidney transplant recipients (KTRs) [168,170,171].
Therapeutic drug monitoring (TDM) remains essential for personalizing tacrolimus dosages to optimize immunosuppressive efficacy while minimizing adverse effects [6]. Emerging biomarkers, such as the torque teno virus levels, show promise for guiding immunosuppression and enhancing therapeutic precision [172].

3.6.4. mTOR Inhibitors: A Perilous Alternative

Experimental studies suggest that mTOR inhibitors may slow the progression of atherosclerosis, although they frequently induce dyslipidemia [73]. While mTOR inhibitors may address allograft vasculopathy in heart transplant recipients [173], their safety and efficacy as CNI replacements in kidney transplantation remain questionable. A meta-analysis of 2323 patients showed a nonsignificant trend toward increased new-onset diabetes after transplantation (NODAT) and significant increases in hypercholesterolemia, acute rejection, proteinuria, and anemia [73].

3.6.5. Belatacept: A Promising Alternative

Belatacept, a costimulation blocker, has emerged as a promising alternative to CNIs in kidney transplantation. The BENEFIT study demonstrated belatacept’s advantages in improving patient and graft survival while preserving renal function and slowing the decline in GFR [174]. Experimental studies also suggest that belatacept attenuates progressive renal injury, pressure overload-induced heart failure, and accelerated atherosclerosis [175,176,177].
In clinical trials, belatacept was associated with fewer cardiac disorders than cyclosporine (2% vs. 12%) in a five-year extension study [178]. In the seven-year extension, serious vascular disorders were less frequent with belatacept (3.5%) compared with cyclosporine (5.4%) [179]. Additionally, belatacept, alone or combined with minimized-dose tacrolimus, reduced the incidence of PTDM compared with tacrolimus alone (1.7%, 2.2%, and 3.8%, respectively) [74]. These findings suggest belatacept as a valuable strategy, particularly for high-risk cardiovascular patients, warranting further clinical trials to refine its use, but weighed against the risk of infection in some patients.

3.6.6. Early Steroid Withdrawal: A Beneficial Strategy for Select Patients

Corticosteroid withdrawal after kidney transplantation is a common strategy in the United States, with approximately 30% of patients following a steroid-free regimen one year post-transplant [180]. A five-year randomized controlled trial showed reduced PTDM incidence and improved lipid profiles with early steroid withdrawal (ESW), though there was no significant impact on cardiovascular mortality. However, ESW increased biopsy-proven cellular rejection, albeit without affecting graft survival [181].
A retrospective study in deceased-donor KTRs reported better overall survival, graft survival, and fewer cardiac, metabolic, and bone complications in patients receiving reduced corticosteroid doses [182]. Conversely, a large U.S. registry found that in patients with DGF, corticosteroid discontinuation was associated with higher rejection rates and worse graft survival [183].
Bae et al. analyzed 210,086 adult deceased-donor KTRs, including 26,248 retransplant recipients, and reported mixed results. Among all recipients, ESW had no significant difference in acute rejection, a slightly higher hazard of graft failure, and slightly lower mortality compared with continuous steroid maintenance. However, retransplant recipients experienced higher acute rejection and graft failure risks with ESW [184].
A propensity score-matched study of 304 older KTRs (≥65 years) revealed that ESW significantly reduced the PTDM rates at one and three years compared with continuous steroid immunosuppression (22.36% vs. 30.37% and 34.89% vs. 44.29%, respectively). ESW also correlated with lower myocardial infarction rates at five years (6.75% vs. 14.39%) and fewer infection-related complications, suggesting improved patient and graft survival in this population [185].
While ESW shows promise for carefully selected patients, identifying reliable biomarkers and conducting targeted studies are urgently needed to refine corticosteroid minimization strategies in kidney transplantation.

3.7. Vascular Access Management After Transplantation: What Is the Best Option?

The creation of an arteriovenous fistula (AVF) for hemodialysis is a cornerstone in the management of ESRD [186]. While it is preferred for its long-term durability and lower risk of infection compared with central venous catheters or grafts, AVF creation significantly impacts the cardiovascular dynamics and poses unique challenges for KTRs [187].

3.7.1. Cardiovascular Implications of AVF

AVFs, due to their high-flow, low-resistance nature, increase the venous return and cardiac output [32]. This additional strain on the heart can exacerbate pre-existing cardiovascular conditions. Increased cardiac output and pressure changes can contribute to pulmonary hypertension and left ventricular hypertrophy, while heightened preload and afterload may worsen heart failure, particularly in patients with reduced left ventricular function [188,189]. Additionally, elevated myocardial oxygen demand may exacerbate ischemia in individuals with coronary artery disease, further amplifying cardiovascular risk [190].
Notably, some of these cardiovascular effects may be reversible with AVF flow reduction in carefully selected patients. Studies have shown that reducing AVF flow significantly decreases the left ventricular mass, end-diastolic volume, end-diastolic diameter, and left atrial volume, with corresponding improvements in diastolic function. Reductions have also been observed in right ventricular diameter and right atrial volume, alongside a notable decrease in the estimated pulmonary artery systolic pressure [191,192].

3.7.2. AVF Management Post-Transplantation

The management of AVFs after kidney transplantation is a topic of ongoing debate (Table 4). Approximately 50% of KTRs return to dialysis within a decade, highlighting the importance of maintaining AVF patency [192]. A retrospective study in Slovenia involving 626 KTRs found that 23.9% used AVF for delayed graft function, 8.4% for graft failure, and 5.4% for plasma exchanges. However, 29% experienced AVF-related complications including aneurysms, thrombosis, high-flow AVFs (HF-AVFs), distal hypoperfusion, venous hypertension, arm trauma, or pain [193]. Another study by Manca et al. reported spontaneous AVF closure in 167 out of 365 KTRs, while 42 patients required surgical intervention due to complications, emphasizing the risk of spontaneous thrombosis post-transplantation [194].
Table 4. HF-AVF management and cardiovascular and kidney outcomes.
Table 4. HF-AVF management and cardiovascular and kidney outcomes.
StudyDesignPatients (N)
Period
InterventionEchographic FindingsClinical OutcomesRef.
Stoumpos et al.Observational study1330
(2010–2020)
None, analysis of existing AVFsIncreased left ventricular mass, elevated cardiac outputHigher risk of new-onset heart failure post-transplant (adjusted Hazard Ratio 2.14)[68]
Rao et al.Randomized controlled trial64
(2013–2017)
AVF ligationDecrease in left ventricular mass, atrial volumes, and NT-proBNP levelsImproved cardiac remodeling, no impact on eGFR[195]
Hetz et al.Randomized controlled trial28
(2013–2018)
Prophylactic ligationReduction in cardiac volumes and pulmonary systolic pressuresPrevention of high-output heart failure, decrease of NT pro BNP, no serum creatinine differences.[196]
Keller et al.Observational Prospective study49
(2013–2015)
None, comparative analysisHigher LVEDD and LVESD in high-flow AVF compared with normal-flow AVFAssociated with hemodynamic abnormalities[67]
Janeckova et al.Observational study40
(2018–2023)
Flow reduction/ligationSignificant reduction in arterial flow and cardiac output post-ligation88.3% primary patency after flow reduction, lower dyspnea according NYHA grade III, lower creatininemia and improve GFR[197]
HF-AVFs—defined as a flow of 2 L/min or exceeding 20% of cardiac output—are relatively common post-transplantation, with a prevalence of 43–69% among functional AVFs in KTRs [67,197]. These HF-AVFs are associated with pathological cardiac remodeling and complications, as demonstrated by observational studies from Stoumpos et al. and Keller et al. [67,68,197].

3.7.3. Evidence for AVF Ligation

RCTs by Rao et al. and Hetz et al. showed that AVF ligation can significantly reduce the left ventricular mass, improve cardiac remodeling, and prevent high-output heart failure without adversely affecting renal function [195,196]. However, these trials lack data on mortality and MACE. A study by Janeckova et al. uniquely highlighted the impact of AVF on both kidney and graft function, offering valuable insights into the cardiorenal interplay, though these findings require confirmation through larger RCTs [197].
A systematic review by Yasir et al. confirmed that ligating symptomatic AVFs in high-output heart failure is both safe and effective. The review also identified an AVF flow-to-cardiac output ratio >0.3 as a predictive marker for acute heart failure risk [198].

3.7.4. Individualized AVF Management

These findings underscore the importance of individualized AVF management for KTRs. While preserving AVF patency is crucial for patients with unstable graft function, ligation should be considered in cases of HF-AVFs to improve cardiovascular outcomes. Clinicians must carefully balance the need to maintain vascular access for future dialysis with the potential cardiovascular benefits of AVF closure, tailoring decisions to the specific needs of each patient.

4. Management of Post-Transplant Cardiovascular Disease

4.1. Management of Coronary Artery Diseases in Kidney Transplant Recipients

The benefit of early revascularization in asymptomatic KTRs remains a debated issue within the scientific community, particularly given the inconsistent results observed over recent decades. Current evidence suggests no significant reduction in the risk of MACE or post-transplant mortality. This debate is increasingly relevant considering the advancements in therapies, diagnostic techniques, monitoring, and a deeper understanding of the pathophysiology of CAD. Studies advocating widespread revascularization for asymptomatic CAD in CKD patients are often small, retrospective, or outdated, making them incompatible with modern treatment strategies and tools [199]. No recent randomized prospective studies have conclusively demonstrated a benefit to this approach.
The ISCHEMIA-CKD trial [200] provided pivotal insights. It included patients with an estimated glomerular filtration rate (eGFR) < 30 mL/min/1.73 m2 who tested positive for ischemia (via SPECT, stress echocardiography, or exercise testing with moderate to severe results). Participants were randomized to either an invasive strategy (angiography ± revascularization) or conservative management (medical therapy and monitoring) and followed for a median of 2.2 years. Among those undergoing angiography, 57% had multivessel disease, 26% had no significant lesions, and 50% were revascularized. However, the primary outcome (cumulative incidence of nonfatal myocardial infarction and death) was 36% in both groups, demonstrating that invasive strategies do not improve outcomes compared with optimized medical therapy. This finding was supported by a post hoc analysis in KTRs [201].
When revascularization is indicated, the 2024 ESC Guidelines [84] recommend coronary artery bypass grafting (CABG) over percutaneous coronary intervention (PCI) for multivessel disease, particularly in CKD patients. Although PCI carries a lower short-term risk of vascular complications, death, and stroke, CABG offers superior long-term outcomes, with lower rates of myocardial infarction and repeat revascularizations [199,202,203].
An optimized medical strategy remains the cornerstone of CAD management. This includes antiplatelet agents, RAAS inhibitors, and beta-blockers where applicable. The use of statins in this population has been debated. Large randomized controlled trials in hemodialysis patients showed no significant reduction in cardiovascular risk with statin therapy, and a subgroup analysis from the SHARP study found no benefit of lowering LDL cholesterol in patients with a GFR < 30 mL/min [204]. Nevertheless, combining medical therapy with lifestyle modifications remains at least as effective in reducing MACE and improving survival, emphasizing the role of microvascular disease, which cannot be directly assessed or treated via coronary angiography.
The necessity of CAD screening itself has come under scrutiny. A retrospective study by Lee et al. [205] of 22,687 CKD patients found that transplant candidates underwent 5–6 times more non-invasive ischemia tests and 16% more coronary angiograms than dialysis patients, but had lower revascularization rates. No difference in one-year post-transplant mortality was observed between the screened and unscreened patients. However, screened patients experienced longer waitlist times (median delay of 599 days), attributed to comorbidities, the need for additional tests, delays from dual antiplatelet therapy, and logistical or financial burdens [206]. Nonetheless, these screenings allowed for better MACE risk stratification, aiding clinicians in tailoring management strategies.
No study to date has demonstrated a reduction in cardiovascular risk through screening. The ongoing CARSK study [207], a randomized trial, aims to test the hypothesis that discontinuing CAD screening after waitlisting is not inferior to standard MACE screening. It will also evaluate the transplant rates, safety, and cost-effectiveness.
The early post-transplant period poses the highest cardiovascular risk, with a 4.3% incidence of MI within the first six months [20]. Cheng et al. [23], in a quasi-experimental study, found no correlation between CAD screening and reduced MACE or early mortality, especially within the critical 30-day post-transplant period. Interestingly, some data even suggest that screening may increase event risks during this window, potentially due to confounding factors like inherent high risk among the screened patients. Randomized trials comparing screening and non-screening groups are needed to address these biases, including those identified by Kopparam et al. [208], which highlighted the lack of outcomes data for screened patients who were not selected for transplantation.
The inability to demonstrate a benefit of screening for early perioperative risks may also reflect the influence of immediate post-transplant factors. Inflammatory markers released during graft reperfusion, such as galectin-3 [209] and IL-33 [210], have been implicated in cardiac remodeling following acute kidney injury (AKI). Evidence from various populations (e.g., post-cardiac surgery [211], hospitalized patients [212], CKD patients [213]) shows that AKI increases cardiovascular risk. This “butterfly effect” describes how acute kidney insults can lead to long-term cardiovascular remodeling [170], partially explaining the increased post-transplant risk and the limited utility of screening. Other contributors include high-dose immunosuppressive therapy and de novo diabetes, which may play as significant a role as pretransplant cardiovascular risk factors in explaining post-transplant outcomes.
Cardiac screening for asymptomatic CAD has not demonstrated clear benefits in improving survival or reducing the incidence of MACE after transplantation. Similarly, many coronary angiograms performed in this context could likely be avoided, as the benefits of revascularization in these patients remain uncertain. Therefore, it is imperative to refine the current screening algorithm by integrating novel biomarkers and conducting robust, well-designed studies to provide stronger evidence and potentially reshape large-scale management strategies. The current stagnation in this area is largely due to insufficient data and the absence of universally accepted recommendations. Additionally, the contribution of cardiovascular risk carried by the graft itself remains undefined in this population and warrants further investigation.

4.2. Management of Cerebrovascular Disease in Kidney Transplant Recipients

The risk of stroke progressively increases in individuals with advanced CKD and those undergoing dialysis compared with the general population. Additionally, albuminuria independently contributes to this heightened risk, regardless of blood pressure and diabetes status [214]. The pathophysiology is complex, involving traditional cardiovascular risk factors, dialysis-related factors, and non-traditional risk factors [215] such as increased blood–brain barrier permeability [216]. Emerging research suggests that uremic toxins may play a critical role in stroke pathogenesis, presenting a promising avenue for further investigation [217].
The reported prevalence of stroke ranges from 1.3% to 8%, with ischemic strokes being more common (63–91.4%) than hemorrhagic strokes. Incidence rates vary between 5.26 per 1000 person-years to 5.34 per 1000 person-years, depending on the study design and follow-up duration. Stroke-related mortality remains high, ranging from 29.7% to 41.7%, with 10.3% to 47.4% of patients dying within three months of the event [218,219,220,221,222,223,224,225,226]. Predictive factors for stroke prior to transplantation include advanced age, diabetes mellitus, atrial fibrillation, and hypertension. Patient-specific factors, such as tobacco use, obesity, and prior cardiovascular events, further elevate risk. Post-transplantation, complications like graft failure or rejection are associated with a higher stroke incidence. These findings underscore the importance of rigorous cardiovascular risk management before and after transplantation to mitigate stroke-related morbidity and mortality.
Compared with waitlisted patients with ESRD, KT significantly reduces the incidence of stroke. Huang et al. [219] reported an incidence density of 5.34 per 1000 person-years in KTRs compared with 14.5 per 1000 person-years in ESRD patients, suggesting a substantial reduction. This benefit is likely due to the resolution of non-traditional and dialysis-related risk factors post-transplantation. However, potential selection bias exists, as KTRs are typically younger and have fewer comorbidities.
Lentine et al. [220] demonstrated that the cumulative three-year incidence of de novo cerebrovascular disease events post-transplantation was 6.8%, significantly lower than the adjusted three-year estimates of 11.8% in waitlisted patients and 11.2% in those with graft loss. Time-dependent regression analysis revealed that transplantation predicted a 34% reduction in cerebrovascular disease risk, while graft failure increased the risk by over 150%. Weng et al. [226] corroborated these findings, reporting a 60% reduction in overall stroke risk among the KTRs compared with the non-recipients, with ischemic and hemorrhagic stroke risks reduced by 48% and 74%, respectively. Regarding ischemic stroke mortality, Zhang et al. [224] found no significant difference in in-hospital mortality between the KTRs and a CKD-free population, whereas patients with CKD G5D faced markedly higher in-hospital mortality (adjusted odds ratio [aOR]: 2.04, 95% CI: 1.93–2.15). These results emphasize the protective effects of kidney transplantation on cerebrovascular outcomes and the detrimental impact of graft loss and dialysis dependence.
Stroke prevention strategies in KTRs are similar to those in CKD patients, focusing on managing traditional stroke risk factors, particularly atherosclerosis, using antiplatelet therapy and treating symptomatic severe carotid stenosis [215]. The NASCET study [227] demonstrated that CKD patients benefited more from carotid endarterectomy than non-CKD patients. However, the benefit of this procedure for asymptomatic carotid stenosis remains unclear in both CKD patients and KTRs due to the challenges of assessing surgical risks in these populations [228]. The SPACE-2 randomized controlled trial [229], with a five-year follow-up, found no benefit from carotid revascularization via stenting or endarterectomy. Additionally, an observational study reported a fourfold increase in in-hospital mortality associated with both techniques in ESRD patients [230].
The initiation of dialysis represents a particularly high-risk period, with stroke incidence peaking during this time [229]. While the mechanisms underlying this increased risk are not fully understood, it underscores the potential value of preemptive kidney transplantation as a preventative measure. However, current data on preemptive transplantation’s impact on cerebrovascular outcomes are sparse, highlighting a critical need for further research.

4.3. Management of Atrial Fibrillation in Kidney Transplant Recipients

AF accounts for approximately 25% of cardioembolic strokes [225]. Watschinger et al. identified pretransplant AF as a significant predictor of stroke in KTRs [5]. In the general population, AF is associated with an elevated risk of stroke and adverse outcomes including increased mortality, cognitive decline, and left ventricular dysfunction [231]. Anticoagulation is recommended for high-risk individuals with a CHADS-VASc score >2 (or >1 for men), a category encompassing many CKD patients who are particularly vulnerable to AF due to mechanisms like inflammasome pathway activation [232,233,234].
However, anticoagulation in ESRD patients remains controversial, as the bleeding risk often outweighs its benefits. The RENAL-AF trial highlighted this concern, reporting that major bleeding events occurred ten times more frequently than strokes or systemic embolisms in ESRD patients [235]. While direct oral anticoagulants (DOACs) are associated with a lower bleeding risk compared with vitamin K antagonists in ESRD patients [236], their overall benefit in terms of reducing embolic risk and mortality in this population remains debated [237]. Moreover, DOAC use is limited in dialysis patients on the kidney transplant waiting list due to the complexities of managing perioperative bleeding risks during transplantation [238]. Emerging approaches like left atrial appendage closure may provide an alternative for this unique population [239]. A recent meta-analysis demonstrated that DOACs are probably a safer option than vitamin K antagonists after kidney transplantation, with a similar efficacy [240].
A meta-analysis of eight cohort studies involving 137,709 KTRs provided key insights into the epidemiology and outcomes of AF in this population. Pre-existing AF had a prevalence of 7.0% (95% CI: 5.6–8.8%), while the incidence of post-transplant AF was 4.9% (95% CI: 1.7–13.0%). Despite transplantation, the burden of AF remained substantial, with a 2.5-fold increased risk of stroke and a 1.9-fold increased risk of mortality. Additionally, AF was associated with adverse post-transplant outcomes including a 1.5-fold increase in death-censored allograft loss [241].
These findings highlight the critical need for further research to guide the effective management of AF in kidney transplant recipients. Developing tailored strategies to balance thromboembolic prevention with bleeding risk will be essential for optimizing outcomes in this high-risk population.

4.4. Management of Peripheral Artery Disease in Kidney Transplant Recipients

The epidemiology of peripheral arterial disease (PAD) remains challenging to accurately define due to the polymorphic clinical presentation of chronic limb ischemia and intermittent claudication, which increases the likelihood of underdiagnosis. However, in high-income countries, the prevalence of PAD is estimated at approximately 5% among individuals aged 40–44 years and around 12% among those aged 70–74 years [242]. The prognosis for critical limb ischemia (CLI) is particularly grim, with an all-cause mortality rate of 22% (95% confidence interval [CI]: 12–33%) and a major amputation rate of 22% (95% CI: 2–42%) [243].
PAD is significantly more prevalent among individuals with CKD, with estimates ranging from 12% to 38%, even in mild to moderate CKD stages [244]. CKD patients face a higher risk of severe PAD complications including amputation. Despite its severity, PAD in CKD is frequently underdiagnosed and undertreated, leading to poor outcomes such as low amputation-free survival rates (~80% at one year) and a high risk of amputation in dialysis patients [245]. Diagnostic tools like the ankle-brachial index (ABI) have limitations in CKD due to vascular calcification, which may cause false-normal or elevated results. Performing ABI dynamically during a one-minute treadmill walking test can improve its diagnostic accuracy [246].
The pathogenesis of PAD in CKD involves both traditional risk factors (e.g., diabetes, hypertension) and non-traditional mechanisms such as vascular calcification, inflammation, impaired angiogenesis, uremic toxins, and microvascular disease. These mechanisms accelerate vascular aging and contribute to both atherosclerosis and medial arterial calcification, complicating diagnosis and treatment [244,247,248].
In KTRs, the risk of PAD is lower compared with dialysis patients. However, PAD remains associated with poor outcomes, including reduced allograft survival and higher mortality, with a reported threefold increase in the relative risk of death in affected KTRs [249]. Data from the U.S. Renal Data System highlight that symptomatic PAD at waitlisting or post-transplant amputation correlates with diminished allograft and patient survival [250,251]. Furthermore, Patel et al. demonstrated that even asymptomatic PAD, identified by a low ABI, is linked to a threefold higher risk of graft failure [252].
Despite these findings, routine screening for PAD in asymptomatic patients awaiting kidney transplantation remains controversial. While ensuring adequate vascular access is essential for successful anastomosis during transplantation, performing aortic bypass surgery is associated with high perioperative and postoperative mortality and morbidity in ESRD patients [253]. As a result, this procedure should be reserved for treating aneurysmal disease or symptomatic PAD, as per the current guidelines [254]. Conversely, evidence suggests that transplanting a kidney to the iliac artery does not significantly worsen ipsilateral lower extremity ischemia in adults with PAD [255,256]. Additionally, in high-risk populations, such as individuals with type 1 diabetes, proposing a simultaneous kidney-pancreas transplantation may help mitigate the development and progression of PAD [257].
These observations highlight the critical need to develop tailored diagnostic and therapeutic strategies to improve outcomes in the CKD and kidney transplant populations. A better understanding of PAD pathophysiology and management in these groups will ultimately lead to improved patient and graft survival.

5. Limitations

This narrative review had some inherent limitations. We used a general set of criteria to select studies, which was based on their overall quality and pertinence rather than a predefined protocol. Therefore, this may have introduced a selection bias despite our efforts to minimize it. Additionally, we aimed to describe the incidence and impact of post-transplant cardiovascular diseases, but most of the available data were derived from North American and European epidemiological studies. Our study may not adequately reflect the situation in other countries, such as Asian countries, or even peculiar European countries, due to the variations across populations. Although we aimed to provide a comprehensive overview, certain subjects on the topic may not have been comprehensively addressed due to the variation in the study designs and potential residual confounding bias in the observational studies such as treatment adherence or socioeconomic status. Obviously, this narrative review was subject to a risk of publication bias, since studies with statistically significant findings are more likely to be published. Future systematic reviews and meta-analyses are therefore needed for accuracy and to validate our data.

6. Conclusions

In conclusion, CVD remains a significant challenge for kidney transplant recipients, influencing both graft and patient survival. The complex interplay between traditional and transplant-specific risk factors necessitates a tailored approach to risk assessment, prevention, and management. While advancements in diagnostic tools and therapeutic strategies have improved outcomes, there are still gaps in our understanding of the optimal management of CVD in this population. Integrating novel biomarkers, refining screening algorithms, and exploring emerging therapies such as SGLT2 inhibitors and GLP-1 receptor agonists offer promising avenues for reducing cardiovascular complications. Future research should prioritize robust, large-scale studies to establish evidence-based protocols that can guide clinical practice and improve the quality of life for kidney transplant recipients. By addressing these challenges, we can move closer to optimizing the long-term outcomes and reducing the burden of cardiovascular morbidity and mortality in this vulnerable population.

Author Contributions

Conceptualization, T.B., D.B., C.W. and N.F.; Validation, S.C. and N.F.; Writing—original draft preparation, T.B., D.B., C.W. and N.F.; Writing—review and editing, S.C. and N.F.; Supervision, N.F.; Project administration, N.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Jankowski, J.; Floege, J.; Fliser, D.; Böhm, M.; Marx, N. Cardiovascular Disease in Chronic Kidney Disease. Circulation 2021, 143, 1157–1172. [Google Scholar] [CrossRef]
  2. Matsushita, K.; Ballew, S.H.; Wang, A.Y.-M.; Kalyesubula, R.; Schaeffner, E.; Agarwal, R. Epidemiology and risk of cardiovascular disease in populations with chronic kidney disease. Nat. Rev. Nephrol. 2022, 18, 696–707. [Google Scholar] [CrossRef] [PubMed]
  3. Cozzolino, M.; Mangano, M.; Stucchi, A.; Ciceri, P.; Conte, F.; Galassi, A. Cardiovascular disease in dialysis patients. Nephrol. Dial. Transplant. 2018, 33, iii28–iii34. [Google Scholar] [CrossRef]
  4. Strohmaier, S.; Wallisch, C.; Kammer, M.; Geroldinger, A.; Heinze, G.; Oberbauer, R.; Haller, M.C. Survival Benefit of First Single-Organ Deceased Donor Kidney Transplantation Compared with Long-term Dialysis Across Ages in Transplant-Eligible Patients With Kidney Failure. JAMA Netw. Open 2022, 5, e2234971. [Google Scholar] [CrossRef] [PubMed]
  5. Wolfe, R.A.; Ashby, V.B.; Milford, E.L.; Ojo, A.O.; Ettenger, R.E.; Agodoa, L.Y.C.; Held, P.J.; Port, F.K. Comparison of Mortality in All Patients on Dialysis, Patients on Dialysis Awaiting Transplantation, and Recipients of a First Cadaveric Transplant. N. Engl. J. Med. 1999, 341, 1725–1730. [Google Scholar] [CrossRef]
  6. Foley, R.N.; Parfrey, P.S.; Sarnak, M.J. Clinical epidemiology of cardiovascular disease in chronic renal disease. Am. J. Kidney Dis. 1998, 32, S112–S119. [Google Scholar] [CrossRef] [PubMed]
  7. Disney, A.P.S. Demography and survival of patients receiving treatment for chronic renal failure in Australia and New Zealand: Report on dialysis and renal transplantation treatment from the Australia and New Zealand dialysis and transplant registry. Am. J. Kidney Dis. 1995, 25, 165–175. [Google Scholar] [CrossRef]
  8. Ojo, A.O.; Hanson, J.A.; Wolfe, R.A.; Leichtman, A.B.; Agodoa, L.Y.; Port, F.K. Long-term survival in renal transplant recipients with graft function. Kidney Int. 2000, 57, 307–313. [Google Scholar] [CrossRef]
  9. Awan, A.A.; Niu, J.; Pan, J.S.; Erickson, K.F.; Mandayam, S.; Winkelmayer, W.C.; Navaneethan, S.D.; Ramanathan, V. Trends in the Causes of Death among Kidney Transplant Recipients in the United States (1996–2014). Am. J. Nephrol. 2018, 48, 472–481. [Google Scholar] [CrossRef]
  10. Ying, T.; Shi, B.; Kelly, P.J.; Pilmore, H.; Clayton, P.A.; Chadban, S.J. Death after Kidney Transplantation: An Analysis by Era and Time Post-Transplant. J. Am. Soc. Nephrol. 2020, 31, 2887–2899. [Google Scholar] [CrossRef]
  11. Wyld, M.L.R.; De La Mata, N.L.; Masson, P.; O’Lone, E.; Kelly, P.J.; Webster, A.C. Cardiac Mortality in Kidney Transplant Patients: A Population-based Cohort Study 1988–2013 in Australia and New Zealand. Transplantation 2021, 105, 413–422. [Google Scholar] [CrossRef] [PubMed]
  12. Helve, S.; Helanterä, I.; Laine, M.; Nieminen, T.; Finne, P.; Helve, J. Trends and Specific Causes of Cardiovascular Mortality after Kidney Transplantation in Finland. Clin. J. Am. Soc. Nephrol. 2023, 19, 355–363. [Google Scholar] [CrossRef]
  13. Acosta, E.; Mehta, N.; Myrskylä, M.; Ebeling, M. Cardiovascular Mortality Gap Between the United States and Other High Life Expectancy Countries in 2000–2016. J. Gerontol. Ser. B 2022, 77, S148–S157. [Google Scholar] [CrossRef] [PubMed]
  14. Mayrdorfer, M.; Liefeldt, L.; Osmanodja, B.; Naik, M.G.; Schmidt, D.; Duettmann, W.; Hammett, C.; Schrezenmeier, E.; Friedersdorff, F.; Wu, K.; et al. A single centre in-depth analysis of death with a functioning kidney graft and reasons for overall graft failure. Nephrol. Dial. Transplant. 2022, 38, 1857–1866. [Google Scholar] [CrossRef]
  15. Carpenter, M.A.; Weir, M.R.; Adey, D.B.; House, A.A.; Bostom, A.G.; Kusek, J.W. Inadequacy of cardiovascular risk factor management in chronic kidney transplantation—Evidence from the FAVORIT study. Clin. Transplant. 2012, 26, E438–E446. [Google Scholar] [CrossRef] [PubMed]
  16. Chukwu, C.A.; Rao, A.; Middleton, R.; Kalra, P.A. Post-Transplant Cardiovascular Disease in Kidney Transplant Recipients: Incidence, Risk Factors, and Outcomes in the Era of Modern Immunosuppression. J. Clin. Med. 2024, 13, 2734. [Google Scholar] [CrossRef]
  17. Andersson, C.; Hansen, D.; Sørensen, S.S.; McGrath, M.; McCausland, F.R.; Torp-Pedersen, C.; Schou, M.; Køber, L.; Pfeffer, M.A. Long-term cardiovascular events, graft failure, and mortality in kidney transplant recipients. Eur. J. Intern. Med. 2024, 121, 109–113. [Google Scholar] [CrossRef]
  18. Aakhus, S.; Dahl, K.; Widerøe, T.E. Cardiovascular disease in stable renal transplant patients in Norway: Morbidity and mortality during a 5-yr follow-up. Clin. Transplant. 2004, 18, 596–604. [Google Scholar] [CrossRef]
  19. Mathur, A.K.; Chang, Y.-H.; Steidley, D.E.; Heilman, R.; Khurmi, N.; Wasif, N.; Etzioni, D.; Moss, A.A. Patterns of Care and Outcomes in Cardiovascular Disease After Kidney Transplantation in the United States. Transplant. Direct 2017, 3, e126. [Google Scholar] [CrossRef]
  20. Lentine, K.L.; Brennan, D.C.; Schnitzler, M.A. Incidence and predictors of myocardial infarction after kidney transplantation. J. Am. Soc. Nephrol. 2005, 16, 496–506. [Google Scholar] [CrossRef]
  21. Israni, A.K.; Snyder, J.J.; Skeans, M.A.; Peng, Y.; Maclean, J.R.; Weinhandl, E.D.; Kasiske, B.L.; PORT Investigators. Predicting coronary heart disease after kidney transplantation: Patient Outcomes in Renal Transplantation (PORT) Study. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2010, 10, 338–353. [Google Scholar] [CrossRef] [PubMed]
  22. Kahn, M.R.; Fallahi, A.; Kim, M.C.; Esquitin, R.; Robbins, M.J. Coronary artery disease in a large renal transplant population: Implications for management. Am. J. Transplant. 2011, 11, 2665–2674. [Google Scholar] [CrossRef] [PubMed]
  23. Cheng, X.S.; Liu, S.; Han, J.; Stedman, M.R.; Baiocchi, M.; Tan, J.C.; Chertow, G.M.; Fearon, W.F. Association of Pretransplant Coronary Heart Disease Testing with Early Kidney Transplant Outcomes. JAMA Intern. Med. 2023, 183, 134–141. [Google Scholar] [CrossRef] [PubMed]
  24. House, A.A.; Wanner, C.; Sarnak, M.J.; Piña, I.L.; McIntyre, C.W.; Komenda, P.; Kasiske, B.L.; Deswal, A.; deFilippi, C.R.; Cleland, J.G.F.; et al. Heart failure in chronic kidney disease: Conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int. 2019, 95, 1304–1317. [Google Scholar] [CrossRef]
  25. de Mattos, A.M.; Siedlecki, A.; Gaston, R.S.; Perry, G.J.; Julian, B.A.; Kew, C.E.I.; Deierhoi, M.H.; Young, C.; Curtis, J.J.; Iskandrian, A.E. Systolic Dysfunction Portends Increased Mortality among Those Waiting for Renal Transplant. J. Am. Soc. Nephrol. 2008, 19, 1191. [Google Scholar] [CrossRef]
  26. Wali, R.K.; Wang, G.S.; Gottlieb, S.S.; Bellumkonda, L.; Hansalia, R.; Ramos, E.; Drachenberg, C.; Papadimitriou, J.; Brisco, M.A.; Blahut, S.; et al. Effect of kidney transplantation on left ventricular systolic dysfunction and congestive heart failure in patients with end-stage renal disease. J. Am. Coll. Cardiol. 2005, 45, 1051–1060. [Google Scholar] [CrossRef]
  27. Lenihan, C.R.; Montez-Rath, M.E.; Scandling, J.D.; Turakhia, M.P.; Winkelmayer, W.C. Outcomes after kidney transplantation of patients previously diagnosed with atrial fibrillation. Am. J. Transplant. 2013, 13, 1566–1575. [Google Scholar] [CrossRef]
  28. Lentine, K.L.; Schnitzler, M.A.; Abbott, K.C.; Li, L.; Xiao, H.; Burroughs, T.E.; Takemoto, S.K.; Willoughby, L.M.; Gavard, J.A.; Brennan, D.C. Incidence, predictors, and associated outcomes of atrial fibrillation after kidney transplantation. Clin. J. Am. Soc. Nephrol. 2006, 1, 288–296. [Google Scholar] [CrossRef]
  29. Lentine, K.L.; Villines, T.C.; Axelrod, D.; Kaviratne, S.; Weir, M.R.; Costa, S.P. Evaluation and Management of Pulmonary Hypertension in Kidney Transplant Candidates and Recipients: Concepts and Controversies. Transplantation 2017, 101, 166. [Google Scholar] [CrossRef]
  30. Alhamad, E.H.; Al-Ghonaim, M.; Alfaleh, H.F.; Cal, J.P.; Said, N. Pulmonary hypertension in end-stage renal disease and post renal transplantation patients. J. Thorac. Dis. 2014, 6, 606–616. [Google Scholar] [CrossRef]
  31. Reddy, Y.N.V.; Lunawat, D.; Abraham, G.; Matthew, M.; Mullasari, A.; Nagarajan, P.; Reddy, Y.N.V. Progressive pulmonary hypertension: Another criterion for expeditious renal transplantation. Saudi J. Kidney Dis. Transplant. 2013, 24, 925–929. [Google Scholar] [CrossRef]
  32. Basile, C.; Lomonte, C.; Vernaglione, L.; Casucci, F.; Antonelli, M.; Losurdo, N. The relationship between the flow of arteriovenous fistula and cardiac output in haemodialysis patients. Nephrol. Dial. Transplant. 2008, 23, 282–287. [Google Scholar] [CrossRef] [PubMed]
  33. Daragó, A.; Szabó, E.; Barkó, D.; Schwegler, G.; Szabó, R.P.; Nagy, A.C.; Nemes, B. Effects of Kidney Transplantation on Valvular Heart Diseases. Transplant. Proc. 2021, 53, 1433–1438. [Google Scholar] [CrossRef] [PubMed]
  34. Abbott, K.C.; Hshieh, P.; Cruess, D.; Agodoa, L.Y.C.; Welch, P.G.; Taylor, A.J.; Yuan, C.M. Hospitalized valvular heart disease in patients on renal transplant waiting list: Incidence, clinical correlates and outcomes. Clin. Nephrol. 2003, 59, 79–87. [Google Scholar] [CrossRef]
  35. Fox, H.; Büttner, S.; Hemmann, K.; Asbe-Vollkopf, A.; Doss, M.; Beiras-Fernandez, A.; Moritz, A.; Zeiher, A.M.; Scheuermann, E.; Geiger, H.; et al. Transcatheter aortic valve implantation improves outcome compared to open-heart surgery in kidney transplant recipients requiring aortic valve replacement. J. Cardiol. 2013, 61, 423–427. [Google Scholar] [CrossRef] [PubMed]
  36. Marcassi, A.P.; Yasbek, D.C.; Pestana, J.O.M.; Fachini, F.C.; De Lira Filho, E.B.; Cassiolato, J.L.; Canziani, M.E.F. Ventricular arrhythmia in incident kidney transplant recipients: Prevalence and associated factors. Transpl. Int. 2011, 24, 67–72. [Google Scholar] [CrossRef]
  37. Roufosse, C.; Simmonds, N.; Clahsen-van Groningen, M.; Haas, M.; Henriksen, K.J.; Horsfield, C.; Loupy, A.; Mengel, M.; Perkowska-Ptasińska, A.; Rabant, M.; et al. A 2018 Reference Guide to the Banff Classification of Renal Allograft Pathology. Transplantation 2018, 102, 1795–1814. [Google Scholar] [CrossRef]
  38. Hill, G.S.; Nochy, D.; Loupy, A. Accelerated arteriosclerosis: A form of transplant arteriopathy. Curr. Opin. Organ Transplant. 2010, 15, 11. [Google Scholar] [CrossRef]
  39. Freedman, B.I.; Cohen, A.H. Hypertension-attributed nephropathy: What’s in a name? Nat. Rev. Nephrol. 2016, 12, 27–36. [Google Scholar] [CrossRef]
  40. Loupy, A.; Vernerey, D.; Viglietti, D.; Aubert, O.; Duong Van Huyen, J.-P.; Empana, J.-P.; Bruneval, P.; Glotz, D.; Legendre, C.; Jouven, X.; et al. Determinants and Outcomes of Accelerated Arteriosclerosis. Circ. Res. 2015, 117, 470–482. [Google Scholar] [CrossRef]
  41. Kasiske, B.L.; Maclean, J.R.; Snyder, J.J. Acute Myocardial Infarction and Kidney Transplantation. J. Am. Soc. Nephrol. 2006, 17, 900. [Google Scholar] [CrossRef] [PubMed]
  42. De Lima, J.J.G.; Gowdak, L.H.W.; David-Neto, E.; Bortolotto, L.A. Early cardiovascular events and cardiovascular death after renal transplantation: Role of pretransplant risk factors. Clin. Exp. Nephrol. 2021, 25, 545–553. [Google Scholar] [CrossRef] [PubMed]
  43. Dunn, T.; Saeed, M.J.; Shpigel, A.; Novak, E.; Alhamad, T.; Stwalley, D.; Rich, M.W.; Brown, D.L. The association of preoperative cardiac stress testing with 30-day death and myocardial infarction among patients undergoing kidney transplantation. PLoS ONE 2019, 14, e0211161. [Google Scholar] [CrossRef] [PubMed]
  44. Nimmo, A.; Forsyth, J.L.; Oniscu, G.C.; Robb, M.; Watson, C.; Fotheringham, J.; Roderick, P.J.; Ravanan, R.; Taylor, D.M. A propensity score–matched analysis indicates screening for asymptomatic coronary artery disease does not predict cardiac events in kidney transplant recipients. Kidney Int. 2021, 99, 431–442. [Google Scholar] [CrossRef]
  45. Pilmore, H.; Dent, H.; Chang, S.; McDonald, S.P.; Chadban, S.J. Reduction in cardiovascular death after kidney transplantation. Transplantation 2010, 89, 851–857. [Google Scholar] [CrossRef]
  46. Carpenter, M.A.; John, A.; Weir, M.R.; Smith, S.R.; Hunsicker, L.; Kasiske, B.L.; Kusek, J.W.; Bostom, A.; Ivanova, A.; Levey, A.S.; et al. BP, cardiovascular disease, and death in the Folic Acid for Vascular Outcome Reduction in Transplantation trial. J. Am. Soc. Nephrol. 2014, 25, 1554–1562. [Google Scholar] [CrossRef]
  47. Kasiske, B.L.; Anjum, S.; Shah, R.; Skogen, J.; Kandaswamy, C.; Danielson, B.; O’Shaughnessy, E.A.; Dahl, D.C.; Silkensen, J.R.; Sahadevan, M.; et al. Hypertension after kidney transplantation. Am. J. Kidney Dis. 2004, 43, 1071–1081. [Google Scholar] [CrossRef]
  48. Speer, C.; Benning, L.; Morath, C.; Zeier, M.; Frey, N.; Opelz, G.; Döhler, B.; Tran, T.H. Blood Pressure Goals and Outcomes in Kidney Transplant Recipients in an Analysis of the Collaborative Transplant Study. Kidney Int. Rep. 2024, 10, 780–790. [Google Scholar] [CrossRef]
  49. Liu, J.; Chen, S.; Gao, W. Gender differences in cardiovascular outcomes of kidney transplant recipients: A retrospective cohort study. Medicine 2024, 103, e39568. [Google Scholar] [CrossRef]
  50. Roodnat, J.I.; Mulder, P.G.; Zietse, R.; Rischen-Vos, J.; van Riemsdijk, I.C.; IJzermans, J.N.; Weimar, W. Cholesterol as an independent predictor of outcome after renal transplantation. Transplantation 2000, 69, 1704–1710. [Google Scholar] [CrossRef]
  51. Holdaas, H.; Fellström, B.; Jardine, A.G.; Holme, I.; Nyberg, G.; Fauchald, P.; Grönhagen-Riska, C.; Madsen, S.; Neumayer, H.-H.; Cole, E.; et al. Effect of fluvastatin on cardiac outcomes in renal transplant recipients: A multicentre, randomised, placebo-controlled trial. Lancet Lond. Engl. 2003, 361, 2024–2031. [Google Scholar] [CrossRef]
  52. Holdaas, H.; Fellström, B.; Cole, E.; Nyberg, G.; Olsson, A.G.; Pedersen, T.R.; Madsen, S.; Grönhagen-Riska, C.; Neumayer, H.-H.; Maes, B.; et al. Long-term Cardiac Outcomes in Renal Transplant Recipients Receiving Fluvastatin: The ALERT Extension Study. Am. J. Transplant. 2005, 5, 2929–2936. [Google Scholar] [CrossRef] [PubMed]
  53. Kasiske, B.L.; Snyder, J.J.; Gilbertson, D.; Matas, A.J. Diabetes mellitus after kidney transplantation in the United States. Am. J. Transplant. 2003, 3, 178–185. [Google Scholar] [CrossRef] [PubMed]
  54. Vincenti, F.; Friman, S.; Scheuermann, E.; Rostaing, L.; Jenssen, T.; Campistol, J.M.; Uchida, K.; Pescovitz, M.D.; Marchetti, P.; Tuncer, M.; et al. Results of an international, randomized trial comparing glucose metabolism disorders and outcome with cyclosporine versus tacrolimus. Am. J. Transplant. 2007, 7, 1506–1514. [Google Scholar] [CrossRef] [PubMed]
  55. Cosio, F.G.; Kudva, Y.; van der Velde, M.; Larson, T.S.; Textor, S.C.; Griffin, M.D.; Stegall, M.D. New onset hyperglycemia and diabetes are associated with increased cardiovascular risk after kidney transplantation. Kidney Int. 2005, 67, 2415–2421. [Google Scholar] [CrossRef]
  56. Kuo, H.-T.; Sampaio, M.S.; Vincenti, F.; Bunnapradist, S. Associations of Pretransplant Diabetes Mellitus, New-Onset Diabetes After Transplant, and Acute Rejection With Transplant Outcomes: An Analysis of the Organ Procurement and Transplant Network/United Network for Organ Sharing (OPTN/UNOS) Database. Am. J. Kidney Dis. 2010, 56, 1127–1139. [Google Scholar] [CrossRef]
  57. Caillard, S.; Eprinchard, L.; Perrin, P.; Braun, L.; Heibel, F.; Moreau, F.; Kessler, L.; Moulin, B. Incidence and risk factors of glucose metabolism disorders in kidney transplant recipients: Role of systematic screening by oral glucose tolerance test. Transplantation 2011, 91, 757–764. [Google Scholar] [CrossRef]
  58. Lim, W.H.; Lok, C.E.; Kim, S.J.; Knoll, G.; Shah, B.R.; Naylor, K.; Luo, B.; Vinegar, M.; Dixon, S.N.; Hawley, C.; et al. Impact of Pretransplant and New-Onset Diabetes After Transplantation on the Risk of Major Adverse Cardiovascular Events in Kidney Transplant Recipients: A Population-based Cohort Study. Transplantation 2021, 105, 2470–2481. [Google Scholar] [CrossRef]
  59. Kasiske, B.L.; Klinger, D. Cigarette smoking in renal transplant recipients. J. Am. Soc. Nephrol. 2000, 11, 753–759. [Google Scholar] [CrossRef]
  60. Hurst, F.P.; Altieri, M.; Patel, P.P.; Jindal, T.R.; Guy, S.R.; Sidawy, A.N.; Agodoa, L.Y.; Abbott, K.C.; Jindal, R.M. Effect of smoking on kidney transplant outcomes: Analysis of the United States Renal Data System. Transplantation 2011, 92, 1101–1107. [Google Scholar] [CrossRef]
  61. Lentine, K.L.; Rey, L.A.R.; Bacchi, G.; Wasi, N.; Schmitz, L.; Salvalaggio, P.R.; Abbott, K.C.; Schnitzler, M.A.; Neri, L.; Brennan, D.C. Obesity and cardiac risk after kidney transplantation: Experience at one center and comprehensive literature review. Transplantation 2008, 86, 303. [Google Scholar] [CrossRef] [PubMed]
  62. Gwon, J.G.; Choi, J.; Jung, C.W.; Lee, C.H.; Oh, S.W.; Jo, S.-K.; Cho, W.Y.; Park, J.B.; Huh, K.H.; Ro, H.; et al. Impact of changes in waist-to-hip ratio after kidney transplantation on cardiovascular outcomes. Sci. Rep. 2021, 11, 783. [Google Scholar] [CrossRef]
  63. Soveri, I.; Abedini, S.; Holdaas, H.; Jardine, A.; Eriksson, N.; Fellström, B. Metabolic syndrome and cardiovascular risk in renal transplant recipients: Effects of statin treatment. Clin. Transplant. 2009, 23, 914–920. [Google Scholar] [CrossRef]
  64. Helanterä, I.; Salmela, K.; Kyllönen, L.; Koskinen, P.; Grönhagen-Riska, C.; Finne, P. Pretransplant Dialysis Duration and Risk of Death After Kidney Transplantation in the Current Era. Transplantation 2014, 98, 458. [Google Scholar] [CrossRef] [PubMed]
  65. Ivarsson, K.M.; Akaberi, S.; Isaksson, E.; Reihnér, E.; Czuba, T.; Prütz, K.-G.; Clyne, N.; Almquist, M. Cardiovascular and Cerebrovascular Events After Parathyroidectomy in Patients on Renal Replacement Therapy. World J. Surg. 2019, 43, 1981–1988. [Google Scholar] [CrossRef]
  66. Tapiawala, S.N.; Tinckam, K.J.; Cardella, C.J.; Schiff, J.; Cattran, D.C.; Cole, E.H.; Kim, S.J. Delayed graft function and the risk for death with a functioning graft. J. Am. Soc. Nephrol. 2010, 21, 153–161. [Google Scholar] [CrossRef]
  67. Keller, N.; Monnier, A.; Caillard, S.; Cognard, N.; Geny, B.; Moulin, B.; Talha, S. High-flow arteriovenous fistula and hemodynamic consequences at 1 year after kidney transplantation. Semin. Dial. 2022, 35, 171–180. [Google Scholar] [CrossRef] [PubMed]
  68. Stoumpos, S.; Van Rhijn, P.; Mangion, K.; Thomson, P.C.; Mark, P.B. Arteriovenous fistula for haemodialysis as a predictor of de novo heart failure in kidney transplant recipients. Clin. Kidney J. 2024, 17, sfae105. [Google Scholar] [CrossRef]
  69. Kasiske, B.L.; Chakkera, H.A.; Roel, J. Explained and unexplained ischemic heart disease risk after renal transplantation. J. Am. Soc. Nephrol. 2000, 11, 1735–1743. [Google Scholar] [CrossRef]
  70. Fernández-Fresnedo, G.; Escallada, R.; Rodrigo, E.; De Francisco, A.L.M.; Cotorruelo, J.G.; Sanz De Castro, S.; Zubimendi, J.A.; Ruiz, J.C.; Arias, M. The risk of cardiovascular disease associated with proteinuria in renal transplant patients. Transplantation 2002, 73, 1345–1348. [Google Scholar] [CrossRef]
  71. Hiremath, S.; Fergusson, D.A.; Fergusson, N.; Bennett, A.; Knoll, G.A. Renin-Angiotensin System Blockade and Long-term Clinical Outcomes in Kidney Transplant Recipients: A Meta-analysis of Randomized Controlled Trials. Am. J. Kidney Dis. 2017, 69, 78–86. [Google Scholar] [CrossRef] [PubMed]
  72. Weiner, D.E.; Carpenter, M.A.; Levey, A.S.; Ivanova, A.; Cole, E.H.; Hunsicker, L.; Kasiske, B.L.; Kim, S.J.; Kusek, J.W.; Bostom, A.G. Kidney function and risk of cardiovascular disease and mortality in kidney transplant recipients: The favorit trial. Am. J. Transplant. 2012, 12, 2437–2445. [Google Scholar] [CrossRef] [PubMed]
  73. Murakami, N.; Riella, L.V.; Funakoshi, T. Risk of metabolic complications in kidney transplantation after conversion to mTOR inhibitor: A systematic review and meta-analysis. Am. J. Transplant. 2014, 14, 2317–2327. [Google Scholar] [CrossRef]
  74. Wen, X.; Casey, M.J.; Santos, A.H.; Hartzema, A.; Womer, K.L. Comparison of Utilization and Clinical Outcomes for Belatacept- and Tacrolimus-Based Immunosuppression in Renal Transplant Recipients. Am. J. Transplant. 2016, 16, 3202–3211. [Google Scholar] [CrossRef]
  75. Divard, G.; Aubert, O.; Debiais-Deschamp, C.; Raynaud, M.; Goutaudier, V.; Sablik, M.; Sayeg, C.; Legendre, C.; Obert, J.; Anglicheau, D.; et al. Long-Term Outcomes after Conversion to a Belatacept-Based Immunosuppression in Kidney Transplant Recipients. Clin. J. Am. Soc. Nephrol. 2024, 19, 628. [Google Scholar] [CrossRef]
  76. Bakri, R.S.; Afzali, B.; Covic, A.; Sriskantharan, R.; Bharma-Ariza, P.; Park, W.-H.; Sriharan, M.; Dalton, N.; Wierzbicki, A.S.; Crook, M.A.; et al. Cardiovascular disease in renal allograft recipients is associated with elevated sialic acid or markers of inflammation. Clin. Transplant. 2004, 18, 201–204. [Google Scholar] [CrossRef] [PubMed]
  77. Rigatto, C.; Foley, R.; Jeffery, J.; Negrijn, C.; Tribula, C.; Parfrey, P. Electrocardiographic Left Ventricular Hypertrophy in Renal Transplant Recipients: Prognostic Value and Impact of Blood Pressure and Anemia. J. Am. Soc. Nephrol. 2003, 14, 462. [Google Scholar] [CrossRef]
  78. Ibernon, M.; Moreso, F.; Ruiz-Majoral, A.; Sarrias, X.; Sarrias, M.; Grinyó, J.M.; Serón, D. Contribution of anemia and hypertension to left ventricular hypertrophy during the initial 2 years after renal transplantation. Transplant. Proc. 2011, 43, 2199–2204. [Google Scholar] [CrossRef]
  79. Go, A.S.; Chertow, G.M.; Fan, D.; McCulloch, C.E.; Hsu, C. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N. Engl. J. Med. 2004, 351, 1296–1305. [Google Scholar] [CrossRef]
  80. Writing Group for the CKD Prognosis Consortium; Grams, M.E.; Coresh, J.; Matsushita, K.; Ballew, S.H.; Sang, Y.; Surapaneni, A.; Alencar de Pinho, N.; Anderson, A.; Appel, L.J.; et al. Estimated Glomerular Filtration Rate, Albuminuria, and Adverse Outcomes: An Individual-Participant Data Meta-Analysis. JAMA 2023, 330, 1266–1277. [Google Scholar] [CrossRef]
  81. Sarnak, M.J.; Amann, K.; Bangalore, S.; Cavalcante, J.L.; Charytan, D.M.; Craig, J.C.; Gill, J.S.; Hlatky, M.A.; Jardine, A.G.; Landmesser, U.; et al. Chronic Kidney Disease and Coronary Artery Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 74, 1823–1838. [Google Scholar] [CrossRef] [PubMed]
  82. Herzog, C.A.; Littrell, K.; Arko, C.; Frederick, P.D.; Blaney, M. Clinical characteristics of dialysis patients with acute myocardial infarction in the United States: A collaborative project of the United States Renal Data System and the National Registry of Myocardial Infarction. Circulation 2007, 116, 1465–1472. [Google Scholar] [CrossRef] [PubMed]
  83. Sosnov, J.; Lessard, D.; Goldberg, R.J.; Yarzebski, J.; Gore, J.M. Differential symptoms of acute myocardial infarction in patients with kidney disease: A community-wide perspective. Am. J. Kidney Dis. 2006, 47, 378–384. [Google Scholar] [CrossRef]
  84. Vrints, C.; Andreotti, F.; Koskinas, K.C.; Rossello, X.; Adamo, M.; Ainslie, J.; Banning, A.P.; Budaj, A.; Buechel, R.R.; Chiariello, G.A.; et al. 2024 ESC Guidelines for the management of chronic coronary syndromes. Eur. Heart J. 2024, 45, 3415–3537. [Google Scholar] [CrossRef]
  85. Cheng, X.S.; Mohanty, S.; Turner, V.; Mastrodicasa, D.; Winther, S.; Fleischmann, D.; Tan, J.C.; Fearon, W.F. Coronary Computed Tomography Angiography in Diagnosing Obstructive Coronary Artery Disease in Patients with Advanced Chronic Kidney Disease: A Systematic Review and Meta-Analysis. Cardiorenal Med. 2021, 11, 44–51. [Google Scholar] [CrossRef] [PubMed]
  86. Winther, S.; Svensson, M.; Jørgensen, H.S.; Bouchelouche, K.; Gormsen, L.C.; Pedersen, B.B.; Holm, N.R.; Bøtker, H.E.; Ivarsen, P.; Bøttcher, M. Diagnostic Performance of Coronary CT Angiography and Myocardial Perfusion Imaging in Kidney Transplantation Candidates. JACC Cardiovasc. Imaging 2015, 8, 553–562. [Google Scholar] [CrossRef]
  87. El-Tallawi, K.C.; Aljizeeri, A.; Nabi, F.; Al-Mallah, M.H. Myocardial Perfusion Imaging Using Positron Emission Tomography. Methodist DeBakey Cardiovasc. J. 2020, 16, 114–121. [Google Scholar] [CrossRef]
  88. Winther, S.; Svensson, M.; Jørgensen, H.S.; Rasmussen, L.D.; Holm, N.R.; Gormsen, L.C.; Bouchelouche, K.; Bøtker, H.E.; Ivarsen, P.; Bøttcher, M. Prognostic Value of Risk Factors, Calcium Score, Coronary CTA, Myocardial Perfusion Imaging, and Invasive Coronary Angiography in Kidney Transplantation Candidates. JACC Cardiovasc. Imaging 2018, 11, 842–854. [Google Scholar] [CrossRef]
  89. Dahl, J.N.; Nielsen, M.B.; Rasmussen, L.D.; Ivarsen, P.; Williams, M.C.; Svensson, M.H.S.; Birn, H.; Bøttcher, M.; Winther, S. Coronary Plaque Characteristics in Patients With Chronic Kidney Failure: Impact on Cardiovascular Events and Mortality. Circ. Cardiovasc. Imaging 2024, 17, e017066. [Google Scholar] [CrossRef]
  90. Cheng, X.S.; VanWagner, L.B.; Costa, S.P.; Axelrod, D.A.; Bangalore, S.; Norman, S.P.; Herzog, C.A.; Lentine, K.L.; On behalf of the American Heart Association Council on the Kidney in Cardiovascular Disease and Council on Cardiovascular Radiology and Intervention. Emerging Evidence on Coronary Heart Disease Screening in Kidney and Liver Transplantation Candidates: A Scientific Statement From the American Heart Association. Circulation 2022, 146, e299–e324. [Google Scholar] [CrossRef]
  91. Lange, S.; Mędrzycka-Dąbrowska, W.; Zorena, K.; Dąbrowski, S.; Ślęzak, D.; Malecka-Dubiela, A.; Rutkowski, P. Nephrogenic Systemic Fibrosis as a Complication after Gadolinium-Containing Contrast Agents: A Rapid Review. Int. J. Environ. Res. Public. Health 2021, 18, 3000. [Google Scholar] [CrossRef] [PubMed]
  92. Dilsizian, V.; Gewirtz, H.; Marwick, T.H.; Kwong, R.Y.; Raggi, P.; Al-Mallah, M.H.; Herzog, C.A. Cardiac Imaging for Coronary Heart Disease Risk Stratification in Chronic Kidney Disease. JACC Cardiovasc. Imaging 2021, 14, 669–682. [Google Scholar] [CrossRef] [PubMed]
  93. Dundon, B.K.; Pisaniello, A.D.; Nelson, A.J.; Maia, M.; Teo, K.S.L.; Worthley, S.G.; Coates, P.T.; Russ, G.R.; Faull, R.J.; Bannister, K.; et al. Dobutamine stress cardiac MRI for assessment of coronary artery disease prior to kidney transplantation. Am. J. Kidney Dis. 2015, 65, 808–809. [Google Scholar] [CrossRef]
  94. Ripley, D.P.; Kannoly, S.; Gosling, O.E.; Hossain, E.; Chawner, R.R.; Moore, J.; Shore, A.C.; Bellenger, N.G. Safety and feasibility of dobutamine stress cardiac magnetic resonance for cardiovascular assessment prior to renal transplantation. J. Cardiovasc. Med. Hagerstown Md 2014, 15, 288–294. [Google Scholar] [CrossRef]
  95. Golzar, Y.; Doukky, R. Stress SPECT Myocardial Perfusion Imaging in End-Stage Renal Disease. Curr. Cardiovasc. Imaging Rep. 2017, 10, 13. [Google Scholar] [CrossRef]
  96. Wang, L.W.; Fahim, M.A.; Hayen, A.; Mitchell, R.L.; Baines, L.; Lord, S.; Craig, J.C.; Webster, A.C. Cardiac testing for coronary artery disease in potential kidney transplant recipients. Cochrane Database Syst. Rev. 2011, 2011, CD008691. [Google Scholar] [CrossRef] [PubMed]
  97. Kelderman, J.R.; Jolink, F.E.J.; Benjamens, S.; Monroy-Gonzalez, A.G.; Pol, R.A.; Slart, R.H.J.A. Diagnostic accuracy of myocardial perfusion imaging in patients evaluated for kidney transplantation: A systematic review and meta-analysis. J. Nucl. Cardiol. 2022, 29, 3405–3415. [Google Scholar] [CrossRef]
  98. Ives, C.W.; AlJaroudi, W.A.; Kumar, V.; Farag, A.; Rizk, D.V.; Oparil, S.; Iskandrian, A.E.; Hage, F.G. Prognostic value of myocardial perfusion imaging performed pre-renal transplantation: Post-transplantation follow-up and outcomes. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 1998–2008. [Google Scholar] [CrossRef]
  99. Helve, S.; Laine, M.; Sinisalo, J.; Helanterä, I.; Hänninen, H.; Lammintausta, O.; Lehtonen, J.; Finne, P.; Nieminen, T. Even mild reversible myocardial perfusion defects predict mortality in patients evaluated for kidney transplantation. Eur. Heart J. Cardiovasc. Imaging 2018, 19, 1019–1025. [Google Scholar] [CrossRef]
  100. Poli, F.E.; Gulsin, G.S.; McCann, G.P.; Burton, J.O.; Graham-Brown, M.P. The assessment of coronary artery disease in patients with end-stage renal disease. Clin. Kidney J. 2019, 12, 721–734. [Google Scholar] [CrossRef] [PubMed]
  101. Wang, L.W.; Masson, P.; Turner, R.M.; Lord, S.W.; Baines, L.A.; Craig, J.C.; Webster, A.C. Prognostic value of cardiac tests in potential kidney transplant recipients: A systematic review. Transplantation 2015, 99, 731–745. [Google Scholar] [CrossRef] [PubMed]
  102. Phawanawichian, C.; Kaolawanich, Y.; Skulratanasak, P.; Ratanasit, N. Prognostic value of dobutamine stress echocardiography for the long-term outcomes in kidney transplant candidates. Cardiovasc. Diagn. Ther. 2024, 14, 899–910. [Google Scholar] [CrossRef] [PubMed]
  103. Bloemendal, N.T.; Prakken, N.H.J.; Gareb, B.; Benjamens, S.; Sanders, J.S.F.; Slart, R.H.J.A.; Pol, R.A. Prognostic value of single photon emission computed tomography myocardial perfusion imaging for the prediction of MACE in pre- kidney transplant recipients: A systematic review and meta-analysis. Transplant. Rev. Orlando Fla 2024, 38, 100879. [Google Scholar] [CrossRef] [PubMed]
  104. Del Buono, M.G.; Montone, R.A.; Camilli, M.; Carbone, S.; Narula, J.; Lavie, C.J.; Niccoli, G.; Crea, F. Coronary Microvascular Dysfunction Across the Spectrum of Cardiovascular Diseases: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2021, 78, 1352–1371. [Google Scholar] [CrossRef]
  105. Huck, D.M.; Weber, B.; Schreiber, B.; Pandav, J.; Parks, S.; Hainer, J.; Brown, J.M.; Divakaran, S.; Blankstein, R.; Dorbala, S.; et al. Comparative Effectiveness of PET and SPECT MPI for Predicting Cardiovascular Events After Kidney Transplant. Circ. Cardiovasc. Imaging 2024, 17, e015858. [Google Scholar] [CrossRef]
  106. Eggers, K.M.; Lindahl, B.; Carrero, J.J.; Evans, M.; Szummer, K.; Jernberg, T. Cardiac Troponins and Their Prognostic Importance in Patients with Suspected Acute Coronary Syndrome and Renal Dysfunction. Clin. Chem. 2017, 63, 1409–1417. [Google Scholar] [CrossRef]
  107. Szramowska, A.; Bielecki, M.; Grzeszczyk, M.; Łabyk, A.; Kurnicka, K.; Pruszczyk, P.; Roik, M. High-sensitivity cardiac troponin T in detecting obstructive coronary artery disease in hemodialysis patients listed for kidney transplantation. Kardiol. Pol. 2024, 82, 285–291. [Google Scholar] [CrossRef]
  108. Palamuthusingam, D.; Pascoe, E.M.; Hawley, C.M.; Johnson, D.W.; Fahim, M. Revised cardiac risk index in predicting cardiovascular complications in patients receiving chronic kidney replacement therapy undergoing elective general surgery. Perioper. Med. Lond. Engl. 2024, 13, 70. [Google Scholar] [CrossRef]
  109. Silver, S.A.; Huang, M.; Nash, M.M.; Prasad, G.V.R. Framingham risk score and novel cardiovascular risk factors underpredict major adverse cardiac events in kidney transplant recipients. Transplantation 2011, 92, 183–189. [Google Scholar] [CrossRef]
  110. Chadban, S.J.; Ahn, C.; Axelrod, D.A.; Foster, B.J.; Kasiske, B.L.; Kher, V.; Kumar, D.; Oberbauer, R.; Pascual, J.; Pilmore, H.L.; et al. Summary of the Kidney Disease: Improving Global Outcomes (KDIGO) Clinical Practice Guideline on the Evaluation and Management of Candidates for Kidney Transplantation. Transplantation 2020, 104, 708–714. [Google Scholar] [CrossRef]
  111. Nielsen, M.B.; Dahl, J.N.; Jespersen, B.; Ivarsen, P.; Birn, H.; Winther, S. External Validation of Proposed American Heart Association Algorithm for Cardiovascular Screening Before Kidney Transplantation. J. Am. Heart Assoc. 2023, 12, e031150. [Google Scholar] [CrossRef] [PubMed]
  112. Cross, N.B.; Webster, A.C.; Masson, P.; O’connell, P.J.; Craig, J.C. Antihypertensives for kidney transplant recipients: Systematic review and meta-analysis of randomized controlled trials. Transplantation 2009, 88, 7–18. [Google Scholar] [CrossRef] [PubMed]
  113. Whelton, P.K.; Carey, R.M.; Aronow, W.S.; Casey, D.E.; Collins, K.J.; Dennison Himmelfarb, C.; DePalma, S.M.; Gidding, S.; Jamerson, K.A.; Jones, D.W.; et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension 2018, 71, 1269–1324. [Google Scholar] [CrossRef]
  114. Wanner, C.; Tonelli, M. Kidney Disease: Improving Global Outcomes Lipid Guideline Development Work Group Members KDIGO Clinical Practice Guideline for Lipid Management in CKD: Summary of recommendation statements and clinical approach to the patient. Kidney Int. 2014, 85, 1303–1309. [Google Scholar] [CrossRef] [PubMed]
  115. Kohnle, M.; Pietruck, F.; Kribben, A.; Philipp, T.; Heemann, U.; Witzke, O. Ezetimibe for the Treatment of Uncontrolled Hypercholesterolemia in Patients with High-Dose Statin Therapy After Renal Transplantation. Am. J. Transplant. 2006, 6, 205–208. [Google Scholar] [CrossRef]
  116. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A.; et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk: The Task Force for the management of dyslipidaemias of the European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS). Eur. Heart J. 2020, 41, 111–188. [Google Scholar] [CrossRef]
  117. Lemahieu, W.P.D.; Hermann, M.; Asberg, A.; Verbeke, K.; Holdaas, H.; Vanrenterghem, Y.; Maes, B.D. Combined therapy with atorvastatin and calcineurin inhibitors: No interactions with tacrolimus. Am. J. Transplant. 2005, 5, 2236–2243. [Google Scholar] [CrossRef]
  118. Migliozzi, D.R.; Asal, N.J. Clinical Controversy in Transplantation: Tacrolimus Versus Cyclosporine in Statin Drug Interactions. Ann. Pharmacother. 2020, 54, 171–177. [Google Scholar] [CrossRef]
  119. Wiggins, B.S.; Saseen, J.J.; Page, R.L.; Reed, B.N.; Sneed, K.; Kostis, J.B.; Lanfear, D.; Virani, S.; Morris, P.B.; On behalf of the American Heart Association Clinical Pharmacology Committee of the Council on Clinical Cardiology; et al. Recommendations for Management of Clinically Significant Drug-Drug Interactions With Statins and Select Agents Used in Patients With Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation 2016, 134, e468–e495. [Google Scholar] [CrossRef]
  120. Hecking, M.; Haidinger, M.; Döller, D.; Werzowa, J.; Tura, A.; Zhang, J.; Tekoglu, H.; Pleiner, J.; Wrba, T.; Rasoul-Rockenschaub, S.; et al. Early basal insulin therapy decreases new-onset diabetes after renal transplantation. J. Am. Soc. Nephrol. 2012, 23, 739–749. [Google Scholar] [CrossRef]
  121. Zinman, B.; Wanner, C.; Lachin, J.M.; Fitchett, D.; Bluhmki, E.; Hantel, S.; Mattheus, M.; Devins, T.; Johansen, O.E.; Woerle, H.J.; et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N. Engl. J. Med. 2015, 373, 2117–2128. [Google Scholar] [CrossRef] [PubMed]
  122. Heerspink, H.J.L.; Stefánsson, B.V.; Correa-Rotter, R.; Chertow, G.M.; Greene, T.; Hou, F.-F.; Mann, J.F.E.; McMurray, J.J.V.; Lindberg, M.; Rossing, P.; et al. Dapagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2020, 383, 1436–1446. [Google Scholar] [CrossRef] [PubMed]
  123. Sheu, J.-Y.; Chang, L.-Y.; Chen, J.-Y.; Pan, H.-C.; Tseng, C.-S.; Chueh, J.S.; Wu, V.-C. The outcomes of SGLT-2 inhibitor utilization in diabetic kidney transplant recipients. Nat. Commun. 2024, 15, 10043. [Google Scholar] [CrossRef]
  124. Lim, J.-H.; Kwon, S.; Jeon, Y.; Kim, Y.H.; Kwon, H.; Kim, Y.S.; Lee, H.; Kim, Y.-L.; Kim, C.-D.; Park, S.-H.; et al. The Efficacy and Safety of SGLT2 Inhibitor in Diabetic Kidney Transplant Recipients. Transplantation 2022, 106, e404. [Google Scholar] [CrossRef] [PubMed]
  125. Marso, S.P.; Bain, S.C.; Consoli, A.; Eliaschewitz, F.G.; Jódar, E.; Leiter, L.A.; Lingvay, I.; Rosenstock, J.; Seufert, J.; Warren, M.L.; et al. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N. Engl. J. Med. 2016, 375, 1834–1844. [Google Scholar] [CrossRef]
  126. Perkovic, V.; Tuttle, K.R.; Rossing, P.; Mahaffey, K.W.; Mann, J.F.E.; Bakris, G.; Baeres, F.M.M.; Idorn, T.; Bosch-Traberg, H.; Lausvig, N.L.; et al. Effects of Semaglutide on Chronic Kidney Disease in Patients with Type 2 Diabetes. N. Engl. J. Med. 2024, 391, 109–121. [Google Scholar] [CrossRef]
  127. Krisanapan, P.; Suppadungsuk, S.; Sanpawithayakul, K.; Thongprayoon, C.; Pattharanitima, P.; Tangpanithandee, S.; Mao, M.A.; Miao, J.; Cheungpasitporn, W. Safety and efficacy of glucagon-like peptide-1 receptor agonists among kidney transplant recipients: A systematic review and meta-analysis. Clin. Kidney J. 2024, 17, sfae018. [Google Scholar] [CrossRef]
  128. Kidney Disease: Improving Global Outcomes (KDIGO) Transplant Work Group KDIGO clinical practice guideline for the care of kidney transplant recipients. Am. J. Transplant. 2009, 9 (Suppl. S3), S1–S155. [CrossRef]
  129. Pirsch, J.D.; Henning, A.K.; First, M.R.; Fitzsimmons, W.; Gaber, A.O.; Reisfield, R.; Shihab, F.; Woodle, E.S. New-Onset Diabetes After Transplantation: Results From a Double-Blind Early Corticosteroid Withdrawal Trial. Am. J. Transplant. 2015, 15, 1982–1990. [Google Scholar] [CrossRef]
  130. Devresse, A.; Gohy, S.; Robert, A.; Kanaan, N. How to manage cigarette smoking in kidney transplant candidates and recipients? Clin. Kidney J. 2021, 14, 2295. [Google Scholar] [CrossRef]
  131. Rangaswami, J.; Mathew, R.O.; Parasuraman, R.; Tantisattamo, E.; Lubetzky, M.; Rao, S.; Yaqub, M.S.; Birdwell, K.A.; Bennett, W.; Dalal, P.; et al. Cardiovascular disease in the kidney transplant recipient: Epidemiology, diagnosis and management strategies. Nephrol. Dial. Transplant. 2019, 34, 760–773. [Google Scholar] [CrossRef] [PubMed]
  132. el-Agroudy, A.E.; Wafa, E.W.; Gheith, O.E.; Shehab el-Dein, A.B.; Ghoneim, M.A. Weight gain after renal transplantation is a risk factor for patient and graft outcome. Transplantation 2004, 77, 1381–1385. [Google Scholar] [CrossRef] [PubMed]
  133. Camilleri, B.; Bridson, J.M.; Sharma, A.; Halawa, A. From chronic kidney disease to kidney transplantation: The impact of obesity and its treatment modalities. Transplant. Rev. Orlando Fla 2016, 30, 203–211. [Google Scholar] [CrossRef]
  134. Lincoff, A.M.; Brown-Frandsen, K.; Colhoun, H.M.; Deanfield, J.; Emerson, S.S.; Esbjerg, S.; Hardt-Lindberg, S.; Hovingh, G.K.; Kahn, S.E.; Kushner, R.F.; et al. Semaglutide and Cardiovascular Outcomes in Obesity without Diabetes. N. Engl. J. Med. 2023, 389, 2221–2232. [Google Scholar] [CrossRef]
  135. Opelz, G.; Döhler, B. Cardiovascular death in kidney recipients treated with renin-angiotensin system blockers. Transplantation 2014, 97, 310–315. [Google Scholar] [CrossRef] [PubMed]
  136. Lubetzky, M.; Tantisattamo, E.; Molnar, M.Z.; Lentine, K.L.; Basu, A.; Parsons, R.F.; Woodside, K.J.; Pavlakis, M.; Blosser, C.D.; Singh, N.; et al. The failing kidney allograft: A review and recommendations for the care and management of a complex group of patients. Am. J. Transplant. 2021, 21, 2937–2949. [Google Scholar] [CrossRef]
  137. Montgomery, R.A.; Loupy, A.; Segev, D.L. Antibody-mediated rejection: New approaches in prevention and management. Am. J. Transplant. 2018, 18, 3–17. [Google Scholar] [CrossRef] [PubMed]
  138. Ho, J.; Okoli, G.N.; Rabbani, R.; Lam, O.L.T.; Reddy, V.K.; Askin, N.; Rampersad, C.; Trachtenberg, A.; Wiebe, C.; Nickerson, P.; et al. Effectiveness of T cell–mediated rejection therapy: A systematic review and meta-analysis. Am. J. Transplant. 2022, 22, 772–785. [Google Scholar] [CrossRef]
  139. Arnett, D.K.; Blumenthal, R.S.; Albert, M.A.; Buroker, A.B.; Goldberger, Z.D.; Hahn, E.J.; Himmelfarb, C.D.; Khera, A.; Lloyd-Jones, D.; McEvoy, J.W.; et al. 2019 ACC/AHA Guideline on the Primary Prevention of Cardiovascular Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019, 140, e596–e646. [Google Scholar] [CrossRef]
  140. Zhang, D.; Yu, L.; Xia, B.; Zhang, X.; Liang, P.; Hu, X. Systematic review and meta-analysis of the efficacy of exercise intervention in kidney transplant recipients. World J. Urol. 2023, 41, 3449–3469. [Google Scholar] [CrossRef]
  141. Painter, P.L.; Hector, L.; Ray, K.; Lynes, L.; Paul, S.M.; Dodd, M.; Tomlanovich, S.L.; Ascher, N.L. Effects of exercise training on coronary heart disease risk factors in renal transplant recipients. Am. J. Kidney Dis. 2003, 42, 362–369. [Google Scholar] [CrossRef] [PubMed]
  142. Gomes-Neto, A.W.; Osté, M.C.J.; Sotomayor, C.G.; van den Berg, E.; Geleijnse, J.M.; Berger, S.P.; Gans, R.O.B.; Bakker, S.J.L.; Navis, G.J. Mediterranean Style Diet and Kidney Function Loss in Kidney Transplant Recipients. Clin. J. Am. Soc. Nephrol. 2020, 15, 238–246. [Google Scholar] [CrossRef]
  143. Osté, M.C.J.; Gomes-Neto, A.W.; Corpeleijn, E.; Gans, R.O.B.; de Borst, M.H.; van den Berg, E.; Soedamah-Muthu, S.S.; Kromhout, D.; Navis, G.J.; Bakker, S.J.L. Dietary Approach to Stop Hypertension (DASH) diet and risk of renal function decline and all-cause mortality in renal transplant recipients. Am. J. Transplant. 2018, 18, 2523–2533. [Google Scholar] [CrossRef]
  144. Palmer, S.C.; Maggo, J.K.; Campbell, K.L.; Craig, J.C.; Johnson, D.W.; Sutanto, B.; Ruospo, M.; Tong, A.; Strippoli, G.F. Dietary interventions for adults with chronic kidney disease. Cochrane Database Syst. Rev. 2017, 4, CD011998. [Google Scholar] [CrossRef] [PubMed]
  145. Pedrollo, E.F.; Corrêa, C.; Nicoletto, B.B.; Corrêa Souza, G.; Leitão, C.B. What is Known About Dietary Interventions and Body Weight Management After Kidney Transplantation? A Scoping Review. J. Ren. Nutr. 2023, 33, 4–11. [Google Scholar] [CrossRef] [PubMed]
  146. Brenner, B.M.; Cooper, M.E.; de Zeeuw, D.; Keane, W.F.; Mitch, W.E.; Parving, H.H.; Remuzzi, G.; Snapinn, S.M.; Zhang, Z.; Shahinfar, S.; et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N. Engl. J. Med. 2001, 345, 861–869. [Google Scholar] [CrossRef]
  147. Midtvedt, K.; Hartmann, A.; Foss, A.; Fauchald, P.; Nordal, K.P.; Rootwelt, K.; Holdaas, H. Sustained improvement of renal graft function for two years in hypertensive renal transplant recipients treated with nifedipine as compared to lisinopril. Transplantation 2001, 72, 1787–1792. [Google Scholar] [CrossRef]
  148. Heinze, G.; Mitterbauer, C.; Regele, H.; Kramar, R.; Winkelmayer, W.C.; Curhan, G.C.; Oberbauer, R. Angiotensin-converting enzyme inhibitor or angiotensin II type 1 receptor antagonist therapy is associated with prolonged patient and graft survival after renal transplantation. J. Am. Soc. Nephrol. 2006, 17, 889–899. [Google Scholar] [CrossRef]
  149. Opelz, G.; Zeier, M.; Laux, G.; Morath, C.; Döhler, B. No improvement of patient or graft survival in transplant recipients treated with angiotensin-converting enzyme inhibitors or angiotensin II type 1 receptor blockers: A collaborative transplant study report. J. Am. Soc. Nephrol. 2006, 17, 3257–3262. [Google Scholar] [CrossRef]
  150. Knoll, G.A.; Fergusson, D.; Chassé, M.; Hebert, P.; Wells, G.; Tibbles, L.A.; Treleaven, D.; Holland, D.; White, C.; Muirhead, N.; et al. Ramipril versus placebo in kidney transplant patients with proteinuria: A multicentre, double-blind, randomised controlled trial. Lancet Diabetes Endocrinol. 2016, 4, 318–326. [Google Scholar] [CrossRef]
  151. Cheungpasitporn, W.; Thongprayoon, C.; Mao, M.A.; Kittanamongkolchai, W.; Sathick, I.J.J.; Erickson, S.B. The Effect of Renin-angiotensin System Inhibitors on Kidney Allograft Survival: A Systematic Review and Meta-analysis. N. Am. J. Med. Sci. 2016, 8, 291–296. [Google Scholar] [CrossRef] [PubMed]
  152. Kovarik, J.J.; Kaltenecker, C.C.; Kopecky, C.; Domenig, O.; Antlanger, M.; Werzowa, J.; Eskandary, F.; Kain, R.; Poglitsch, M.; Schmaldienst, S.; et al. Intrarenal Renin-Angiotensin-System Dysregulation after Kidney Transplantation. Sci. Rep. 2019, 9, 9762. [Google Scholar] [CrossRef]
  153. Ujjawal, A.; Schreiber, B.; Verma, A. Sodium-glucose cotransporter-2 inhibitors (SGLT2i) in kidney transplant recipients: What is the evidence? Ther. Adv. Endocrinol. Metab. 2022, 13, 20420188221090001. [Google Scholar] [CrossRef] [PubMed]
  154. Sánchez Fructuoso, A.I.; Bedia Raba, A.; Banegas Deras, E.; Vigara Sánchez, L.A.; Valero San Cecilio, R.; Franco Esteve, A.; Cruzado Vega, L.; Gavela Martínez, E.; González Garcia, M.E.; Saurdy Coronado, P.; et al. Sodium-glucose cotransporter-2 inhibitor therapy in kidney transplant patients with type 2 or post-transplant diabetes: An observational multicentre study. Clin. Kidney J. 2023, 16, 1022–1034. [Google Scholar] [CrossRef]
  155. Girerd, S.; Jaisser, F. Mineralocorticoid receptor antagonists in kidney transplantation: Time to consider? Nephrol. Dial. Transplant. 2018, 33, 2080–2091. [Google Scholar] [CrossRef]
  156. Mahzari, M.M.; Alluhayyan, O.B.; Almutairi, M.H.; Bayounis, M.A.; Alrayani, Y.H.; Omair, A.A.; Alshahrani, A.S. Safety and efficacy of semaglutide in post kidney transplant patients with type 2 diabetes or Post-Transplant diabetes. J. Clin. Transl. Endocrinol. 2024, 36, 100343. [Google Scholar] [CrossRef] [PubMed]
  157. Polychronopoulou, E.; Bourdon, F.; Teta, D. SGLT2 inhibitors in diabetic and non-diabetic kidney transplant recipients: Current knowledge and expectations. Front. Nephrol. 2024, 4, 1332397. [Google Scholar] [CrossRef]
  158. Birdwell, K.A.; Park, M. Post-Transplant Cardiovascular Disease. Clin. J. Am. Soc. Nephrol. 2021, 16, 1878. [Google Scholar] [CrossRef]
  159. Schiffrin, E.L. T lymphocytes: A role in hypertension? Curr. Opin. Nephrol. Hypertens. 2010, 19, 181–186. [Google Scholar] [CrossRef]
  160. Carney, E.F. Acute kidney injury: Targeting Treg cells to protect the kidney. Nat. Rev. Nephrol. 2017, 13, 444. [Google Scholar] [CrossRef]
  161. Lichtenstein, K.A.; Armon, C.; Buchacz, K.; Chmiel, J.S.; Buckner, K.; Tedaldi, E.M.; Wood, K.; Holmberg, S.D.; Brooks, J.T. HIV Outpatient Study (HOPS) Investigators Low CD4+ T cell count is a risk factor for cardiovascular disease events in the HIV outpatient study. Clin. Infect. Dis. 2010, 51, 435–447. [Google Scholar] [CrossRef] [PubMed]
  162. Roy, P.; Orecchioni, M.; Ley, K. How the immune system shapes atherosclerosis: Roles of innate and adaptive immunity. Nat. Rev. Immunol. 2022, 22, 251–265. [Google Scholar] [CrossRef] [PubMed]
  163. Sabapathy, V.; Price, A.; Cheru, N.T.; Venkatadri, R.; Dogan, M.; Costlow, G.; Mohammad, S.; Sharma, R. ST2+ T-Regulatory Cells in Renal Inflammation and Fibrosis after Ischemic Kidney Injury. J. Am. Soc. Nephrol. 2024, 36, 73–86. [Google Scholar] [CrossRef]
  164. Feng, G.; Bajpai, G.; Ma, P.; Koenig, A.; Bredemeyer, A.; Lokshina, I.; Lai, L.; Förster, I.; Leuschner, F.; Kreisel, D.; et al. CCL17 Aggravates Myocardial Injury by Suppressing Recruitment of Regulatory T Cells. Circulation 2022, 145, 765–782. [Google Scholar] [CrossRef]
  165. Ducloux, D.; Courivaud, C.; Bamoulid, J.; Crepin, T.; Chalopin, J.-M.; Tiberghien, P.; Saas, P. Polyclonal antithymocyte globulin and cardiovascular disease in kidney transplant recipients. J. Am. Soc. Nephrol. 2014, 25, 1349–1356. [Google Scholar] [CrossRef]
  166. Müller, T.F.; Grebe, S.O.; Neumann, M.C.; Heymanns, J.; Radsak, K.; Sprenger, H.; Lange, H. Persistent long-term changes in lymphocyte subsets induced by polyclonal antibodies. Transplantation 1997, 64, 1432–1437. [Google Scholar] [CrossRef]
  167. Sandal, S.; Bae, S.; McAdams-DeMarco, M.; Massie, A.B.; Lentine, K.L.; Cantarovich, M.; Segev, D.L. Induction immunosuppression agents as risk factors for incident cardiovascular events and mortality after kidney transplantation. Am. J. Transplant. 2019, 19, 1150–1159. [Google Scholar] [CrossRef]
  168. Opałka, B.; Żołnierczuk, M.; Grabowska, M. Immunosuppressive Agents-Effects on the Cardiovascular System and Selected Metabolic Aspects: A Review. J. Clin. Med. 2023, 12, 6935. [Google Scholar] [CrossRef]
  169. Javadov, S.; Jang, S.; Parodi-Rullán, R.; Khuchua, Z.; Kuznetsov, A.V. Mitochondrial permeability transition in cardiac ischemia-reperfusion: Whether cyclophilin D is a viable target for cardioprotection? Cell. Mol. Life Sci. 2017, 74, 2795–2813. [Google Scholar] [CrossRef]
  170. Bedo, D.; Beaudrey, T.; Florens, N. Unraveling Chronic Cardiovascular and Kidney Disorder through the Butterfly Effect. Diagnostics 2024, 14, 463. [Google Scholar] [CrossRef]
  171. Elezaby, A.; Dexheimer, R.; Sallam, K. Cardiovascular effects of immunosuppression agents. Front. Cardiovasc. Med. 2022, 9, 981838. [Google Scholar] [CrossRef]
  172. Solis, M.; Velay, A.; Gantner, P.; Bausson, J.; Filipputtu, A.; Freitag, R.; Moulin, B.; Caillard, S.; Fafi-Kremer, S. Torquetenovirus viremia for early prediction of graft rejection after kidney transplantation. J. Infect. 2019, 79, 56–60. [Google Scholar] [CrossRef]
  173. Hirt, S.W.; Bara, C.; Barten, M.J.; Deuse, T.; Doesch, A.O.; Kaczmarek, I.; Schulz, U.; Stypmann, J.; Haneya, A.; Lehmkuhl, H.B. Everolimus in heart transplantation: An update. J. Transplant. 2013, 2013, 683964. [Google Scholar] [CrossRef]
  174. Vincenti, F.; Rostaing, L.; Grinyo, J.; Rice, K.; Steinberg, S.; Gaite, L.; Moal, M.-C.; Mondragon-Ramirez, G.A.; Kothari, J.; Polinsky, M.S.; et al. Belatacept and Long-Term Outcomes in Kidney Transplantation. N. Engl. J. Med. 2016, 374, 333–343. [Google Scholar] [CrossRef]
  175. Ekperikpe, U.S.; Mandal, S.; Bhopatkar, A.A.; Shields, C.A.; Coley, C.A.; Chambers, C.L.; Johnson, T.D.; Cornelius, D.C.; Williams, J.M. Abatacept Decreases Renal T-cell Infiltration and Renal Inflammation and Ameliorates Progressive Renal Injury in Obese Dahl Salt-sensitive Rats Before Puberty. J. Cardiovasc. Pharmacol. 2024, 83, 635–645. [Google Scholar] [CrossRef]
  176. Kallikourdis, M.; Martini, E.; Carullo, P.; Sardi, C.; Roselli, G.; Greco, C.M.; Vignali, D.; Riva, F.; Ormbostad Berre, A.M.; Stølen, T.O.; et al. T cell costimulation blockade blunts pressure overload-induced heart failure. Nat. Commun. 2017, 8, 14680. [Google Scholar] [CrossRef]
  177. Ewing, M.M.; Karper, J.C.; Abdul, S.; de Jong, R.C.M.; Peters, H.A.B.; de Vries, M.R.; Redeker, A.; Kuiper, J.; Toes, R.E.M.; Arens, R.; et al. T-cell co-stimulation by CD28-CD80/86 and its negative regulator CTLA-4 strongly influence accelerated atherosclerosis development. Int. J. Cardiol. 2013, 168, 1965–1974. [Google Scholar] [CrossRef]
  178. Vincenti, F.; Blancho, G.; Durrbach, A.; Friend, P.; Grinyo, J.; Halloran, P.F.; Klempnauer, J.; Lang, P.; Larsen, C.P.; Mühlbacher, F.; et al. Five-year safety and efficacy of belatacept in renal transplantation. J. Am. Soc. Nephrol. 2010, 21, 1587–1596. [Google Scholar] [CrossRef]
  179. Durrbach, A.; Pestana, J.M.; Florman, S.; Del Carmen Rial, M.; Rostaing, L.; Kuypers, D.; Matas, A.; Wekerle, T.; Polinsky, M.; Meier-Kriesche, H.U.; et al. Long-Term Outcomes in Belatacept—Versus Cyclosporine-Treated Recipients of Extended Criteria Donor Kidneys: Final Results From BENEFIT-EXT, a Phase III Randomized Study. Am. J. Transplant. 2016, 16, 3192–3201. [Google Scholar] [CrossRef]
  180. Hart, A.; Lentine, K.L.; Smith, J.M.; Miller, J.M.; Skeans, M.A.; Prentice, M.; Robinson, A.; Foutz, J.; Booker, S.E.; Israni, A.K.; et al. OPTN/SRTR 2019 Annual Data Report: Kidney. Am. J. Transplant. 2021, 21, 21–137. [Google Scholar] [CrossRef]
  181. Woodle, E.S.; First, M.R.; Pirsch, J.; Shihab, F.; Gaber, A.O.; Van Veldhuisen, P. Astellas Corticosteroid Withdrawal Study Group A prospective, randomized, double-blind, placebo-controlled multicenter trial comparing early (7 day) corticosteroid cessation versus long-term, low-dose corticosteroid therapy. Ann. Surg. 2008, 248, 564–577. [Google Scholar] [CrossRef]
  182. Serrano, O.K.; Kandaswamy, R.; Gillingham, K.; Chinnakotla, S.; Dunn, T.B.; Finger, E.; Payne, W.; Ibrahim, H.; Kukla, A.; Spong, R.; et al. Rapid Discontinuation of Prednisone in Kidney Transplant Recipients: 15-Year Outcomes From the University of Minnesota. Transplantation 2017, 101, 2590–2598. [Google Scholar] [CrossRef]
  183. Bae, S.; Garonzik Wang, J.M.; Massie, A.B.; Jackson, K.R.; McAdams-DeMarco, M.A.; Brennan, D.C.; Lentine, K.L.; Coresh, J.; Segev, D.L. Early Steroid Withdrawal in Deceased-Donor Kidney Transplant Recipients with Delayed Graft Function. J. Am. Soc. Nephrol. 2020, 31, 175–185. [Google Scholar] [CrossRef]
  184. Bae, S.; Chen, Y.; Sandal, S.; Lentine, K.L.; Schnitzler, M.; Segev, D.L.; McAdams DeMarco, M.A. Association of early steroid withdrawal with kidney transplant outcomes in first-transplant and retransplant recipients. Nephrol. Dial. Transplant. 2024, gfae218. [Google Scholar] [CrossRef]
  185. Johnson, J.C.; Malik, M.; Engebretsen, T.L.; Mujtaba, M.; Lea, A.S.; Stevenson, H.L.; Kueht, M.L. Assessing Long-Term Adverse Outcomes in Older Kidney Transplant Recipients: A Propensity Score-Matched Comparison of Early Steroid Withdrawal Versus Continuous Steroid Immunosuppression Using a Large Real-World Database. Drugs Aging 2024, 41, 915–927. [Google Scholar] [CrossRef]
  186. Lok, C.E.; Huber, T.S.; Lee, T.; Shenoy, S.; Yevzlin, A.S.; Abreo, K.; Allon, M.; Asif, A.; Astor, B.C.; Glickman, M.H.; et al. KDOQI Clinical Practice Guideline for Vascular Access: 2019 Update. Am. J. Kidney Dis. 2020, 75, S1–S164. [Google Scholar] [CrossRef]
  187. Robinson, B.M.; Bieber, B.; Pisoni, R.L.; Port, F.K. Dialysis Outcomes and Practice Patterns Study (DOPPS): Its strengths, limitations, and role in informing practices and policies. Clin. J. Am. Soc. Nephrol. 2012, 7, 1897–1905. [Google Scholar] [CrossRef]
  188. Stoumpos, S.; Rankin, A.; Hall Barrientos, P.; Mangion, K.; McGregor, E.; Thomson, P.C.; Stevenson, K.; Welsh, P.; Kasthuri, R.; Kingsmore, D.B.; et al. Interrogating the haemodynamic effects of haemodialysis arteriovenous fistula on cardiac structure and function. Sci. Rep. 2021, 11, 18102. [Google Scholar] [CrossRef]
  189. Liu, Z.; Hilbelink, D.R.; Crockett, W.B.; Gerdes, A.M. Regional changes in hemodynamics and cardiac myocyte size in rats with aortocaval fistulas. 1. Developing and established hypertrophy. Circ. Res. 1991, 69, 52–58. [Google Scholar] [CrossRef]
  190. Aala, A.; Sharif, S.; Parikh, L.; Gordon, P.C.; Hu, S.L. High-Output Cardiac Failure and Coronary Steal With an Arteriovenous Fistula. Am. J. Kidney Dis. 2018, 71, 896–903. [Google Scholar] [CrossRef]
  191. Maresca, B.; Filice, F.B.; Orlando, S.; Ciavarella, G.M.; Scrivano, J.; Volpe, M.; Pirozzi, N. Early echocardiographic modifications after flow reduction by proximal radial artery ligation in patients with high-output heart failure due to high-flow forearm arteriovenous fistula. J. Vasc. Access 2020, 21, 753–759. [Google Scholar] [CrossRef]
  192. Wilmink, T.; Hollingworth, L.; Dasgupta, I. Access ligation in transplant patients. J. Vasc. Access 2016, 17 (Suppl. S1), S64–S68. [Google Scholar] [CrossRef]
  193. Vajdič Trampuž, B.; Arnol, M.; Gubenšek, J.; Ponikvar, R.; Buturović Ponikvar, J. A national cohort study on hemodialysis arteriovenous fistulas after kidney transplantation—Long-term patency, use and complications. BMC Nephrol. 2021, 22, 344. [Google Scholar] [CrossRef]
  194. Manca, O.; Pisano, G.L.; Carta, P.; Manca, E.M.; Piredda, G.B.; Pili, G.; Logias, F.; Lo Jacono, F.; Barracca, A. The management of hemodialysis arteriovenous fistulas in well functioning renal transplanted patients: Many doubts, few certainties. J. Vasc. Access 2005, 6, 182–186. [Google Scholar] [CrossRef]
  195. Rao, N.N.; Stokes, M.B.; Rajwani, A.; Ullah, S.; Williams, K.; King, D.; Macaulay, E.; Russell, C.H.; Olakkengil, S.; Carroll, R.P.; et al. Effects of Arteriovenous Fistula Ligation on Cardiac Structure and Function in Kidney Transplant Recipients. Circulation 2019, 139, 2809–2818. [Google Scholar] [CrossRef]
  196. Hetz, P.; Pirklbauer, M.; Müller, S.; Posch, L.; Gummerer, M.; Tiefenthaler, M. Prophylactic Ligature of AV Fistula Prevents High Output Heart Failure after Kidney Transplantation. Am. J. Nephrol. 2020, 51, 511–519. [Google Scholar] [CrossRef]
  197. Janeckova, J.; Bachleda, P.; Utikal, P.; Orsag, J. Management of Arteriovenous Fistula After Successful Kidney Transplantation in Long-Term Follow-Up. Transpl. Int. 2024, 37, 12841. [Google Scholar] [CrossRef]
  198. Yasir, M.B.; Man, R.K.; Gogikar, A.; Nanda, A.; Niharika Janga, L.S.; Sambe, H.G.; Mohamed, L. A Systematic Review Exploring the Impact of Arteriovenous Fistula Ligature on High-Output Heart Failure in Renal Transplant Recipients. Ann. Vasc. Surg. 2024, 100, 67–80. [Google Scholar] [CrossRef]
  199. Bangalore, S.; Guo, Y.; Samadashvili, Z.; Blecker, S.; Xu, J.; Hannan, E.L. Revascularization in Patients With Multivessel Coronary Artery Disease and Chronic Kidney Disease: Everolimus-Eluting Stents Versus Coronary Artery Bypass Graft Surgery. J. Am. Coll. Cardiol. 2015, 66, 1209–1220. [Google Scholar] [CrossRef]
  200. Bangalore, S.; Maron, D.J.; O’Brien, S.M.; Fleg, J.L.; Kretov, E.I.; Briguori, C.; Kaul, U.; Reynolds, H.R.; Mazurek, T.; Sidhu, M.S.; et al. Management of Coronary Disease in Patients with Advanced Kidney Disease. N. Engl. J. Med. 2020, 382, 1608–1618. [Google Scholar] [CrossRef]
  201. Herzog, C.A.; Simegn, M.A.; Xu, Y.; Costa, S.P.; Mathew, R.O.; El-Hajjar, M.C.; Gulati, S.; Maldonado, R.A.; Daugas, E.; Madero, M.; et al. Kidney Transplant List Status and Outcomes in the ISCHEMIA-CKD Trial. J. Am. Coll. Cardiol. 2021, 78, 348–361. [Google Scholar] [CrossRef] [PubMed]
  202. Chan, W.; Ivanov, J.; Ko, D.; Fremes, S.; Rao, V.; Jolly, S.; Cantor, W.J.; Lavi, S.; Overgaard, C.B.; Ruel, M.; et al. Clinical outcomes of treatment by percutaneous coronary intervention versus coronary artery bypass graft surgery in patients with chronic kidney disease undergoing index revascularization in Ontario. Circ. Cardiovasc. Interv. 2015, 8, e001973. [Google Scholar] [CrossRef]
  203. Li, X.; Xiao, F.; Zhang, S. Coronary revascularisation in patients with chronic kidney disease and end-stage renal disease: A meta-analysis. Int. J. Clin. Pract. 2021, 75, e14506. [Google Scholar] [CrossRef] [PubMed]
  204. SHARP Collaborative Group Study of Heart and Renal Protection (SHARP): Randomized trial to assess the effects of lowering low-density lipoprotein cholesterol among 9,438 patients with chronic kidney disease. Am. Heart J. 2010, 160, 785–794.e10. [CrossRef]
  205. Lee, M.-S.; Batiste, C.; Onwuzurike, J.; Elkoustaf, R.; Wu, Y.-L.; Chen, W.; Kahwaji, J.; Sahota, A.; Lee, R.L. Pretransplant cardiac stress testing and transplant wait time in kidney transplantation candidates. Open Heart 2024, 11, e002738. [Google Scholar] [CrossRef] [PubMed]
  206. Pullen, L.C. Rethinking coronary heart disease tests in pretransplant evaluation: Cardiologists no longer screen asymptomatic patients for coronary artery disease-so why are transplant centers still doing it? Am. J. Transplant. 2023, 23, 1087–1089. [Google Scholar] [CrossRef]
  207. Ying, T.; Gill, J.; Webster, A.; Kim, S.J.; Morton, R.; Klarenbach, S.W.; Kelly, P.; Ramsay, T.; Knoll, G.A.; Pilmore, H.; et al. Canadian-Australasian Randomised trial of screening kidney transplant candidates for coronary artery disease-A trial protocol for the CARSK study. Am. Heart J. 2019, 214, 175–183. [Google Scholar] [CrossRef]
  208. Kopparam, R.V.; Grady, D.; Redberg, R.F. Coronary Heart Disease Testing Before Kidney Transplant-A Call for Revised Guidance. JAMA Intern. Med. 2023, 183, 287–288. [Google Scholar] [CrossRef]
  209. Prud’homme, M.; Coutrot, M.; Michel, T.; Boutin, L.; Genest, M.; Poirier, F.; Launay, J.-M.; Kane, B.; Kinugasa, S.; Prakoura, N.; et al. Acute Kidney Injury Induces Remote Cardiac Damage and Dysfunction Through the Galectin-3 Pathway. JACC Basic Transl. Sci. 2019, 4, 717–732. [Google Scholar] [CrossRef]
  210. Florens, N.; Kasam, R.K.; Rudman-Melnick, V.; Lin, S.-C.; Prasad, V.; Molkentin, J.D. Interleukin-33 Mediates Cardiomyopathy After Acute Kidney Injury by Signaling to Cardiomyocytes. Circulation 2023, 147, 746–758. [Google Scholar] [CrossRef]
  211. Odutayo, A.; Wong, C.X.; Farkouh, M.; Altman, D.G.; Hopewell, S.; Emdin, C.A.; Hunn, B.H. AKI and Long-Term Risk for Cardiovascular Events and Mortality. J. Am. Soc. Nephrol. 2017, 28, 377–387. [Google Scholar] [CrossRef] [PubMed]
  212. Go, A.S.; Hsu, C.-Y.; Yang, J.; Tan, T.C.; Zheng, S.; Ordonez, J.D.; Liu, K.D. Acute Kidney Injury and Risk of Heart Failure and Atherosclerotic Events. Clin. J. Am. Soc. Nephrol. 2018, 13, 833–841. [Google Scholar] [CrossRef]
  213. Florens, N.; Aymes, E.; Gauthier, V.; Frimat, L.; Laville, M.; Bedo, D.; Beaudrey, T.; Amouyel, P.; Mansencal, N.; Lange, C.; et al. Acute kidney injury as a key predictor of cardiovascular events in chronic kidney disease patients: The CKD-REIN study. Clin. Kidney J. 2024, 17, sfae337. [Google Scholar] [CrossRef] [PubMed]
  214. Masson, P.; Webster, A.C.; Hong, M.; Turner, R.; Lindley, R.I.; Craig, J.C. Chronic kidney disease and the risk of stroke: A systematic review and meta-analysis. Nephrol. Dial. Transplant. 2015, 30, 1162–1169. [Google Scholar] [CrossRef]
  215. Kelly, D.M.; Ademi, Z.; Doehner, W.; Lip, G.Y.H.; Mark, P.; Toyoda, K.; Wong, C.X.; Sarnak, M.; Cheung, M.; Herzog, C.A.; et al. Chronic Kidney Disease and Cerebrovascular Disease: Consensus and Guidance From a KDIGO Controversies Conference. Stroke 2021, 52, e328–e346. [Google Scholar] [CrossRef] [PubMed]
  216. Bobot, M.; Suissa, L.; Hak, J.-F.; Burtey, S.; Guillet, B.; Hache, G. Kidney disease and stroke: Epidemiology and potential mechanisms of susceptibility. Nephrol. Dial. Transplant. 2023, 38, 1940–1951. [Google Scholar] [CrossRef]
  217. Bobot, M.; Guedj, E.; Resseguier, N.; Faraut, J.; Garrigue, P.; Nail, V.; Hache, G.; Gonzalez, S.; McKay, N.; Vial, R.; et al. Increased Blood-Brain Barrier Permeability and Cognitive Impairment in Patients With ESKD. Kidney Int. Rep. 2024, 9, 2988–2995. [Google Scholar] [CrossRef]
  218. Aull-Watschinger, S.; Konstantin, H.; Demetriou, D.; Schillinger, M.; Habicht, A.; Hörl, W.H.; Watschinger, B. Pre-transplant predictors of cerebrovascular events after kidney transplantation. Nephrol. Dial. Transplant. 2008, 23, 1429–1435. [Google Scholar] [CrossRef]
  219. Huang, S.-T.; Yu, T.-M.; Chuang, Y.-W.; Chung, M.-C.; Wang, C.-Y.; Fu, P.-K.; Ke, T.-Y.; Li, C.-Y.; Lin, C.-L.; Wu, M.-J.; et al. The Risk of Stroke in Kidney Transplant Recipients with End-Stage Kidney Disease. Int. J. Environ. Res. Public. Health 2019, 16, 326. [Google Scholar] [CrossRef]
  220. Lentine, K.L.; Rocca Rey, L.A.; Kolli, S.; Bacchi, G.; Schnitzler, M.A.; Abbott, K.C.; Xiao, H.; Brennan, D.C. Variations in the risk for cerebrovascular events after kidney transplant compared with experience on the waiting list and after graft failure. Clin. J. Am. Soc. Nephrol. 2008, 3, 1090–1101. [Google Scholar] [CrossRef]
  221. Oliveras, A.; Roquer, J.; Puig, J.M.; Rodríguez, A.; Mir, M.; Orfila, M.A.; Masramon, J.; Lloveras, J. Stroke in renal transplant recipients: Epidemiology, predictive risk factors and outcome. Clin. Transplant. 2003, 17, 1–8. [Google Scholar] [CrossRef] [PubMed]
  222. de Mattos, A.M.; Prather, J.; Olyaei, A.J.; Shibagaki, Y.; Keith, D.S.; Mori, M.; Norman, D.J.; Becker, T. Cardiovascular events following renal transplantation: Role of traditional and transplant-specific risk factors. Kidney Int. 2006, 70, 757–764. [Google Scholar] [CrossRef] [PubMed]
  223. Kasiske, B.L.; Guijarro, C.; Massy, Z.A.; Wiederkehr, M.R.; Ma, J.Z. Cardiovascular disease after renal transplantation. J. Am. Soc. Nephrol. 1996, 7, 158–165. [Google Scholar] [CrossRef]
  224. Zhang, L.; Wang, Z.; Lv, J.; Zheng, M.; Zhu, Y. Outcomes of acute ischemic stroke in kidney transplant recipients: An analysis of US Nationwide inpatient sample. Transl. Neurosci. 2022, 13, 327–334. [Google Scholar] [CrossRef] [PubMed]
  225. Kwon, H.; Shin, S.; Baek, C.H.; Chang, J.Y.; Kang, D.-W.; Kwon, S.U.; Kim, J.S.; Kim, B.J. Characteristics of stroke after liver and kidney transplantation. Front. Neurol. 2023, 14, 1123518. [Google Scholar] [CrossRef]
  226. Weng, S.-F.; Shen, Y.-C.; Wang, J.-J.; Tien, K.-J. Reduced risk of new onset stroke after kidney transplantation in Asian dialysis patients: A propensity score-matched, competing risk study in Taiwan. QJM Mon. J. Assoc. Physicians 2019, 112, 489–495. [Google Scholar] [CrossRef]
  227. Mathew, A.; Eliasziw, M.; Devereaux, P.J.; Merino, J.G.; Barnett, H.J.M.; Garg, A.X. North American Symptomatic Carotid Endarterectomy Trial (NASCET) Collaborators Carotid endarterectomy benefits patients with CKD and symptomatic high-grade stenosis. J. Am. Soc. Nephrol. 2010, 21, 145–152. [Google Scholar] [CrossRef]
  228. Paraskevas, K.I.; AbuRahma, A.F. A comparison of the 2022 Society for Vascular Surgery and the 2023 European Society for Vascular Surgery guidelines for the management of patients with asymptomatic and symptomatic carotid stenosis. J. Vasc. Surg. 2024, 79, 1272–1275. [Google Scholar] [CrossRef]
  229. Reiff, T.; Eckstein, H.-H.; Mansmann, U.; Jansen, O.; Fraedrich, G.; Mudra, H.; Böckler, D.; Böhm, M.; Debus, E.S.; Fiehler, J.; et al. Carotid endarterectomy or stenting or best medical treatment alone for moderate-to-severe asymptomatic carotid artery stenosis: 5-year results of a multicentre, randomised controlled trial. Lancet Neurol. 2022, 21, 877–888. [Google Scholar] [CrossRef]
  230. Adil, M.M.; Saeed, F.; Chaudhary, S.A.; Malik, A.; Qureshi, A.I. Comparative Outcomes of Carotid Artery Stent Placement and Carotid Endarterectomy in Patients with Chronic Kidney Disease and End-Stage Renal Disease. J. Stroke Cerebrovasc. Dis. 2016, 25, 1721–1727. [Google Scholar] [CrossRef]
  231. Hindricks, G.; Potpara, T.; Dagres, N.; Arbelo, E.; Bax, J.J.; Blomström-Lundqvist, C.; Boriani, G.; Castella, M.; Dan, G.-A.; Dilaveris, P.E.; et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): The Task Force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur. Heart J. 2021, 42, 373–498. [Google Scholar] [CrossRef] [PubMed]
  232. Wetmore, J.B.; Mahnken, J.D.; Rigler, S.K.; Ellerbeck, E.F.; Mukhopadhyay, P.; Spertus, J.A.; Hou, Q.; Shireman, T.I. The prevalence of and factors associated with chronic atrial fibrillation in Medicare/Medicaid-eligible dialysis patients. Kidney Int. 2012, 81, 469–476. [Google Scholar] [CrossRef] [PubMed]
  233. Guo, Y.; Gao, J.; Ye, P.; Xing, A.; Wu, Y.; Wu, S.; Luo, Y. Comparison of atrial fibrillation in CKD and non-CKD populations: A cross-sectional analysis from the Kailuan study. Int. J. Cardiol. 2019, 277, 125–129. [Google Scholar] [CrossRef]
  234. Song, J.; Navarro-Garcia, J.A.; Wu, J.; Saljic, A.; Abu-Taha, I.; Li, L.; Lahiri, S.K.; Keefe, J.A.; Aguilar-Sanchez, Y.; Moore, O.M.; et al. Chronic kidney disease promotes atrial fibrillation via inflammasome pathway activation. J. Clin. Investig. 2023, 133, e167517. [Google Scholar] [CrossRef]
  235. Pokorney, S.D.; Chertow, G.M.; Al-Khalidi, H.R.; Gallup, D.; Dignacco, P.; Mussina, K.; Bansal, N.; Gadegbeku, C.A.; Garcia, D.A.; Garonzik, S.; et al. Apixaban for Patients With Atrial Fibrillation on Hemodialysis: A Multicenter Randomized Controlled Trial. Circulation 2022, 146, 1735–1745. [Google Scholar] [CrossRef]
  236. Fu, E.L.; Desai, R.J.; Paik, J.M.; Kim, D.H.; Zhang, Y.; Mastrorilli, J.M.; Cervone, A.; Lin, K.J. Comparative Safety and Effectiveness of Warfarin or Rivaroxaban Versus Apixaban in Patients With Advanced CKD and Atrial Fibrillation: Nationwide US Cohort Study. Am. J. Kidney Dis. 2024, 83, 293–305.e1. [Google Scholar] [CrossRef]
  237. Elenjickal, E.J.; Travlos, C.K.; Marques, P.; Mavrakanas, T.A. Anticoagulation in Patients with Chronic Kidney Disease. Am. J. Nephrol. 2024, 55, 146–164. [Google Scholar] [CrossRef]
  238. Albaladejo, P.; Bonhomme, F.; Blais, N.; Collet, J.-P.; Faraoni, D.; Fontana, P.; Godier, A.; Llau, J.; Longrois, D.; Marret, E.; et al. Management of direct oral anticoagulants in patients undergoing elective surgeries and invasive procedures: Updated guidelines from the French Working Group on Perioperative Hemostasis (GIHP)—September 2015. Anaesth. Crit. Care Pain Med. 2017, 36, 73–76. [Google Scholar] [CrossRef]
  239. Wazni, O.M.; Saliba, W.I.; Nair, D.G.; Marijon, E.; Schmidt, B.; Hounshell, T.; Ebelt, H.; Skurk, C.; Oza, S.; Patel, C.; et al. Left Atrial Appendage Closure after Ablation for Atrial Fibrillation. N. Engl. J. Med. 2024. [Google Scholar] [CrossRef] [PubMed]
  240. Mora, M.M.R.; Reis, A.M.; Tavares, F.P.; Oliveira, L.S.; Godoi, A.; Viana, P.; Riella, J. Safety and efficacy of direct oral anticoagulants in kidney transplant recipients: A systematic review and meta-analysis. Transplant. Rev. 2024, 39, 100899. [Google Scholar] [CrossRef]
  241. Thongprayoon, C.; Chokesuwattanaskul, R.; Bathini, T.; Khoury, N.J.; Sharma, K.; Ungprasert, P.; Prasitlumkum, N.; Aeddula, N.R.; Watthanasuntorn, K.; Salim, S.A.; et al. Epidemiology and Prognostic Importance of Atrial Fibrillation in Kidney Transplant Recipients: A Meta-Analysis. J. Clin. Med. 2018, 7, 370. [Google Scholar] [CrossRef]
  242. Criqui, M.H.; Matsushita, K.; Aboyans, V.; Hess, C.N.; Hicks, C.W.; Kwan, T.W.; McDermott, M.M.; Misra, S.; Ujueta, F. Lower Extremity Peripheral Artery Disease: Contemporary Epidemiology, Management Gaps, and Future Directions: A Scientific Statement From the American Heart Association. Circulation 2021, 144, e171–e191. [Google Scholar] [CrossRef]
  243. Abu Dabrh, A.M.; Steffen, M.W.; Undavalli, C.; Asi, N.; Wang, Z.; Elamin, M.B.; Conte, M.S.; Murad, M.H. The natural history of untreated severe or critical limb ischemia. J. Vasc. Surg. 2015, 62, 1642–1651.e3. [Google Scholar] [CrossRef]
  244. Johansen, K.L.; Garimella, P.S.; Hicks, C.W.; Kalra, P.A.; Kelly, D.M.; Martens, S.; Matsushita, K.; Sarafidis, P.; Sood, M.M.; Herzog, C.A.; et al. Central and peripheral arterial diseases in chronic kidney disease: Conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int. 2021, 100, 35–48. [Google Scholar] [CrossRef]
  245. Bourrier, M.; Ferguson, T.W.; Embil, J.M.; Rigatto, C.; Komenda, P.; Tangri, N. Peripheral Artery Disease: Its Adverse Consequences With and Without CKD. Am. J. Kidney Dis. 2020, 75, 705–712. [Google Scholar] [CrossRef]
  246. Tsuyuki, K.; Kohno, K.; Ebine, K.; Obara, T.; Aoki, T.; Muto, A.; Ninomiya, K.; Kumagai, K.; Yokouchi, I.; Yazaki, Y.; et al. Exercise-ankle brachial pressure index with one-minute treadmill walking in patients on maintenance hemodialysis. Ann. Vasc. Dis. 2013, 6, 52–56. [Google Scholar] [CrossRef]
  247. Arinze, N.V.; Gregory, A.; Francis, J.M.; Farber, A.; Chitalia, V.C. Unique aspects of peripheral artery disease in patients with chronic kidney disease. Vasc. Med. Lond. Engl. 2019, 24, 251–260. [Google Scholar] [CrossRef]
  248. Hernández, D.; Vázquez, T.; Armas-Padrón, A.M.; Alonso-Titos, J.; Casas, C.; Gutiérrez, E.; Jironda, C.; Cabello, M.; López, V. Peripheral Vascular Disease and Kidney Transplant Outcomes: Rethinking an Important Ongoing Complication. Transplantation 2021, 105, 1188–1202. [Google Scholar] [CrossRef]
  249. Snyder, J.J.; Kasiske, B.L.; Maclean, R. Peripheral arterial disease and renal transplantation. J. Am. Soc. Nephrol. 2006, 17, 2056–2068. [Google Scholar] [CrossRef] [PubMed]
  250. Brar, A.; Jindal, R.M.; Sumrani, N.; John, D.; Mondal, Z.; Tedla, F.; Salifu, M.O. Impact of renal posttransplantation amputation on allograft outcomes: A study of United States renal data system. Transplantation 2013, 95, 1249–1253. [Google Scholar] [CrossRef] [PubMed]
  251. Brar, A.; Jindal, R.M.; Elster, E.A.; Tedla, F.; John, D.; Sumrani, N.; Salifu, M.O. Effect of peripheral vascular disease on kidney allograft outcomes: A study of U.S. Renal data system. Transplantation 2013, 95, 810–815. [Google Scholar] [CrossRef]
  252. Patel, S.I.; Chakkera, H.A.; Wennberg, P.W.; Liedl, D.A.; Alrabadi, F.; Cha, S.S.; Hooley, D.D.; Amer, H.; Wadei, H.M.; Shamoun, F.E. Peripheral arterial disease preoperatively may predict graft failure and mortality in kidney transplant recipients. Vasc. Med. Lond. Engl. 2017, 22, 225–230. [Google Scholar] [CrossRef] [PubMed]
  253. Franquet, Q.; Terrier, N.; Pirvu, A.; Rambeaud, J.-J.; Long, J.-A.; Janbon, B.; Tetaz, R.; Malvezzi, P.; Jouve, T.; Descotes, J.-L.; et al. Aortic bypass surgery for asymptomatic patients awaiting a kidney transplant: A word of caution. Clin. Transplant. 2018, 32, e13218. [Google Scholar] [CrossRef]
  254. Kasiske, B.L.; Cangro, C.B.; Hariharan, S.; Hricik, D.E.; Kerman, R.H.; Roth, D.; Rush, D.N.; Vazquez, M.A.; Weir, M.R. American Society of Transplantation The evaluation of renal transplantation candidates: Clinical practice guidelines. Am. J. Transplant. 2001, 1 (Suppl. S2), 3–95. [Google Scholar] [PubMed]
  255. Northcutt, A.; Zibari, G.; Tan, T.-W.; Coulter, A.H.; Zhang, W.W. Does kidney transplantation to iliac artery deteriorate ischemia in the ipsilateral lower extremity with peripheral arterial disease? Vascular 2015, 23, 490–493. [Google Scholar] [CrossRef] [PubMed]
  256. Monteiro, S.S.; Santos, T.S.; Pereira, C.A.; Duarte, D.B.; Neto, H.; Gomes, A.; Loureiro, L.; Martins, J.; Silva, F.; Martins, L.S.; et al. The influence of simultaneous pancreas-kidney transplantation on the evolution of diabetic foot lesions and peripheral arterial disease. J. Endocrinol. Investig. 2023, 46, 1459–1464. [Google Scholar] [CrossRef]
  257. Sucher, R.; Rademacher, S.; Jahn, N.; Brunotte, M.; Wagner, T.; Alvanos, A.; Sucher, E.; Seehofer, D.; Scheuermann, U.; Hau, H.-M. Effects of simultaneous pancreas-kidney transplantation and kidney transplantation alone on the outcome of peripheral vascular diseases. BMC Nephrol. 2019, 20, 453. [Google Scholar] [CrossRef]
Figure 1. Evolution of classical and non-classical risk factors over time and their impact on CVD occurrence. (A) Distribution of cardiovascular risk factors based on their impact on cardiovascular disease (CVD) risk and their timing relative to transplantation. The vertical axis represents the magnitude of impact on cardiovascular risk, while the horizontal axis indicates the time before or after transplantation (zero = day of transplantation). Classical risk factors are shown in light blue, whereas non-classical risk factors are depicted in dark blue. (B) Evolution of cardiovascular risk over time. Periods of gradual increase in CVD risk are represented in green, periods of significant increase are shown in orange, and the phase with the highest impact on CVD risk is highlighted in red. Abbreviations: MACE, major adverse cardiovascular event; CVD, cardiovascular disease; CNI, calcineurin inhibitor; DGF, delayed graft function; ESRD, end-stage renal disease; CV risk, cardiovascular risk.
Figure 1. Evolution of classical and non-classical risk factors over time and their impact on CVD occurrence. (A) Distribution of cardiovascular risk factors based on their impact on cardiovascular disease (CVD) risk and their timing relative to transplantation. The vertical axis represents the magnitude of impact on cardiovascular risk, while the horizontal axis indicates the time before or after transplantation (zero = day of transplantation). Classical risk factors are shown in light blue, whereas non-classical risk factors are depicted in dark blue. (B) Evolution of cardiovascular risk over time. Periods of gradual increase in CVD risk are represented in green, periods of significant increase are shown in orange, and the phase with the highest impact on CVD risk is highlighted in red. Abbreviations: MACE, major adverse cardiovascular event; CVD, cardiovascular disease; CNI, calcineurin inhibitor; DGF, delayed graft function; ESRD, end-stage renal disease; CV risk, cardiovascular risk.
Diagnostics 15 00802 g001
Figure 2. Simplified algorithm for cardiac screening in potential asymptomatic kidney recipients, adapted from Cheng et al. [90]. This algorithm does not apply to cases involving symptoms or clinical signs of cardiac involvement, which require specialized evaluation. Guideline-directed medical therapy (GDMT) remains indicated in all scenarios. (1) For non-ischemic abnormalities, pulmonary hypertension, or valvular disease: specific management tailored to the condition is necessary. (2) If GDMT proves inadequate, referral to a specialist is recommended, or reconsideration of transplant listing may be warranted. (3) If left ventricular ejection fraction (LVEF) remains < 40% despite optimal treatment, referral to a specialist or reconsideration of transplant listing is advised. Abbreviations: TTE, transthoracic echocardiography; ECG, electrocardiogram; CAD, coronary artery disease; LVEF, left ventricular ejection fraction; PAD, peripheral artery disease; MI, myocardial infarction; GDMT, guideline-directed medical therapy.
Figure 2. Simplified algorithm for cardiac screening in potential asymptomatic kidney recipients, adapted from Cheng et al. [90]. This algorithm does not apply to cases involving symptoms or clinical signs of cardiac involvement, which require specialized evaluation. Guideline-directed medical therapy (GDMT) remains indicated in all scenarios. (1) For non-ischemic abnormalities, pulmonary hypertension, or valvular disease: specific management tailored to the condition is necessary. (2) If GDMT proves inadequate, referral to a specialist is recommended, or reconsideration of transplant listing may be warranted. (3) If left ventricular ejection fraction (LVEF) remains < 40% despite optimal treatment, referral to a specialist or reconsideration of transplant listing is advised. Abbreviations: TTE, transthoracic echocardiography; ECG, electrocardiogram; CAD, coronary artery disease; LVEF, left ventricular ejection fraction; PAD, peripheral artery disease; MI, myocardial infarction; GDMT, guideline-directed medical therapy.
Diagnostics 15 00802 g002
Table 3. Main cardiovascular and kidney adverse effects of maintenance therapies in kidney transplantation adapted from KDIGO 2009, Opalka et al. [168] and Elezaby et al. [168] Abbreviations: CTC, corticosteroid, CsA, cyclosporine A, TAC, tacrolimus, imTor, mTOR inhibitors, MMF, mycophenolate mofetil; Bela, belatacept, HTN, hypertension, GFR, glomerular filtration rate, ↓: protective, ↑: mild to moderate side effect, ↑↑: moderate to severe side effect.
Table 3. Main cardiovascular and kidney adverse effects of maintenance therapies in kidney transplantation adapted from KDIGO 2009, Opalka et al. [168] and Elezaby et al. [168] Abbreviations: CTC, corticosteroid, CsA, cyclosporine A, TAC, tacrolimus, imTor, mTOR inhibitors, MMF, mycophenolate mofetil; Bela, belatacept, HTN, hypertension, GFR, glomerular filtration rate, ↓: protective, ↑: mild to moderate side effect, ↑↑: moderate to severe side effect.
Side Effects of Immunosuppressive TreatmentCTCCsATACimTorMMFBelatacept
Post-transplantation diabetes melitus↑↑↑↑
Dyslipidemia↑↑↑↑
Hypertension (HTN)↑↑↑↑
Glomerular proteinuria ↑↑
Kidney fibrosis ↑↑↑↑
Cardiac hypertrophy, fibrosis↑↑↑↑↑↑
Vascular remodeling
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Beaudrey, T.; Bedo, D.; Weschler, C.; Caillard, S.; Florens, N. From Risk Assessment to Management: Cardiovascular Complications in Pre- and Post-Kidney Transplant Recipients: A Narrative Review. Diagnostics 2025, 15, 802. https://doi.org/10.3390/diagnostics15070802

AMA Style

Beaudrey T, Bedo D, Weschler C, Caillard S, Florens N. From Risk Assessment to Management: Cardiovascular Complications in Pre- and Post-Kidney Transplant Recipients: A Narrative Review. Diagnostics. 2025; 15(7):802. https://doi.org/10.3390/diagnostics15070802

Chicago/Turabian Style

Beaudrey, Thomas, Dimitri Bedo, Célia Weschler, Sophie Caillard, and Nans Florens. 2025. "From Risk Assessment to Management: Cardiovascular Complications in Pre- and Post-Kidney Transplant Recipients: A Narrative Review" Diagnostics 15, no. 7: 802. https://doi.org/10.3390/diagnostics15070802

APA Style

Beaudrey, T., Bedo, D., Weschler, C., Caillard, S., & Florens, N. (2025). From Risk Assessment to Management: Cardiovascular Complications in Pre- and Post-Kidney Transplant Recipients: A Narrative Review. Diagnostics, 15(7), 802. https://doi.org/10.3390/diagnostics15070802

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

Article Metrics

Back to TopTop