Review Open Access
Copyright ©2014 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Transplant. Jun 24, 2014; 4(2): 93-101
Published online Jun 24, 2014. doi: 10.5500/wjt.v4.i2.93
Coronary microvasculopathy in heart transplantation: Consequences and therapeutic implications
Alessandra Vecchiati, Sara Tellatin, Annalisa Angelini, Sabino Iliceto, Francesco Tona, Department of Cardiac, Thoracic and Vascular Sciences, Padova University Hospital, 35128 Padova, Italy
Author contributions: Vecchiati A wrote the manuscript; Tona F supervised and discussed this manuscript; all authors contributed to this work.
Correspondence to: Francesco Tona, MD, PhD, Department of Cardiac, Thoracic and Vascular Sciences, Padova University Hospital, via Giustiniani 2, 35128 Padova, Italy. francesco.tona@unipd.it
Telephone: +39-49-8211844  Fax: +39-49-8211802
Received: November 26, 2013
Revised: January 11, 2014
Accepted: March 11, 2014
Published online: June 24, 2014
Processing time: 236 Days and 14.8 Hours

Abstract

Despite the progress made in the prevention and treatment of rejection of the transplanted heart, cardiac allograft vasculopathy (CAV) remains the main cause of death in late survival transplanted patients. CAV consists of a progressive diffuse intimal hyperplasia and the proliferation of vascular smooth muscle cells, ending in wall thickening of epicardial vessels, intramyocardial arteries (50-20 μm), arterioles (20-10 μm), and capillaries (< 10 μm). The etiology of CAV remains unclear; both immunologic and non-immunologic mechanisms contribute to endothelial damage with a sustained inflammatory response. The immunological factors involved are Human Leukocyte Antigen compatibility between donor and recipient, alloreactive T cells and the humoral immune system. The non-immunological factors are older donor age, ischemia-reperfusion time, hyperlipidemia and CMV infections. Diagnostic techniques that are able to assess microvascular function are lacking. Intravascular ultrasound and fractional flow reserve, when performed during coronary angiography, are able to detect epicardial coronary artery disease but are not sensitive enough to assess microvascular changes. Some authors have proposed an index of microcirculatory resistance during maximal hyperemia, which is calculated by dividing pressure by flow (distal pressure multiplied by the hyperemic mean transit time). Non-invasive methods to assess coronary physiology are stress echocardiography, coronary flow reserve by transthoracic Doppler echocardiography, single photon emission computed tomography, and perfusion cardiac magnetic resonance. In this review, we intend to analyze the mechanisms, consequences and therapeutic implications of microvascular dysfunction, including an extended citation of relevant literature data.

Key Words: Heart transplantation, Cardiac allograft vasculopathy, Microvascular function, Coronary flow reserve, Endothelial dysfunction

Core tip: In this review, we intend to analyze the mechanisms, consequences and therapeutic implications of microvascular dysfunction in heart transplantation recipients, including an extended citation of relevant data from the literature. We think that this manuscript could be of interest for many research workers and physicians working in the field of cardiovascular surgery, cardiology and transplant medicine.



INTRODUCTION

Heart transplantation (HT) is the most effective treatment for patients with end-stage heart failure. Recently, early survival after HT has been improved through the use of immunosuppressive therapy and updated surgical procedures. Unfortunately, late survival is still limited by the onset of malignancies and cardiac allograft vasculopathy (CAV). CAV is a specific form of coronary artery disease that affects heart transplanted patients and is characterized by an early, diffuse intimal proliferation of both the epicardial and microvascular vessels, resulting in epicardial coronary artery stenosis and small vessel occlusion[1]. The 29th Official Adult Heart Transplant Report, edited by the Registry of Heart and Lung Transplantation, noted a relatively small decrease in the cumulative incidence of CAV: at 7 years after transplant, 37% of the patients transplanted between 2003 and June 2010 had CAV, compared with 42% of those transplanted between April 1994 and 2002. In fact, CAV affects 8% by year 1, 30% by year 5 and 50% by year 10 after transplant[2]. This decrease seems to be related to newer approaches to CAV treatment, such as targeting lower low-density lipoproteins (LDL)-cholesterol levels or the use of mammalian target of rapamycin (mTOR) inhibitors or drug-eluting coronary stents[3]. The 1-year survival rate after HT is 81%, and the 5-year survival rate is 69%, with a median survival of 11 years for all HT patients and 13 years for those surviving the first year. CAV causes approximately 10%-15% of the deaths between years 1 and 3 after HT and contributes to potentially more deaths resulting from graft dysfunction[4]. Epicardial coronary artery disease is detectable by intravascular ultrasound (IVUS) during coronary angiography. Coronary microvascular function can be assessed by transthoracic Doppler echocardiography (TDE) measuring coronary flow reserve (CFR)[5]. Understanding the physiopathology of endothelial and microvascular dysfunction in CAV plays a crucial role in the development of new therapies.

THE ROLE OF ENDOTHELIAL FUNCTION

Coronary endothelial vasodilator dysfunction is a common finding in HT recipients and is an early marker for the development of intimal thickening and graft atherosclerosis. Since 1988, a paradoxical coronary vasoconstriction to acetylcholine in allograft recipients with and without angiographic evidence of CAV has been observed[6]. Subsequently, other investigators have observed abnormal responses (vasoconstriction and/or impairment in coronary blood flow response) to serotonin, substance P, cold-pressor testing, and exercise[7-10]. The impairment of endothelial function is time-dependent. Endothelial dysfunction is caused by both immunological and non-immunological risk factors[11]. The immunological response is the principal initiating stimulus and results in endothelial injury and dysfunction and altered endothelial permeability, with consequent myo-intimal hyperplasia and extracellular matrix synthesis. Non-immunological events, including ischemia/reperfusion time, donor age, donor brain death, infections (i.e., Cytomegalovirus, CMV) and traditional risk factors such as hypertension, dyslipidemia and diabetes, contribute to maintaining inflammatory responses and to extend vessel damage[12-14].

Immunological response

Alloimmune injury is initiated when donor major histocompatibility antigens expressed on the surface of graft endothelial cells interact with recipient dendridic cells, resulting in a chronic immune response[15]. Recipient CD4+ lymphocytes recognize donor major histocompatibility complex (MHC) class II antigens on the cell’s surface (HLA-DR, DP and DQ) and are activated. This process leads to a cascade of cytokines, such as Interleukin-2 (IL-2), IL-4, IL-5, IL-6, interferon-γ (IFN-γ), and tumor necrosis factor α and β (TNF-α, TNF-β), which promote the proliferation of alloreactive T cells and stimulate the expression of other cytokines and adhesion molecules (i.e., intercellular adhesion molecule-1, ICAM, and vascular cell adhesion molecule-1, VCAM) by the endothelium with leukocyte adhesion to the vessel wall. As a result, the activated macrophages and lymphocytes in the intima of the artery secrete platelet-derived growth factor and transforming growth factor, which stimulate the proliferation of smooth muscle cells (SMCs) and vascular remodeling[16]. Non-human leukocyte antigen (HLA) allo- and auto-antibodies are an increasingly recognized component of the immune response. They are often directed against angiotensin type-1 receptor and the endothelin-1 type A receptor and may alone induce endothelial activation, trigger proinflammatory, and both proproliferative and profibrotic responses[17-19].

Nitric oxide pathway

Cytokines and growth factors lead to coronary endothelial vasodilator dysfunction through the dysregulation of the L-arginine-nitric oxide pathway, resulting in the reduced synthesis and bioactivity of the vasodilators in favor of endothelium-derived vasoconstrictors such as endothelin (ET) and thromboxane. Endothelium-derived nitric oxide (NO) is the most potent endogenous vasodilator known. It induces vasodilatation by stimulating soluble guanylate cyclase to produce cyclic guanosine monophosphate and inhibits platelet and leukocyte adherence to the vessel wall. IFN-γ is the determinant mediator, linking endothelial dysfunction to structural changes in transplanted human arteries through the down-regulation of endothelial NO synthase (eNOS) expression, inducible-NOS (iNOS) activation and potentiating growth-factor-induced SMC mitogenesis. The iNOS is not a normal constituent of quiescent healthy cells but is expressed in a wide variety of cell types that have been exposed to bacterial endotoxin or combinations of inflammatory cytokines. Under conditions of reduced availability of L-arginine (the NO precursor), the product of iNOS is the superoxide anion, which can increase local oxidative stress and exacerbate the inflammatory process[10,20,21]. The increased production of reactive oxygen species (ROS) is considered a major determinant of reduced levels of NO[22]. In human cardiac allografts, enhanced endomyocardial iNOS mRNA expression is accompanied by the expression of nitrotyrosine protein, suggesting peroxynitrite-mediated vessel damage. Importantly, dietary L-arginine has been shown to attenuate the structural changes of CAV in vivo and has been associated with the down-regulation of insulin-like growth factor-I and IL-6[10]. Recently, great importance has been attributed to the ratio of L-arginine/asymmetric dimethylarginine (ADMA), which is an endogenous NO synthase inhibitor. ADMA is normally produced by the hydrolysis of proteins and degraded by the oxidant-sensitive enzyme dimethylarginine dimethyl aminohydrolase (DDAH)[23]. An increase in the ADMA levels of HT patients has been observed due to an oxidative impairment of the DDAH. The loss of endothelium-derived NO permits the increased activity of the pro-inflammatory transcription nuclear factor kappa B (NF-κB), resulting in the expression of leukocyte adhesion molecules[22].

Non-immunological mechanisms

Non-immunological risk factors for endothelial dysfunction are the same as those observed in non-transplanted patients, such as CMV infections, diabetes and dyslipidemia. CMV infection of seronegative HT recipients plays an important role in CAV development. It increases the ADMA levels, generates ROS and, through NF-κB activation and TNF-α production, induces proinflammatory cytokines and destabilizes the mRNA message for eNOS[24]. Donor- or recipient-related factors (e.g., age/gender, pre-transplant diagnosis) and factors related to surgery (e.g., ischemia-reperfusion injury) also increase the risk of CAV[25,26]. Diabetes mellitus is present in 28% of recipients at 1 year after HT and in 40% of patients at 5 years after HT[4]. Risk factors for new-onset diabetes include pre-transplant blood glucose of > 5.6 mmol/L, a family history of diabetes, being overweight, and the pre-transplant use of immunosuppressive drugs, particularly calcineurin inhibitors and corticosteroids[27].

Insulin resistance impedes the removal of triglycerides (TG) from very-low-density lipoproteins (VLDL) that are in circulation, resulting in hypertriglyceridemia and high VLDL concentrations. This impedance increases the transfer of cholesterol from high-density lipoproteins (HDL), thus decreasing the HDL concentrations and forming small cholesterol-depleted LDL[28]. These small dense LDL particles are rich in TG but contain relatively little cholesterol and are not readily cleared by the physiological LDL receptor; these particles are highly atherogenic[29]. Markers of metabolic syndrome such as a TG/HDL ratio of ≥ 3 and levels of C-reactive protein (CRP) > 3 mg/L are considered markers of insulin resistance and may lead to endothelial dysfunction and the development of CAV[28]. Hyperlipidemia occurs frequently in HT recipients, with pre-existing or similar conditions to treatment with calcineurin inhibitors and corticosteroids. Hyperlipidemia leads to an increased intimal thickening, but there is only limited evidence that shows its direct association with CAV development[28]. Importantly, the benefits from statin therapy are well documented. Early treatment has been reported to be beneficial to first-year survival and has helped reduce severe rejection, thereby decreasing the development of CAV[30]. Statins inhibit MHC II induction by IFN-γ on primary human endothelial cells and monocytes-macrophages and may exert a dampening effect on MHC II-mediated T lymphocyte activation[31].

HISTOPATHOLOGICAL FEATURES

The precise interaction between host and donor endothelium remains unclear, but there is a significant amount of data showing a partial re-endothelization from recipient-derived cells, possibly as a response to allogenic stimuli causing vascular injury[32-34]. Endothelial chimerism (the coexistence of both donor and recipient endothelial cells) has been shown to be much higher in the microcirculation than in larger vessels, with a predilection for small epicardial and intramyocardial vessels, which had a notable 3- to 5-fold-greater chimerism than their larger counterparts. The high degree of endothelial chimerism may have immune implications for myocardial rejection or graft vasculopathy[33-37]. It has been hypothesized that this replacement could lead to a decrease in alloreactivity with a positive influence on graft outcome, but further studies are needed[38].

A study conducted by our group investigated the correlation between levels of human endothelial circulating progenitor cells (EPCs) and microvascular dysfunction, as evaluated by CFR. We demonstrated that EPCs in both the circulation and the graft decrease significantly in HT recipients with microvascular damage. A possible explanation for this may involve humoral factors that occur in a chronic low-grade rejection and influence mobilization, migration, and cell survival[39,40].

Hiemann et al[41] established a grading system of microvasculopathy in post-transplantation biopsies by light microscopy. The endothelial layer was defined as the mono-cell layer at the inner part of the blood vessel wall. The presence of a thin layer of cells whose diameter was less than the diameter of the endothelial cell cores was considered normal. Endothelial cells were graded as thickened if the diameter of the cell layer was at least as thick as the endothelial cell cores. The wall layer (media) was defined as the poly-cell layer adjacent to the endothelium. The wall was graded as normal if its diameter was less than the luminal radius. Wall thickening was classified as non-stenotic if the ratio of the luminal radius to wall thickness was < 3 but ≥ 1, and stenotic wall thickening was graded if this ratio was < 1 (Table 1). Stenotic microvasculopathy was diagnosed if there was evidence of microvascular stenosis due to either endothelial thickening or wall thickening in at least one blood vessel per field of view on endomyocardial biopsies[41].

Table 1 Different definitions of microvasculopathy.
AuthorMicrovesselsdiameter (μm)Microvasculopathy assessment
Drakos et al[97]< 60Microvascular density (number of microvessels/total tissue analysis area)
Escaned et al[96]< 100Arteriolar density, capillary and arteriolar obliteration index
Hiemann et al[41]10-20Luminal radius/medial thickness < 1
MICROVASCULOPATHY: DIAGNOSTIC TOOLS

Microvascular disease can be detected in HT recipients using both invasive and non-invasive techniques. The international society of heart and lung transplantation (ISHLT) guidelines has suggested CFR during coronary angiography as an option for detecting microvascular disease in HT recipients who are suspected of having CAV, but its routine use has not yet been widely instituted[31,42]. CFR is the ratio of the maximum stress flow (during intravenous adenosine vasodilator stress) to the rest flow for a given arterial distribution with or without a stenosis or diffuse narrowing, and it could be performed in more quickly and less expensively using TDE[43,44]. Our group demonstrated that microvascular dysfunction, as evaluated by CFR measured in the distal portion of the left anterior descending coronary artery (LAD), correlates with intimal hyperplasia measured by IVUS in patients with physiologically normal epicardial coronary arteries[45-47].

Dobutamine stress echocardiography (DSE) is a useful technique for HT recipients unable to undergo an angiogram for CAV detection. For CAV detection, the sensitivity and specificity of DSE have been shown in different studies to vary from 67% to 95% and from 55% to 91%, respectively[48-50]. However, its ability in detecting microvascular graft disease is still uncertain[51].

Another noninvasive test is dual-source computed tomography, which showed a sensitivity of 100%, a specificity of 92%, a positive predictive value of 50%, a negative predictive value of 100%, and a global accuracy of 93% in detecting CAV. Similar to DSE, its predictive value in microvascular dysfunction is not well established[52].

Magnetic resonance perfusion imaging with myocardial perfusion reserve (MPR) analysis showed a significant correlation with CFR when invasively evaluated.

Muehling and colleagues analyzed the resting endomyocardial/epimyocardial perfusion ratio (Endo/Epi ratio), which is decreased in impaired coronary circulation. CAV can be excluded by an MPR of > 2.3 with a sensitivity and specificity of 100% and 85%, respectively, and an Endo/Epi ratio of > 1.3 with a sensitivity and specificity of 100% and 80%, respectively[53,54].

MEDICAL TREATMENT

CAV prevention requires a combination of immunosuppressant agents, the prevention of CMV infection and a reduction in common cardiovascular risk factors[25,42,55].

Endothelial dysfunction is an early marker and contributes to the development of CAV[6,56-58]. Standard immunosuppression after cardiac transplantation includes a calcineurin inhibitor (CNIs, such as cyclosporin or tacrolimus) in combination with an antiproliferative agent [mycophenolate mofetil (MMF) or azathioprine (AZA)] with or without corticosteroids[59]. Cyclosporin (Cy-A) was the first immunosuppressive drug that had an important impact on the result of clinical HT by reducing the incidence and severity of rejection. Cy-A is known to impair endothelial function by increasing the release and response to vasoconstrictors, impairing the synthesis of NO, and generating free radicals. It may also result in increased ET levels and an impaired vascular response to NO[60-63]. Kobashigawa et al[64] showed that the five-year survival and incidence of angiographic CAV were similar between groups treated with microemulsion Cy-A- or tacrolimus. In a study by Meiser et al[65], a more pronounced intimal proliferation was detected in the group treated with Cy-A and MMF than in the tacrolimus-MMF-treated group. Moreover, microvascular endothelial function deteriorates more in Cy-A-treated patients than in tacrolimus-treated patients, a finding that correlates with the enhanced ET-1 concentration and reduced vascular remodeling[65-67]. The progression of CAV is slower in patients randomized to receive MMF instead of AZA. The combination of Cy-A and MMF was associated with a 35% reduction in 3-year mortality or graft loss compared with patients treated with Cy-A and AZA[68]. MMF-treated HT patients, when compared to AZA-treated patients, both treated concurrently on Cy-A and corticosteroids, have significantly less progression of first-year intimal thickening[69]. In terms of CAV prevention, MMF is superior to AZA in both combinations. A trend toward improved survival in MMF patients was noted. The lower number of rejection episodes in the MMF groups may have contributed to these results.

MMF is associated with the reduction of leukocyte adhesion to the graft endothelium and inhibition of the proliferation of SMCs[70-72]. Rapamycin therapy has been associated with improved coronary artery physiology at the level of both the epicardial artery and the microvasculature soon after HT[73]. Proliferation signal inhibitors (PSIs), e.g., sirolimus and everolimus, may have the potential to reduce the incidence of microvasculopathy and, later, of CAV. In a 2-year randomized clinical trial, the use of sirolimus was associated with fewer acute rejection episodes and a significant absence of the progression of intimal plus medial proliferation compared with the use of AZA[74,75]. These drugs were also associated with a lower rate of CMV infection[76,77]. The occurrence of malignancies after HT is a well-described consequence of immunosuppression that affects the long-term prognosis of HT recipients. Patients on mTOR inhibitors, a class of drugs that has been experimentally proven to have both immunosuppressive and potent antitumor effects, developed significantly fewer malignancies, as expected due to the drug’s mechanism of action[78]. In a recent retrospective study, Fröhlich et al[79] demonstrated that statin use is also protective against malignancies. Hypercholesterolemia and hypertriglyceridemia may occur in HT recipients who are treated with sirolimus, but the presence of these side effects did not appear to impair its ability to slow the progression of CAV[80]. Everolimus is an analog of sirolimus. Several studies demonstrated a decreased severity and incidence of CAV in HT recipients receiving immunosuppressive therapy with everolimus. It was compared with AZA in the largest trial conducted thus far for HT, which randomized 634 patients. This study showed that both average intimal thickening by IVUS and the incidence of acute rejection at 6 mo after HT were significantly lower in patients receiving everolimus[74,81,82]. Prophylaxis consisting of CMV hyperimmune globulin plus ganciclovir has been associated with decreased intimal thickening and reduced coronary artery disease[83].

Of the recommendations made by the ISHLT regarding CAV management, only statin therapy had a level of evidence A[42]. In several studies, cholesterol and TG have been proven to directly correlate with the development and progression of CAV[84]. It is currently advocated that statins should be given soon after HT, when the most rapid expansion of intimal hyperplasia occurs. Different statins have been associated with the reduced progression of CAV. Simvastatin improved the 8-year survival in HT recipients[85]. A one-year trial in 92 patients randomized to pravastatin or no 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor showed not only lower mean cholesterol levels but also less intimal thickening by IVUS as well as less frequent high-grade acute rejections and rejections with hemodynamic compromise[86].

The vasculoprotective effects of statins are likely mediated by multiple immunogenic effects. The immunomodulating effects of statins in the presence of Cy-A include the suppression of T-cell responses[87], the reduction of chemokine synthesis by mononuclear cells in the peripheral bloodstream, and the inhibition of the expression of MHC-II genes[88]. Simvastatin inhibits the proliferation of SMCs, which is an important process in the pathogenesis of the atherosclerotic lesion. Moreover, simvastatin has been shown to have a direct influence on the gene expression of ET-1 in cultivated endothelial cells, leading to improved endothelial function and thus protecting against atherosclerosis and microvasculopathy[89]. Another direct positive effect of simvastatin in the atherogenesis process is that it reduces monocyte adhesion to endothelial cells, which is one of the initial steps in the development of atherosclerotic plaques[90].

The use of calcium channel blockers or angiotensin-converting enzyme inhibitors (ACE-Is) decrease the incidence of CAV detected by IVUS[91]. Additionally, the use of calcium channel blockers decreases angiographically detected CAV 2-years after HT[92]. ACE-Is partially improve allograft microvascular endothelial dysfunction, reduce oxidative stress, and down-regulate endothelial ET-1 release[93], and their use has been associated with plaque regression[94] and improved graft survival[30]. The combined use of an ACE-I and a calcium-antagonist is more effective than the individual use of either drug alone on CAV development. Large randomized clinical trials are warranted to evaluate the possibility of this synergistic efficacy[95].

CONCLUSION

Coronary microvascular function has an impact on long-term graft survival after HT. Microvascular vessel disease has been demonstrated by histological findings of stenotic microvasculopathy and evaluated by non-invasive CFR[41,45,96]. The potential influence of combined immunosuppressive regimens, lipid-lowering agents, or ACE-Is and/or calcium-antagonists on microvessel response is therefore of major interest. More trials are needed for microvasculopathy prevention and/or CFR preservation and to reduce the negative prognostic impact on the survival of HT recipients.

Footnotes

P- Reviewer: Manginas A S- Editor: Wen LL L- Editor: A E- Editor: Wu HL

References
1.  Mehra MR, Crespo-Leiro MG, Dipchand A, Ensminger SM, Hiemann NE, Kobashigawa JA, Madsen J, Parameshwar J, Starling RC, Uber PA. International Society for Heart and Lung Transplantation working formulation of a standardized nomenclature for cardiac allograft vasculopathy-2010. J Heart Lung Transplant. 2010;29:717-727.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 563]  [Cited by in F6Publishing: 625]  [Article Influence: 44.6]  [Reference Citation Analysis (0)]
2.  Lund LH, Edwards LB, Kucheryavaya AY, Dipchand AI, Benden C, Christie JD, Dobbels F, Kirk R, Rahmel AO, Yusen RD. The Registry of the International Society for Heart and Lung Transplantation: Thirtieth Official Adult Heart Transplant Report--2013; focus theme: age. J Heart Lung Transplant. 2013;32:951-964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 460]  [Cited by in F6Publishing: 458]  [Article Influence: 45.8]  [Reference Citation Analysis (0)]
3.  Stehlik J, Edwards LB, Kucheryavaya AY, Benden C, Christie JD, Dipchand AI, Dobbels F, Kirk R, Rahmel AO, Hertz MI. The Registry of the International Society for Heart and Lung Transplantation: 29th official adult heart transplant report--2012. J Heart Lung Transplant. 2012;31:1052-1064.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 452]  [Cited by in F6Publishing: 426]  [Article Influence: 38.7]  [Reference Citation Analysis (0)]
4.  Stehlik J, Edwards LB, Kucheryavaya AY, Benden C, Christie JD, Dobbels F, Kirk R, Rahmel AO, Hertz MI. The Registry of the International Society for Heart and Lung Transplantation: Twenty-eighth Adult Heart Transplant Report--2011. J Heart Lung Transplant. 2011;30:1078-1094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 385]  [Cited by in F6Publishing: 397]  [Article Influence: 33.1]  [Reference Citation Analysis (0)]
5.  Osto E, Tona F, Angelini A, Montisci R, Ruscazio M, Vinci A, Tarantini G, Ramondo A, Gambino A, Thiene G. Determinants of coronary flow reserve in heart transplantation: a study performed with contrast-enhanced echocardiography. J Heart Lung Transplant. 2009;28:453-460.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
6.  Fish RD, Nabel EG, Selwyn AP, Ludmer PL, Mudge GH, Kirshenbaum JM, Schoen FJ, Alexander RW, Ganz P. Responses of coronary arteries of cardiac transplant patients to acetylcholine. J Clin Invest. 1988;81:21-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 189]  [Cited by in F6Publishing: 197]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
7.  Mügge A, Heublein B, Kuhn M, Nolte C, Haverich A, Warnecke J, Forssmann WG, Lichtlen PR. Impaired coronary dilator responses to substance P and impaired flow-dependent dilator responses in heart transplant patients with graft vasculopathy. J Am Coll Cardiol. 1993;21:163-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
8.  Benvenuti C, Aptecar E, Mazzucotelli JP, Jouannot P, Loisance D, Nitenberg A. Coronary artery response to cold-pressor test is impaired early after operation in heart transplant recipients. J Am Coll Cardiol. 1995;26:446-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
9.  Preumont N, Lenaers A, Goldman S, Vachiery JL, Wikler D, Damhaut P, Degré S, Berkenboom G. Coronary vasomotility and myocardial blood flow early after heart transplantation. Am J Cardiol. 1996;78:550-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
10.  Weis M, Cooke JP. Cardiac allograft vasculopathy and dysregulation of the NO synthase pathway. Arterioscler Thromb Vasc Biol. 2003;23:567-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
11.  Vassalli G, Gallino A, Weis M, von Scheidt W, Kappenberger L, von Segesser LK, Goy JJ. Alloimmunity and nonimmunologic risk factors in cardiac allograft vasculopathy. Eur Heart J. 2003;24:1180-1188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 104]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
12.  Rahmani M, Cruz RP, Granville DJ, McManus BM. Allograft vasculopathy versus atherosclerosis. Circ Res. 2006;99:801-815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in F6Publishing: 239]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
13.  Caforio AL, Tona F, Fortina AB, Angelini A, Piaserico S, Gambino A, Feltrin G, Ramondo A, Valente M, Iliceto S. Immune and nonimmune predictors of cardiac allograft vasculopathy onset and severity: multivariate risk factor analysis and role of immunosuppression. Am J Transplant. 2004;4:962-970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 118]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
14.  Valantine HA. Cardiac allograft vasculopathy: central role of endothelial injury leading to transplant “atheroma”. Transplantation. 2003;76:891-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 128]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
15.  Rogers NJ, Lechler RI. Allorecognition. Am J Transplant. 2001;1:97-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 154]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
16.  Behrendt D, Ganz P, Fang JC. Cardiac allograft vasculopathy. Curr Opin Cardiol. 2000;15:422-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 52]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
17.  Hiemann NE, Meyer R, Wellnhofer E, Schoenemann C, Heidecke H, Lachmann N, Hetzer R, Dragun D. Non-HLA antibodies targeting vascular receptors enhance alloimmune response and microvasculopathy after heart transplantation. Transplantation. 2012;94:919-924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 102]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
18.  Schneider MP, Boesen EI, Pollock DM. Contrasting actions of endothelin ET(A) and ET(B) receptors in cardiovascular disease. Annu Rev Pharmacol Toxicol. 2007;47:731-759.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
19.  Reinsmoen NL. Role of angiotensin II type 1 receptor-activating antibodies in solid organ transplantation. Hum Immunol. 2013;74:1474-1477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
20.  Wildhirt SM, Weis M, Schulze C, Conrad N, Pehlivanli S, Rieder G, Enders G, von Scheidt W, Reichart B. Expression of endomyocardial nitric oxide synthase and coronary endothelial function in human cardiac allografts. Circulation. 2001;104:I336-I343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
21.  Wildhirt SM, Weis M, Schulze C, Conrad N, Pehlivanli S, Rieder G, Enders G, von Scheidt W, Reichart B. Coronary flow reserve and nitric oxide synthases after cardiac transplantation in humans. Eur J Cardiothorac Surg. 2001;19:840-847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
22.  Osto E, Tona F, De Bon E, Iliceto S, Cella G. Endothelial dysfunction in cardiac allograft vasculopathy: potential pharmacological interventions. Curr Vasc Pharmacol. 2010;8:169-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 7]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
23.  Cooke JP. Does ADMA cause endothelial dysfunction? Arterioscler Thromb Vasc Biol. 2000;20:2032-2037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 384]  [Cited by in F6Publishing: 373]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
24.  Petrakopoulou P, Kübrich M, Pehlivanli S, Meiser B, Reichart B, von Scheidt W, Weis M. Cytomegalovirus infection in heart transplant recipients is associated with impaired endothelial function. Circulation. 2004;110:II207-II212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 51]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
25.  Valantine H. Cardiac allograft vasculopathy after heart transplantation: risk factors and management. J Heart Lung Transplant. 2004;23:S187-S193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 187]  [Cited by in F6Publishing: 180]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
26.  Tilney NL, Paz D, Ames J, Gasser M, Laskowski I, Hancock WW. Ischemia-reperfusion injury. Transplant Proc. 2001;33:843-844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
27.  Potena L, Valantine HA. Cardiac allograft vasculopathy and insulin resistance--hope for new therapeutic targets. Endocrinol Metab Clin North Am. 2007;36:965-981; ix.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
28.  Raichlin ER, McConnell JP, Lerman A, Kremers WK, Edwards BS, Kushwaha SS, Clavell AL, Rodeheffer RJ, Frantz RP. Systemic inflammation and metabolic syndrome in cardiac allograft vasculopathy. J Heart Lung Transplant. 2007;26:826-833.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 43]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
29.  Valantine H, Rickenbacker P, Kemna M, Hunt S, Chen YD, Reaven G, Stinson EB. Metabolic abnormalities characteristic of dysmetabolic syndrome predict the development of transplant coronary artery disease: a prospective study. Circulation. 2001;103:2144-2152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 91]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
30.  Kobashigawa JA, Starling RC, Mehra MR, Kormos RL, Bhat G, Barr ML, Sigouin CS, Kolesar J, Fitzsimmons W. Multicenter retrospective analysis of cardiovascular risk factors affecting long-term outcome of de novo cardiac transplant recipients. J Heart Lung Transplant. 2006;25:1063-1069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 56]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
31.  Colvin-Adams M, Agnihotri A. Cardiac allograft vasculopathy: current knowledge and future direction. Clin Transplant. 2011;25:175-184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 78]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
32.  Ross R. The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature. 1993;362:801-809.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7939]  [Cited by in F6Publishing: 7649]  [Article Influence: 246.7]  [Reference Citation Analysis (0)]
33.  Minami E, Laflamme MA, Saffitz JE, Murry CE. Extracardiac progenitor cells repopulate most major cell types in the transplanted human heart. Circulation. 2005;112:2951-2958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 125]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
34.  Kapessidou Y, Habran C, Buonocore S, Flamand V, Barvais L, Goldman M, Braun MY. The replacement of graft endothelium by recipient-type cells conditions allograft rejection mediated by indirect pathway CD4(+) T cells. Transplantation. 2006;81:726-735.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
35.  Hillebrands JL, Klatter FA, van den Hurk BM, Popa ER, Nieuwenhuis P, Rozing J. Origin of neointimal endothelium and alpha-actin-positive smooth muscle cells in transplant arteriosclerosis. J Clin Invest. 2001;107:1411-1422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 229]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
36.  Quaini F, Urbanek K, Beltrami AP, Finato N, Beltrami CA, Nadal-Ginard B, Kajstura J, Leri A, Anversa P. Chimerism of the transplanted heart. N Engl J Med. 2002;346:5-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 926]  [Cited by in F6Publishing: 956]  [Article Influence: 43.5]  [Reference Citation Analysis (0)]
37.  Simper D, Wang S, Deb A, Holmes D, McGregor C, Frantz R, Kushwaha SS, Caplice NM. Endothelial progenitor cells are decreased in blood of cardiac allograft patients with vasculopathy and endothelial cells of noncardiac origin are enriched in transplant atherosclerosis. Circulation. 2003;108:143-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 113]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
38.  Koopmans M, Kremer Hovinga IC, Baelde HJ, de Heer E, Bruijn JA, Bajema IM. Endothelial chimerism in transplantation: Looking for needles in a haystack. Transplantation. 2006;82:S25-S29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
39.  Osto E, Castellani C, Fadini GP, Baesso I, Gambino A, Agostini C, Avogaro A, Gerosa G, Thiene G, Iliceto S. Impaired endothelial progenitor cell recruitment may contribute to heart transplant microvasculopathy. J Heart Lung Transplant. 2011;30:70-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
40.  Angelini A, Castellani C, Tona F, Gambino A, Caforio AP, Feltrin G, Della Barbera M, Valente M, Gerosa G, Thiene G. Continuous engraftment and differentiation of male recipient Y-chromosome-positive cardiomyocytes in donor female human heart transplants. J Heart Lung Transplant. 2007;26:1110-1118.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
41.  Hiemann NE, Wellnhofer E, Knosalla C, Lehmkuhl HB, Stein J, Hetzer R, Meyer R. Prognostic impact of microvasculopathy on survival after heart transplantation: evidence from 9713 endomyocardial biopsies. Circulation. 2007;116:1274-1282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 124]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
42.  Costanzo MR, Dipchand A, Starling R, Anderson A, Chan M, Desai S, Fedson S, Fisher P, Gonzales-Stawinski G, Martinelli L. The International Society of Heart and Lung Transplantation Guidelines for the care of heart transplant recipients. J Heart Lung Transplant. 2010;29:914-956.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1100]  [Cited by in F6Publishing: 1157]  [Article Influence: 82.6]  [Reference Citation Analysis (0)]
43.  Caiati C, Montaldo C, Zedda N, Bina A, Iliceto S. New noninvasive method for coronary flow reserve assessment: contrast-enhanced transthoracic second harmonic echo Doppler. Circulation. 1999;99:771-778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 217]  [Cited by in F6Publishing: 233]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
44.  Caiati C, Montaldo C, Zedda N, Montisci R, Ruscazio M, Lai G, Cadeddu M, Meloni L, Iliceto S. Validation of a new noninvasive method (contrast-enhanced transthoracic second harmonic echo Doppler) for the evaluation of coronary flow reserve: comparison with intracoronary Doppler flow wire. J Am Coll Cardiol. 1999;34:1193-1200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 157]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
45.  Tona F, Caforio AL, Montisci R, Angelini A, Ruscazio M, Gambino A, Ramondo A, Thiene G, Gerosa G, Iliceto S. Coronary flow reserve by contrast-enhanced echocardiography: a new noninvasive diagnostic tool for cardiac allograft vasculopathy. Am J Transplant. 2006;6:998-1003.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
46.  Tona F, Caforio AL, Montisci R, Gambino A, Angelini A, Ruscazio M, Toscano G, Feltrin G, Ramondo A, Gerosa G. Coronary flow velocity pattern and coronary flow reserve by contrast-enhanced transthoracic echocardiography predict long-term outcome in heart transplantation. Circulation. 2006;114:I49-I55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 52]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
47.  Tona F, Osto E, Tarantini G, Gambino A, Cavallin F, Feltrin G, Montisci R, Caforio AL, Gerosa G, Iliceto S. Coronary flow reserve by transthoracic echocardiography predicts epicardial intimal thickening in cardiac allograft vasculopathy. Am J Transplant. 2010;10:1668-1676.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
48.  Eroglu E, D’hooge J, Sutherland GR, Marciniak A, Thijs D, Droogne W, Herbots L, Van Cleemput J, Claus P, Bijnens B. Quantitative dobutamine stress echocardiography for the early detection of cardiac allograft vasculopathy in heart transplant recipients. Heart. 2008;94:e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 37]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
49.  Spes CH, Klauss V, Mudra H, Schnaack SD, Tammen AR, Rieber J, Siebert U, Henneke KH, Uberfuhr P, Reichart B. Diagnostic and prognostic value of serial dobutamine stress echocardiography for noninvasive assessment of cardiac allograft vasculopathy: a comparison with coronary angiography and intravascular ultrasound. Circulation. 1999;100:509-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 142]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
50.  Cai Q, Rangasetty UC, Barbagelata A, Fujise K, Koerner MM. Cardiac allograft vasculopathy: advances in diagnosis. Cardiol Rev. 2011;19:30-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
51.  Jackson PA, Akosah KO, Kirchberg DJ, Mohanty PK, Minisi AJ. Relationship between dobutamine-induced regional wall motion abnormalities and coronary flow reserve in heart transplant patients without angiographic coronary artery disease. J Heart Lung Transplant. 2002;21:1080-1089.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
52.  Mastrobuoni S, Bastarrika G, Ubilla M, Castaño S, Azcarate P, Barrero EA, Castellano JM, Herreros J, Rabago G. Dual-source CT coronary angiogram in heart transplant recipients in comparison with dobutamine stress echocardiography for detection of cardiac allograft vasculopathy. Transplantation. 2009;87:587-590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
53.  Lim YJ, Nanto S, Masuyama T, Kodama K, Ikeda T, Kitabatake A, Kamada T. Visualization of subendocardial myocardial ischemia with myocardial contrast echocardiography in humans. Circulation. 1989;79:233-244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
54.  Muehling OM, Wilke NM, Panse P, Jerosch-Herold M, Wilson BV, Wilson RF, Miller LW. Reduced myocardial perfusion reserve and transmural perfusion gradient in heart transplant arteriopathy assessed by magnetic resonance imaging. J Am Coll Cardiol. 2003;42:1054-1060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 86]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
55.  Gustafsson F, Ross HJ. Proliferation signal inhibitors in cardiac transplantation. Curr Opin Cardiol. 2007;22:111-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
56.  Hollenberg SM, Klein LW, Parrillo JE, Scherer M, Burns D, Tamburro P, Oberoi M, Johnson MR, Costanzo MR. Coronary endothelial dysfunction after heart transplantation predicts allograft vasculopathy and cardiac death. Circulation. 2001;104:3091-3096.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 117]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
57.  Davis SF, Yeung AC, Meredith IT, Charbonneau F, Ganz P, Selwyn AP, Anderson TJ. Early endothelial dysfunction predicts the development of transplant coronary artery disease at 1 year posttransplant. Circulation. 1996;93:457-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 172]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
58.  Tona F, Marra MP, Fedrigo M, Famoso G, Bellu R, Thiene G, Gerosa G, Angelini A, Iliceto S. Recent developments on coronary microvasculopathy after heart transplantation: a new target in the therapy of cardiac allograft vasculopathy. Curr Vasc Pharmacol. 2012;10:206-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
59.  Raichlin E, Prasad A, Kremers WK, Edwards BS, Rihal CS, Lerman A, Kushwaha SS. Sirolimus as primary immunosuppression is associated with improved coronary vasomotor function compared with calcineurin inhibitors in stable cardiac transplant recipients. Eur Heart J. 2009;30:1356-1363.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
60.  Sudhir K, MacGregor JS, DeMarco T, De Groot CJ, Taylor RN, Chou TM, Yock PG, Chatterjee K. Cyclosporine impairs release of endothelium-derived relaxing factors in epicardial and resistance coronary arteries. Circulation. 1994;90:3018-3023.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 62]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
61.  Diederich D, Skopec J, Diederich A, Dai FX. Cyclosporine produces endothelial dysfunction by increased production of superoxide. Hypertension. 1994;23:957-961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 92]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
62.  Jeanmart H, Malo O, Carrier M, Nickner C, Desjardins N, Perrault LP. Comparative study of cyclosporine and tacrolimus vs newer immunosuppressants mycophenolate mofetil and rapamycin on coronary endothelial function. J Heart Lung Transplant. 2002;21:990-998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 72]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
63.  Barton M, Haudenschild CC, d’Uscio LV, Shaw S, Münter K, Lüscher TF. Endothelin ETA receptor blockade restores NO-mediated endothelial function and inhibits atherosclerosis in apolipoprotein E-deficient mice. Proc Natl Acad Sci USA. 1998;95:14367-14372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 271]  [Cited by in F6Publishing: 275]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
64.  Kobashigawa JA, Patel J, Furukawa H, Moriguchi JD, Yeatman L, Takemoto S, Marquez A, Shaw J, Oeser BT, Subherwal S. Five-year results of a randomized, single-center study of tacrolimus vs microemulsion cyclosporine in heart transplant patients. J Heart Lung Transplant. 2006;25:434-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 76]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
65.  Meiser BM, Groetzner J, Kaczmarek I, Landwehr P, Müller M, Jung S, Uberfuhr P, Fraunberger P, Stempfle HU, Weis M. Tacrolimus or cyclosporine: which is the better partner for mycophenolate mofetil in heart transplant recipients? Transplantation. 2004;78:591-598.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 59]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
66.  Petrakopoulou P, Anthopoulou L, Muscholl M, Klauss V, von Scheidt W, Uberfuhr P, Meiser BM, Reichart B, Weis M. Coronary endothelial vasomotor function and vascular remodeling in heart transplant recipients randomized for tacrolimus or cyclosporine immunosuppression. J Am Coll Cardiol. 2006;47:1622-1629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
67.  Tepperman ED, Tumiati LC, Ramzy D, Badiwala MV, Sheshgiri R, Prodger JL, Ross HJ, Rao V. Tacrolimus preserves vasomotor function and maintains vascular homeostasis. J Heart Lung Transplant. 2011;30:583-588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
68.  Eisen HJ, Kobashigawa J, Keogh A, Bourge R, Renlund D, Mentzer R, Alderman E, Valantine H, Dureau G, Mancini D. Three-year results of a randomized, double-blind, controlled trial of mycophenolate mofetil versus azathioprine in cardiac transplant recipients. J Heart Lung Transplant. 2005;24:517-525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 172]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
69.  Kobashigawa JA, Tobis JM, Mentzer RM, Valantine HA, Bourge RC, Mehra MR, Smart FW, Miller LW, Tanaka K, Li H. Mycophenolate mofetil reduces intimal thickness by intravascular ultrasound after heart transplant: reanalysis of the multicenter trial. Am J Transplant. 2006;6:993-997.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 72]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
70.  Kobashigawa J, Miller L, Renlund D, Mentzer R, Alderman E, Bourge R, Costanzo M, Eisen H, Dureau G, Ratkovec R. A randomized active-controlled trial of mycophenolate mofetil in heart transplant recipients. Mycophenolate Mofetil Investigators. Transplantation. 1998;66:507-515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 462]  [Cited by in F6Publishing: 413]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
71.  Cantarovich M, Giannetti N, Cecere R. Impact of cyclosporine 2-h level and mycophenolate mofetil dose on clinical outcomes in de novo heart transplant patients receiving anti-thymocyte globulin induction. Clin Transplant. 2003;17:144-150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
72.  Kaczmarek I, Ertl B, Schmauss D, Sadoni S, Knez A, Daebritz S, Meiser B, Reichart B. Preventing cardiac allograft vasculopathy: long-term beneficial effects of mycophenolate mofetil. J Heart Lung Transplant. 2006;25:550-556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 58]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
73.  Sinha SS, Pham MX, Vagelos RH, Perlroth MG, Hunt SA, Lee DP, Valantine HA, Yeung AC, Fearon WF. Effect of rapamycin therapy on coronary artery physiology early after cardiac transplantation. Am Heart J. 2008;155:889.e1-889.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
74.  Eisen HJ, Tuzcu EM, Dorent R, Kobashigawa J, Mancini D, Valantine-von Kaeppler HA, Starling RC, Sørensen K, Hummel M, Lind JM. Everolimus for the prevention of allograft rejection and vasculopathy in cardiac-transplant recipients. N Engl J Med. 2003;349:847-858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 936]  [Cited by in F6Publishing: 859]  [Article Influence: 40.9]  [Reference Citation Analysis (0)]
75.  Keogh A, Richardson M, Ruygrok P, Spratt P, Galbraith A, O’Driscoll G, Macdonald P, Esmore D, Muller D, Faddy S. Sirolimus in de novo heart transplant recipients reduces acute rejection and prevents coronary artery disease at 2 years: a randomized clinical trial. Circulation. 2004;110:2694-2700.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in F6Publishing: 330]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
76.  Eisen H, Kobashigawa J, Starling RC, Valantine H, Mancini D. Improving outcomes in heart transplantation: the potential of proliferation signal inhibitors. Transplant Proc. 2005;37:4S-17S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 31]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
77.  Weill D. Role of cytomegalovirus in cardiac allograft vasculopathy. Transpl Infect Dis. 2001;3 Suppl 2:44-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
78.  Doesch AO, Müller S, Konstandin M, Celik S, Kristen A, Frankenstein L, Ehlermann P, Sack FU, Katus HA, Dengler TJ. Malignancies after heart transplantation: incidence, risk factors, and effects of calcineurin inhibitor withdrawal. Transplant Proc. 2010;42:3694-3699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
79.  Fröhlich GM, Rufibach K, Enseleit F, Wolfrum M, von Babo M, Frank M, Berli R, Hermann M, Holzmeister J, Wilhelm M. Statins and the risk of cancer after heart transplantation. Circulation. 2012;126:440-447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 57]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
80.  Massy ZA. Hyperlipidemia and cardiovascular disease after organ transplantation. Transplantation. 2001;72:S13-S15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 45]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
81.  Gurk-Turner C, Manitpisitkul W, Cooper M. A comprehensive review of everolimus clinical reports: a new mammalian target of rapamycin inhibitor. Transplantation. 2012;94:659-668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
82.  Patel JK, Kobashigawa JA. Everolimus for cardiac allograft vasculopathy--every patient, at any time? Transplantation. 2011;92:127-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
83.  Valantine HA, Gao SZ, Menon SG, Renlund DG, Hunt SA, Oyer P, Stinson EB, Brown BW, Merigan TC, Schroeder JS. Impact of prophylactic immediate posttransplant ganciclovir on development of transplant atherosclerosis: a post hoc analysis of a randomized, placebo-controlled study. Circulation. 1999;100:61-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 235]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
84.  Hoang K, Chen YD, Reaven G, Zhang L, Ross H, Billingham M, Valantine H. Diabetes and dyslipidemia. A new model for transplant coronary artery disease. Circulation. 1998;97:2160-2168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
85.  Wenke K, Meiser B, Thiery J, Nagel D, von Scheidt W, Krobot K, Steinbeck G, Seidel D, Reichart B. Simvastatin initiated early after heart transplantation: 8-year prospective experience. Circulation. 2003;107:93-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 182]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
86.  Kobashigawa JA, Katznelson S, Laks H, Johnson JA, Yeatman L, Wang XM, Chia D, Terasaki PI, Sabad A, Cogert GA. Effect of pravastatin on outcomes after cardiac transplantation. N Engl J Med. 1995;333:621-627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 972]  [Cited by in F6Publishing: 900]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
87.  Kurakata S, Kada M, Shimada Y, Komai T, Nomoto K. Effects of different inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, pravastatin sodium and simvastatin, on sterol synthesis and immunological functions in human lymphocytes in vitro. Immunopharmacology. 1996;34:51-61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 72]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
88.  Kwak B, Mulhaupt F, Myit S, Mach F. Statins as a newly recognized type of immunomodulator. Nat Med. 2000;6:1399-1402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 975]  [Cited by in F6Publishing: 973]  [Article Influence: 40.5]  [Reference Citation Analysis (0)]
89.  Hernández-Perera O, Pérez-Sala D, Navarro-Antolín J, Sánchez-Pascuala R, Hernández G, Díaz C, Lamas S. Effects of the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors, atorvastatin and simvastatin, on the expression of endothelin-1 and endothelial nitric oxide synthase in vascular endothelial cells. J Clin Invest. 1998;101:2711-2719.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 536]  [Cited by in F6Publishing: 532]  [Article Influence: 20.5]  [Reference Citation Analysis (0)]
90.  Teupser D, Bruegel M, Stein O, Stein Y, Thiery J. HMG-CoA reductase inhibitors reduce adhesion of human monocytes to endothelial cells. Biochem Biophys Res Commun. 2001;289:838-844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 32]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
91.  Mehra MR, Ventura HO, Smart FW, Collins TJ, Ramee SR, Stapleton DD. An intravascular ultrasound study of the influence of angiotensin-converting enzyme inhibitors and calcium entry blockers on the development of cardiac allograft vasculopathy. Am J Cardiol. 1995;75:853-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 88]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
92.  Schroeder JS, Gao SZ, Alderman EL, Hunt SA, Johnstone I, Boothroyd DB, Wiederhold V, Stinson EB. A preliminary study of diltiazem in the prevention of coronary artery disease in heart-transplant recipients. N Engl J Med. 1993;328:164-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 251]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
93.  Steinhauff S, Pehlivanli S, Bakovic-Alt R, Meiser BM, Becker BF, von Scheidt W, Weis M. Beneficial effects of quinaprilat on coronary vasomotor function, endothelial oxidative stress, and endothelin activation after human heart transplantation. Transplantation. 2004;77:1859-1865.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
94.  Bae JH, Rihal CS, Edwards BS, Kushwaha SS, Mathew V, Prasad A, Holmes DR, Lerman A. Association of angiotensin-converting enzyme inhibitors and serum lipids with plaque regression in cardiac allograft vasculopathy. Transplantation. 2006;82:1108-1111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 56]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
95.  Erinc K, Yamani MH, Starling RC, Crowe T, Hobbs R, Bott-Silverman C, Rincon G, Young JB, Feng J, Cook DJ. The effect of combined Angiotensin-converting enzyme inhibition and calcium antagonism on allograft coronary vasculopathy validated by intravascular ultrasound. J Heart Lung Transplant. 2005;24:1033-1038.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 39]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
96.  Escaned J, Flores A, García-Pavía P, Segovia J, Jimenez J, Aragoncillo P, Salas C, Alfonso F, Hernández R, Angiolillo DJ. Assessment of microcirculatory remodeling with intracoronary flow velocity and pressure measurements: validation with endomyocardial sampling in cardiac allografts. Circulation. 2009;120:1561-1568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 65]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
97.  Drakos SG, Kfoury AG, Hammond EH, Reid BB, Revelo MP, Rasmusson BY, Whitehead KJ, Salama ME, Selzman CH, Stehlik J. Impact of mechanical unloading on microvasculature and associated central remodeling features of the failing human heart. J Am Coll Cardiol. 2010;56:382-391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 112]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]