1
|
Lo Cicero L, Lentini P, Sessa C, Castellino N, D'Anca A, Torrisi I, Marcantoni C, Castellino P, Santoro D, Zanoli L. Inflammation and Arterial Stiffness as Drivers of Cardiovascular Risk in Kidney Disease. Cardiorenal Med 2024; 15:29-40. [PMID: 39631378 DOI: 10.1159/000542965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) have an increased cardiovascular (CV) risk. The lower the glomerular filtration rate, the higher the CV risk. SUMMARY Current data suggest that several uremic toxins lead to vascular inflammation and oxidative stress that, in turn, lead to endothelial dysfunction, changes in smooth muscle cells' phenotype, and increased degradation of elastin and collagen fibers. These processes lead to both functional and structural arterial stiffening and explain part of the increased risk of acute myocardial infarction and stroke reported in patients with CKD. Considering that, at least in patients with end-stage kidney disease, the reduction of arterial stiffness is associated with a parallel decrease of the CV risk; vascular function is a potential target for therapy to reduce the CV risk. KEY MESSAGES In this review, we explore mechanisms of vascular dysfunction in CKD, paying particular attention to inflammation, reporting current data in other models of mild and severe inflammation, and discussing the vascular effect of several drugs currently used in nephrology.
Collapse
Affiliation(s)
- Lorenzo Lo Cicero
- School of Nephrology, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Paolo Lentini
- Nephrology and Dialysis, San Bassiano Hospital, Bassano del Grappa, Italy
| | - Concetto Sessa
- Nephrology and Dialysis, ASP Ragusa, Ragusa, Italy
- Departement of Nephrology, University of Catania, Catania, Italy
| | | | - Ambra D'Anca
- Nephrology and Dialysis, San Marco Hospital, Catania, Italy
| | - Irene Torrisi
- Nephrology and Dialysis, San Marco Hospital, Catania, Italy
| | | | | | - Domenico Santoro
- School of Nephrology, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
- Nephrology and Dialysis, University of Messina, Messina, Italy
| | - Luca Zanoli
- School of Nephrology, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
- Departement of Nephrology, University of Catania, Catania, Italy
- Nephrology and Dialysis, San Marco Hospital, Catania, Italy
| |
Collapse
|
2
|
Dumont A, Hamzaoui M, Groussard D, Iacob M, Bertrand D, Remy-Jouet I, Hanoy M, Le Roy F, Chevalier L, Enzensperger C, Arndt HD, Renet S, Dumesnil A, Lévêque E, Duflot T, Brunel V, Michel-Després A, Audrézet MP, Richard V, Joannidès R, Guerrot D, Bellien J. Chronic endothelial dopamine receptor stimulation improves endothelial function and hemodynamics in autosomal dominant polycystic kidney disease. Kidney Int 2024; 106:1158-1169. [PMID: 39216660 DOI: 10.1016/j.kint.2024.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 07/19/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Altered polycystin-mediated endothelial flow mechanosensitivity contributes to the development of hypertension and cardiovascular complications in patients with autosomal dominant polycystic kidney disease (ADPKD). Stimulation of endothelial type 5 dopamine receptors (DR5) can acutely compensate for the endothelial consequences of polycystin deficiency, but the chronic impact of this approach must be evaluated in ADPKD. Nineteen patients with ADPKD on standard of care therapy were randomized to receive a 2-month treatment with the DR agonist rotigotine using transdermal patches, nine at 2 mg/24hours and ten at 4 mg/24hours or while ten were untreated. Rotigotine at the dose of 4 mg/24hours significantly increased nitric oxide release (nitrite levels from 10±30 to 46±34 nmol/L) and radial artery endothelium-dependent flow-mediated dilatation (from 16.4±6.3 to 22.5±7.3%) in response to hand skin heating. Systemic hemodynamics were not significantly modified but aplanation tonometry showed that rotigotine at 4 mg/24hours reduced aortic augmentation index and pulse pressure without affecting carotid-to femoral pulse wave velocity. Plasma creatinine and urea, urinary cyclic AMP, which contributes to cyst growth in ADPKD and copeptin, a surrogate marker of vasopressin, were not affected by rotigotine. In mice with a specific deletion of polycystin-1 in endothelial cells, chronic infusion of the peripheral DR5 agonist fenoldopam also improved mesenteric artery flow-mediated dilatation and reduced blood pressure. Thus, our study demonstrates that in patients with ADPKD, chronic administration of rotigotine improves conduit artery endothelial function through the restoration of flow-induced nitric oxide release as well as hemodynamics suggesting that endothelial DR5 activation may represent a promising pharmacological approach to prevent cardiovascular complications of ADPKD.
Collapse
Affiliation(s)
- Audrey Dumont
- University Rouen Normandie, Institut national de la santé et de la recherche médicale (INSERM) U1096, Endothélium, Valvulopathies, Insuffisance cardiaque (EnVI), Rouen, France; Department of Pharmacology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France; Centre d'Investigation CLinique-Centre de Ressources Biologiques (CIC-CRB) 1404, Rouen, France
| | - Mouad Hamzaoui
- Department of Nephrology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France
| | - Déborah Groussard
- University Rouen Normandie, Institut national de la santé et de la recherche médicale (INSERM) U1096, Endothélium, Valvulopathies, Insuffisance cardiaque (EnVI), Rouen, France
| | - Michèle Iacob
- Department of Pharmacology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France
| | - Dominique Bertrand
- Department of Nephrology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France
| | - Isabelle Remy-Jouet
- University Rouen Normandie, Institut national de la santé et de la recherche médicale (INSERM) U1096, Endothélium, Valvulopathies, Insuffisance cardiaque (EnVI), Rouen, France
| | - Mélanie Hanoy
- Department of Nephrology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France
| | - Frank Le Roy
- Department of Nephrology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France
| | - Laurence Chevalier
- University Rouen Normandie, Centre national de la recherche scientifique (CNRS), Institut national des sciences appliquées (INSA) Rouen Normandie-Normandie Université-Groupe de Physique des Matériaux-Unité Mixte de Recherche (GPM-UMR) 6634, Rouen, France
| | - Christoph Enzensperger
- Friedrich Schiller University Jena, Institute for Organic Chemistry and Macromolecular Chemistry, Jena, Germany
| | - Hans-Dieter Arndt
- Friedrich Schiller University Jena, Institute for Organic Chemistry and Macromolecular Chemistry, Jena, Germany
| | - Sylvanie Renet
- University Rouen Normandie, Institut national de la santé et de la recherche médicale (INSERM) U1096, Endothélium, Valvulopathies, Insuffisance cardiaque (EnVI), Rouen, France
| | - Anaïs Dumesnil
- University Rouen Normandie, Institut national de la santé et de la recherche médicale (INSERM) U1096, Endothélium, Valvulopathies, Insuffisance cardiaque (EnVI), Rouen, France
| | - Emilie Lévêque
- Department of Biostatistics, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France
| | - Thomas Duflot
- University Rouen Normandie, Institut national de la santé et de la recherche médicale (INSERM) U1096, Endothélium, Valvulopathies, Insuffisance cardiaque (EnVI), Rouen, France; Department of Pharmacology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France
| | - Valéry Brunel
- Department of General Biochemistry, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France
| | - Aurore Michel-Després
- Centre Hospitalier Régional Universitaire (CHRU) Brest, University Brest, Institut national de la santé et de la recherche médicale (Inserm), Unité Mixte de Recherche (UMR) 1078, Génétique, Génomique fonctionnelle et Biotechnologies (GGB), Brest, France
| | - Marie-Pierre Audrézet
- Centre Hospitalier Régional Universitaire (CHRU) Brest, University Brest, Institut national de la santé et de la recherche médicale (Inserm), Unité Mixte de Recherche (UMR) 1078, Génétique, Génomique fonctionnelle et Biotechnologies (GGB), Brest, France
| | - Vincent Richard
- Department of Pharmacology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France; Centre d'Investigation CLinique-Centre de Ressources Biologiques (CIC-CRB) 1404, Rouen, France
| | - Robinson Joannidès
- University Rouen Normandie, Institut national de la santé et de la recherche médicale (INSERM) U1096, Endothélium, Valvulopathies, Insuffisance cardiaque (EnVI), Rouen, France; Department of Pharmacology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France
| | - Dominique Guerrot
- University Rouen Normandie, Institut national de la santé et de la recherche médicale (INSERM) U1096, Endothélium, Valvulopathies, Insuffisance cardiaque (EnVI), Rouen, France; Centre d'Investigation CLinique-Centre de Ressources Biologiques (CIC-CRB) 1404, Rouen, France; Department of Nephrology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France
| | - Jérémy Bellien
- University Rouen Normandie, Institut national de la santé et de la recherche médicale (INSERM) U1096, Endothélium, Valvulopathies, Insuffisance cardiaque (EnVI), Rouen, France; Department of Pharmacology, Centre Hospitalier Universitaire (CHU) Rouen, Rouen, France; Centre d'Investigation CLinique-Centre de Ressources Biologiques (CIC-CRB) 1404, Rouen, France.
| |
Collapse
|
3
|
Harinstein ME, Gandolfo C, Gruttadauria S, Accardo C, Crespo G, VanWagner LB, Humar A. Cardiovascular disease assessment and management in liver transplantation. Eur Heart J 2024; 45:4399-4413. [PMID: 39152050 DOI: 10.1093/eurheartj/ehae502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/21/2024] [Accepted: 07/25/2024] [Indexed: 08/19/2024] Open
Abstract
The prevalence and mortality related to end-stage liver disease (ESLD) continue to rise globally. Liver transplant (LT) recipients continue to be older and have inherently more comorbidities. Among these, cardiac disease is one of the three main causes of morbidity and mortality after LT. Several reasons exist including the high prevalence of associated risk factors, which can also be attributed to the rise in the proportion of patients undergoing LT for metabolic dysfunction-associated steatohepatitis (MASH). Additionally, as people age, the prevalence of now treatable cardiac conditions, including coronary artery disease (CAD), cardiomyopathies, significant valvular heart disease, pulmonary hypertension, and arrhythmias rises, making the need to treat these conditions critical to optimize outcomes. There is an emerging body of literature regarding CAD screening in patients with ESLD, however, there is a paucity of strong evidence to support the guidance regarding the management of cardiac conditions in the pre-LT and perioperative settings. This has resulted in significant variations in assessment strategies and clinical management of cardiac disease in LT candidates between transplant centres, which impacts LT candidacy based on a transplant centre's risk tolerance and comfort level for caring for patients with concomitant cardiac disease. Performing a comprehensive assessment and understanding the potential approaches to the management of ESLD patients with cardiac conditions may increase the acceptance of patients, who appear too complex, but rather require extra evaluation and may be reasonable candidates for LT. The unique physiology of ESLD can profoundly influence preoperative assessment, perioperative management, and outcomes associated with underlying cardiac pathology, and requires a thoughtful multidisciplinary approach. The strategies proposed in this manuscript attempt to review the latest expert experience and opinions and provide guidance to practicing clinicians who assess and treat patients being considered for LT. These topics also highlight the gaps that exist in the comprehensive care of LT patients and the need for future investigations in this field.
Collapse
Affiliation(s)
- Matthew E Harinstein
- Division of Cardiology, Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Caterina Gandolfo
- Unit of Interventional Cardiology, Department of Cardiothoracic Surgery, UPMC IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Palermo, Italy
| | - Salvatore Gruttadauria
- Department for the Treatment and Study of Abdominal Diseases and Abdominal Transplantation, UPMC IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Palermo, Italy
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Caterina Accardo
- Department for the Treatment and Study of Abdominal Diseases and Abdominal Transplantation, UPMC IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Palermo, Italy
| | - Gonzalo Crespo
- Liver Transplant Unit, Hospital Clínic, IDIBAPS, CIBERehd, University of Barcelona, Barcelona, Spain
| | - Lisa B VanWagner
- Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Abhinav Humar
- Division of Transplantation, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Islam MT, Hall SA, Dutson T, Bloom SI, Bramwell RC, Kim J, Tucker JR, Machin DR, Donato AJ, Lesniewski LA. Endothelial cell-specific reduction in mTOR ameliorates age-related arterial and metabolic dysfunction. Aging Cell 2024; 23:e14040. [PMID: 38017701 PMCID: PMC10861194 DOI: 10.1111/acel.14040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 10/04/2023] [Accepted: 10/23/2023] [Indexed: 11/30/2023] Open
Abstract
Systemic inhibition of the mammalian target of rapamycin (mTOR) delays aging and many age-related conditions including arterial and metabolic dysfunction. However, the mechanisms and tissues involved in these beneficial effects remain largely unknown. Here, we demonstrate that activation of S6K, a downstream target of mTOR, is increased in arteries with advancing age, and that this occurs preferentially in the endothelium compared with the vascular smooth muscle. Induced endothelial cell-specific deletion of mTOR reduced protein expression by 60-70%. Although this did not significantly alter arterial and metabolic function in young mice, endothelial mTOR reduction reversed arterial stiffening and improved endothelium-dependent dilation (EDD) in old mice, indicating an improvement in age-related arterial dysfunction. Improvement in arterial function in old mice was concomitant with reductions in arterial cellular senescence, inflammation, and oxidative stress. The reduction in endothelial mTOR also improved glucose tolerance in old mice, and this was associated with attenuated hepatic gluconeogenesis and improved lipid tolerance, but was independent of alterations in peripheral insulin sensitivity, pancreatic beta cell function, or fasted plasma lipids in old mice. Lastly, we found that endothelial mTOR reduction suppressed gene expression of senescence and inflammatory markers in endothelial-rich (i.e., lung) and metabolically active organs (i.e., liver and adipose tissue), which may have contributed to the improvement in metabolic function in old mice. This is the first evidence demonstrating that reducing endothelial mTOR in old age improves arterial and metabolic function. These findings have implications for future drug development.
Collapse
Affiliation(s)
- Md Torikul Islam
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
| | - Shelby A. Hall
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
| | - Tavia Dutson
- Division of Geriatrics, Department of Internal MedicineThe University of Utah School of MedicineSalt Lake CityUtahUSA
| | - Samuel I. Bloom
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
| | - R. Colton Bramwell
- Division of Geriatrics, Department of Internal MedicineThe University of Utah School of MedicineSalt Lake CityUtahUSA
| | - John Kim
- Nora Eccles Harrison Cardiovascular Research and Training InstituteThe University of UtahSalt Lake CityUtahUSA
| | - Jordan R. Tucker
- Division of Geriatrics, Department of Internal MedicineThe University of Utah School of MedicineSalt Lake CityUtahUSA
| | - Daniel R. Machin
- Division of Geriatrics, Department of Internal MedicineThe University of Utah School of MedicineSalt Lake CityUtahUSA
| | - Anthony J. Donato
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
- Division of Geriatrics, Department of Internal MedicineThe University of Utah School of MedicineSalt Lake CityUtahUSA
- Nora Eccles Harrison Cardiovascular Research and Training InstituteThe University of UtahSalt Lake CityUtahUSA
- Geriatric Research Education and Clinical CenterVeteran's Affairs Medical CenterSalt Lake CityUtahUSA
- Department of BiochemistryThe University of UtahSalt Lake CityUtahUSA
| | - Lisa A. Lesniewski
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
- Division of Geriatrics, Department of Internal MedicineThe University of Utah School of MedicineSalt Lake CityUtahUSA
- Nora Eccles Harrison Cardiovascular Research and Training InstituteThe University of UtahSalt Lake CityUtahUSA
- Geriatric Research Education and Clinical CenterVeteran's Affairs Medical CenterSalt Lake CityUtahUSA
| |
Collapse
|
5
|
Cheng DCY, Climie RE, Shu M, Grieve SM, Kozor R, Figtree GA. Vascular aging and cardiovascular disease: pathophysiology and measurement in the coronary arteries. Front Cardiovasc Med 2023; 10:1206156. [PMID: 38089775 PMCID: PMC10715672 DOI: 10.3389/fcvm.2023.1206156] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2024] Open
Abstract
Age is a key risk factor for cardiovascular disease, including atherosclerosis. However, pathophysiological disease processes in the arteries are not an inevitable feature of aging. Large cohort studies with arterial phenotyping along with clinical and demographic data are essential to better understand factors related to the susceptibility or resilience to age-related vascular pathophysiology in humans. This review explores the mechanisms by which vascular structure and function alters with age, and how these changes relate to cardiovascular pathophysiology and disease. Features of vascular aging in the coronary arteries have historically been difficult to quantify pre-mortem due to their size and location. However, non-invasive imaging modalities including CT Coronary Angiogram are now being used to assess coronary vascular age, and further advances in imaging analysis such as the CT Fat Attenuation Index will help provide further measurement of features associated with coronary vascular aging. Currently, markers of vascular aging are not used as therapeutic targets in routine clinical practice, but non-pharmacological interventions including aerobic exercise and low salt diet, as well as anti-hypertensives have been demonstrated to reduce arterial stiffness. Advances in imaging technology, both in acquisition and advanced analysis, as well as harmonisation of measurements for researchers across the globe will be invaluable in understanding what constitutes healthy vascular aging and in identifying features of vascular aging that are associated with coronary artery disease and its adverse outcomes. Assessing such images in large cohorts can facilitate improved definitions of resilient and susceptible phenotypes to vascular aging in the coronary arteries. This is a critical step in identifying further risk factors and biomarkers within these groups and driving forward the development of novel therapies aimed at slowing or stopping age-related vascular changes in the coronary arteries.
Collapse
Affiliation(s)
- Daniel C. Y. Cheng
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Rachel E. Climie
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Matthew Shu
- Northern Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Stuart M. Grieve
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Rebecca Kozor
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Gemma A. Figtree
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| |
Collapse
|
6
|
Li A, Yan J, Zhao Y, Yu Z, Tian S, Khan AH, Zhu Y, Wu A, Zhang C, Tian XL. Vascular Aging: Assessment and Intervention. Clin Interv Aging 2023; 18:1373-1395. [PMID: 37609042 PMCID: PMC10441648 DOI: 10.2147/cia.s423373] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/06/2023] [Indexed: 08/24/2023] Open
Abstract
Vascular aging represents a collection of structural and functional changes in a blood vessel with advancing age, including increased stiffness, vascular wall remodeling, loss of angiogenic ability, and endothelium-dependent vasodilation dysfunction. These age-related alterations may occur earlier in those who are at risk for or have cardiovascular diseases, therefore, are defined as early or premature vascular aging. Vascular aging contributes independently to cardio-cerebral vascular diseases (CCVDs). Thus, early diagnosis and interventions targeting vascular aging are of paramount importance in the delay or prevention of CCVDs. Here, we review the direct assessment of vascular aging by examining parameters that reflect changes in structure, function, or their compliance with age including arterial wall thickness and lumen diameter, endothelium-dependent vasodilation, arterial stiffness as well as indirect assessment through pathological studies of biomarkers including endothelial progenitor cell, lymphocytic telomeres, advanced glycation end-products, and C-reactive protein. Further, we evaluate how different types of interventions including lifestyle mediation, such as caloric restriction and salt intake, and treatments for hypertension, diabetes, and hyperlipidemia affect age-related vascular changes. As a single parameter or intervention targets only a certain vascular physiological change, it is recommended to use multiple parameters to evaluate and design intervention approaches accordingly to prevent systemic vascular aging in clinical practices or population-based studies.
Collapse
Affiliation(s)
- Ao Li
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330031, People’s Republic of China
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Jinhua Yan
- Department of Geriatrics, Institute of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ya Zhao
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Zhenping Yu
- Institute of Translational Medicine, School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Shane Tian
- Department of Biochemistry/Chemistry, Ohio State University, Columbus, OH, USA
| | - Abdul Haseeb Khan
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Yuanzheng Zhu
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Andong Wu
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| | - Cuntai Zhang
- Department of Geriatrics, Institute of Gerontology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, Jiangxi, 330031, People’s Republic of China
| |
Collapse
|
7
|
Sommerer C, Legendre C, Citterio F, Watarai Y, Oberbauer R, Basic-Jukic N, Han J, Gawai A, Bernhardt P, Chadban S. Cardiovascular Outcomes in De Novo Kidney Transplant Recipients Receiving Everolimus and Reduced Calcineurin Inhibitor or Standard Triple Therapy: 24-month Post Hoc Analysis From TRANSFORM Study. Transplantation 2023; 107:1593-1604. [PMID: 36959121 DOI: 10.1097/tp.0000000000004555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
BACKGROUND The comparative impact of everolimus (EVR)-based regimens versus standard of care (mycophenolic acid+standard calcineurin inhibitor [MPA+sCNI]) on cardiovascular outcomes in de novo kidney transplant recipients (KTRs) is poorly understood. The incidence of major adverse cardiac events (MACEs) in KTRs receiving EVR+reduced CNI (rCNI) or MPA+sCNI from the TRANSplant eFficacy and safety Outcomes with an eveRolimus-based regiMen study was evaluated. METHODS The incidence of MACE was determined for all randomized patients receiving at least 1 dose of the study drug. Factors associated with MACEs were determined by logistic regression. Risk of MACE out to 3 y post-study was calculated using the Patient Outcome in Renal Transplantation equation. RESULTS MACE occurred in 81 of 1014 (8.0%; EVR+rCNI) versus 89 of 1012 (8.8%; MPA+sCNI) KTRs (risk ratio, 0.91 [95% confidence interval [CI], 0.68-1.21]). The incidence of circulatory death, myocardial infarction, revascularization, or angina was similar between the arms. Incidence of MACE was similar between EVR+rCNI and MPA+sCNI arms with a higher incidence in prespecified risk groups: older age, pretransplant diabetes (15.1% versus 15.9%), statin use (8.5% versus 10.8%), and low estimated glomerular filtration rate (Month 2 estimated glomerular filtration rate <30 versus >60 mL/min/1.73 m 2 ; odds ratio, 2.23 [95% CI, 1.02-4.86]; P = 0.044), respectively. Predicted risk of MACE within 3 y of follow-up did not differ between the treatment arms. CONCLUSIONS Cardiovascular morbidity and mortality were similar between de novo KTRs receiving EVR+rCNI and MPA+sCNI. EVR+rCNI is a viable alternative to the current standard of care in KTRs.
Collapse
Affiliation(s)
- Claudia Sommerer
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christophe Legendre
- Department of Adult Kidney Transplantation, Hôpital Necker, Université de Paris, Paris, France
| | - Franco Citterio
- Agostino Gemelli University Polyclinic Foundation, Catholic University of the Sacred Heart, Rome, Italy
| | - Yoshihiko Watarai
- Department of Transplant Surgery, Nagoya Daini Red Cross Hospital, Nagoya-City, Aichi, Japan
| | - Rainer Oberbauer
- Department of Nephrology and Dialysis, University Clinic for Internal Medicine III, Medical University Vienna, Vienna, Austria
| | | | - Jackie Han
- Novartis Pharmaceuticals, East Hanover, NJ
| | | | | | - Steve Chadban
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney Local Health District, NSW, Australia
| |
Collapse
|
8
|
Izzy M, Fortune BE, Serper M, Bhave N, deLemos A, Gallegos-Orozco JF, Guerrero-Miranda C, Hall S, Harinstein ME, Karas MG, Kriss M, Lim N, Palardy M, Sawinski D, Schonfeld E, Seetharam A, Sharma P, Tallaj J, Dadhania DM, VanWagner LB. Management of cardiac diseases in liver transplant recipients: Comprehensive review and multidisciplinary practice-based recommendations. Am J Transplant 2022; 22:2740-2758. [PMID: 35359027 PMCID: PMC9522925 DOI: 10.1111/ajt.17049] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/10/2022] [Accepted: 03/27/2022] [Indexed: 01/25/2023]
Abstract
Cardiac diseases are one of the most common causes of morbidity and mortality following liver transplantation (LT). Prior studies have shown that cardiac diseases affect close to one-third of liver transplant recipients (LTRs) long term and that their incidence has been on the rise. This rise is expected to continue as more patients with advanced age and/or non-alcoholic steatohepatitis undergo LT. In view of the increasing disease burden, a multidisciplinary initiative was developed to critically review the existing literature (between January 1, 1990 and March 17, 2021) surrounding epidemiology, risk assessment, and risk mitigation of coronary heart disease, arrhythmia, heart failure, and valvular heart disease and formulate practice-based recommendations accordingly. In this review, the expert panel emphasizes the importance of optimizing management of metabolic syndrome and its components in LTRs and highlights the cardioprotective potential for the newer diabetes medications (e.g., sodium glucose transporter-2 inhibitors) in this high-risk population. Tailoring the multidisciplinary management of cardiac diseases in LTRs to the cardiometabolic risk profile of the individual patient is critical. The review also outlines numerous knowledge gaps to pave the road for future research in this sphere with the ultimate goal of improving clinical outcomes.
Collapse
Affiliation(s)
- Manhal Izzy
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Vanderbilt University, Nashville, TN, USA
| | - Brett E Fortune
- Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, NY, USA
| | - Marina Serper
- Department of Medicine, Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicole Bhave
- Department of Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrew deLemos
- Department of Medicine, Division of Hepatology, Atrium Health, Charlotte, NC, USA
| | - Juan F. Gallegos-Orozco
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Utah School, Salt Lake City, UT, USA
| | - Cesar Guerrero-Miranda
- Center for Advanced Heart and Lung Disease, Baylor University Medical Center, Dallas, TX, USA
| | - Shelley Hall
- Center for Advanced Heart and Lung Disease, Baylor University Medical Center, Dallas, TX, USA
| | - Matthew E Harinstein
- Department of Medicine, Division of Cardiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maria G. Karas
- Department of Medicine, Division of Cardiology, Weill Cornell Medical College, New York, NY, USA
| | - Michael Kriss
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado, Aurora, CO, USA
| | - Nicholas Lim
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN, USA
| | - Maryse Palardy
- Department of Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Deirdre Sawinski
- Department of Medicine, Division of Nephrology and Hypertension, Weill Cornell Medical College, New York, NY, USA
| | - Emily Schonfeld
- Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, NY, USA
| | - Anil Seetharam
- Department of Medicine, Division of Gastroenterology and Hepatology, Banner - University Medical Center Phoenix, Phoenix, AZ, USA
| | - Pratima Sharma
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
| | - Jose Tallaj
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama, Birmingham, AL, USA
| | - Darshana M Dadhania
- Department of Medicine, Division of Nephrology and Hypertension, Weill Cornell Medical College, New York, NY, USA
| | - Lisa B. VanWagner
- Department of Medicine, Division of Gastroenterology & Hepatology, and Department of Preventive Medicine, Division of Epidemiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
9
|
Elezaby A, Dexheimer R, Sallam K. Cardiovascular effects of immunosuppression agents. Front Cardiovasc Med 2022; 9:981838. [PMID: 36211586 PMCID: PMC9534182 DOI: 10.3389/fcvm.2022.981838] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/30/2022] [Indexed: 11/26/2022] Open
Abstract
Immunosuppressive medications are widely used to treat patients with neoplasms, autoimmune conditions and solid organ transplants. Key drug classes, namely calcineurin inhibitors, mammalian target of rapamycin (mTOR) inhibitors, and purine synthesis inhibitors, have direct effects on the structure and function of the heart and vascular system. In the heart, immunosuppressive agents modulate cardiac hypertrophy, mitochondrial function, and arrhythmia risk, while in vasculature, they influence vessel remodeling, circulating lipids, and blood pressure. The aim of this review is to present the preclinical and clinical literature examining the cardiovascular effects of immunosuppressive agents, with a specific focus on cyclosporine, tacrolimus, sirolimus, everolimus, mycophenolate, and azathioprine.
Collapse
Affiliation(s)
- Aly Elezaby
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States
| | - Ryan Dexheimer
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Karim Sallam
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, United States
- *Correspondence: Karim Sallam
| |
Collapse
|
10
|
Maassen H, Said MY, Frenay ARS, Koning A, Post A, Riphagen IJ, Heiner-Fokkema MR, Drabert K, Fernandez BO, Gans ROB, van den Berg E, Navis G, Tsikas D, Feelisch M, Bakker SJL, van Goor H. Nitric oxide and long-term outcomes after kidney transplantation: Results of the TransplantLines cohort study. Nitric Oxide 2022; 125-126:1-11. [PMID: 35660109 DOI: 10.1016/j.niox.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/22/2022] [Accepted: 05/31/2022] [Indexed: 11/12/2022]
Abstract
Impaired endogenous nitric oxide (NO) production may contribute to graft failure and premature mortality in kidney transplant recipients (KTR). We investigated potential associations of 24-h urinary NOx (NO3- + NO2-) excretion (uNOx) with long-term outcomes. uNOx was determined by HPLC and GC-MS in 698 KTR and in 132 kidney donors before and after donation. Additionally, we measured urinary nitroso species (RXNO) by gas-phase chemiluminescence. Median uNOx was lower in KTR compared to kidney donors (688 [393-1076] vs. 1301 [868-1863] before donation and 1312 [982-1853] μmol/24h after donation, P < 0.001). During median follow-up of 5.4 [4.8-6.1] years, 150 KTR died (61 due to cardiovascular disease) and 83 experienced graft failure. uNOx was inversely associated with all-cause mortality (HR per doubling of uNOx: 0.84 [95% CI 0.75-0.93], P < 0.001) and cardiovascular mortality (HR 0.78 [95% CI 0.67-0.92], P = 0.002). The association of uNOx with graft failure was lost when adjusted for renal function (HR per doubling of uNOx: 0.89 [95% CI 0.76-1.05], P = 0.17). There were no significant associations of urinary RXNO with outcomes. Our study suggests that KTR have lower NO production than healthy subjects and that lower uNOx is associated with a higher risk of all-cause and cardiovascular mortality.
Collapse
Affiliation(s)
- Hanno Maassen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - M Yusof Said
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Anne-Roos S Frenay
- Department of Gynecology and Obstetrics, Amsterdam University Medical Center, University of Amsterdam, the Netherlands
| | - Anne Koning
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Adrian Post
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Ineke J Riphagen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M Rebecca Heiner-Fokkema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kathrin Drabert
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, Hannover, Germany
| | - Bernadette O Fernandez
- Clinical & Experimental Sciences, Faculty of Medicine and NIHR Southampton Biomedical Research Centre, Southampton General Hospital, United Kingdom
| | - Reinold O B Gans
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Else van den Berg
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Gerjan Navis
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Dimitrios Tsikas
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, Hannover, Germany
| | - Martin Feelisch
- Clinical & Experimental Sciences, Faculty of Medicine and NIHR Southampton Biomedical Research Centre, Southampton General Hospital, United Kingdom
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, the Netherlands.
| |
Collapse
|
11
|
Cardiovascular protection associated with cilostazol, colchicine and target of rapamycin inhibitors. J Cardiovasc Pharmacol 2022; 80:31-43. [PMID: 35384911 DOI: 10.1097/fjc.0000000000001276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/06/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT An alteration in extracellular matrix production by vascular smooth muscle cells is a crucial event in the pathogenesis of vascular diseases such as aging-related, atherosclerosis and allograft vasculopathy. The human target of rapamycin (TOR) is involved in the synthesis of extracellular matrix by vascular smooth muscle cells. TOR inhibitors reduce arterial stiffness, blood pressure, and left ventricle hypertrophy and decrease cardiovascular risk in kidney graft recipients and patients with coronary artery disease and heart allograft vasculopathy. Other drugs that modulate extracellular matrix production such as cilostazol and colchicine have also demonstrated a beneficial cardiovascular effect. Clinical studies have consistently shown that cilostazol confers cardiovascular protection in peripheral vascular disease, coronary artery disease, and cerebrovascular disease. In patients with type 2 diabetes, cilostazol prevents the progression of subclinical coronary atherosclerosis. Colchicine reduces arterial stiffness in patients with Familial Mediterranean Fever and patients with coronary artery disease. Pathophysiological mechanisms underlying the cardioprotective effect of these drugs may be related to interactions between the cytoskeleton, TOR signaling and cyclic AMP synthesis that remain to be fully elucidated. Adult vascular smooth muscle cells exhibit a contractile phenotype and produce little extracellular matrix. Conditions that upregulate extracellular matrix synthesis induce a phenotypic switch toward a synthetic phenotype. TOR inhibition with rapamycin reduces extracellular matrix production by promoting the change to the contractile phenotype. Cilostazol increases the cytosolic level of cyclic AMP, which in turn leads to a reduction in extracellular matrix synthesis. Colchicine is a microtubule-destabilizing agent that may enhance the synthesis of cyclic AMP.
Collapse
|
12
|
Potential Effects of Immunosuppression on Oxidative Stress and Atherosclerosis in Kidney Transplant Recipients. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6660846. [PMID: 33688391 PMCID: PMC7920738 DOI: 10.1155/2021/6660846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/06/2021] [Accepted: 02/13/2021] [Indexed: 01/16/2023]
Abstract
Chronic kidney disease is a public health problem that, depending on the country, affects approximately 8-13% of the population, involving both males and females of all ages. Renal replacement therapy remains one of the most costly procedures. It is assumed that one of the factors influencing the course of chronic kidney disease might be oxidative stress. It is believed that the main mediators of oxidative stress are reactive oxygen species (ROS). Transiently increased concentrations of ROS play a significant role in maintaining an organism's homeostasis, as they are part of the redox-related signaling, and in the immune defense system, as they are produced in high amounts in inflammation. Systemic oxidative stress can significantly contribute to endothelial dysfunction along with exaggeration of atherosclerosis and development of cardiovascular disease, the leading cause of mortality in patients with kidney disease. Moreover, the progression of chronic kidney disease is strictly associated with the atherosclerotic process. Transplantation is the optimal method for renal replacement therapy. It improves better quality of life and prolongs survival compared with hemodialysis and peritoneal dialysis; however, even a successful transplantation does not correct the abnormalities found in chronic kidney disease. As transplantation reduces the concentration of uremic toxins, which are a factor of inflammation per se, both the procedure itself and the subsequent immunosuppressive treatment may be a factor that increases oxidative stress and hence vascular sclerosis and atherosclerotic cardiovascular disease. In the current work, we review the effect of several risk factors in kidney transplant recipients as well as immunosuppressive therapy on oxidative stress.
Collapse
|
13
|
Hamczyk MR, Nevado RM, Barettino A, Fuster V, Andrés V. Biological Versus Chronological Aging: JACC Focus Seminar. J Am Coll Cardiol 2020; 75:919-930. [PMID: 32130928 DOI: 10.1016/j.jacc.2019.11.062] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 01/13/2023]
Abstract
Aging is the main risk factor for vascular disease and ensuing cardiovascular and cerebrovascular events, the leading causes of death worldwide. In a progressively aging population, it is essential to develop early-life biomarkers that efficiently identify individuals who are at high risk of developing accelerated vascular damage, with the ultimate goal of improving primary prevention and reducing the health care and socioeconomic impact of age-related cardiovascular disease. Studies in experimental models and humans have identified 9 highly interconnected hallmark processes driving mammalian aging. However, strategies to extend health span and life span require understanding of interindividual differences in age-dependent functional decline, known as biological aging. This review summarizes the current knowledge on biological age biomarkers, factors influencing biological aging, and antiaging interventions, with a focus on vascular aspects of the aging process and its cardiovascular disease related manifestations.
Collapse
Affiliation(s)
- Magda R Hamczyk
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain. https://twitter.com/HamczykMagda
| | - Rosa M Nevado
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Ana Barettino
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Valentín Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Spain; The Zena and Michael A. Wiener Cardiovascular Institute/Marie-Josée and Henry R. Kravis Center for Cardiovascular Health, Mount Sinai School of Medicine, New York, New York
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain.
| |
Collapse
|
14
|
Liberale L, Kraler S, Camici GG, Lüscher TF. Ageing and longevity genes in cardiovascular diseases. Basic Clin Pharmacol Toxicol 2020; 127:120-131. [DOI: 10.1111/bcpt.13426] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/22/2022]
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology University of Zürich Schlieren Switzerland
- Department of Internal Medicine First Clinic of Internal Medicine University of Genoa Genoa Italy
| | - Simon Kraler
- Center for Molecular Cardiology University of Zürich Schlieren Switzerland
| | - Giovanni G. Camici
- Center for Molecular Cardiology University of Zürich Schlieren Switzerland
- Department of Cardiology University Heart Center University Hospital Zurich Zurich Switzerland
- Department of Research and Education University Hospital Zurich Zurich Switzerland
| | - Thomas F. Lüscher
- Center for Molecular Cardiology University of Zürich Schlieren Switzerland
- Heart Division Royal Brompton and Harefield Hospitals and National Heart and Lung Institute Imperial College London UK
| |
Collapse
|
15
|
Saliba F, Duvoux C, Dharancy S, Dumortier J, Calmus Y, Gugenheim J, Kamar N, Salamé E, Neau‐Cransac M, Vanlemmens C, Durand F, Pageaux G, Leroy V, Hardwigsen J, Gharbi H, Masson C, Tindel M, Conti F. Early Switch From Tacrolimus to Everolimus After Liver Transplantation: Outcomes at 2 Years. Liver Transpl 2019; 25:1822-1832. [PMID: 31631501 PMCID: PMC7383505 DOI: 10.1002/lt.25664] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022]
Abstract
The observational CERTITUDE study follows liver transplant patients who completed the SIMCER trial. SIMCER randomized patients at month 1 after transplant to everolimus (EVR) with stepwise tacrolimus (TAC) withdrawal or to standard TAC, both with basiliximab induction and mycophenolic acid ± steroids. After completing SIMCER at 6 months after transplant, 65 EVR-treated patients and 78 TAC-treated patients entered CERTITUDE. At month 24 after transplant, 34/65 (52.3%) EVR-treated patients remained calcineurin inhibitor (CNI) free. Mean estimated glomerular filtration rate (eGFR) was significantly higher with EVR versus TAC during months 3-12. At month 24, eGFR values were 83.6 versus 75.3 mL/minute/1.73 m2 , respectively (P = 0.90) and adjusted mean change in eGFR from randomization was -8.0 versus -13.5 mL/minute/1.73 m2 (P = 0.15). At month 24, 45.9%, 31.1%, and 23.0% of EVR-treated patients had chronic kidney disease stages 1, 2, and 3, respectively, versus 25.7%, 45.7%, and 28.6% of TAC-treated patients (P = 0.05). Treated biopsy-proven acute rejection affected 4 EVR-treated patients and 2 TAC patients during months 6-24. Adverse events led to study discontinuation in 15.4% and 7.7% of EVR-treated and TAC-treated patients, respectively. Grade 3 or 4 hematological events were rare in both groups. A CNI-free EVR-based maintenance regimen appears feasible in approximately half of liver transplant patients. It preserves renal function effectively with good efficacy without compromising safety or hematological tolerance.
Collapse
Affiliation(s)
- Faouzi Saliba
- Centre Hépato‐BiliaireHôpital Paul Brousse, AP‐HPVillejuifFrance
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Paoletti E, Citterio F, Corsini A, Potena L, Rigotti P, Sandrini S, Bussalino E, Stallone G. Everolimus in kidney transplant recipients at high cardiovascular risk: a narrative review. J Nephrol 2019; 33:69-82. [DOI: 10.1007/s40620-019-00609-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/05/2019] [Indexed: 12/20/2022]
|
17
|
Saliba F, Fischer L, de Simone P, Bernhardt P, Bader G, Fung J. Association Between Renal Dysfunction and Major Adverse Cardiac Events After Liver Transplantation: Evidence from an International Randomized Trial of Everolimus-Based Immunosuppression. Ann Transplant 2018; 23:751-757. [PMID: 30361470 PMCID: PMC6248043 DOI: 10.12659/aot.911030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Prospective evidence is lacking regarding the association between renal dysfunction and cardiovascular events after liver transplantation. Material/Methods Data were analyzed post hoc regarding renal function and major adverse cardiac events in a two-year prospective trial of de novo liver transplant recipients randomized at 30 days post-transplant to (i) everolimus [EVR]/reduced tacrolimus [EVR/rTAC] (ii) EVR with tacrolimus discontinued [TAC Elimination] or (iii) standard tacrolimus [TAC Control]. Results By month 24 post-transplant, 32/716 patients had experienced a first major cardiac event (4.5%): 4.1% (10/245), 2.2% (5/229) and 7.0% (17/242) of patients in the EVR/rTAC, TAC Elimination and TAC Control groups, respectively (p=0.043). The cumulative eGFR area under the curve (AUC) from randomization to month 24 was 119 706, 123 082, and 105 946 mL in the EVR/rTAC, TAC Elimination, and TAC Control groups, respectively, corresponding to a mean eGFR AUC of 82.4, 83.0, and 71.9 mL/min/1.73 m2. Cox regression modeling showed that mean eGFR AUC was inversely associated with time to first major cardiac event: the hazard ratio per mL/min/1.73 m2 was −0.0000015 [95% CI −0.00000078; −0.0000024] (p<0.001). Conclusions These findings confirm retrospective evidence that the risk of major cardiac events increases with deteriorating renal function after liver transplantation and demonstrate the need for careful cardiovascular risk management in patients with renal impairment. Immunosuppression based on everolimus with tacrolimus withdrawal, or to a lesser extent tacrolimus reduction, improves both renal function and the risk of major cardiac events compared to standard tacrolimus therapy in liver transplant recipients.
Collapse
Affiliation(s)
- Faouzi Saliba
- Hepato-Biliary Center, AP-HP Hôpital Paul Brousse, Université Paris-Sud, Villejuif, France
| | - Lutz Fischer
- Department of Hepatobiliary Surgery and Transplantation, University Medical Center Eppendorf, Hamburg, Germany
| | - Paolo de Simone
- Hepatobiliary Surgery and Liver Transplantation, University of Pisa, Pisa, Italy
| | | | | | - John Fung
- Transplantation Center, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
18
|
Abstract
Advancing age promotes cardiovascular disease (CVD), the leading cause of death in the United States and many developed nations. Two major age-related arterial phenotypes, large elastic artery stiffening and endothelial dysfunction, are independent predictors of future CVD diagnosis and likely are responsible for the development of CVD in older adults. Not limited to traditional CVD, these age-related changes in the vasculature also contribute to other age-related diseases that influence mammalian health span and potential life span. This review explores mechanisms that influence age-related large elastic artery stiffening and endothelial dysfunction at the tissue level via inflammation and oxidative stress and at the cellular level via Klotho and energy-sensing pathways (AMPK [AMP-activated protein kinase], SIRT [sirtuins], and mTOR [mammalian target of rapamycin]). We also discuss how long-term calorie restriction-a health span- and life span-extending intervention-can prevent many of these age-related vascular phenotypes through the prevention of deleterious alterations in these mechanisms. Lastly, we discuss emerging novel mechanisms of vascular aging, including senescence and genomic instability within cells of the vasculature. As the population of older adults steadily expands, elucidating the cellular and molecular mechanisms of vascular dysfunction with age is critical to better direct appropriate and measured strategies that use pharmacological and lifestyle interventions to reduce risk of CVD within this population.
Collapse
Affiliation(s)
- Anthony J. Donato
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| | - Daniel R. Machin
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| | - Lisa A. Lesniewski
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| |
Collapse
|
19
|
Affiliation(s)
- Kristen L Nowak
- From the University of Colorado Anschutz Medical Campus, Aurora (K.L.N., M.C.); and University of Colorado Boulder (M.J.R., D.R.S.).
| | - Matthew J Rossman
- From the University of Colorado Anschutz Medical Campus, Aurora (K.L.N., M.C.); and University of Colorado Boulder (M.J.R., D.R.S.)
| | - Michel Chonchol
- From the University of Colorado Anschutz Medical Campus, Aurora (K.L.N., M.C.); and University of Colorado Boulder (M.J.R., D.R.S.)
| | - Douglas R Seals
- From the University of Colorado Anschutz Medical Campus, Aurora (K.L.N., M.C.); and University of Colorado Boulder (M.J.R., D.R.S.)
| |
Collapse
|
20
|
Jones-Hughes T, Snowsill T, Haasova M, Coelho H, Crathorne L, Cooper C, Mujica-Mota R, Peters J, Varley-Campbell J, Huxley N, Moore J, Allwood M, Lowe J, Hyde C, Hoyle M, Bond M, Anderson R. Immunosuppressive therapy for kidney transplantation in adults: a systematic review and economic model. Health Technol Assess 2018; 20:1-594. [PMID: 27578428 DOI: 10.3310/hta20620] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND End-stage renal disease is a long-term irreversible decline in kidney function requiring renal replacement therapy: kidney transplantation, haemodialysis or peritoneal dialysis. The preferred option is kidney transplantation, followed by immunosuppressive therapy (induction and maintenance therapy) to reduce the risk of kidney rejection and prolong graft survival. OBJECTIVES To review and update the evidence for the clinical effectiveness and cost-effectiveness of basiliximab (BAS) (Simulect(®), Novartis Pharmaceuticals UK Ltd) and rabbit anti-human thymocyte immunoglobulin (rATG) (Thymoglobulin(®), Sanofi) as induction therapy, and immediate-release tacrolimus (TAC) (Adoport(®), Sandoz; Capexion(®), Mylan; Modigraf(®), Astellas Pharma; Perixis(®), Accord Healthcare; Prograf(®), Astellas Pharma; Tacni(®), Teva; Vivadex(®), Dexcel Pharma), prolonged-release tacrolimus (Advagraf(®) Astellas Pharma), belatacept (BEL) (Nulojix(®), Bristol-Myers Squibb), mycophenolate mofetil (MMF) (Arzip(®), Zentiva; CellCept(®), Roche Products; Myfenax(®), Teva), mycophenolate sodium (MPS) (Myfortic(®), Novartis Pharmaceuticals UK Ltd), sirolimus (SRL) (Rapamune(®), Pfizer) and everolimus (EVL) (Certican(®), Novartis) as maintenance therapy in adult renal transplantation. METHODS Clinical effectiveness searches were conducted until 18 November 2014 in MEDLINE (via Ovid), EMBASE (via Ovid), Cochrane Central Register of Controlled Trials (via Wiley Online Library) and Web of Science (via ISI), Cochrane Database of Systematic Reviews, Database of Abstracts of Reviews of Effects and Health Technology Assessment (The Cochrane Library via Wiley Online Library) and Health Management Information Consortium (via Ovid). Cost-effectiveness searches were conducted until 18 November 2014 using a costs or economic literature search filter in MEDLINE (via Ovid), EMBASE (via Ovid), NHS Economic Evaluation Database (via Wiley Online Library), Web of Science (via ISI), Health Economic Evaluations Database (via Wiley Online Library) and the American Economic Association's electronic bibliography (via EconLit, EBSCOhost). Included studies were selected according to predefined methods and criteria. A random-effects model was used to analyse clinical effectiveness data (odds ratios for binary data and mean differences for continuous data). Network meta-analyses were undertaken within a Bayesian framework. A new discrete time-state transition economic model (semi-Markov) was developed, with acute rejection, graft function (GRF) and new-onset diabetes mellitus used to extrapolate graft survival. Recipients were assumed to be in one of three health states: functioning graft, graft loss or death. RESULTS Eighty-nine randomised controlled trials (RCTs), of variable quality, were included. For induction therapy, no treatment appeared more effective than another in reducing graft loss or mortality. Compared with placebo/no induction, rATG and BAS appeared more effective in reducing biopsy-proven acute rejection (BPAR) and BAS appeared more effective at improving GRF. For maintenance therapy, no treatment was better for all outcomes and no treatment appeared most effective at reducing graft loss. BEL + MMF appeared more effective than TAC + MMF and SRL + MMF at reducing mortality. MMF + CSA (ciclosporin), TAC + MMF, SRL + TAC, TAC + AZA (azathioprine) and EVL + CSA appeared more effective than CSA + AZA and EVL + MPS at reducing BPAR. SRL + AZA, TAC + AZA, TAC + MMF and BEL + MMF appeared to improve GRF compared with CSA + AZA and MMF + CSA. In the base-case deterministic and probabilistic analyses, BAS, MMF and TAC were predicted to be cost-effective at £20,000 and £30,000 per quality-adjusted life-year (QALY). When comparing all regimens, only BAS + TAC + MMF was cost-effective at £20,000 and £30,000 per QALY. LIMITATIONS For included trials, there was substantial methodological heterogeneity, few trials reported follow-up beyond 1 year, and there were insufficient data to perform subgroup analysis. Treatment discontinuation and switching were not modelled. FUTURE WORK High-quality, better-reported, longer-term RCTs are needed. Ideally, these would be sufficiently powered for subgroup analysis and include health-related quality of life as an outcome. CONCLUSION Only a regimen of BAS induction followed by maintenance with TAC and MMF is likely to be cost-effective at £20,000-30,000 per QALY. STUDY REGISTRATION This study is registered as PROSPERO CRD42014013189. FUNDING The National Institute for Health Research Health Technology Assessment programme.
Collapse
Affiliation(s)
- Tracey Jones-Hughes
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Tristan Snowsill
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Marcela Haasova
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Helen Coelho
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Louise Crathorne
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Chris Cooper
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Ruben Mujica-Mota
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Jaime Peters
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Jo Varley-Campbell
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Nicola Huxley
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Jason Moore
- Exeter Kidney Unit, Royal Devon and Exeter Foundation Trust Hospital, Exeter, UK
| | - Matt Allwood
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Jenny Lowe
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Chris Hyde
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Martin Hoyle
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Mary Bond
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| | - Rob Anderson
- Peninsula Technology Assessment Group (PenTAG), University of Exeter, Exeter, UK
| |
Collapse
|
21
|
|
22
|
Melilli E, Manonelles A, Montero N, Grinyo J, Martinez-Castelao A, Bestard O, Cruzado J. Impact of immunosuppressive therapy on arterial stiffness in kidney transplantation: are all treatments the same? Clin Kidney J 2017; 11:413-421. [PMID: 29988241 PMCID: PMC6007381 DOI: 10.1093/ckj/sfx120] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 09/06/2017] [Indexed: 02/07/2023] Open
Abstract
Arterial stiffness is a biologic process related to ageing and its relationship with cardiovascular risk is well established. Several methods are currently available for non-invasive measurement of arterial stiffness that provide valuable information to further assess patients’ vascular status in real time. In kidney transplantation recipients, several factors could accelerate the stiffness process, such as the use of calcineurin inhibitors (CNIs), the presence of chronic kidney disease and other classical cardiovascular factors, which would explain, at least in part, the high cardiovascular mortality and morbidity. Despite the importance of arterial stiffness as a biomarker of cardiovascular risk, and unlike other cardiovascular risk factors (e.g. left ventricular hypertrophy), only a few clinical trials or retrospective studies of kidney recipients have evaluated its impact. In this review we describe the clinical impact of arterial stiffness as a prognostic marker of cardiovascular disease and the effects of different immunosuppressive regimens on its progression, focusing on the potential benefits of CNI-sparing protocols and supporting the rationale for individualization of immunosuppression in patients with lower arterial elasticity. Among the immunosuppressive drugs, a belatacept-based regimen seems to offer better vascular protection compared with CNIs, although further studies are needed to confirm the preliminary positive results.
Collapse
Affiliation(s)
- Edoardo Melilli
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
- Correspondence and offprint requests to: Edoardo Melilli; E-mail:
| | - Anna Manonelles
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | - Nuria Montero
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | - Josep Grinyo
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | | | - Oriol Bestard
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| | - Josep Cruzado
- Department of Nephrology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Cataluny, Spain
| |
Collapse
|
23
|
Cardiovascular Parameters to 2 years After Kidney Transplantation Following Early Switch to Everolimus Without Calcineurin Inhibitor Therapy: An Analysis of the Randomized ELEVATE Study. Transplantation 2017; 101:2612-2620. [PMID: 28333860 DOI: 10.1097/tp.0000000000001739] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Mammalian target of rapamycin inhibitors may confer cardioprotective advantages, but clinical data are limited. METHODS In the open-label ELEVATE trial, kidney transplant patients were randomized at 10 to 14 weeks after transplant to convert from calcineurin inhibitor (CNI) to everolimus or remain on standard CNI therapy. Prespecified end points included left ventricular mass index and, in a subpopulation of patients, arterial stiffness as measured by pulse wave velocity. RESULTS The mean change in left ventricular mass index from randomization was similar with everolimus versus CNI (month 24, -4.37 g/m versus -5.26 g/m; mean difference, 0.89 [p = 0.392]). At month 24, left ventricular hypertrophy was present in 41.7% versus 37.7% of everolimus and CNI patients, respectively. Mean pulse wave velocity remained stable with both everolimus (mean change from randomization to month 12, -0.24 m/s; month 24, -0.03 m/s) and CNI (month 12, 0.11 m/s; month 24, 0.16 m/s). The change in mean ambulatory nighttime blood pressure from randomization showed a benefit for diastolic pressure at month 12 (P = 0.039) but not at month 24. Major adverse cardiac events occurred in 1.1% and 4.2% of everolimus-treated and CNI-treated patients, respectively, by month 12 (P = 0.018) and 2.3% (8/353) and 4.5% by month 24 (P = 0.145). CONCLUSIONS Overall, these data do not suggest a clinically relevant effect on cardiac end points after early conversion from CNI to a CNI-free everolimus-based regimen.
Collapse
|
24
|
Jiao Y, Li G, Li Q, Ali R, Qin L, Li W, Qyang Y, Greif DM, Geirsson A, Humphrey JD, Tellides G. mTOR (Mechanistic Target of Rapamycin) Inhibition Decreases Mechanosignaling, Collagen Accumulation, and Stiffening of the Thoracic Aorta in Elastin-Deficient Mice. Arterioscler Thromb Vasc Biol 2017; 37:1657-1666. [PMID: 28751568 DOI: 10.1161/atvbaha.117.309653] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 07/17/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Elastin deficiency because of heterozygous loss of an ELN allele in Williams syndrome causes obstructive aortopathy characterized by medial thickening and fibrosis and consequent aortic stiffening. Previous work in Eln-null mice with a severe arterial phenotype showed that inhibition of mTOR (mechanistic target of rapamycin), a key regulator of cell growth, lessened the aortic obstruction but did not prevent early postnatal death. We investigated the effects of mTOR inhibition in Eln-null mice partially rescued by human ELN that manifest a less severe arterial phenotype and survive long term. APPROACH AND RESULTS Thoracic aortas of neonatal and juvenile mice with graded elastin deficiency exhibited increased signaling through both mTOR complex 1 and 2. Despite lower predicted wall stress, there was increased phosphorylation of focal adhesion kinase, suggestive of greater integrin activation, and increased transforming growth factor-β-signaling mediators, associated with increased collagen expression. Pharmacological blockade of mTOR by rapalogs did not improve luminal stenosis but reduced mechanosignaling (in delayed fashion after mTOR complex 1 inhibition), medial collagen accumulation, and stiffening of the aorta. Rapalog administration also retarded somatic growth, however, and precipitated neonatal deaths. Complementary, less-toxic strategies to inhibit mTOR via altered growth factor and nutrient responses were not effective. CONCLUSIONS In addition to previously demonstrated therapeutic benefits of rapalogs decreasing smooth muscle cell proliferation in the absence of elastin, we find that rapalogs also prevent aortic fibrosis and stiffening attributable to partial elastin deficiency. Our findings suggest that mTOR-sensitive perturbation of smooth muscle cell mechanosensing contributes to elastin aortopathy.
Collapse
Affiliation(s)
- Yang Jiao
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Guangxin Li
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Qingle Li
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Rahmat Ali
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Lingfeng Qin
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Wei Li
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Yibing Qyang
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Daniel M Greif
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Arnar Geirsson
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - Jay D Humphrey
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.)
| | - George Tellides
- From the Department of Surgery (Y.J., G.L., Q.L., R.A., L.Q., W.L., A.G., G.T.), Department of Internal Medicine (Y.Q., D.M.G.), and Vascular Biology and Therapeutics Program (Y.Q., D.M.G., A.G., J.D.H., G.T.), Yale University School of Medicine, New Haven, CT; Department of Vascular Surgery, Peking University People's Hospital, Beijing, P. R. China (Y.J., Q.L., W.L.); Department of Biomedical Engineering, Yale University, New Haven, CT (J.D.H.); and Veterans Affairs Connecticut Healthcare System, West Haven (G.T.).
| |
Collapse
|
25
|
Karpe KM, Talaulikar GS, Walters GD. Calcineurin inhibitor withdrawal or tapering for kidney transplant recipients. Cochrane Database Syst Rev 2017; 7:CD006750. [PMID: 28730648 PMCID: PMC6483545 DOI: 10.1002/14651858.cd006750.pub2] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Calcineurin inhibitors (CNI) can reduce acute transplant rejection and immediate graft loss but are associated with significant adverse effects such as hypertension and nephrotoxicity which may contribute to chronic rejection. CNI toxicity has led to numerous studies investigating CNI withdrawal and tapering strategies. Despite this, uncertainty remains about minimisation or withdrawal of CNI. OBJECTIVES This review aimed to look at the benefits and harms of CNI tapering or withdrawal in terms of graft function and loss, incidence of acute rejection episodes, treatment-related side effects (hypertension, hyperlipidaemia) and death. SEARCH METHODS We searched the Cochrane Kidney and Transplant Specialised Register to 11 October 2016 through contact with the Information Specialist using search terms relevant to this review. Studies contained in the Specialised Register are identified through search strategies specifically designed for CENTRAL, MEDLINE, and EMBASE; handsearching conference proceedings; and searching the International Clinical Trials Register (ICTRP) Search Portal and ClinicalTrials.gov. SELECTION CRITERIA All randomised controlled trials (RCTs) where drug regimens containing CNI were compared to alternative drug regimens (CNI withdrawal, tapering or low dose) in the post-transplant period were included, without age or dosage restriction. DATA COLLECTION AND ANALYSIS Two authors independently assessed studies for eligibility, risk of bias, and extracted data. Results were expressed as risk ratio (RR) or mean difference (MD) with 95% confidence intervals (CI). MAIN RESULTS We included 83 studies that involved 16,156 participants. Most were open-label studies; less than 30% of studies reported randomisation method and allocation concealment. Studies were analysed as intent-to-treat in 60% and all pre-specified outcomes were reported in 54 studies. The attrition and reporting bias were unclear in the remainder of the studies as factors used to judge bias were reported inconsistently. We also noted that 50% (47 studies) of studies were funded by the pharmaceutical industry.We classified studies into four groups: CNI withdrawal or avoidance with or without substitution with mammalian target of rapamycin inhibitors (mTOR-I); and low dose CNI with or without mTOR-I. The withdrawal groups were further stratified as avoidance and withdrawal subgroups for major outcomes.CNI withdrawal may lead to rejection (RR 2.54, 95% CI 1.56 to 4.12; moderate certainty evidence), may make little or no difference to death (RR 1.09, 95% CI 0.96 to 1.24; moderate certainty), and probably slightly reduces graft loss (RR 0.85, 95% CI 0.74 to 0.98; low quality evidence). Hypertension was probably reduced in the CNI withdrawal group (RR 0.82, 95% CI 0.71 to 0.95; low certainty), while CNI withdrawal may make little or no difference to malignancy (RR 1.10, 95% CI 0.93 to 1.30; low certainty), and probably makes little or no difference to cytomegalovirus (CMV) (RR 0.87, 95% CI 0.52 to 1.45; low certainty)CNI avoidance may result in increased acute rejection (RR 2.16, 95% CI 0.85 to 5.49; low certainty) but little or no difference in graft loss (RR 0.96, 95% CI 0.79 to 1.16; low certainty). Late CNI withdrawal increased acute rejection (RR 3.21, 95% CI 1.59 to 6.48; moderate certainty) but probably reduced graft loss (RR 0.84, 95% CI 0.72 to 0.97, low certainty).Results were similar when CNI avoidance or withdrawal was combined with the introduction of mTOR-I; acute rejection was probably increased (RR 1.43; 95% CI 1.15 to 1.78; moderate certainty) and there was probably little or no difference in death (RR 0.96; 95% CI 0.69 to 1.36, moderate certainty). mTOR-I substitution may make little or no difference to graft loss (RR 0.94, 95% CI 0.75 to 1.19; low certainty), probably makes little of no difference to hypertension (RR 0.86, 95% CI 0.64 to 1.15; moderate), and probably reduced the risk of cytomegalovirus (CMV) (RR 0.60, 95% CI 0.44 to 0.82; moderate certainty) and malignancy (RR 0.69, 95% CI 0.47 to 1.00; low certainty). Lymphoceles were increased with mTOR-I substitution (RR 1.45, 95% CI 0.95 to 2.21; low certainty).Low dose CNI combined with mTOR-I probably increased glomerular filtration rate (GFR) (MD 6.24 mL/min, 95% CI 3.28 to 9.119; moderate certainty), reduced graft loss (RR 0.75, 95% CI 0.55 to 1.02; moderate certainty), and made little or no difference to acute rejection (RR 1.13 ; 95% CI 0.91 to 1.40; moderate certainty). Hypertension was decreased (RR 0.98, 95% CI 0.80 to 1.20; low certainty) as was CMV (RR 0.41, 95% CI 0.16 to 1.06; low certainty). Low dose CNI plus mTOR-I makes probably makes little of no difference to malignancy (RR 1.22, 95% CI 0.42 to 3.53; low certainty) and may make little of no difference to death (RR 1.16, 95% CI 0.71 to 1.90; moderate certainty). AUTHORS' CONCLUSIONS CNI avoidance increased acute rejection and CNI withdrawal increases acute rejection but reduced graft loss at least over the short-term. Low dose CNI with induction regimens reduced acute rejection and graft loss with no major adverse events, also in the short-term. The use of mTOR-I reduced CMV infections but increased the risk of acute rejection. These conclusions must be tempered by the lack of long-term data in most of the studies, particularly with regards to chronic antibody-mediated rejection, and the suboptimal methodological quality of the included studies.
Collapse
Affiliation(s)
- Krishna M Karpe
- Canberra HospitalRenal ServicesYamba DriveGarranACTAustralia2605
- Australian National University Medical SchoolActonACTAustralia2601
| | - Girish S Talaulikar
- Canberra HospitalRenal ServicesYamba DriveGarranACTAustralia2605
- Australian National University Medical SchoolActonACTAustralia2601
| | - Giles D Walters
- Canberra HospitalRenal ServicesYamba DriveGarranACTAustralia2605
- Australian National University Medical SchoolActonACTAustralia2601
| | | |
Collapse
|
26
|
de Fijter JW, Holdaas H, Øyen O, Sanders JS, Sundar S, Bemelman FJ, Sommerer C, Pascual J, Avihingsanon Y, Pongskul C, Oppenheimer F, Toselli L, Russ G, Wang Z, Lopez P, Kochuparampil J, Cruzado JM, van der Giet M. Early Conversion From Calcineurin Inhibitor- to Everolimus-Based Therapy Following Kidney Transplantation: Results of the Randomized ELEVATE Trial. Am J Transplant 2017; 17:1853-1867. [PMID: 28027625 DOI: 10.1111/ajt.14186] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 12/20/2016] [Indexed: 01/25/2023]
Abstract
In a 24-month, multicenter, open-label, randomized trial, 715 de novo kidney transplant recipients were randomized at 10-14 weeks to convert to everolimus (n = 359) or remain on standard calcineurin inhibitor (CNI) therapy (n = 356; 231 tacrolimus; 125 cyclosporine), all with mycophenolic acid and steroids. The primary endpoint, change in estimated glomerular filtration rate (eGFR) from randomization to month 12, was similar for everolimus versus CNI: mean (standard error) 0.3(1.5) mL/min/1.732 versus -1.5(1.5) mL/min/1.732 (p = 0.116). Biopsy-proven acute rejection (BPAR) at month 12 was more frequent under everolimus versus CNI overall (9.7% vs. 4.8%, p = 0.014) and versus tacrolimus-treated patients (2.6%, p < 0.001) but similar to cyclosporine-treated patients (8.8%, p = 0.755). Reporting on de novo donor-specific antibodies (DSA) was limited but suggested more frequent anti-HLA Class I DSA under everolimus. Change in left ventricular mass index was similar. Discontinuation due to adverse events was more frequent with everolimus (23.6%) versus CNI (8.4%). In conclusion, conversion to everolimus at 10-14 weeks posttransplant was associated with renal function similar to that with standard therapy overall. Rates of BPAR were low in all groups, but lower with tacrolimus than everolimus.
Collapse
Affiliation(s)
- J W de Fijter
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - H Holdaas
- Section of Nephrology, Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - O Øyen
- Section of Transplant Surgery, Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - J-S Sanders
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - S Sundar
- Department of Nephrology, Columbia Asia Hospitals, Malleshwaram West, Bangalore, India
| | - F J Bemelman
- Department of Nephrology, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - C Sommerer
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - J Pascual
- Department of Nephrology, Hospital del Mar, Barcelona, Spain
| | - Y Avihingsanon
- Faculty of Medicine, Chulalongkorn University and Excellent Center of Organ Transplantation, King Chulalongkorn Memorial Hospital, Patumwan, Bangkok, Thailand
| | - C Pongskul
- Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - F Oppenheimer
- Department of Nephrology and Kidney Transplantation, Hospital Clínic de Barcelona, Barcelona, Spain
| | - L Toselli
- Kidney, Liver and Pancreas Transplant Unit, CRAI Norte, Ministry of Health, Buenos Aires, Argentina
| | - G Russ
- University of Adelaide and Central and Northern Adelaide Renal and Transplant Service, Royal Adelaide Hospital, North Terrace, Adelaide, Australia
| | - Z Wang
- Biometrics and Statistical Science, Novartis Pharmaceuticals, East Hanover, NJ
| | - P Lopez
- Research and Development, Novartis Pharma AG, Basel, Switzerland
| | - J Kochuparampil
- Research and Development, Novartis Pharma AG, Basel, Switzerland
| | - J M Cruzado
- Department of Nephrology, University Hospital of Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - M van der Giet
- Department of Nephrology, Charité - Universitätsmedizin, Berlin, Germany
| | | |
Collapse
|
27
|
Effect of Immunosuppressive Treatment on Carotid Atherosclerosis in Renal Transplant Recipients. Transplant Proc 2017; 48:1626-9. [PMID: 27496459 DOI: 10.1016/j.transproceed.2016.03.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/17/2016] [Accepted: 03/01/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND The aim of this study was to compare the effect of immunosuppressive regimens using either mammalian target of rapamycin inhibitors (mTORi) or calcineurin inhibitors (CNI) on the risk of atherosclerosis in renal transplant patients. MATERIALS AND METHODS The study involved a group of 24 recipients treated with mTORi (mTORi group) and a group of 20 recipients treated with immunosuppressive regimen based on CNI (CNI group). Laboratory and clinical markers of cardiovascular risk in both groups were investigated. Carotid atherosclerosis was evaluated by measurement of the intima media thickness (IMT) of the common and internal carotid artery walls and detection of carotid plaques by a high-resolution ultrasonography. The study was performed 3-24 years after transplantation. RESULTS The mTORi group showed higher level of total cholesterol (242 vs 201 mg/dL; P < .004), low-density lipoprotein cholesterol (140 vs 116 mg/dL; P < .05), and triglycerides (226 vs 168 mg/dL; P < .01). Posttransplant diabetes developed in 34% of mTORi group compared with 25% in the CNI group. The mean of IMT (left and right) of common and internal carotid arteries was similar in both groups. Carotid plaques were detected in 46% of patients from the mTORi group and 25% from CNI group (P < .02). The presence of carotid plaques combined with an IMT of >0.9 mm were associated with male gender, mTORi treatment (P = .03), and cardiovascular events. The incidence of coronary heart disease was higher in mTORi group than in CNI group (53% vs 20%; P = .03). CONCLUSIONS There was not beneficial effect of immunosuppressive treatment with mTORi on carotid atherosclerosis in renal transplant patients.
Collapse
|
28
|
Unique Considerations When Managing Hypertension in the Transplant Patient. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016. [PMID: 27815930 DOI: 10.1007/5584_2016_87] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
For the select fortunate recipients of organ transplants, transplantation affords the rare opportunity for a new life. Given the scarcity of organs for transplantation, it is imperative that the health of transplant recipients be optimized in order to fully benefit from this gift of life. Unfortunately, hypertension is highly prevalent in the transplant population and it is considered a major cardiovascular risk factor contributing to mortality and morbidity in this population. In this chapter, we expound on the epidemiology, unique pathophysiology, evaluation, and management of hypertension as it pertains to the solid organ transplant recipient. In addition, a brief commentary is made on the subject of hypertension following living kidney donation, and practical aspects of management of hypertension in the solid organ recipient are summarized at the end of the chapter.
Collapse
|
29
|
Roles of mTOR complexes in the kidney: implications for renal disease and transplantation. Nat Rev Nephrol 2016; 12:587-609. [PMID: 27477490 DOI: 10.1038/nrneph.2016.108] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The mTOR pathway has a central role in the regulation of cell metabolism, growth and proliferation. Studies involving selective gene targeting of mTOR complexes (mTORC1 and mTORC2) in renal cell populations and/or pharmacologic mTOR inhibition have revealed important roles of mTOR in podocyte homeostasis and tubular transport. Important advances have also been made in understanding the role of mTOR in renal injury, polycystic kidney disease and glomerular diseases, including diabetic nephropathy. Novel insights into the roles of mTORC1 and mTORC2 in the regulation of immune cell homeostasis and function are helping to improve understanding of the complex effects of mTOR targeting on immune responses, including those that impact both de novo renal disease and renal allograft outcomes. Extensive experience in clinical renal transplantation has resulted in successful conversion of patients from calcineurin inhibitors to mTOR inhibitors at various times post-transplantation, with excellent long-term graft function. Widespread use of this practice has, however, been limited owing to mTOR-inhibitor- related toxicities. Unique attributes of mTOR inhibitors include reduced rates of squamous cell carcinoma and cytomegalovirus infection compared to other regimens. As understanding of the mechanisms by which mTORC1 and mTORC2 drive the pathogenesis of renal disease progresses, clinical studies of mTOR pathway targeting will enable testing of evolving hypotheses.
Collapse
|
30
|
Granata S, Dalla Gassa A, Carraro A, Brunelli M, Stallone G, Lupo A, Zaza G. Sirolimus and Everolimus Pathway: Reviewing Candidate Genes Influencing Their Intracellular Effects. Int J Mol Sci 2016; 17:ijms17050735. [PMID: 27187382 PMCID: PMC4881557 DOI: 10.3390/ijms17050735] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/21/2016] [Accepted: 05/06/2016] [Indexed: 02/07/2023] Open
Abstract
Sirolimus (SRL) and everolimus (EVR) are mammalian targets of rapamycin inhibitors (mTOR-I) largely employed in renal transplantation and oncology as immunosuppressive/antiproliferative agents. SRL was the first mTOR-I produced by the bacterium Streptomyces hygroscopicus and approved for several medical purposes. EVR, derived from SRL, contains a 2-hydroxy-ethyl chain in the 40th position that makes the drug more hydrophilic than SRL and increases oral bioavailability. Their main mechanism of action is the inhibition of the mTOR complex 1 and the regulation of factors involved in a several crucial cellular functions including: protein synthesis, regulation of angiogenesis, lipid biosynthesis, mitochondrial biogenesis and function, cell cycle, and autophagy. Most of the proteins/enzymes belonging to the aforementioned biological processes are encoded by numerous and tightly regulated genes. However, at the moment, the polygenic influence on SRL/EVR cellular effects is still not completely defined, and its comprehension represents a key challenge for researchers. Therefore, to obtain a complete picture of the cellular network connected to SRL/EVR, we decided to review major evidences available in the literature regarding the genetic influence on mTOR-I biology/pharmacology and to build, for the first time, a useful and specific “SRL/EVR genes-focused pathway”, possibly employable as a starting point for future in-depth research projects.
Collapse
Affiliation(s)
- Simona Granata
- Renal Unit, Department of Medicine, University/Hospital of Verona, 37126 Verona, Italy.
| | | | - Amedeo Carraro
- Liver Transplant Unit, Department of General Surgery and Odontoiatrics, University/Hospital of Verona, 37126 Verona, Italy.
| | - Matteo Brunelli
- Department of Pathology and Diagnostics, University of Verona, Azienda Ospedaliera Universitaria Integrata, 37126 Verona, Italy.
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, University of Foggia, 71122 Foggia, Italy.
| | - Antonio Lupo
- Renal Unit, Department of Medicine, University/Hospital of Verona, 37126 Verona, Italy.
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University/Hospital of Verona, 37126 Verona, Italy.
| |
Collapse
|
31
|
Tsai JP, Lee MC, Le MC, Chen YC, Ho GJ, Shih MH, Hsu BG. Hyperleptinemia Is a Risk Factor for the Development of Central Arterial Stiffness in Kidney Transplant Patients. Transplant Proc 2016; 47:1825-30. [PMID: 26293058 DOI: 10.1016/j.transproceed.2015.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/13/2015] [Accepted: 06/02/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Arterial stiffness could cause adverse outcomes in kidney transplant (KT) patients. Leptin has a role in influencing vascular smooth muscle that may contribute to atherosclerosis. The aim of this study was to evaluate the relationship between fasting serum leptin concentration and carotid-femoral pulse wave velocity (cfPWV) in KT patients. MATERIALS AND METHODS Fasting blood samples were obtained from 55 KT patients and 65 subjects from the outpatient department were enrolled as the control group. The cfPWV values of >10 m/s were used to define as the high arterial stiffness group and <10 m/s as the low arterial stiffness group. The predictive ability of leptin for arterial stiffness of KT was assessed using receiver operating characteristic (ROC) curve and multivariate logistic regression analyses. RESULTS Kidney transplant patients had lower hemoglobin, but higher blood urea nitrogen, creatinine, total cholesterol, diastolic blood pressure, intact parathyroid hormone levels, and leptin levels than controls. Although cfPWV levels were higher in KT patients, there is no difference of cfPWV levels between KT patients and control (P = .595). Fifteen KT patients (27.3%) were defined in the high arterial stiffness group, and serum leptin level was higher in the high arterial stiffness group compared with the low arterial stiffness group in KT patients (P < .001). Multivariate logistic regression analysis showed that leptin (odds ratio: 1.044, 95% confidence interval [CI]: 1.016-1.072, P = .002) was an independent predictor of arterial stiffness in KT patients. The sensitivity, specificity, positive predictive value, negative predictive value, and area under the ROC curve predicting arterial stiffness in KT patients were 73.33%, 87.5%, 68.7%, 89.7%, and 0.828 (95% CI: 0.703-0.917, P < .001), and the leptin cut-off value was 74.14 ng/mL. CONCLUSION Serum fasting leptin level could predict the development of central arterial stiffness of KT patients.
Collapse
Affiliation(s)
- J P Tsai
- Division of Nephrology, Buddhist Tzu Chi General Hospital, Dalin Branch, Chiayi, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan
| | | | - M C Le
- School of Medicine, Tzu Chi University, Hualien, Taiwan; Department of Surgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Y C Chen
- Department of Surgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - G J Ho
- Department of Surgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - M H Shih
- Department of Nursing, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - B G Hsu
- School of Medicine, Tzu Chi University, Hualien, Taiwan; Division of Nephrology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan.
| |
Collapse
|
32
|
Witzke O, Sommerer C, Arns W. Everolimus immunosuppression in kidney transplantation: What is the optimal strategy? Transplant Rev (Orlando) 2016; 30:3-12. [DOI: 10.1016/j.trre.2015.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 08/31/2015] [Accepted: 09/01/2015] [Indexed: 12/19/2022]
|
33
|
Cho YE, Kim HS, Lai C, Stanfill A, Cashion A. Oxidative stress is associated with weight gain in recipients at 12-months following kidney transplantation. Clin Biochem 2015; 49:237-42. [PMID: 26545907 DOI: 10.1016/j.clinbiochem.2015.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 10/28/2015] [Accepted: 11/01/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Weight gain after kidney transplantation (Tx) is considered a risk factor for poor outcomes. Increased oxidative stress is associated with not only chronic renal disease and Tx, but also obesity and cardiovascular disease. The aim of this pilot study was to test whether oxidative stress is related to weight gain at 12-months after kidney Tx and to obtain preliminary insight into potential mechanisms involved. DESIGN & METHODS Recipients (n=33) were classified into two groups; weight loss and weight gain, based on their weight changes at 12-months post-transplant. Total antioxidant capacity (TAOC) and lipid peroxidation (TBARS) were measured to evaluate oxidative stress from plasma at baseline and 12-months. A secondary data analysis was conducted to identify potential gene regulation. RESULTS Seventeen recipients lost (-6.63±5.52kg), and sixteen recipients gained weight (8.94±6.18kg). TAOC was significantly decreased at 12-months compared to baseline for the total group, however, there was no significant difference between groups at either time point. TBARS was higher in weight gain group, at both time points, and it was significantly higher at 12-months (p=0.012). Gene expression profiling analysis showed that 7 transcripts annotated to reactive oxygen species related genes in adipose tissue were expressed significantly lower in weight gain group at baseline, which might be a negative feedback mechanism to reduce oxidative stress. CONCLUSION These results may indicate that elevated oxidative stress (TBARS) is associated with weight gain after kidney Tx and that incorporating early clinical prevention strategies known to decrease oxidative stress could be recommended.
Collapse
Affiliation(s)
- Young-Eun Cho
- National Institute of Nursing Research, National Institutes of Health, 3 Center Drive, Building 3, Room 5E26, Bethesda, MD 20892-2178, USA.
| | - Hyung-Suk Kim
- National Institute of Nursing Research, National Institutes of Health, 3 Center Drive, Building 3, Room 5E26, Bethesda, MD 20892-2178, USA.
| | - Chen Lai
- National Institute of Nursing Research, National Institutes of Health, 3 Center Drive, Building 3, Room 5E26, Bethesda, MD 20892-2178, USA.
| | - Ansley Stanfill
- University of Pittsburgh, 440 Victoria Building, 3500 Victoria St, Pittsburgh, PA 15261, USA.
| | - Ann Cashion
- National Institute of Nursing Research, National Institutes of Health, 3 Center Drive, Building 3, Room 5E26, Bethesda, MD 20892-2178, USA.
| |
Collapse
|
34
|
Aortic Stiffness in a Mortality Risk Calculator for Kidney Transplant Recipients. Transplantation 2015; 99:1730-7. [DOI: 10.1097/tp.0000000000000660] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
35
|
Birdwell KA, Jaffe G, Bian A, Wu P, Ikizler TA. Assessment of arterial stiffness using pulse wave velocity in tacrolimus users the first year post kidney transplantation: a prospective cohort study. BMC Nephrol 2015; 16:93. [PMID: 26133166 PMCID: PMC4489361 DOI: 10.1186/s12882-015-0092-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/23/2015] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The leading cause of death in end stage renal disease is cardiovascular disease (CVD). Kidney transplantation is associated with improved survival over dialysis. We hypothesized that arterial stiffness, a marker of CVD, would improve in patients post kidney transplant, potentially explaining one mechanism of survival benefit from transplant. METHODS After obtaining Institutional Review Board approval and informed consent, we performed a longitudinal prospective cohort study of 66 newly transplanted adult kidney transplant recipients, using aortic pulse wave velocity (PWV) to assess arterial stiffness over a 12 month period. All patients were assessed within one month of transplant (baseline) and 12 months post transplant. The primary outcome was change in PWV score at 12 months which we assessed using Wilcoxon Signed Rank test. Secondary analyses included correlation of predictors with PWV score at both time points. RESULTS The median age of the cohort was 49.7 years at transplant, with 27 % Black and 27 % female. At baseline, 43 % had tobacco use, 30 % had a history of CVD, and 42 % had diabetes. Median baseline calcium was 9.1 mg/dL and median phosphorus was 5.1 mg/dL. Median PWV score was 9.25 and 8.97 m/s at baseline versus month 12, respectively, showing no significant change (median change of -0.07, p = 0.7). In multivariable regression, subjects with increased age at transplant (p = 0.008), diabetes (p = 0.002), and a higher baseline PWV score (p < 0.001) were at increased risk of having a high PWV score 12 months post transplant. CONCLUSION Aortic arterial stiffness does not progress in the first year post kidney transplant. Increasing age, diabetes, and higher baseline PWV score identify patients at risk for increased arterial stiffness. Further research that assesses patients for greater than one year and includes a control dialysis group would be helpful in further understanding the change in arterial stiffness post transplantation.
Collapse
Affiliation(s)
- Kelly Ann Birdwell
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, 1161 21st Avenue, S-3223 MCN, Nashville, TN, 37232, USA.
| | - Gilad Jaffe
- University at Buffalo School of Medicine and Biomedical Sciences, 3435 Main Street, Buffalo, NY, 14260, USA.
| | - Aihua Bian
- Department of Biostatistics, Vanderbilt University Medical Center, 2525 West End Avenue, Suite 11000, Nashville, TN, 37203, USA.
| | - Pingsheng Wu
- Department of Biostatistics, Vanderbilt University Medical Center, 2525 West End Avenue, Suite 11000, Nashville, TN, 37203, USA.
| | - Talat Alp Ikizler
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, 1161 21st Avenue, S-3223 MCN, Nashville, TN, 37232, USA.
| |
Collapse
|
36
|
Van Laecke S, Van Biesen W. Hypomagnesaemia in kidney transplantation. Transplant Rev (Orlando) 2015; 29:154-60. [PMID: 26001746 DOI: 10.1016/j.trre.2015.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 05/03/2015] [Indexed: 01/14/2023]
Abstract
In the era of calcineurin inhibitors, hypomagnesaemia is a very common finding in kidney transplant recipients. Especially the first weeks after transplantation it is the rule rather than the exception. Hypomagnesaemia or low magnesium intake have been associated with a higher mortality or more cardiovascular events in the general population, but this association has never been explored in kidney transplant recipients, despite their increased cardiovascular risk. Kidney transplant recipients with pre- or post-transplant hypomagnesaemia seem to have an aberrant glucose metabolism and develop diabetes mellitus more frequently. Moreover, observations from alternate study populations, animal experiments or in vitro studies suggest a possible role of magnesium deficiency in graft dysfunction, bone metabolism and transplant immunology. Future observational and especially interventional studies should further define whether and to what extent we should make effort to correct this electrolyte disturbance in transplant recipients. Considering the mechanism of renal magnesium wasting, normalizing the serum magnesium concentration by oral supplementation alone might turn out to be cumbersome in kidney transplant recipients.
Collapse
Affiliation(s)
| | - Wim Van Biesen
- Renal Division, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
37
|
Benhamou Y, Bellien J, Armengol G, Brakenhielm E, Adriouch S, Iacob M, Remy-Jouet I, Le Cam-Duchez V, Monteil C, Renet S, Jouen F, Drouot L, Menard JF, Borg JY, Thuillez C, Boyer O, Levesque H, Richard V, Joannidès R. Role of Toll-like receptors 2 and 4 in mediating endothelial dysfunction and arterial remodeling in primary arterial antiphospholipid syndrome. Arthritis Rheumatol 2015; 66:3210-20. [PMID: 25047402 DOI: 10.1002/art.38785] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 07/10/2014] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To assess the role of Toll-like receptors (TLRs) in antiphospholipid antibody (aPL)-mediated vascular abnormalities in patients with primary arterial antiphospholipid syndrome (APS). METHODS Forty-eight subjects participated in the study. Arterial function and structure and TLR pathway activation were determined in patients with primary arterial APS and matched controls. The pathogenic effects of aPL isolated from patients were assessed in wild-type (WT) and TLR-knockout mice. RESULTS APS patients had endothelial dysfunction, arterial stiffening, and hypertrophy, as evidenced by decreased brachial artery endothelium-dependent flow-mediated dilation (FMD) and increased aortic pulse wave velocity and carotid intima-media thickness (IMT), as compared with controls. Plasma samples from APS patients revealed decreased nitric oxide (NO) availability and a pro-oxidative, proinflammatory, and prothrombotic state illustrated by a decrease in nitrite and an increase in lipid peroxidation, tumor necrosis factor α levels, and tissue factor (TF) levels. Furthermore, TLR pathway activation was found in APS patients with increased TLR-2 and TLR-4 messenger RNA expression and increased protein levels of the activated TLR transduction protein interleukin-1 receptor-associated kinase 1 in peripheral blood mononuclear cells. Moreover, agonist-stimulated cell-surface expression of TLR-2 and TLR-4 in circulating monocytes was higher in APS patients than in controls. These changes were positively associated with IMT and negatively associated with FMD. Finally, aPL injection decreased mesenteric endothelium-dependent relaxation and increased TF expression in WT mice but not in TLR-2- or TLR-4-knockout mice. CONCLUSION This translational study supports the notion that TLR-2 and TLR-4 play a role in mediating vascular abnormalities in patients with primary arterial APS. TLRs thus constitute a promising pharmacologic target for preventing cardiovascular complications in APS.
Collapse
Affiliation(s)
- Ygal Benhamou
- Rouen University Hospital, INSERM U1096, University of Rouen, and Centre d'Investigation Clinique, INSERM 1404, Rouen, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lee MC, Chen YC, Ho GJ, Shih MH, Chou KC, Hsu BG. Serum leptin levels positively correlate with peripheral arterial stiffness in kidney transplantation patients. Transplant Proc 2014; 46:353-8. [PMID: 24655962 DOI: 10.1016/j.transproceed.2013.11.145] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 10/06/2013] [Accepted: 11/27/2013] [Indexed: 12/31/2022]
Abstract
BACKGROUND Leptin is a protein predominantly produced by adipocytes that plays a pathophysiologic role in the pathogenesis of hypertension and cardiovascular diseases. The aim of this study was to evaluate the relationship between fasting serum leptin levels and peripheral arterial stiffness among kidney transplant (KT) patients. METHODS Fasting blood samples were obtained from 74 KT patients. Brachial-ankle pulse wave velocity (baPWV) was measured in the right or left brachial artery to the ankle segments using an automatic pulse wave analyzer (VaSera VS-1000). Plasma leptin levels were measured using a commercial enzyme-linked immunosorbent assay kit. In this study, left or right baPWV values of less than 14.0 m/s were used to define the high arterial stiffness group. RESULTS Forty KT patients (54.1%) were defined in high arterial stiffness group. Hypertension (P < .010), diabetes (P < .010), age (P = .010), KT duration (P = .013), triglyceride levels (P = .016), systolic blood pressure (P < .001), waist circumference (P = .031), and leptin level (P < .001) were higher, whereas serum high-density lipoprotein cholesterol level (P = .030) was lower in the high arterial stiffness group compared with the low arterial stiffness group. Multivariate logistic regression analysis showed that leptin (odds ratio, 1.033; 95% CI, 1.004-1.062; P = .023), KT duration (odds ratio, 1.023; 95% CI, 1.004-1.044; P = .020), and high-density lipoprotein cholesterol level (odds ratio, 0.925; 95% CI, 0.872-0.982; P = .010) were the independent predictors of peripheral arterial stiffness in KT patients. CONCLUSIONS Serum fasting leptin level was positively associated with peripheral arterial stiffness among KT patients.
Collapse
Affiliation(s)
- M-C Lee
- School of Medicine, Tzu Chi University, Hualien, Taiwan; Department of Surgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Y-C Chen
- Department of Surgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - G-J Ho
- Department of Surgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - M-H Shih
- Department of Nursing, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - K-C Chou
- Department of Nursing, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - B-G Hsu
- School of Medicine, Tzu Chi University, Hualien, Taiwan; Department of Nephrology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan.
| |
Collapse
|
39
|
|
40
|
Placental, Lipid, and Glucidic Effects of Mammalian Target of Rapamycin Inhibitors: Impact on Fetal Growth and Metabolic Disorders During Pregnancy After Solid Organ Transplantation. Transplant Proc 2014; 46:2254-8. [DOI: 10.1016/j.transproceed.2014.07.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
41
|
Holdaas H, Potena L, Saliba F. mTOR inhibitors and dyslipidemia in transplant recipients: a cause for concern? Transplant Rev (Orlando) 2014; 29:93-102. [PMID: 25227328 DOI: 10.1016/j.trre.2014.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 07/19/2014] [Accepted: 08/22/2014] [Indexed: 02/06/2023]
Abstract
Post-transplant dyslipidemia is exacerbated by mammalian target of rapamycin (mTOR) inhibitors. Early clinical trials of mTOR inhibitors used fixed dosing with no concomitant reduction in calcineurin inhibitor (CNI) exposure, leading to concerns when consistent and marked dyslipidemia was observed. With use of modern concentration-controlled mTOR inhibitor regimens within CNI-free or reduced-exposure CNI regimens, however, the dyslipidemic effect persists but is less pronounced. Typically, total cholesterol levels are at the upper end of normal, or indicate borderline risk, in kidney and liver transplant recipients, and are lower in heart transplant patients under near-universal statin therapy. Of note, it is possible that mTOR inhibitors may offer a cardioprotective effect. Experimental evidence for delayed progression of atherosclerosis is consistent with evidence from heart transplantation that coronary artery intimal thickening and the incidence of cardiac allograft vasculopathy are reduced with everolimus versus cyclosporine therapy. Preliminary data also indicate that mTOR inhibitors may improve arterial stiffness, a predictor of cardiovascular events, and may reduce ventricular remodeling and decrease left ventricular mass through an anti-fibrotic effect. Post-transplant dyslipidemia under mTOR inhibitor therapy should be monitored and managed closely, but unless unresponsive to therapy should not be regarded as a barrier to its use.
Collapse
Affiliation(s)
- Hallvard Holdaas
- Section of Nephrology, Department of Transplant Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
| | - Luciano Potena
- Heart Failure and Heart Transplant Program, Academic Hospital S. Orsola-Malpighi, Alma-Mater University of Bologna, Bologna, Italy
| | - Faouzi Saliba
- AP-HP Hôpital Paul Brousse, Centre Hépato-Biliaire, Villejuif, France
| |
Collapse
|
42
|
Ho GJ, Lee MC, Lee CJ, Chen YC, Hsu BG. Hypoadiponectinemia correlates with arterial stiffness in kidney transplantation patients. Clin Exp Nephrol 2014; 19:534-41. [PMID: 25037242 DOI: 10.1007/s10157-014-1010-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 07/03/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND Adiponectin is a fat-derived hormone produced and secreted exclusively by adipocytes that have anti-atherosclerotic effects. The aim of this study was to evaluate the relationship between fasting serum adiponectin levels and arterial stiffness among kidney transplant (KT) patients. METHODS Fasting blood samples were obtained from 69 KT patients. Brachial-ankle pulse wave velocity (baPWV) was measured in the right or left brachial artery to the ankle segments using an automatic pulse wave analyzer. Plasma adiponectin levels were measured using a commercial enzyme-linked immunosorbent assay kit. Left or right baPWV values of >14.0 m/s were used to define the high arterial stiffness group. RESULTS Thirty-five KT patients (35/69; 50.7 %) were defined in high arterial stiffness group. Diabetes (P = 0.013), smoking (P = 0.001), KT duration (P < 0.001), body weight (P = 0.013), waist circumference (P = 0.013), body mass index (P = 0.001), fasting glucose (P = 0.013), systolic blood pressure (P < 0.001), diastolic blood pressure (P = 0.008), and pulse pressure (P = 0.003) were higher, while serum adiponectin level (P = 0.004) was lower in high arterial stiffness group compared with low arterial stiffness group. Multivariate logistic regression analysis showed that adiponectin (odds ratio 0.90, 95 % confidence interval 0.81-0.99, P = 0.034) was still the independent predictors of arterial stiffness among the KT patients. CONCLUSION Serum fasting adiponectin level was inversely associated with arterial stiffness among KT patients.
Collapse
Affiliation(s)
- Guan-Jin Ho
- Department of Surgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Ming-Che Lee
- Department of Surgery, Buddhist Tzu Chi General Hospital, and School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chung-Jen Lee
- Department of Nursing, Tzu Chi College of Technology, Hualien, Taiwan
| | - Yen-Cheng Chen
- Department of Surgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Bang-Gee Hsu
- Division of Nephrology, Buddhist Tzu Chi General Hospital, and School of Medicine, Tzu Chi University, No. 707, Section 3, Chung-Yang Raod, Hualien, Taiwan.
| |
Collapse
|
43
|
Zaza G, Granata S, Tomei P, Masola V, Gambaro G, Lupo A. mTOR inhibitors and renal allograft: Yin and Yang. J Nephrol 2014; 27:495-506. [PMID: 24804854 DOI: 10.1007/s40620-014-0103-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 04/22/2014] [Indexed: 02/06/2023]
Abstract
Mammalian target of rapamycin inhibitors (mTOR-I), everolimus and sirolimus, are immunosuppressive drugs extensively used in renal transplantation. Their main mechanism of action is the inhibition of cell signaling through the PI3 K/Akt/mTOR pathway. This interesting mechanism of action confers to these medications both great immunosuppressive potential and important anti-neoplastic properties. Although the clinical utility of this drug category, as with other antineoplastic/immunosuppressants, is clear, the use of mTOR-I commonly results in the development of several complications. In particular, these agents may determine severe renal toxicity that, as recent studies report, seems clearly correlated to dose and duration of drug use. The mTOR-I-induced renal allograft spectrum of toxicity includes the enhanced incidence of delayed graft function, nephrotoxicity in particular when co-administered with calcineurin inhibitors (CNI) and onset of proteinuria. The latter effect appears highly frequent in patients undergoing mTOR-I treatment and significantly associated with a rapid graft lost. The damage leading to this complication interests both the glomerular and tubular area. mTOR-I cause an inhibition of proliferation in podocytes and the epithelial-to-mesenchymal transition in tubular cells. Interestingly, all these side effects are mostly reversible and dose related. Therefore, it is unquestionable that these particular drugs should be administered at the lowest dose able to maintain relatively low trough levels, in order to maximize their important and specific therapeutic effects while minimizing or avoiding drug toxicities. Utilization of low dosages of mTOR-I should be encouraged not only in CNI-combined schemas, but also when administered alone in a CNI-free immunosuppressive protocol.
Collapse
Affiliation(s)
- Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy,
| | | | | | | | | | | |
Collapse
|
44
|
Alpay N, Ozkok A, Caliskan Y, Akagun T, Cinar SA, Deniz G, Sariyar M, Yildiz A. Influence of conversion from calcineurin inhibitors to everolimus on fibrosis, inflammation, tubular damage and vascular function in renal transplant patients. Clin Exp Nephrol 2014; 18:961-7. [PMID: 24515306 DOI: 10.1007/s10157-014-0939-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 01/17/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND Conversion from calcineurin inhibitor (CNI) to mTOR inhibitors may reduce and even halt the progression of chronic allograft dysfunction (CAD) which is the most important cause of renal allograft loss. We aimed to investigate the effects of conversion from CNI to everolimus on parameters of fibrosis, inflammation, glomerulotubular damage and vascular functions in renal transplant recipients. METHODS Fifteen stable renal transplant recipients who were under CNI treatment (male/female 13/2, mean age 41 ± 10 years) were enrolled and switched to everolimus. Serum and urinary transforming growth factor-β (TGF-β), urinary neutrophil gelatinase-associated lipocalin (NGAL) and monocyte chemoattractant protein-1 (MCP-1) were measured as markers of fibrosis, tubular damage and inflammation. As parameters of vascular functions, pulse wave velocity (PWV), augmentation index (AIx), serum asymmetric dimethyl-arginine and fibroblast growth factor-23 (FGF-23) were measured. All these measurements were repeated at the 3rd month of conversion. RESULTS Estimated GFR (52 ± 7-57 ± 11 ml/min/l.73 m(2), p = 0.02) (was increased after conversion to everolimus. However, serum uric acid levels were significantly decreased (6.21 ± 1.21-5.50 ± 1.39 mg/dL, p = 0.01). Serum TGF-β levels (8727 ± 2897-1943 ± 365 pg/mL, p = 0.03) and urinary NGAL levels (26 ± 10-12 ± 2 ng/mg creatinine, p = 0.05) were significantly decreased. However, urinary MCP-1, FGF-23, PWV and AIx did not change. Urinary TGF-β was associated with urinary NGAL (r = 0.62, p = 0.01), urinary MCP-1 (r = 0.68, p = 0.005) and proteinuria (r = 0.50, p = 0.05). CONCLUSION Conversion from CNI to everolimus resulted in significant decreases of serum TGF-β and urinary NGAL which may represent less fibrosis and tubular damage. Association of urinary TGF-β with NGAL and MCP-1 suggests that tubular damage, fibrosis and inflammation may act together for progression of CAD.
Collapse
Affiliation(s)
- Nadir Alpay
- Division of Nephrology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul School of Medicine, Istanbul University, Fatih, 34093, Istanbul, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Benhamou Y, Bellien J, Armengol G, Gomez E, Richard V, Lévesque H, Joannidès R. [Assessment of endothelial function in autoimmune diseases]. Rev Med Interne 2014; 35:512-23. [PMID: 24412013 DOI: 10.1016/j.revmed.2013.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 11/07/2013] [Accepted: 12/02/2013] [Indexed: 11/15/2022]
Abstract
Numerous autoimmune-inflammatory rheumatic diseases have been associated with accelerated atherosclerosis or other types of vasculopathy leading to an increase in cardiovascular disease incidence. In addition to traditional cardiovascular risk factors, endothelial dysfunction is an important early event in the pathogenesis of atherosclerosis, contributing to plaque initiation and progression. Endothelial dysfunction is characterized by a shift of the actions of the endothelium toward reduced vasodilation, a proinflammatory and a proadhesive state, and prothrombic properties. Therefore, assessment of endothelial dysfunction targets this vascular phenotype using several biological markers as indicators of endothelial dysfunction. Measurements of soluble adhesion molecules (ICAM-1, VCAM-1, E-selectin), pro-thrombotic factors (thrombomodulin, von Willebrand factor, plasminogen activator inhibitor-1) and inflammatory cytokines are most often performed. Regarding the functional assessment of the endothelium, the flow-mediated dilatation of conduit arteries is a non-invasive method widely used in pathophysiological and interventional studies. In this review, we will briefly review the most relevant information upon endothelial dysfunction mechanisms and explorations. We will summarize the similarities and differences in the biological and functional assessments of the endothelium in different autoimmune diseases.
Collapse
Affiliation(s)
- Y Benhamou
- Département de médecine interne, CHU de Rouen, 1, rue de Germont, 76031 Rouen cedex, France; Service de pharmacologie, CHU de Rouen, 1, rue de Germont, 76031 Rouen cedex, France; Inserm U 1096, faculté de médecine de Rouen, 22, boulevard Gambetta, 76183 Rouen cedex, France.
| | - J Bellien
- Département de médecine interne, CHU de Rouen, 1, rue de Germont, 76031 Rouen cedex, France; Service de pharmacologie, CHU de Rouen, 1, rue de Germont, 76031 Rouen cedex, France
| | - G Armengol
- Département de médecine interne, CHU de Rouen, 1, rue de Germont, 76031 Rouen cedex, France
| | - E Gomez
- Inserm U 1096, faculté de médecine de Rouen, 22, boulevard Gambetta, 76183 Rouen cedex, France
| | - V Richard
- Inserm U 1096, faculté de médecine de Rouen, 22, boulevard Gambetta, 76183 Rouen cedex, France
| | - H Lévesque
- Département de médecine interne, CHU de Rouen, 1, rue de Germont, 76031 Rouen cedex, France; Inserm U 1096, faculté de médecine de Rouen, 22, boulevard Gambetta, 76183 Rouen cedex, France
| | - R Joannidès
- Service de pharmacologie, CHU de Rouen, 1, rue de Germont, 76031 Rouen cedex, France; Inserm U 1096, faculté de médecine de Rouen, 22, boulevard Gambetta, 76183 Rouen cedex, France
| |
Collapse
|
46
|
Gatault P, Lebranchu Y. Conversion to mTOR-inhibitor-based immunosuppression: which patients and when? Transplant Res 2013; 2:S3. [PMID: 24565231 PMCID: PMC3834549 DOI: 10.1186/2047-1440-2-s1-s3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) inhibitors are currently considered an alternative immunosuppressive treatment that can prevent the nephrotoxicity, viral infections and malignancies that are associated with calcineurin inhibitor-based immunosuppressive regimens. However, the side effects of mTOR-inhibitor-based regimens lead to frequent treatment discontinuations, and not all patients seem to have the same benefits from conversion to mTOR inhibitors. This review focuses on long-term results of trials that have assessed early and late conversion to sirolimus or everolimus. The renal benefit of late conversion (≥1 year post transplantation) is limited, except in patients with good renal function and without proteinuria. Early conversion to mTOR inhibitors in the first 6 months, in combination with mycophenolate mofetil, could be an appropriate strategy for maintenance therapy in renal transplant recipients with a low immunological risk after careful screening at the time of conversion. Good renal function (glomerular filtration rate >40 ml/ minute), weak proteinuria (<1 g/day), an absence of previous acute rejection and subclinical rejection, and appearance of donor-specific anti-human leukocyte antigen antibodies appear to be the most important criteria in identifying patients for whom conversion to an mTOR inhibitor may improve renal function at 5 years.
Collapse
|
47
|
Bellien J, Fréguin-Bouilland C, Joannidès R, Hanoy M, Rémy-Jouet I, Monteil C, Iacob M, Martin L, Renet S, Vendeville C, Godin M, Thuillez C, Le Roy F. High-efficiency on-line haemodiafiltration improves conduit artery endothelial function compared with high-flux haemodialysis in end-stage renal disease patients. Nephrol Dial Transplant 2013; 29:414-22. [PMID: 24235073 DOI: 10.1093/ndt/gft448] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Middle molecular weight uraemic toxins are considered to play an important role in vascular dysfunction and cardiovascular outcomes in end-stage renal disease (ESRD) patients. Recent dialysis techniques based on convection, specifically high-efficiency on-line haemodiafiltration (HDF), enhance the removal of middle molecular weight toxins and reduce all-cause mortality in haemodialysis (HD) patients. However, the mechanisms of these improved outcomes remain to be established. METHODS This prospective study randomly assigned 42 ESRD patients to switch from high-flux HD to high-efficiency on-line HDF (n=22) or to continue HD (n=20). Brachial artery endothelium-dependent flow-mediated dilatation, central pulse pressure, carotid artery intima-media thickness (IMT), internal diastolic diameter and distensibility and circulating markers of uraemia, inflammation and oxidative stress were blindly assessed before and after a 4-month follow-up. RESULTS Brachial flow-mediated dilatation and carotid artery distensibility increased significantly in the HDF group compared with HD, while carotid IMT and diameter remained similar. HDF decreased predialysis levels of the uraemic toxins β2-microglobulin, phosphate and blood TNFα mRNA expression. Oxidative stress markers were not different between the HD and HDF groups. Blood mRNA expression of protein kinase C β2, an endothelial NO-synthase (eNOS) inhibitor, decreased significantly with HDF. CONCLUSIONS High-efficiency on-line HDF prevents the endothelial dysfunction and stiffening of the conduit arteries in ESRD patients compared with high-flux HD. HDF decreases uraemic toxins, vascular inflammation, and is associated with subsequent improvement in eNOS functionality. These results suggest that reduced endothelial dysfunction may be an intermediate mechanism explaining the beneficial outcomes associated with HDF.
Collapse
Affiliation(s)
- Jérémy Bellien
- Department of Pharmacology, Rouen University Hospital, Rouen, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Systemic and nonrenal adverse effects occurring in renal transplant patients treated with mTOR inhibitors. Clin Dev Immunol 2013; 2013:403280. [PMID: 24151517 PMCID: PMC3789319 DOI: 10.1155/2013/403280] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 07/08/2013] [Accepted: 08/04/2013] [Indexed: 02/07/2023]
Abstract
The mammalian target of rapamycin inhibitors (mTOR-I), sirolimus and everolimus, are immunosuppressive drugs largely used in renal transplantation. The main mechanism of action of these drugs is the inhibition of the mammalian target of rapamycin (mTOR), a regulatory protein kinase involved in lymphocyte proliferation. Additionally, the inhibition of the crosstalk among mTORC1, mTORC2, and PI3K confers the antineoplastic activities of these drugs. Because of their specific pharmacological characteristics and their relative lack of nephrotoxicity, these inhibitors are valid option to calcineurine inhibitors (CNIs) for maintenance immunosuppression in renal transplant recipients with chronic allograft nephropathy. However, as other immunosuppressive drugs, mTOR-I may induce the development of several adverse effects that need to be early recognized and treated to avoid severe illness in renal transplant patients. In particular, mTOR-I may induce systemic nonnephrological side effects including pulmonary toxicity, hematological disorders, dysmetabolism, lymphedema, stomatitis, cutaneous adverse effects, and fertility/gonadic toxicity. Although most of the adverse effects are dose related, it is extremely important for clinicians to early recognize them in order to reduce dosage or discontinue mTOR-I treatment avoiding the onset and development of severe clinical complications.
Collapse
|
49
|
Mechanistic target of rapamycin inhibitors in solid organ transplantation: from benchside to clinical use. Curr Opin Organ Transplant 2013; 17:626-33. [PMID: 23080066 DOI: 10.1097/mot.0b013e32835a4be2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Here, we review recent advances and new insights in mechanistic target of rapamycin (mTOR) biology (signalling pathway, kidney biology and immune system), and recent clinical data on mTOR inhibitors related to solid organ transplantation. RECENT FINDINGS The mTOR pathway is a major integrator of signals governing protein and lipid biosynthesis and growth factor-driven cell cycle progression. Recent findings have emphasized a critical role of mTOR in cellular homeostasis with a crucial role in podocyte function. Beyond CD8(+) and regulatory T-cell control, mTOR protein is involved in critical biological functions of T helper cells or dendritic cells. New specific inhibitors of mTORC1/C2 are available and shed new light on mTOR functions. Finally, clinical trials have better defined the use of mTOR inhibitors and emphasized their role in cancer prevention. SUMMARY The mTOR pathway is considered as a key integrator of multiple inputs that drives numerous biological processes in cell biology. mTOR inhibitors are potent immunosuppressive drugs for solid organ transplantation. Newly designed specific inhibitors of mTOR complex 1 and 2 offer promising therapeutic effects and a better understanding of the pathway. Many conditions may benefit from mTOR inhibition for a short period, but tolerance of treatment in a chronic setting remains a major concern.
Collapse
|
50
|
Jankowska I, Czubkowski P, Socha P, Wierzbicka A, Teisseyre M, Teisseyre J, Pawłowska J. Lipid metabolism and oxidative stress in children after liver transplantation treated with sirolimus. Pediatr Transplant 2012; 16:901-6. [PMID: 23131059 DOI: 10.1111/petr.12007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lipid disturbances are one of the most frequent side effects of SRL; however, clinical consequences are not known. The aim of the study was to evaluate the risk of AS in children after LTx treated with SRL. In 17 children with median age 13.2 yr (1.9-17.9) who received SRL on average for 4.1 yr (s.d. ± 2.9) we measured and compared with age-matched healthy control group (n = 45) lipid parameters and markers of AS: ADMA, oxyLDL, GSH, GPx, TC, TG, HDL cholesterol, LDL cholesterol, VLDL cholesterol, ApoAI, ApoB, ApoE, lipoprotein (a) (Lp(a)). We found no major differences in cholesterol, cholesterol in lipoprotein fractions and TG concentrations between patients receiving SRL and the control group. ApoE was markedly increased in the study group (19.1 g/L [±1.8]) when compared to controls (9.8 [±3.9]). ApoA1 was decreased in the study group: 1.30 g/L (±0.2) vs. 1.45 (±0.25), p = 0.04. ApoB and Lpa concentrations were similar in both groups. There were differences in oxidative stress markers: GSH 743 (±66.2) mol/mL vs. 780 (±48.2), p = 0.02 and GPx 32.8 (±5.5) U/gHb vs. 34.3(±2.6), p = 0.01. Markers of AS: ADMA did not differ between groups and oxidized LDLc was significantly lower in SRL group: 190 mU/mL (±113) vs. 237 (±107) in control, p < 0.05. SRL does not significantly disturb lipid metabolism and oxidative status in children after LTx.
Collapse
Affiliation(s)
- Irena Jankowska
- Department of Gastroenterology, Hepatology and Eating Disorders, The Children's Memorial Health Institute, Warsaw, Poland
| | | | | | | | | | | | | |
Collapse
|