1
|
Chienwichai K, Phirom S, Wuttiputhanun T, Leelahavanichkul A, Townamchai N, Avihingsanon Y, Udomkarnjananun S. A systematic review and meta-analysis of factors contributing to post-kidney transplant anemia and the effect of erythropoietin-stimulating agents. Syst Rev 2024; 13:278. [PMID: 39533400 PMCID: PMC11556001 DOI: 10.1186/s13643-024-02709-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The effects of various risk and associated factors on post-kidney transplant anemia (PTA) have not been fully compared and estimated. This meta-analysis aims to elucidate factors contributing to PTA and determine the influence of erythropoietin-stimulating agents (ESAs) on renal outcomes, thus offering potential pathways for enhanced management strategies post-transplant. METHODS A systematic review was conducted in electronical database. Studies reporting on risk factors (with cause-effect relationships) and associated factors (without definite cause-effect relationships) of PTA, and the effects of ESAs on post-kidney transplant outcomes, were included. Pooled odds ratios (ORs) and weighted mean differences (WMDs) were analyzed using random-effects models. RESULTS This systematic review encompassed 38,233 patients from 85 studies. Factors increased PTA risk included African American, older donor age, human antigen leukocyte mismatches, and low pre-transplant hemoglobin levels. Poor allograft function, high interleukine-6, Cytomegalovirus, delayed graft function, allograft rejections, immunosuppressive medications, and renin-angiotensin system blockades were associated with PTA. Native autosomal dominant polycystic kidney disease was a protective factor against PTA. Administration of ESAs with the aim of normalizing hemoglobin levels in patients with chronic allograft dysfunction slowed the decline in eGFR and reduce the risk of death, with a pooled OR of 0.36 (95% CI: 0.14 to 0.89; p = 0.040). CONCLUSIONS The risks and associated factors for PTA have been elucidated, underscoring the need for individualized treatment approaches. Late ESA therapy, aimed at hemoglobin normalization, suggests a renal-protective effect and reduced mortality, which should be considered in the management of PTA. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42024545330.
Collapse
Affiliation(s)
| | - Supitchaya Phirom
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Thunyatorn Wuttiputhanun
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Excellence Center for Organ Transplantation (ECOT), King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Immunology Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Center of Excellence On Translational Research in Inflammation and Immunology (CETRII), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Natavudh Townamchai
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Excellence Center for Organ Transplantation (ECOT), King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Renal Immunology and Renal Transplantation, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yingyos Avihingsanon
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Excellence Center for Organ Transplantation (ECOT), King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Renal Immunology and Renal Transplantation, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Suwasin Udomkarnjananun
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand.
- Excellence Center for Organ Transplantation (ECOT), King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand.
- Department of Microbiology, Immunology Unit, Chulalongkorn University, Bangkok, Thailand.
- Department of Microbiology, Center of Excellence On Translational Research in Inflammation and Immunology (CETRII), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Center of Excellence in Renal Immunology and Renal Transplantation, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
2
|
Chung EY, Palmer SC, Saglimbene VM, Craig JC, Tonelli M, Strippoli GF. Erythropoiesis-stimulating agents for anaemia in adults with chronic kidney disease: a network meta-analysis. Cochrane Database Syst Rev 2023; 2:CD010590. [PMID: 36791280 PMCID: PMC9924302 DOI: 10.1002/14651858.cd010590.pub3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
BACKGROUND Erythropoiesis-stimulating agents (ESAs) are commonly used to treat anaemia in people with chronic kidney disease (CKD). However, their use has been associated with cardiovascular events. This is an update of a Cochrane review first published in 2014. OBJECTIVES To compare the efficacy and safety of ESAs (epoetin alfa, epoetin beta, darbepoetin alfa, methoxy polyethylene glycol-epoetin beta, and biosimilar ESAs against each other, placebo, or no treatment) to treat anaemia in adults with CKD. SEARCH METHODS In this update, we searched the Cochrane Kidney and Transplant Register of Studies up to 29 April 2022 through contact with the Information Specialist using search terms relevant to this review. Studies in the Register are identified through searches of CENTRAL, MEDLINE, and EMBASE, conference proceedings, the International Clinical Trials Register (ICTRP) Search Portal and ClinicalTrials.gov. SELECTION CRITERIA Randomised controlled trials (RCTs) that included a comparison of an ESA (epoetin alfa, epoetin beta, darbepoetin alfa, methoxy polyethylene glycol-epoetin beta, a biosimilar epoetin or a biosimilar darbepoetin alfa) with another ESA, placebo or no treatment in adults with CKD were considered for inclusion. DATA COLLECTION AND ANALYSIS Two independent authors screened the search results and extracted data. Data synthesis was performed using random-effects pairwise meta-analysis (expressed as odds ratios (OR) and their 95% confidence intervals (CI)) and network meta-analysis. We assessed for heterogeneity and inconsistency within meta-analyses using standard techniques and planned subgroup and meta-regression to explore sources of heterogeneity or inconsistency. We assessed certainty in treatment estimates for the primary outcomes (preventing blood transfusions and death (any cause)) using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. MAIN RESULTS Sixty-two new studies (9237 participants) were included in this update, so the review now includes 117 studies with 25,237 participants. Most studies were at high or unclear risk of bias in most methodological domains. Overall, results remain similar in this update compared to our previous review in 2014. For preventing blood transfusion, epoetin alfa (OR 0.28, 95% CI 0.13 to 0.61; low certainty evidence) and epoetin beta (OR 0.19, 95% CI 0.08 to 0.47; low certainty evidence) may be superior to placebo, and darbepoetin alfa was probably superior to placebo (OR 0.27, 95% CI 0.11 to 0.67; moderate certainty evidence). Methoxy polyethylene glycol-epoetin beta (OR 0.33, 95% CI 0.11 to 1.02; very low certainty evidence), a biosimilar epoetin (OR 0.34, 95% CI 0.11 to 1.03; very low certainty evidence) and a biosimilar darbepoetin alfa (OR 0.37, 95% CI 0.07 to 1.91; very low certainty evidence) had uncertain effects on preventing blood transfusion compared to placebo. The comparative effects of ESAs compared with another ESA on preventing blood transfusions were uncertain, in low to very low certainty evidence. Effects on death (any cause) were uncertain for epoetin alfa (OR 0.79, 95% CI 0.51 to 1.22; low certainty evidence), epoetin beta (OR 0.69, 95% CI 0.40 to 1.20; low certainty evidence), methoxy polyethylene glycol-epoetin beta (OR 1.07, 95% CI 0.67 to 1.71; very low certainty evidence), a biosimilar epoetin (OR 0.80, 95% CI 0.47 to 1.36; low certainty evidence) and a biosimilar darbepoetin alfa (OR 1.63, 95% CI 0.51 to 5.23; very low certainty evidence) compared to placebo. There was probably no difference between darbepoetin alfa and placebo on the odds of death (any cause) (OR 0.99, 95% CI 0.81 to 1.21; moderate certainty evidence). The comparative effects of ESAs compared with another ESA on death (any cause) were uncertain in low to very low certainty evidence. Epoetin beta probably increased the odds of hypertension when compared to placebo (OR 2.17, 95% CI 1.17 to 4.00; moderate certainty evidence). Compared to placebo, epoetin alfa (OR 2.10, 95% CI 1.22 to 3.59; very low certainty evidence), darbepoetin alfa (OR 1.88, 95% CI 1.12 to 3.14; low certainty evidence) and methoxy polyethylene glycol-epoetin beta (OR 1.98, 95% CI 1.05 to 3.74; low certainty evidence) may increase the odds of hypertension, but a biosimilar epoetin (OR 1.88, 95% CI 0.96 to 3.67; low certainty evidence) and biosimilar darbepoetin alfa (OR 1.98, 95% CI 0.84 to 4.66; low certainty evidence) had uncertain effects on hypertension. The comparative effects of all ESAs compared with another ESA, placebo or no treatment on cardiovascular death, myocardial infarction, stroke, vascular access thrombosis, kidney failure, and breathlessness were uncertain. Network analysis for fatigue was not possible due to sparse data. AUTHORS' CONCLUSIONS: The comparative effects of different ESAs on blood transfusions, death (any cause and cardiovascular), major cardiovascular events, myocardial infarction, stroke, vascular access thrombosis, kidney failure, fatigue and breathlessness were uncertain.
Collapse
Affiliation(s)
- Edmund Ym Chung
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| | - Suetonia C Palmer
- Department of Medicine, University of Otago Christchurch, Christchurch, New Zealand
| | - Valeria M Saglimbene
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Jonathan C Craig
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Marcello Tonelli
- Department of Medicine, University of Calgary, Calgary, Canada
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Giovanni Fm Strippoli
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
- Cochrane Kidney and Transplant, Centre for Kidney Research, The Children's Hospital at Westmead, Westmead, Australia
| |
Collapse
|
3
|
Alshamsi I. Extended Literature Review of the role of erythropoietin stimulating agents (ESA) use in the management of post renal transplant anaemia. TRANSPLANTATION REPORTS 2022. [DOI: 10.1016/j.tpr.2022.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
4
|
Park JH, Koo BN, Kim MS, Shin D, Kwak YL. Effects of intraoperative dexmedetomidine infusion on renal function in elective living donor kidney transplantation: a randomized controlled trial. Can J Anaesth 2021; 69:448-459. [PMID: 34931289 DOI: 10.1007/s12630-021-02173-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/16/2021] [Accepted: 10/27/2021] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Ischemia-reperfusion injury is inevitable during donor organ harvest and recipient allograft reperfusion in kidney transplantation, and affects graft outcomes. Dexmedetomidine, an α2-adrenoreceptor agonist, has renoprotective effects against ischemia-reperfusion injury. We investigated the effects of intraoperative dexmedetomidine infusion on renal function and the development of delayed graft function after elective living donor kidney transplantation in a randomized controlled trial. METHODS A total of 104 patients were randomly assigned to receive either an intraoperative infusion of dexmedetomidine 0.4 μg·kg-1·hr-1 or 0.9% saline. The primary outcome was the serum creatinine level on postoperative day (POD) 7. Secondary outcomes were renal function and the degree of inflammation and included the following variables: serum creatinine level and estimated glomerular filtration rate up to six months; incidence of delayed graft function; and levels of serum cystatin C, plasma interleukin (IL)-1β, and IL-18 during the perioperative period. RESULTS The mean (standard deviation) serum creatinine level on POD 7 was comparable between the groups (dexmedetomidine vs control: 1.11 [0.87] mg·dL-1 vs 1.06 [0.73] mg·dL-1; mean difference, 0.05; 95% confidence interval, -0.27 to 0.36; P = 0.77). Delayed graft function occurred in one patient in each group (odds ratio, 1.020; P > 0.99). There were no significant differences in the secondary outcomes between the groups (all P > 0.05). CONCLUSIONS Intraoperative dexmedetomidine infusion did not produce any beneficial effects on renal function or delayed graft function in patients undergoing elective living donor kidney transplantation. STUDY REGISTRATION www.ClinicalTrials.gov (NCT03327389); registered 31 October 2017.
Collapse
Affiliation(s)
- Jin Ha Park
- Department of Anesthesiology and Pain Medicine, and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Bon-Nyeo Koo
- Department of Anesthesiology and Pain Medicine, and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Min-Soo Kim
- Department of Anesthesiology and Pain Medicine, and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Dongkwan Shin
- Department of Anesthesiology and Pain Medicine, and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Young-Lan Kwak
- Department of Anesthesiology and Pain Medicine, and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea. .,Yonsei Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
5
|
Wu Y, Yang B. Erythropoietin Receptor/β Common Receptor: A Shining Light on Acute Kidney Injury Induced by Ischemia-Reperfusion. Front Immunol 2021; 12:697796. [PMID: 34276689 PMCID: PMC8278521 DOI: 10.3389/fimmu.2021.697796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/17/2021] [Indexed: 12/29/2022] Open
Abstract
Acute kidney injury (AKI) is a health problem worldwide, but there is a lack of early diagnostic biomarkers and target-specific treatments. Ischemia-reperfusion (IR), a major cause of AKI, not only induces kidney injury, but also stimulates the self-defense system including innate immune responses to limit injury. One of these responses is the production of erythropoietin (EPO) by adjacent normal tissue, which is simultaneously triggered, but behind the action of its receptors, either by the homodimer EPO receptor (EPOR)2 mainly involved in erythropoiesis or the heterodimer EPOR/β common receptor (EPOR/βcR) which has a broad range of biological protections. EPOR/βcR is expressed in several cell types including tubular epithelial cells at low levels or absent in normal kidneys, but is swiftly upregulated by hypoxia and inflammation and also translocated to cellular membrane post IR. EPOR/βcR mediates anti-apoptosis, anti-inflammation, pro-regeneration, and remodeling via the PI3K/Akt, STAT3, and MAPK signaling pathways in AKI. However, the precise roles of EPOR/βcR in the pathogenesis and progression of AKI have not been well defined, and its potential as an earlier biomarker for AKI diagnosis and monitoring repair or chronic progression requires further investigation. Here, we review biological functions and mechanistic signaling pathways of EPOR/βcR in AKI, and discuss its potential clinical applications as a biomarker for effective diagnosis and predicting prognosis, as well as directing cell target drug delivery.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Basic Medical Research Centre, Medical School, Nantong University, Nantong, China.,Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China
| | - Bin Yang
- Nantong-Leicester Joint Institute of Kidney Science, Nephrology, Affiliated Hospital of Nantong University, Nantong, China.,Department of Cardiovascular Sciences, College of Life Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
6
|
Qiu L, Lai X, Wang JJ, Yeap XY, Han S, Zheng F, Lin C, Zhang Z, Procissi D, Fang D, Li L, Thorp EB, Abecassis MM, Kanwar YS, Zhang ZJ. Kidney-intrinsic factors determine the severity of ischemia/reperfusion injury in a mouse model of delayed graft function. Kidney Int 2020; 98:1489-1501. [PMID: 32822703 PMCID: PMC7814505 DOI: 10.1016/j.kint.2020.07.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 06/12/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023]
Abstract
Delayed graft function due to transplant ischemia/reperfusion injury adversely affects up to 50% of deceased-donor kidney transplant recipients. However, key factors contributing to the severity of ischemia/reperfusion injury remain unclear. Here, using a clinically relevant mouse model of delayed graft function, we demonstrated that donor genetic background and kidney-intrinsic MyD88/Trif-dependent innate immunity were key determinants of delayed graft function. Functional deterioration of kidney grafts directly corresponded with the duration of cold ischemia time. The graft dysfunction became irreversible after cold ischemia time exceeded six hours. When cold ischemia time reached four hours, kidney grafts displayed histological features reflective of delayed graft function seen in clinical kidney transplantation. Notably, kidneys of B6 mice exhibited significantly more severe histological and functional impairment than kidneys of C3H or BALB/c mice, regardless of recipient strains or alloreactivities. Furthermore, allografts of B6 mice also showed an upregulation of IL-6, neutrophil gelatinase-associated lipocalin, and endoplasmic reticulum stress genes, as well as an increased influx of host neutrophils and memory CD8 T-cells. In contrast, donor MyD88/Trif deficiency inhibited neutrophil influx and decreased the expression of IL-6 and endoplasmic reticulum stress genes, along with improved graft function and prolonged allograft survival. Thus, kidney-intrinsic factors involving genetic characteristics and innate immunity serve as critical determinants of the severity of delayed graft function. This preclinical murine model allows for further investigations of the mechanisms underlying delayed graft function.
Collapse
Affiliation(s)
- Longhui Qiu
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Xingqiang Lai
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Organ Transplant Center, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiao-Jing Wang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Xin Yi Yeap
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Shulin Han
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Feibo Zheng
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Charlie Lin
- Weinberg Art and Science College, Northwestern University, Evanston, Illinois, USA
| | - Zhuoli Zhang
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Daniele Procissi
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lin Li
- Department of Pathology and Laboratory Medicine, Cedars Sinai Medical Center, West Hollywood, California, USA
| | - Edward B Thorp
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Michael M Abecassis
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yashpal S Kanwar
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Medicine (Nephrology and Hypertension), Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Zheng J Zhang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
7
|
Prevention of Ischemia-Reperfusion Injury in Human Kidney Transplantation: A Meta-Analysis of Randomized Controlled Trials. Nephrourol Mon 2020. [DOI: 10.5812/numonthly.101590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
8
|
Skrifvars MB, Moore E, Mårtensson J, Bailey M, French C, Presneill J, Nichol A, Little L, Duranteau J, Huet O, Haddad S, Arabi Y, McArthur C, Cooper DJ, Bellomo R. Erythropoietin in traumatic brain injury associated acute kidney injury: A randomized controlled trial. Acta Anaesthesiol Scand 2019; 63:200-207. [PMID: 30132785 DOI: 10.1111/aas.13244] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/29/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Acute kidney injury (AKI) in traumatic brain injury (TBI) is poorly understood and it is unknown if it can be attenuated using erythropoietin (EPO). METHODS Pre-planned analysis of patients included in the EPO-TBI (ClinicalTrials.gov NCT00987454) trial who were randomized to weekly EPO (40 000 units) or placebo (0.9% sodium chloride) subcutaneously up to three doses or until intensive care unit (ICU) discharge. Creatinine levels and urinary output (up to 7 days) were categorized according to the Kidney Disease Improving Global Outcome (KDIGO) classification. Severity of TBI was categorized with the International Mission for Prognosis and Analysis of Clinical Trials in TBI. RESULTS Of 3348 screened patients, 606 were randomized and 603 were analyzed. Of these, 82 (14%) patients developed AKI according to KDIGO (60 [10%] with KDIGO 1, 11 [2%] patients with KDIGO 2, and 11 [2%] patients with KDIGO 3). Male gender (hazard ratio [HR] 4.0 95% confidence interval [CI] 1.4-11.2, P = 0.008) and severity of TBI (HR 1.3 95% CI 1.1-1.4, P < 0.001 for each 10% increase in risk of poor 6 month outcome) predicted time to AKI. KDIGO stage 1 (HR 8.8 95% CI 4.5-17, P < 0.001), KDIGO stage 2 (HR 13.2 95% CI 3.9-45.2, P < 0.001) and KDIGO stage 3 (HR 11.7 95% CI 3.5-39.7, P < 0.005) predicted time to mortality. EPO did not influence time to AKI (HR 1.08 95% CI 0.7-1.67, P = 0.73) or creatinine levels during ICU stay (P = 0.09). CONCLUSIONS Acute kidney injury is more common in male patients and those with severe compared to moderate TBI and appears associated with worse outcome. EPO does not prevent AKI after TBI.
Collapse
Affiliation(s)
- Markus B. Skrifvars
- Australian and New Zealand Intensive Care Research Centre; School of Public Health and Preventive Medicine; Monash University; Melbourne Victoria Australia
- Division of Intensive Care; Department of Anaesthesiology, Intensive Care and Pain Medicine; Helsinki University Hospital and University of Helsinki; Helsinki Finland
- Department of Emergency Medicine and Services; Helsinki University Hospital and University of Helsinki; Helsinki Finland
| | - Elizabeth Moore
- Australian and New Zealand Intensive Care Research Centre; School of Public Health and Preventive Medicine; Monash University; Melbourne Victoria Australia
| | - Johan Mårtensson
- Department of Physiology and Pharmacology; Section of Anaesthesia and Intensive Care; Karolinska Institutet; Stockholm Sweden
| | - Michael Bailey
- Australian and New Zealand Intensive Care Research Centre; School of Public Health and Preventive Medicine; Monash University; Melbourne Victoria Australia
| | - Craig French
- Department of Intensive Care; Western Health; Melbourne Victoria Australia
| | - Jeffrey Presneill
- Department of Intensive Care; Royal Melbourne Hospital; Melbourne Victoria Australia
| | - Alistair Nichol
- Australian and New Zealand Intensive Care Research Centre; School of Public Health and Preventive Medicine; Monash University; Melbourne Victoria Australia
- School of Medicine and Medical Sciences; University College Dublin; Dublin Ireland
- St Vincent's University Hospital; Dublin Ireland
- Department of Intensive Care and Hyperbaric Medicine; The Alfred; Melbourne Victoria Australia
| | - Lorraine Little
- Australian and New Zealand Intensive Care Research Centre; School of Public Health and Preventive Medicine; Monash University; Melbourne Victoria Australia
| | - Jacques Duranteau
- Department of Anaesthesia and Intensive Care; Hôpitaux universitaires Paris Sud (HUPS); Université Paris Sud XI; Orsay France
| | - Olivier Huet
- Departement d'anesthésie-réanimation; Hopital de la Cavale Blanche; Boulevard Tanguy Prigent; CHRU de Brest; Univeristé de Bretagne Occidental; Brest France
| | - Samir Haddad
- King Saud Bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center; Riyadh Saudi Arabia
- G&S Medical Associates; Urgent Care; Paterson New Jersey
| | - Yaseen Arabi
- King Saud Bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center; Riyadh Saudi Arabia
| | - Colin McArthur
- Department of Critical Care Medicine; Auckland City Hospital; Auckland New Zealand
| | - David J. Cooper
- Australian and New Zealand Intensive Care Research Centre; School of Public Health and Preventive Medicine; Monash University; Melbourne Victoria Australia
- Department of Intensive Care and Hyperbaric Medicine; The Alfred; Melbourne Victoria Australia
| | - Rinaldo Bellomo
- Australian and New Zealand Intensive Care Research Centre; School of Public Health and Preventive Medicine; Monash University; Melbourne Victoria Australia
- Department of Intensive Care; Austin Health; Melbourne Victoria Australia
| | | |
Collapse
|
9
|
Elliott S, Tomita D, Endre Z. Erythropoiesis stimulating agents and reno-protection: a meta-analysis. BMC Nephrol 2017; 18:14. [PMID: 28077085 PMCID: PMC5225567 DOI: 10.1186/s12882-017-0438-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 01/04/2017] [Indexed: 02/08/2023] Open
Abstract
Background Erythropoiesis stimulating agents (ESAs) were proposed to enhance survival of renal tissues through direct effects via activation of EPO receptors on renal cells resulting in reduced cell apoptosis, or indirect effects via increased oxygen delivery due to increased numbers of Hb containing red blood cells. Thus through several mechanisms there may be benefit of ESA administration on kidney disease progression and kidney function in renal patients. However conflicting ESA reno-protection outcomes have been reported in both pre-clinical animal studies and human clinical trials. To better understand the potential beneficial effects of ESAs on renal-patients, meta-analyses of clinical trials is needed. Methods Literature searches and manual searches of references lists from published studies were performed. Controlled trials that included ESA treatment on renal patients with relevant renal endpoints were selected. Results Thirty two ESA controlled trials in 3 categories of intervention were identified. These included 7 trials with patients who had a high likelihood of AKI, 7 trials with kidney transplant patients and 18 anemia correction trials with chronic kidney disease (predialysis) patients. There was a trend toward improvement in renal outcomes in the ESA treated arm of AKI and transplant trials, but none reached statistical significance. In 12 of the anemia correction trials, meta-analyses showed no difference in renal outcomes with the anemia correction but both arms received some ESA treatment making it difficult to assess effects of ESA treatment alone. However, in 6 trials the low Hb arm received no ESAs and meta-analysis also showed no difference in renal outcomes, consistent with no benefit of ESA/ Hb increase. Conclusions Most ESA trials were small with modest event rates. While trends tended to favor the ESA treatment arm, these meta-analyses showed no reduction of incidence of AKI, no reduction in DGF or improvement in 1-year graft survival after renal transplantation and no significant delay in progression of CKD. These results do not support significant clinical reno-protection by ESAs.
Collapse
Affiliation(s)
- Steve Elliott
- Amgen Inc, One Amgen Center, Newbury Park, Thousand Oaks, CA, 91320, USA.
| | - Dianne Tomita
- Amgen Inc, One Amgen Center, Newbury Park, Thousand Oaks, CA, 91320, USA
| | - Zoltan Endre
- Department of Nephrology, Prince of Wales Hospital and Clinical School, University of New South Wales, Sydney, NSW, 2031, Australia
| |
Collapse
|
10
|
Kapitsinou PP, Haase VH. Molecular mechanisms of ischemic preconditioning in the kidney. Am J Physiol Renal Physiol 2015; 309:F821-34. [DOI: 10.1152/ajprenal.00224.2015] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/21/2015] [Indexed: 12/26/2022] Open
Abstract
More effective therapeutic strategies for the prevention and treatment of acute kidney injury (AKI) are needed to improve the high morbidity and mortality associated with this frequently encountered clinical condition. Ischemic and/or hypoxic preconditioning attenuates susceptibility to ischemic injury, which results from both oxygen and nutrient deprivation and accounts for most cases of AKI. While multiple signaling pathways have been implicated in renoprotection, this review will focus on oxygen-regulated cellular and molecular responses that enhance the kidney's tolerance to ischemia and promote renal repair. Central mediators of cellular adaptation to hypoxia are hypoxia-inducible factors (HIFs). HIFs play a crucial role in ischemic/hypoxic preconditioning through the reprogramming of cellular energy metabolism, and by coordinating adenosine and nitric oxide signaling with antiapoptotic, oxidative stress, and immune responses. The therapeutic potential of HIF activation for the treatment and prevention of ischemic injuries will be critically examined in this review.
Collapse
Affiliation(s)
- Pinelopi P. Kapitsinou
- Departments of Medicine, Anatomy and Cell Biology, and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Volker H. Haase
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and
- Medicine and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
11
|
Strategies to optimize kidney recovery and preservation in transplantation: specific aspects in pediatric transplantation. Pediatr Nephrol 2015; 30:1243-54. [PMID: 25185880 DOI: 10.1007/s00467-014-2924-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 07/09/2014] [Accepted: 07/21/2014] [Indexed: 01/08/2023]
Abstract
In renal transplantation, live donor kidney grafts are associated with optimum success rates due to the shorter period of ischemia during the surgical procedure. The current shortage of donor organs for adult patients has caused a shift towards deceased donors, often with co-morbidity factors, whose organs are more sensitive to ischemia-reperfusion injury, which is unavoidable during transplantation. Donor management is pivotal to kidney graft survival through the control of the ischemia-reperfusion sequence, which is known to stimulate numerous deleterious or regenerative pathways. Although the key role of endothelial cells has been established, the complexity of the injury, associated with stimulation of different cell signaling pathways, such as unfolded protein response and cell death, prevents the definition of a unique therapeutic target. Preclinical transplant models in large animals are necessary to establish relationships and kinetics and have already contributed to the improvement of organ preservation. Therapeutic strategies using mesenchymal stem cells to induce allograft tolerance are promising advances in the treatment of the pediatric recipient in terms of reducing/withdrawing immunosuppressive therapy. In this review we focus on the different donor management strategies in kidney graft conditioning and on graft preservation consequences by highlighting the role of endothelial cells. We also propose strategies for preventing ischemia-reperfusion, such as cell therapy.
Collapse
|
12
|
Cyclic Helix B Peptide in Preservation Solution and Autologous Blood Perfusate Ameliorates Ischemia-Reperfusion Injury in Isolated Porcine Kidneys. Transplant Direct 2015; 1:e6. [PMID: 27500213 DOI: 10.1097/txd.0000000000000515] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 02/04/2015] [Indexed: 01/20/2023] Open
Abstract
UNLABELLED There is a critical need to better preserve isolated organs before transplantation. We developed a novel nonerythropoiesis cyclic helix B peptide (CHBP) derived from erythropoietin, which has potent tissue protection and prolonged serum stability. The renoprotection and potential mechanism of CHBP were evaluated in a kidney preservation model. MATERIALS AND METHODS Porcine kidneys (n = 5) subjected to 20-minute warm ischemia were retrieved and flushed with hyperosmolar citrate to mimic deceased donation. The kidneys and autologous blood ± 10.56 nmol/L CHBP were placed in cold storage (CS) for 18 hours. These kidneys were then normothermically hemoreperfused for 3 hours using an isolated organ perfusion system. The renal function and structure, apoptosis, inflammation, and expression of caspase-3 and heat shock protein 70 (HSP70) were assessed. RESULTS Cyclic helix B peptide significantly increased the renal blood flow, oxygen consumption, and urine output during reperfusion, but decreased serum potassium and renal tissue damage. Apoptotic cells were significantly decreased in the tubular areas, but increased in the lumens and interstitial areas in the post-CS and postreperfused kidneys, whereas myeloperoxidase+ cells were reduced. In addition, the expression of both caspase-3 precursor and active subunits was downregulated by CHBP in reperfused kidneys. However, HSP70 was upregulated in the post-CS and postreperfused kidneys treated with CHBP. CONCLUSIONS Cyclic helix B peptide administered into preservation and reperfusion solutions ameliorated renal ischemia-reperfusion injury, which might be associated with decreased apoptosis, inflammation and caspase-3, but increased HSP70. This novel preservation approach using CHBP may be applied in a porcine kidney transplant model and potential human donor kidney preservation.
Collapse
|
13
|
Coupes B, de Freitas DG, Roberts SA, Read I, Riad H, Brenchley PEC, Picton ML. rhErythropoietin-b as a tissue protective agent in kidney transplantation: a pilot randomized controlled trial. BMC Res Notes 2015; 8:21. [PMID: 25643790 PMCID: PMC4330593 DOI: 10.1186/s13104-014-0964-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 12/30/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Extended criteria donor (ECD) and donation after circulatory death (DCD) kidneys are at increased risk of delayed graft function (DGF). Experimental evidence suggests that erythropoietin (EPO) attenuates renal damage in acute kidney injury. This study piloted the administration of high dose recombinant human EPO-beta at implantation of ECD and DCD kidneys, and evaluated biomarkers of kidney injury post-transplant. METHODS Forty patients were randomly assigned to receive either rhEPO-b (100,000 iu) (n = 19 in the intervention group, as 1 patient was un-transplantable post randomisation), or placebo (n = 20) in this, double blind, placebo-controlled trial at Manchester Royal Infirmary from August 2007 to June 2009. Participants received either an ECD (n = 17) or DCD (n = 22) kidney. Adverse events, renal function, haematopoietic markers, and rejections were recorded out to 90 days post-transplant. Biomarkers of kidney injury (neutrophil gelatinase-associated lipocalin, Kidney Injury Molecule-1 and IL-18) were measured in blood and urine during the first post-operative week. RESULTS The incidence of DGF (53% vs 55%) (RR = 1.0; CI = 0.5-1.6; p = 0.93) and slow graft function (SGF) (32% vs 25%) (RR = 1.1; CI = 0.5-1.9; p = 0.73) respectively, serum creatinine, eGFR, haemoglobin and haematocrit, blood pressure, and acute rejection were similar in the 2 study arms. High dose rhEPO-b had little effect on the temporal profiles of the biomarkers. CONCLUSIONS High dose rhEPO-b appears to be safe and well tolerated in the early post- transplant period in this study, but has little effect on delayed or slow graft function in recipients of kidneys from DCD and ECD donors. Comparing the profiles of biomarkers of kidney injury (NGAL, IL-18 and KIM-1) showed little difference between the rhEPO-b treated and placebo groups. A meta-analysis of five trials yielded an overall estimate of the RR for DGF of 0.89 (CI = 0.73; 1.07), a modest effect favouring EPO but not a significant difference. A definitive trial based on this estimate would require 1000-2500 patients per arm for populations with base DGF rates of 50-30% and 90% power. Such a trial is clearly unfeasible. TRIAL REGISTRATION EudraCT Number 2006-005373-22 ISRCTN ISRCTN85447324 registered 19/08/09.
Collapse
Affiliation(s)
- Beatrice Coupes
- Department of Renal Medicine, Manchester Royal Infirmary, Oxford Rd, Manchester, M13 9WL, UK.
| | - Declan G de Freitas
- Department of Renal Medicine, Manchester Royal Infirmary, Oxford Rd, Manchester, M13 9WL, UK.
| | - Stephen A Roberts
- Centre for Biostatistics, University of Manchester, Manchester, M13 9PL, UK.
| | - Ian Read
- Department of Renal Medicine, Manchester Royal Infirmary, Oxford Rd, Manchester, M13 9WL, UK.
| | - Hany Riad
- Department of Surgery, Manchester Royal Infirmary, Oxford Rd, Manchester, M13 9WL, UK.
| | - Paul E C Brenchley
- Department of Renal Medicine, Manchester Royal Infirmary, Oxford Rd, Manchester, M13 9WL, UK.
| | - Michael L Picton
- Department of Renal Medicine, Manchester Royal Infirmary, Oxford Rd, Manchester, M13 9WL, UK.
| |
Collapse
|
14
|
Vlachopanos G, Kassimatis TI, Agrafiotis A. Perioperative administration of high-dose recombinant human erythropoietin for delayed graft function prevention in kidney transplantation: a meta-analysis. Transpl Int 2015; 28:330-40. [DOI: 10.1111/tri.12506] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 11/26/2014] [Accepted: 12/12/2014] [Indexed: 12/01/2022]
|
15
|
Lund A, Lundby C, Olsen NV. High-dose erythropoietin for tissue protection. Eur J Clin Invest 2014; 44:1230-8. [PMID: 25345962 DOI: 10.1111/eci.12357] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 10/20/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND The discovery of potential anti-apoptotic and cytoprotective effects of recombinant human erythropoietin (rHuEPO) has led to clinical trials investigating the use of high-dose, short-term rHuEPO therapy for tissue protection in conditions such as stroke and myocardial infarction. Experimental studies have been favourable, but the clinical efficacy has yet to be validated. MATERIALS AND METHODS We have reviewed clinical studies regarding the use of high-dose, short-term rHuEPO therapy for tissue protection in humans with the purpose to detail the safety and efficacy of rHuEPO for this indication. A systematic literature search was performed using the PubMed/MEDLINE database for randomized, placebo-controlled clinical trials. RESULTS Twenty-six randomized controlled trials that enrolled 3176 patients were included. The majority of trials (20 trials including 2724 patients) reported no effect of rHuEPO therapy on measures of tissue protection. Five trials including 1025 patients reported safety concerns in the form of increased mortality or adverse event rates. No studies reported reduced mortality. CONCLUSIONS Evidence is sparse to support a tissue-protective benefit of rHuEPO in humans. Moreover, a number of studies indicate that short-term administration of high-dose rHuEPO is associated with an increased risk of mortality and serious adverse events. Further work is needed to elucidate the mechanisms of toxicity of rHuEPO in humans.
Collapse
Affiliation(s)
- Anton Lund
- Department of Neuroscience and Pharmacology, The Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
16
|
Xin H, Ge YZ, Wu R, Yin Q, Zhou LH, Shen JW, Lu TZ, Hu ZK, Wang M, Zhou CC, Wu JP, Li WC, Zhu JG, Jia RP. Effect of high-dose erythropoietin on graft function after kidney transplantation: a meta-analysis of randomized controlled trials. Biomed Pharmacother 2014; 69:29-33. [PMID: 25661334 DOI: 10.1016/j.biopha.2014.10.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 10/27/2014] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Current evidence suggests that preconditioning with erythropoietin (EPO) can protect against ischemia reperfusion injury in rodents. However, randomized controlled trials (RCTs) assessing the efficacy and safety of high-dose EPO in kidney transplantation have yielded inconclusive results. Herein, we performed a meta-analysis of RCTs to assess whether the administration of high-dose EPO can improve graft function and the potential adverse events. METHODS Relevant RCT studies that investigated high-dose EPO on graft function after kidney transplantation were comprehensively searched in Pubmed, Embase, and Cochrane Library until July 10, 2014. All statistical analyses were performed using Review Manager 5.0 and STATA 12.0. RESULTS A total of 4 RCTs involving 356 patients were identified. Comprehensively, a trend of reduction in the incidence of delayed graft function could be observed in the EPO group (EPO vs. placebo groups: RR=0.88); however, the result did not reach the significance level (95% CI, 0.72-1.08; P=0.21). Furthermore, no significant difference in the incidences of adverse events was observed between the two groups. CONCLUSIONS The current meta-analysis indicates that the administration of high-dose EPO is, to some extent, prone to protect kidney function without increasing the susceptibility to adverse events.
Collapse
Affiliation(s)
- Hui Xin
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China; Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Yu-Zheng Ge
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China; Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Ran Wu
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China; Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Qian Yin
- Department of Pharmacology, Nanjing Medical University School of Pharmacy, 140 Hanzhong Road, 210029 Nanjing, PR China; Central Laboratory, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Liu-Hua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Jiang-Wei Shen
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Tian-Ze Lu
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China; Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Zhi-Kai Hu
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China; Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Min Wang
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China; Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Chang-Cheng Zhou
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China; Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Jian-Ping Wu
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China; Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Wen-Cheng Li
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Jia-Geng Zhu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China
| | - Rui-Peng Jia
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China; Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, 210006 Nanjing, PR China.
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Acute kidney injury (AKI) in transplant recipients is a prevalent condition with a broad list of potential inciting causes. This review highlights recent data describing the epidemiology and long-term consequences of transplant AKI, novel interventions in the management of delayed graft function (DGF), and noninvasive diagnostic strategies. RECENT FINDINGS The incidence and outcomes of nontransplant AKI are well documented, and similar data are emerging in the transplant setting with recent reports suggesting a high incidence rate and significant impact on long-term graft outcomes. DGF represents a 'pure' form of transplant AKI, and many interventional trials aiming to limit ischemia-reperfusion-induced injury have recently been reported or are currently ongoing. The search for accurate noninvasive predictors of DGF and acute rejection is ongoing and recent literature describes novel plasma and urine-based biomarkers as well as transcriptional profiling methods with high potential for clinical applicability. SUMMARY AKI in transplant recipients is a frequent occurrence with significant potential for poor long-term graft outcomes. Recent efforts to limit ischemia-reperfusion injury and diagnose transplant AKI via noninvasive methods may help to minimize the impact of AKI on future graft function.
Collapse
|
18
|
Gardner DS, Welham SJM, Dunford LJ, McCulloch TA, Hodi Z, Sleeman P, O'Sullivan S, Devonald MAJ. Remote conditioning or erythropoietin before surgery primes kidneys to clear ischemia-reperfusion-damaged cells: a renoprotective mechanism? Am J Physiol Renal Physiol 2014; 306:F873-84. [PMID: 24523383 DOI: 10.1152/ajprenal.00576.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Acute kidney injury is common, serious with no specific treatment. Ischemia-reperfusion is a common cause of acute kidney injury (AKI). Clinical trials suggest that preoperative erythropoietin (EPO) or remote ischemic preconditioning may have a renoprotective effect. Using a porcine model of warm ischemia-reperfusion-induced AKI (40-min bilateral cross-clamping of renal arteries, 48-h reperfusion), we examined the renoprotective efficacy of EPO (1,000 iu/kg iv.) or remote ischemic preconditioning (3 cycles, 5-min inflation/deflation to 200 mmHg of a hindlimb sphygmomanometer cuff). Ischemia-reperfusion induced significant kidney injury at 24 and 48 h (χ(2), 1 degree of freedom, >10 for 6/7 histopathological features). At 2 h, a panel of biomarkers including plasma creatinine, neutrophil gelatinase-associated lipocalin, and IL-1β, and urinary albumin:creatinine could be used to predict histopathological injury. Ischemia-reperfusion increased cell proliferation and apoptosis in the renal cortex but, for pretreated groups, the apoptotic cells were predominantly intratubular rather than interstitial. At 48-h reperfusion, plasma IL-1β and the number of subcapsular cells in G2-M arrest were reduced after preoperative EPO, but not after remote ischemic preconditioning. These data suggest an intrarenal mechanism acting within cortical cells that may underpin a renoprotective function for preoperative EPO and, to a limited extent, remote ischemic preconditioning. Despite equivocal longer-term outcomes in clinical studies investigating EPO as a renoprotective agent in AKI, optimal clinical dosing and administration have not been established. Our data suggest further clinical studies on the potential renoprotective effect of EPO and remote ischemic preconditioning are justified.
Collapse
Affiliation(s)
- David S Gardner
- School of Veterinary Medicine and Science, Univ. of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
van Rijt WG, Nieuwenhuijs-Moeke GJ, van Goor H, Ottens PJ, Ploeg RJ, Leuvenink HGD. Renoprotective capacities of non-erythropoietic EPO derivative, ARA290, following renal ischemia/reperfusion injury. J Transl Med 2013; 11:286. [PMID: 24225194 PMCID: PMC3842642 DOI: 10.1186/1479-5876-11-286] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 11/11/2013] [Indexed: 11/13/2022] Open
Abstract
Background ARA290 is a non-erythropoietic EPO derivative which only binds to the cytoprotective receptor complex (EPOR2-βcR2) consisting of two EPO-receptors (EPOR) and two β common receptors (βcR). ARA290 is renoprotective in renal ischemia/reperfusion (I/R). In a renal I/R model we focussed on timing of post-reperfusional administration of ARA290. Furthermore, we investigated the anti-inflammatory properties of ARA290. Methods Twenty-six male Lewis/HanHsd rats were exposed to unilateral ischemia for 30 minutes, with subsequent removal of the contralateral kidney. Post-reperfusion, ARA290 was administered early (one hour), late (four hours) or repetitive (one and four hours). Saline was used as vehicle treatment. Rats were sacrificed after three days. Results Early ARA290 treatment improved renal function. Late- or repetitive treatment tended to improve clinical markers. Furthermore, early ARA290 treatment reduced renal inflammation and acute kidney injury at three days post-reperfusion. Late- or repetitive treatment did not affect inflammation or acute kidney injury. Conclusions ARA290 attenuated renal ischemia/reperfusion injury. This study showed the anti-inflammatory effect of ARA290 and suggests early administration in the post-reperfusional phase is most effective. ARA290 is a candidate drug for protection against ischemic injury following renal transplantation.
Collapse
Affiliation(s)
- Willem G van Rijt
- Department of Surgery, University Medical Center Groningen, Hanzeplein 1, 9713, GZ Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
20
|
van Rijt WG, van Goor H, Ploeg RJ, Leuvenink HGD. Erythropoietin-mediated protection in kidney transplantation: nonerythropoietic EPO derivatives improve function without increasing risk of cardiovascular events. Transpl Int 2013; 27:241-8. [PMID: 23964738 DOI: 10.1111/tri.12174] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 04/29/2013] [Accepted: 07/27/2013] [Indexed: 11/30/2022]
Abstract
The protective, nonerythropoietic effects of erythropoietin (EPO) have become evident in preclinical models in renal ischaemia/reperfusion injury and kidney transplantation. However, four recently published clinical trials using high-dose EPO treatment following renal transplantation did not reveal any protective effect for short-term renal function and even reported an increased risk of thrombosis. This review focusses on the current status of protective pathways mediated by EPO, the safety concerns using high EPO dosage and discusses the discrepancies between pre-clinical and clinical studies. The protective effects are mediated by binding of EPO to a heteromeric receptor complex consisting of two β-common receptors and two EPO receptors. An important role for the activation of endothelial nitric oxide synthase is proposed. EPO-mediated cytoprotection still has enormous potential. However, only nonerythropoietic EPO derivatives may induce protection without increasing the risk of cardiovascular events. In preclinical models, nonerythropoietic EPO derivatives, such as carbamoylated EPO and ARA290, have been tested. These EPO derivatives improve renal function and do not affect erythropoiesis. Therefore, nonerythropoietic EPO derivatives may be able to render EPO-mediated cytoprotection useful and beneficial for clinical transplantation.
Collapse
Affiliation(s)
- Willem G van Rijt
- Department of Surgery, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands; Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | |
Collapse
|
21
|
van Rijt WG, Nieuwenhuijs-Moeke GJ, van Goor H, Jespersen B, Ottens PJ, Ploeg RJ, Leuvenink HGD. ARA290, a non-erythropoietic EPO derivative, attenuates renal ischemia/reperfusion injury. J Transl Med 2013; 11:9. [PMID: 23302512 PMCID: PMC3567997 DOI: 10.1186/1479-5876-11-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 12/21/2012] [Indexed: 11/25/2022] Open
Abstract
Background In contrast with various pre-clinical studies, recent clinical trials suggest that high dose erythropoietin (EPO) treatment following kidney transplantation does not improve short-term outcome and that it even increases the risk of thrombotic events. ARA290 is a non-erythropoietic EPO derivative and does not increase the risk of cardiovascular events, but potentially has cytoprotective capacities in prevention of renal ischemia/reperfusion injury. Methods Eight female Dutch Landrace pigs were exposed to unilateral renal ischemia for 45 minutes with simultaneous cannulation of the ureter of the ischemic kidney. ARA290 or saline was administered by an intravenous injection at 0, 2, 4 and 6 hours post-reperfusion. The animals were sacrificed seven days post-reperfusion. Results ARA290 increased glomerular filtration rate during the observation period of seven days. Furthermore, ARA290 tended to reduce MCP-1 and IL-6 expression 15 minutes post-reperfusion. Seven days post-reperfusion ARA290 reduced interstitial fibrosis. Conclusions The improvement in renal function following renal ischemia/reperfusion and reduced structural damage observed in this study by ARA290 warrants further investigation towards clinical application.
Collapse
Affiliation(s)
- Willem G van Rijt
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | | | | | | | | | | | | |
Collapse
|
22
|
Pallet N, Rabant M, Legendre C, Martinez F, Choukroun G. The nephroprotective properties of recombinant human erythropoietin in kidney transplantation: experimental facts and clinical proofs. Am J Transplant 2012; 12:3184-90. [PMID: 23057777 DOI: 10.1111/j.1600-6143.2012.04287.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Adaptive responses to hypoxia, including hypoxia-inducible factor signaling, allow the cell to satisfy its basal metabolic demand and avoid death, but these responses can also be deleterious by promoting inflammation, cell dedifferentiation and fibrogenesis. Therefore, targeting hypoxia constitutes a promising therapeutic avenue. Recombinant human erythropoietin (rhEPO) appeared as a good candidate therapy because its hematopoietic properties could reverse anemia, and its tissue-protective properties could reduce cell death and limit maladaptive cellular responses to hypoxia. Despite experimental evidence on the nephroprotecive properties of rhEPO, recent clinical trials provided evidence that rhEPO was ineffective in preventing delayed graft function after ischemic acute injury but that the normalization of hemoglobin values preserved kidney function deterioration and reduced graft loss. Our aim here is to provide a survey of the rationale for evaluating the administration of rhEPO in the setting of kidney transplantation. We will discuss the intriguing findings that emerged from the clinical trials and the discrepancies between promising experimental results and negative clinical studies, as well as the differences in terms of the benefits and safety profiles of the normalization of hemoglobin values in chronic kidney disease patients and kidney transplant patients.
Collapse
Affiliation(s)
- N Pallet
- INSERM U775, Centre Universitaire des Saints Pères, et Université Paris Descartes, Paris, France.
| | | | | | | | | |
Collapse
|
23
|
Penny H, Leckström D, Goldsmith D. The Janus faces of ESAs: caveat Chimaera! Int Urol Nephrol 2012; 45:761-7. [PMID: 22972568 DOI: 10.1007/s11255-012-0270-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Accepted: 08/08/2012] [Indexed: 11/30/2022]
Abstract
Patients with chronic kidney disease (CKD) have a Janus quality as they look back whence they came in developing CKD and, in some cases, also look forwards to a potential kidney transplant with the attendant promise of improvement in quality and often quantity of life. Making the most of this often unique opportunity is key-maximising the chance that the engraftment starts as a success, and then later, preserving good kidney transplant function for as long as possible. Two recently published, independently conceived and executed studies are relevant to both aspects of this quest and thus to all kidney transplant recipients (KTRs). Both trials also simultaneously stoke and quench the continuing, heated debates over target haemoglobin (Hb) levels, and the use of erythropoiesis-stimulating agents (ESAs), in CKD patients. One study--of acute, high-dose ESA administration--adds to the plethora of adverse safety signals swirling around the use of ESAs while surprisingly also showing renal function benefits at 12 months. The other study features chronic lower-dose ESA use in stable KTRs with anaemia and impaired renal function and not only purports to show a salutary effect on 2-year renal function outcomes (and thus reducing "return to dialysis" rates), but also rebuts the now widely accepted current notion that by chronic use of ESAs to target full Hb correction/higher Hb values in anaemic CKD patients, we are potentially causing harm.
Collapse
Affiliation(s)
- Hugo Penny
- King's Health Partners AHSC, Wing Guy's Hospital, 6th Floor, Borough, London, SE1 9RT, UK
| | | | | |
Collapse
|
24
|
Sureshkumar KK, Hussain SM, Ko TY, Thai NL, Marcus RJ. Effect of high-dose erythropoietin on graft function after kidney transplantation: a randomized, double-blind clinical trial. Clin J Am Soc Nephrol 2012; 7:1498-1506. [PMID: 22745272 PMCID: PMC3430945 DOI: 10.2215/cjn.01360212] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 05/29/2012] [Indexed: 01/14/2023]
Abstract
BACKGROUND AND OBJECTIVES Delayed graft function (DGF) is associated with adverse long-term outcomes after deceased-donor kidney (DDK) transplantation. Ischemia-reperfusion injury plays a crucial role in the development of DGF. On the basis of promising animal data, this study evaluated any potential benefits of erythropoietin-alfa (EPO-α) given intra-arterially at the time of reperfusion of renal allograft on the degree of allograft function, as well as tubular cell injury measured by urinary biomarkers in the early post-transplant period. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS A prospective, randomized, double-blind, placebo-controlled clinical trial was conducted to evaluate the influence of EPO-α administered intraoperatively on the outcomes of DDK transplantations performed at the study center between March 2007 and July 2009. RESULTS Seventy-two patients were randomly assigned to EPO-α (n=36) or placebo (n=36). The incidences of DGF, slow graft function, and immediate graft function did not significantly differ between the treatment and control groups (41.7% versus 47.2%, 25.0% versus 36.1%, and 33.3% versus 16.7%, respectively; P=0.24). The groups had similar levels of urinary biomarkers, including neutrophil gelatinase-associated lipocalin and IL-18 at multiple times points soon after transplantation; urinary output during the first 3 postoperative days; 1-month renal function; and BP readings, hemoglobin, and adverse effects during the first month. CONCLUSIONS This study did not show any clinically demonstrable beneficial effects of high-dose EPO-α given intra-arterially during the early reperfusion phase in DDK transplant recipients in terms of reducing the incidence of DGF or improving short-term allograft function.
Collapse
Affiliation(s)
- Kalathil K Sureshkumar
- Division of Nephrology and Hypertension, Department of Medicine, Allegheny General Hospital, Pittsburgh, Pennsylvania 15212, USA.
| | | | | | | | | |
Collapse
|
25
|
Aydin Z, Mallat MJK, Schaapherder AFM, van Zonneveld AJ, van Kooten C, Rabelink TJ, de Fijter JW. Randomized trial of short-course high-dose erythropoietin in donation after cardiac death kidney transplant recipients. Am J Transplant 2012; 12:1793-800. [PMID: 22429395 DOI: 10.1111/j.1600-6143.2012.04019.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Eryhropoiesis-stimulating agents have demonstrated tissue-protective effects in experimental models of ischemia-reperfusion injury. PROTECT was a 12-month, randomized, double-blind, placebo-controlled, single center study with high-dose recombinant human erythropoietin-β (Epoetin) in 92 donation after cardiac death (DCD) kidney transplant recipients. Patients were randomized to receive an intravenous bolus of Epoetin (3.3 × 10(4) international unit (IU); n = 45) or placebo (saline 0.9% solution; n = 47) on 3 consecutive days, starting 3-4 h before the transplantation and 24 h and 48 h after reperfusion. The immunosuppressive regimen included an anti-CD25 antibody, steroids, mycophenolate mofetil and delayed introduction of cyclosporine. Primary end point was a composite of the incidence of primary nonfunction and delayed graft function, either defined by spontaneous functional recovery or need for dialysis in the first week. Secondary objectives included duration of delayed function, renal function and proteinuria up to 1 year and thrombotic adverse events. Results showed no differences in the incidence or duration of delayed graft function and/or primary nonfunction (Epoetin 77.8 vs. placebo 78.7%, p = 1.00). Epoetin treatment significantly increased the risk of thrombotic events at 1 month and 1 year (Epoetin 24.4% vs. placebo 6.4%, p = 0.02).
Collapse
Affiliation(s)
- Z Aydin
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Erythropoietin (Epo) is an essential hormone that binds and activates the Epo receptor (EpoR) resident on the surface of erythroid progenitor cells, thereby promoting erythropoiesis. Recombinant human erythropoietin has been used successfully for over 20 years to treat anemia in millions of patients. In addition to erythropoiesis, Epo has also been reported to have other effects, such as tissue protection and promotion of tumor cell growth or survival. This became of significant concern in 2003, when some clinical trials in cancer patients reported increased tumor progression and worse survival outcomes in patients treated with erythropoiesis-stimulating agents (ESAs). One of the potential mechanisms proffered to explain the observed safety issues was that functional EpoR was expressed in tumors and/or endothelial cells, and that ESAs directly stimulated tumor growth and/or antagonized tumor ablative therapies. Since then, numerous groups have performed further research evaluating this potential mechanism with conflicting data and conclusions. Here, we review the biology of endogenous Epo and EpoR expression and function in erythropoiesis, and evaluate the evidence pertaining to the expression of EpoR on normal nonhematopoietic and tumor cells.
Collapse
|
27
|
In this Issue. Kidney Int 2012. [DOI: 10.1038/ki.2012.79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|