1
|
Alsunaid A, Spencer S, Bhandari S. Intravenous iron in chronic kidney disease without anaemia but iron deficiency: A scoping review. World J Nephrol 2025; 14:101576. [PMID: 40134647 PMCID: PMC11755244 DOI: 10.5527/wjn.v14.i1.101576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/30/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025] Open
Abstract
Iron deficiency (ID) is a prevalent complication of chronic kidney disease (CKD), often managed reactively when associated with anaemia. This scoping review evaluates the evidence supporting intravenous (IV) iron therapy in non-anaemic individuals with CKD and ID, focusing on safety, efficacy, and emerging therapeutic implications. Current diagnostic markers, including serum ferritin, transferrin saturation, and reticulocyte haemoglobin content, are reviewed alongside their limitations in the context of inflammation and variability. The pathophysiology of ID in CKD is explored, highlighting the roles of hepcidin, hypoxia-inducible factor pathways, and uraemic toxins. Comparative studies reveal that IV iron offers a more rapid correction of iron stores, improved compliance, and fewer gastrointestinal side effects compared to oral iron. Evidence from trials such as "iron and heart" and "iron and muscle" suggests potential benefits of IV iron on functional capacity and fatigue, though findings were statistically non-significant. Insights from heart failure trials support the safety and efficacy of IV iron in improving quality of life and reducing hospitalizations, with newer formulations like ferric derisomaltose demonstrating favourable safety profiles. This review underscores the need for standardized screening protocols for ID in CKD, even in the absence of anaemia, to facilitate earlier intervention. Future research should prioritise robust outcome measures, larger sample sizes, and person-specific treatment strategies to optimise dosing and administration frequency. Tailored approaches to IV iron therapy have the potential to significantly improve functional outcomes, quality of life, and long-term health in people with CKD.
Collapse
Affiliation(s)
- Abdulrahman Alsunaid
- Department of Medical Science, Hull York Medical School, Kingston Upon Hull HU6 7RU, United Kingdom
| | - Sebastian Spencer
- Department of Medical Science, Hull York Medical School, Kingston Upon Hull HU6 7RU, United Kingdom
- Department of Medical Science, University of Hull, Kingston Upon Hull HU6 7RU, United Kingdom
- Department of Academic Renal, Hull University Teaching Hospitals NHS Trust, Kingston Upon Hull HU3 2JZ, United Kingdom
| | - Sunil Bhandari
- Department of Medical Science, Hull York Medical School, Kingston Upon Hull HU6 7RU, United Kingdom
- Department of Academic Renal, Hull University Teaching Hospitals NHS Trust, Kingston Upon Hull HU3 2JZ, United Kingdom
| |
Collapse
|
2
|
McDonnell T, Kalra PA. Iron in Chronic Kidney Disease and End-Stage Kidney Disease-Current Trends and Future Direction. Br J Hosp Med (Lond) 2025; 86:1-19. [PMID: 39998134 DOI: 10.12968/hmed.2024.0619] [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] [Indexed: 02/26/2025]
Abstract
Anaemia is a frequent and serious complication in chronic kidney disease (CKD), affecting both non-dialysis-dependent (NDD) and dialysis-dependent (DD) patients. While erythropoietin (EPO) deficiency is the primary cause, iron deficiency (ID) also plays a crucial role. ID in CKD can be classified as either absolute, resulting from blood loss, or functional, driven by inflammation and elevated hepcidin levels, which trap iron in macrophages and hepatocytes, preventing its use in erythropoiesis. Elevated hepcidin also reduces dietary iron absorption in the gut, making oral iron supplements ineffective, particularly in advanced CKD. This review summarises the available intravenous (IV) iron formulations, discusses diagnostic definitions and treatment thresholds for ID in NDD and DD CKD, and explores potential future therapeutic directions.
Collapse
Affiliation(s)
- Thomas McDonnell
- Donal O'Donoghue Renal Research Centre, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK
- Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Philip A Kalra
- Donal O'Donoghue Renal Research Centre, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK
- Division of Cardiovascular Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
3
|
Murillo Ortiz BO, Ramírez Emiliano J, Romero Vázquez MJ, Amador Medina LF, Martínez Garza S, Ramos Rodríguez EM. Impact of iron chelation with deferasirox on telomere length and oxidative stress in hemodialysis patients: A randomized study. Nefrologia 2025; 45:68-76. [PMID: 39884801 DOI: 10.1016/j.nefroe.2025.01.003] [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: 02/12/2024] [Accepted: 06/12/2024] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Recent studies have demonstrated the effectiveness, safety, and tolerability of deferasirox in patients in peritoneal dialysis, however, its effect has not been studied in patients undergoing hemodialysis. OBJECTIVE To investigate the impact of iron chelation on telomere length, oxidative stress, and ferritin levels in patients undergoing hemodialysis. METHODS This is an open-label study, with a control group of patients undergoing hemodialysis, who will receive treatment with deferasirox 15mg/kg/day for 6 months for iron chelation. Telomere length was measured using real-time PCR. Serum ferritin levels and oxidation markers were evaluated. To evaluate the pharmacokinetics and safety of deferasirox, plasma concentrations were analyzed by HPLC. RESULTS Fifty-four patients were included to receive deferasirox, and a control group of 50 patients. Significant differences were observed in serum ferritin levels (p<0.0001), TBARS (thiobarbituric acid reactive substances) (p<0.01). Telomere length had a significant increase after chelation (p<0.001). The serum deferasirox concentration at zero time at 48h was maintained within a range of 2.67-23.78mmol/L. CONCLUSIONS Our results demonstrate that iron chelation in hemodialysis patients significantly reduces ferritin and TBARS, resulting in an increase in telomere length. Deferasirox proves to be beneficial for patients with iron overload undergoing hemodialysis.
Collapse
Affiliation(s)
- Blanca Olivia Murillo Ortiz
- Clinical Epidemiology Research Unit, OOAD Guanajuato, Mexican Institute of Social Security, León, Guanajuato, Mexico.
| | - Joel Ramírez Emiliano
- Department of Medical Sciences, Health Sciences Division, University of Guanajuato, León, Guanajuato, Mexico
| | - Marcos Javier Romero Vázquez
- Clinical Epidemiology Research Unit, OOAD Guanajuato, Mexican Institute of Social Security, León, Guanajuato, Mexico
| | - Lauro Fabián Amador Medina
- Clinical Epidemiology Research Unit, OOAD Guanajuato, Mexican Institute of Social Security, León, Guanajuato, Mexico
| | - Sandra Martínez Garza
- Clinical Epidemiology Research Unit, OOAD Guanajuato, Mexican Institute of Social Security, León, Guanajuato, Mexico
| | - Edna Montserrat Ramos Rodríguez
- Department of Hemodialysis, Hospital General Regional No. 58, Mexican Institute of Social Security, León, Guanajuato, Mexico
| |
Collapse
|
4
|
van Lieshout TS, Klerks AK, Mahic O, Vernooij RWM, Eisenga MF, van Jaarsveld BC, Abrahams AC. Comparative iron management in hemodialysis and peritoneal dialysis patients: a systematic review. FRONTIERS IN NEPHROLOGY 2024; 4:1488758. [PMID: 39664943 PMCID: PMC11631840 DOI: 10.3389/fneph.2024.1488758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024]
Abstract
Background Patients with kidney failure undergoing dialysis often suffer from anemia. Iron deficiency, along with a shortage in erythropoietin, is a common cause. Peritoneal dialysis (PD) patients may have a different iron metabolism compared to hemodialysis (HD) patients. This study aims to compare both dialysis modalities regarding their differences in iron management. Methods PubMed (MEDLINE) and Embase were screened for randomized controlled trials and observational studies including both patients on HD or PD with information on iron management. Outcomes for iron management for this systematic review included: prevalence of supplementation, route of administration, dose, frequency and hemoglobin and iron status parameters. Results 15 eligible studies (930,436 patients), of which 8 cohort and 7 cross-sectional, were analyzed. The prevalence of intravenous (IV) iron supplementation ranged from 11.7% to 84.4% in HD patients, compared to 1.6% to 49.0% in PD patients. Ten studies reported that HD patients only received IV iron, while five studies reported this for PD patients. For oral iron supplementation, three studies involved HD patients, whereas seven studies involved PD patients. The cumulative monthly IV iron dose ranged from 108 to 750 mg in the HD group, compared to 65 to 250 mg in the PD group. Hemoglobin levels ranged from 10.0 to 12.0 g/dL in HD patients, versus 9.6 to 11.9 g/dL in PD patients. Conclusion Iron management differs between HD and PD patients, with HD patients receiving higher doses and more frequent IV iron. There was significant heterogeneity in the outcomes between the studies, primarily due to the lack of a uniform global policy on iron management. Despite these differences, hemoglobin levels and iron status parameters were comparable between the two groups. Future research should explore the underlying mechanisms and broader impacts of iron treatment, including patient-reported outcomes, to optimize anemia management and improve quality of life for dialysis patients. Systematic Review Registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022336970.
Collapse
Affiliation(s)
- Thomas S. van Lieshout
- Department of Nephrology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Research Institute Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
- Department of Internal Medicine, Northwest Clinics, Alkmaar, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, Netherlands
| | - Anastasia K. Klerks
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| | - Osman Mahic
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Robin W. M. Vernooij
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Michele F. Eisenga
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Brigit C. van Jaarsveld
- Department of Nephrology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Research Institute Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, Netherlands
- Nephrocare Diapriva Dialysis Center, Amsterdam, Netherlands
| | - Alferso C. Abrahams
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
5
|
Chow K, Trollinger B, Blum M, Alasfar S, Monroy-Trujillo JM, Brown D. Implementation of a Pharmacist-Driven Protocol to Improve Screening and Treatment of Iron Deficiency in Hospitalized Patients with Chronic Kidney Disease. Hosp Pharm 2024; 60:00185787241267730. [PMID: 39558938 PMCID: PMC11569762 DOI: 10.1177/00185787241267730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Purpose: While intravenous (IV) iron repletion is an effective tool to treat anemia and improve outcomes in chronic kidney disease (CKD), guideline laboratory definitions of iron deficiency differ, resulting in variability in screening and repletion strategies. This study sought to describe current practices surrounding identification and treatment of iron deficiency in CKD and then implement a pharmacist-led protocol to optimize care at a tertiary medical center. Methods: This single center, retrospective, pre- and post-protocol implementation study of adults with CKD admitted to the inpatient setting first analyzed historic practices for iron deficiency screening and treatment, followed by deployment of a pharmacist-driven protocol for iron deficiency screening and treatment. Iron deficiency was defined as transferrin saturation of ≤30% and ferritin of ≤500 ng/mL. Improvement in screening and repletion rates was analyzed. Results: Historic pre-protocol practices were reviewed in 7155 admissions of which 2559 (35.8%) included screening for iron deficiency. Over the 2 months intervention (post-protocol) period, 315 admissions were included. The average age of patients in the post-protocol cohort was 64.1 years, 53.7% were female, and 26.4% were dialysis dependent. Compared to pre-protocol, patients were 2.33 (95% CI 2.20-2.47) times more likely to be screened and deficient patients were 2.05 (95% CI 1.46-2.86) times more likely to be treated, with most receiving IV iron therapy (85.4%), in the post-protocol cohort. Patients were 3.58 times (95% CI 1.97-6.48) more likely to receive IV iron versus oral alone in the post-protocol cohort compared to pre-protocol. Conclusion: The frequency of patients with CKD screened and treated with iron increased after implementation of a pharmacist-driven protocol. This study underscores the need for a systematic approach to identification/treatment of iron deficiency in this population.
Collapse
Affiliation(s)
- Karissa Chow
- The Johns Hopkins Hospital Deparment of Pharmacy, Baltimore, MD, USA
| | | | - Matthew Blum
- Johns Hopkins University School of Medicine, Department of Nephrology, Baltimore, MD, USA
| | - Sami Alasfar
- Johns Hopkins University School of Medicine, Department of Nephrology, Baltimore, MD, USA
| | | | - Dannielle Brown
- The Johns Hopkins Hospital Deparment of Pharmacy, Baltimore, MD, USA
| |
Collapse
|
6
|
Chukwu CA, Gilbody H, Wickens O, Carroll C, Bhandari S, Kalra PA. Factors Governing the Erythropoietic Response to Intravenous Iron Infusion in Patients with Chronic Kidney Disease: A Retrospective Cohort Study. Biomedicines 2023; 11:2417. [PMID: 37760860 PMCID: PMC10525177 DOI: 10.3390/biomedicines11092417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Limited knowledge exists about factors affecting parenteral iron response. A study was conducted to determine the factors influencing the erythropoietic response to parenteral iron in iron-deficient anaemic patients whose kidney function ranged from normal through all stages of chronic kidney disease (CKD) severity. METHODS This retrospective cohort study included parenteral iron recipients who did not receive erythropoiesis-stimulating agents (ESA) between 2017 and 2019. The study cohort was derived from two groups of patients: those managed by the CKD team and patients being optimised for surgery in the pre-operative clinic. Patients were categorized based on their kidney function: Patients with normal kidney function [estimated glomerular filtration rate (eGFR) ≥ 60 mL/min/1.73 m2] were compared to those with CKD stages 3-5 (eGFR < 60 mL/min/1.73 m2). Patients were further stratified by the type of iron deficiency [absolute iron deficiency (AID) versus functional iron deficiency (FID)]. The key outcome was change in hemoglobin (∆Hb) between pre- and post-infusion haemoglobin (Hb) values. Parenteral iron response was assessed using propensity-score matching and multivariate linear regression. The impact of kidney impairment versus the nature of iron deficiency (AID vs. FID) in response was explored. RESULTS 732 subjects (mean age 66 ± 17 years, 56% females and 87% White) were evaluated. No significant differences were observed in the time to repeat Hb among CKD stages and FID/AID patients. The Hb rise was significantly lower with lower kidney function (non-CKD and CKD1-2; 13 g/L, CKD3-5; 7 g/L; p < 0.001). When groups with different degrees of renal impairment were propensity-score matched according to whether iron deficiency was due to AID or FID, the level of CKD was found not to be relevant to Hb responses [unmatched (∆Hb) 12.1 vs. 8.7 g/L; matched (∆Hb) 12.4 vs. 12.1 g/L in non-CKD and CKD1-2 versus CKD3-5, respectively]. However, a comparison of patients with AID and FID, while controlling for the degree of CKD, indicated that patients with FID exhibited a diminished Hb response regardless of their level of kidney impairment. CONCLUSION The nature of iron deficiency rather than the severity of CKD has a stronger impact on Hb response to intravenous iron with an attenuated response seen in functional iron deficiency irrespective of the degree of renal impairment.
Collapse
Affiliation(s)
- Chukwuma A. Chukwu
- Department of Nephrology Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK; (O.W.); (C.C.); (P.A.K.)
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| | - Helen Gilbody
- College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK;
| | - Olivia Wickens
- Department of Nephrology Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK; (O.W.); (C.C.); (P.A.K.)
| | - Craig Carroll
- Department of Nephrology Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK; (O.W.); (C.C.); (P.A.K.)
| | - Sunil Bhandari
- Academic Renal Research Department, Hull University Teaching Hospitals NHS Trust and Hull York Medical School, Kingston upon Hull, Hull HU3 2JZ, UK;
| | - Philip A. Kalra
- Department of Nephrology Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK; (O.W.); (C.C.); (P.A.K.)
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
7
|
Hassan S, Mumford L, Robinson S, Foukanelli D, Torpey N, Ploeg RJ, Mamode N, Murphy MF, Brown C, Roberts DJ, Regan F, Willicombe M. Blood transfusions post kidney transplantation are associated with inferior allograft and patient survival-it is time for rigorous patient blood management. FRONTIERS IN NEPHROLOGY 2023; 3:1236520. [PMID: 37675353 PMCID: PMC10479650 DOI: 10.3389/fneph.2023.1236520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/03/2023] [Indexed: 09/08/2023]
Abstract
Background Patient Blood Management (PBM), endorsed by the World Health Organisation is an evidence-based, multi-disciplinary approach to minimise inappropriate blood product transfusions. Kidney transplantation presents a particular challenge to PBM, as comprehensive evidence of the risk of transfusion is lacking. The aim of this study is to investigate the prevalence of post-transplant blood transfusions across multiple centres, to analyse risk factors for transfusion and to compare transplant outcomes by transfusion status. Methods This analysis was co-ordinated via the UK Transplant Registry within NHS Blood and Transplant (NHSBT), and was performed across 4 centres. Patients who had received a kidney transplant over a 1-year period, had their transfusion status identified and linked to data held within the national registry. Results Of 720 patients, 221(30.7%) were transfused, with 214(29.7%) receiving a red blood cell (RBC) transfusion. The proportion of patients transfused at each centre ranged from 20% to 35%, with a median time to transfusion of 4 (IQR:0-12) days post-transplant. On multivariate analysis, age [OR: 1.02(1.01-1.03), p=0.001], gender [OR: 2.11(1.50-2.98), p<0.0001], ethnicity [OR: 1.28(1.28-2.60), p=0.0008], and dialysis dependence pre-transplant [OR: 1.67(1.08-2.68), p=0.02], were associated with transfusion. A risk-adjusted Cox proportional hazards model showed transfusion was associated with inferior 1-year patient survival [HR 7.94(2.08-30.27), p=0.002] and allograft survival [HR: 3.33(1.65-6.71), p=0.0008], and inferior allograft function. Conclusion RBC transfusions are common and are independently associated with inferior transplant outcomes. We urge that further research is needed to understand the mechanisms behind the outcomes, to support the urgent development of transplant-specific anaemia guidelines.
Collapse
Affiliation(s)
- Sevda Hassan
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare National Health Service (NHS) Trust, Hammersmith Hospital, London, United Kingdom
| | - Lisa Mumford
- Statistics and Clinical Studies, National Health Service (NHS) Blood and Transplant, Bristol, United Kingdom
| | - Susan Robinson
- Department of Haematology, Guys, Evelina and St Thomas National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Dora Foukanelli
- Department of Haematology, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Nick Torpey
- Department of Clinical Nephrology and Transplantation, Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, United Kingdom
| | - Rutger J. Ploeg
- Department of Surgery, Nuffield Department of Surgical Science, University of Oxford, Oxford, United Kingdom
| | - Nizam Mamode
- Department of Transplantation, Guys, Evelina and St Thomas National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Michael F. Murphy
- National Health Service (NHS) Blood and Transplant, and Nuffield Department of Clinical and Laboratory Sciences, University of Oxford, Oxford, United Kingdom
| | - Colin Brown
- Haematology, National Health Service (NHS) Blood and Transplant, London, United Kingdom
| | - David J. Roberts
- Haematology, National Health Service (NHS) Blood and Transplant, London, United Kingdom
| | - Fiona Regan
- Haematology, National Health Service (NHS) Blood and Transplant, London, United Kingdom
| | - Michelle Willicombe
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Imperial College Renal and Transplant Centre, Imperial College Healthcare National Health Service (NHS) Trust, Hammersmith Hospital, London, United Kingdom
| |
Collapse
|
8
|
Onder AM, Ansari MAY, Deng F, Grinsell MM, Patterson L, Jetton J, Fathallah-Shaykh S, Ranch D, Aviles D, Copelovitch L, Ellis E, Chadha V, Elmaghrabi A, Lin JJ, Butani L, Haddad M, Marsenic O, Brakeman P, Quigley R, Shin HS, Garro R, Raina R, Langman CB. Persistent Increase in Serum Ferritin Levels despite Converting to Permanent Vascular Access in Pediatric Hemodialysis Patients: Pediatric Nephrology Research Consortium Study. J Clin Med 2023; 12:4251. [PMID: 37445286 DOI: 10.3390/jcm12134251] [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: 05/27/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Our objective was to examine serum ferritin trends after conversion to permanent vascular access (PVA) among children who started hemodialysis (HD) using tunneled cuffed catheters (TCC). Retrospective chart reviews were completed on 98 subjects from 20 pediatric HD centers. Serum ferritin levels were collected at the creation of PVA and for two years thereafter. There were 11 (11%) arteriovenous grafts (AVG) and 87 (89%) arteriovenous fistulae (AVF). Their mean TCC use was 10.4 ± 17.3 months. Serum ferritin at PVA creation was elevated at 562.64 ± 492.34 ng/mL, increased to 753.84 ± 561.54 ng/mL (p = < 0.001) in the first year and remained at 759.60 ± 528.11 ng/mL in the second year (p = 0.004). The serum ferritin levels did not show a statistically significant linear association with respective serum hematocrit values. In a multiple linear regression model, there were three predictors of serum ferritin during the first year of follow-up: steroid-resistant nephrotic syndrome as primary etiology (p = 0.035), being from a center that enrolled >10 cases (p = 0.049) and baseline serum ferritin level (p = 0.017). Increasing serum ferritin after conversion to PVA is concerning. This increase is not associated with serum hematocrit trends. Future studies should investigate the correlation of serum transferrin saturation and ferritin levels in pediatric HD patients.
Collapse
Affiliation(s)
- Ali Mirza Onder
- Division of Pediatric Nephrology, Batson Children's Hospital of Mississippi, University of Mississippi, Jackson, MS 39216, USA
- Division of Pediatric Nephrology, Nemours Children's Hospital, Delaware, Wilmington, DE 19803, USA
| | - Md Abu Yusuf Ansari
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Fang Deng
- Kidney Diseases Division, Feinberg School of Medicine, Northwestern University, Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Matthew M Grinsell
- Division of Pediatric Nephrology, Primary Children's Hospital, University of Utah, Salt Lake City, UT 84112, USA
| | - Larry Patterson
- Division of Pediatric Nephrology, Children's National Health System, Washington, DC 20010, USA
| | - Jennifer Jetton
- Division of Nephrology, Dialysis and Transplantation, University of Iowa Stead Family Children's Hospital, Iowa City, IA 52242, USA
| | - Sahar Fathallah-Shaykh
- Division of Pediatric Nephrology, Children's of Alabama, University of Alabama, Birmingham, AL 35233, USA
| | - Daniel Ranch
- Division of Pediatric Nephrology, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Diego Aviles
- Division of Pediatric Nephrology, Children's Hospital New Orleans, LSU Heath School of Medicine, New Orleans, LA 70118, USA
| | - Lawrence Copelovitch
- Division of Nephrology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eileen Ellis
- Division of Pediatric Nephrology, Arkansas Children's Hospital, Little Rock, AR 72202, USA
| | - Vimal Chadha
- Division of Pediatric Nephrology, Children's Mercy Hospital, Kansas City, MO 64108, USA
| | - Ayah Elmaghrabi
- Division of Pediatric Nephrology, Children's Medical Center Dallas, UT Southwestern, Dallas, TX 75235, USA
| | - Jen-Jar Lin
- Division of Pediatric Nephrology, Brenner Children's Hospital, Wake Forest University, Winston Salem, NC 27157, USA
| | - Lavjay Butani
- Division of Pediatric Nephrology, UC Davis Children's Hospital, Sacramento, CA 95817, USA
| | - Maha Haddad
- Division of Pediatric Nephrology, UC Davis Children's Hospital, Sacramento, CA 95817, USA
| | - Olivera Marsenic
- Division of Pediatric Nephrology, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Paul Brakeman
- Division of Pediatric Nephrology, UCSF Benioff Children's Hospital, San Francisco, CA 94158, USA
| | - Raymond Quigley
- Division of Pediatric Nephrology, Children's Medical Center Dallas, UT Southwestern, Dallas, TX 75235, USA
| | - H Stella Shin
- Division of Pediatric Nephrology, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Rouba Garro
- Division of Pediatric Nephrology, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Rupesh Raina
- Division of Pediatric Nephrology, Akron Children's Hospital, Akron, OH 44308, USA
| | - Craig B Langman
- Kidney Diseases Division, Feinberg School of Medicine, Northwestern University, Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| |
Collapse
|
9
|
Lacquaniti A, Gargano R, Campo S, Casuscelli di Tocco T, Schifilliti S, Monardo P. The Switch from Ferric Gluconate to Ferric Carboxymaltose in Hemodialysis Patients Acts on Iron Metabolism, Erythropoietin, and Costs: A Retrospective Analysis. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1071. [PMID: 37374275 DOI: 10.3390/medicina59061071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/18/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023]
Abstract
Background and Objectives: Iron deficiency and anemia characterize patients on chronic hemodialysis (HD). Available intravenous iron agents, such as ferric gluconate (FG) and ferric carboxymaltose (FCM), vary in dosing regimens and safety profiles. The aim of the present study was to analyze the modification of the iron status, the correction of anemia, and the economic implications after the shift from FG to FCM therapy in chronic HD patients. We evaluated, during the study, the variations in iron metabolism, assessing ferritin and transferrin saturation, erythropoietin-stimulating agent (ESA) doses and the number of administrations, the effects on anemic status, and consequent costs. Materials and Methods: A retrospective study was performed with a follow-up period of 24 months, enrolling forty-two HD patients. The enrolment phase started in January 2015, when patients were treated with iv FG, and continued until December 2015, when FG was discontinued, and, after a wash-out period, the same patients were treated with FCM. Results: The iron switch reduced the administered dose of ESA by 1610.500 UI (31% of reduction; p < 0.001) during the entire study period and reduced the erythropoietin resistance index (ERI) (10.1 ± 0.4 vs. 14.8 ± 0.5; p < 0.0001). The FCM group had the highest percentage of patients who did not require ESA treatment during the study period. The FCM patients were characterized by higher levels of iron (p = 0.04), ferritin (p < 0.001), and TSAT levels (p < 0.001) compared to the FG patients. The annual cost during FG infusion was estimated at EUR 105,390.2, while one year of treatment with FCM had a total cost of EUR 84,180.7 (a difference of EUR 21,209.51 (20%), saving EUR 42.1 per patient/month (p < 0.0001). Conclusions: FCM was a more effective treatment option than FG, reducing ESA dose requirements, increasing Hb levels, and improving iron status. The reduced ESA doses and the decreased number of patients needing ESA were the main factors for reducing overall costs.
Collapse
Affiliation(s)
| | - Romana Gargano
- Department of Economics, University of Messina, 98100 Messina, Italy
| | - Susanna Campo
- Nephrology and Dialysis Unit, Papardo Hospital, 98158 Messina, Italy
| | | | - Silvia Schifilliti
- Faculty of Pharmacy, Clinical Pharmacy Fellowship, University of Messina Annunziata Campus, 98168 Messina, Italy
| | - Paolo Monardo
- Nephrology and Dialysis Unit, Papardo Hospital, 98158 Messina, Italy
| |
Collapse
|
10
|
Czempik PF, Wiórek A. Iron deficiency in sepsis patients managed with divided doses of iron dextran: a prospective cohort study. Sci Rep 2023; 13:5264. [PMID: 37002279 PMCID: PMC10066317 DOI: 10.1038/s41598-023-32002-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
Iron deficiency (ID) impairs hemoglobin (Hb) synthesis and immune function, both crucial for sepsis patients. We assessed the impact of iron dextran on reticulocyte (Ret) Hb equivalent (Ret-He) and Ret subpopulations in iron-deficient sepsis patients. In this prospective clinical study we enrolled patients with sepsis or septic shock with procalcitonin concentration > 0.5 ng/mL, diagnosed with ID based on Ret-He. Study subjects received divided doses of iron dextran until normalization of Ret-He. The study population included 35 subjects. The median Ret-He increase after 2 doses of iron dextran was 3.0 (IQR 1.9-6.1) pg (p < 0.01) with median time to normalization 4 (IQR 3-5) days. Although no change in Ret percentage [Me 1.5 (IQR 1.1-2.1) vs. Me 1.4 (IQR 1.1-2.4) %, p = 0.39] and number [Me 0.05 (IQR 0.04-0.07) vs. Me 0.05 (IQR 0.03-0.06) 106/µL, p = 0.88] was noted, Ret subpopulations changed significantly (p for all < 0.01). Divided doses of iron dextran relatively quickly normalize Ret-He in iron-deficient sepsis patients. Changes in Ret subpopulations suggest increased erythropoietic activity. Further research is needed to explore the role of intravenous iron in this clinical setting.
Collapse
Affiliation(s)
- Piotr F Czempik
- Department of Anaesthesiology and Intensive Care, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 14, 40-752, Katowice, Poland.
- Transfusion Committee, University Clinical Center of Medical University of Silesia in Katowice, Katowice, Poland.
| | - Agnieszka Wiórek
- Department of Anaesthesiology and Intensive Care, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków 14, 40-752, Katowice, Poland
| |
Collapse
|
11
|
[Evaluation of serum iron as a predictor of a hemoglobin response to injectable iron treatment in chronic hemodialysis patients]. Nephrol Ther 2022; 18:634-642. [PMID: 36216731 DOI: 10.1016/j.nephro.2022.03.008] [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: 05/03/2021] [Revised: 03/22/2022] [Accepted: 03/26/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND The detection and correction of iron deficiency are essential for the treatment of anemia in chronic hemodialysis patients. The aim of our study was to assess the ability of serum iron to predict hemoglobin response to intravenous iron supplementation in hemodialysis patients. METHODS It is a retrospective study in 91 hemodialysis patients during 2016 at Clermont-Ferrand University Hospital for whom intravenous iron supplementation had been started. A responder patient was defined as an increase in hemoglobin greater than or equal to 1 g/dL/month and/or a decrease in the dose of erythropoiesis stimulating agent after two months of iron supplementation. RESULTS In responding patients, serum iron was significantly lower (6.7 ± 2.7 μmol/L) compared to non-responding patients (8.9±2.9 μmol/L; P<0.001). The positive response to iron supplementation was significantly associated with low serum iron (odds ratio = 0.58 [0.42-0.81]; P=0.002) in a logistic regression model taking into account ferritin, transferrin saturation coefficient, dose variation monthly iron and erythropoiesis stimulating agent and the duration of dialysis. The area under the receiver operating characteristic curve of serum iron, ferritin and transferrin saturation coefficient to predict the response to iron supplementation were 0.72, 0.51 and 0.64, respectively (serum iron versus ferritin [P=0.006] and serum iron versus transferrin saturation coefficient [P=0.04]). The sensitivity for serum iron below 7.5 μmol/L was better than that for ferritin below 86 ng/mL (P<0.001) and the specificity for serum iron below 7.5 μmol/L was better than that for TSC less than 19% (P=0.02). CONCLUSION Serum iron below 7.5 μmol/L can predict the success of the response to iron supplementation in chronic hemodialysis patients.
Collapse
|
12
|
Chinese Clinical Practice Guideline for the Management of "CKD-PeriDialysis"-the Periods Prior to and in the Early-Stage of Initial Dialysis. Kidney Int Rep 2022; 7:S531-S558. [PMID: 36567827 PMCID: PMC9782818 DOI: 10.1016/j.ekir.2022.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 12/14/2022] Open
Abstract
The National Experts Group on Nephrology have developed these guidelines to improve the management of pre-dialysis and initial dialysis patients with chronic kidney disease (CKD) (two periods contiguous with dialysis initiation termed here 'PeriDialysis CKD'). The pre-dialysis period is variable, whereas the initial dialysis period is more fixed at 3 months to 6 months after initiating dialysis. The new concept and characteristics of 'CKD-PeriDialysis' are proposed in the guideline. During the CKD-PeriDialysis period, the incidence rate of complications, mortality and treatment cost significantly increases and the glomerular filtration rate (GFR) rapidly decreases, which requires intensive management. The guideline systematically and comprehensively elaborates the recommendations for indicators to be used in for disease evaluation, timing and mode selection of renal replacement therapy, dialysis adequacy evaluation, and diagnosis and treatment of common PeriDialysis complications. Finally, future research directions of CKD-PeriDialysis are proposed. CKD-PeriDialysis management is a difficult clinical issue in kidney disease, and the development and implementation of these guidelines is important to improve the management of CKD-PeriDialysis patients in China, which could ultimately improve survival rates and quality of life, and reduce the medical burden.
Collapse
|
13
|
Hildebrand S, Busbridge M, Duncan ND, Tam FWK, Ashby DR. Predictors of iron versus erythropoietin responsiveness in anemic hemodialysis patients. Hemodial Int 2022; 26:519-526. [PMID: 35833334 PMCID: PMC9796788 DOI: 10.1111/hdi.13030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 03/30/2022] [Accepted: 05/30/2022] [Indexed: 01/07/2023]
Abstract
Anemia protocols for hemodialysis patients usually titrate erythropoietin (ESA) according to hemoglobin and iron according to a threshold of ferritin, with variable response seen. A universally optimum threshold for ferritin may be incorrect, and another view is that ESA and iron are alternative anemia treatments, which should be selected based on the likely response to each. Hemodialysis patients developing moderate anemia were randomised to treatment with either an increase in ESA or a course of intravenous iron. Over 2423 patient-months in 197 patients, there were 133 anemia episodes with randomized treatment. Treatment failure was seen in 20/66 patients treated with ESA and 20/67 patients treated with iron (30.3 vs. 29.9%, p = 1.0). Successful ESA treatment was associated with lower C-reactive protein (13.5 vs. 28.6 mg/L, p = 0.038) and lower previous ESA dose (6621 vs. 9273 μg/week, p = 0.097). Successful iron treatment was associated with lower reticulocyte hemoglobin (33.8 vs. 35.5 pg, p = 0.047), lower hepcidin (91.4 vs. 131.0 μg/ml, p = 0.021), and higher C-reactive protein (29.5 vs. 12.6 mg/L, p = 0.085). A four-variable iron preference score was developed to indicate the more favorable treatment, which in a retrospective analysis reduced treatment failure to 17%. Increased ESA and iron are equally effective, though treatment failure occurs in almost 30%. Baseline variables including hepcidin can predict treatment response, and a four-variable score shows promise in allowing directed treatment with improved response rates.
Collapse
Affiliation(s)
- Sarah Hildebrand
- Hammersmith HospitalImperial College Healthcare NHS TrustLondonUK
| | - Mark Busbridge
- Hammersmith HospitalImperial College Healthcare NHS TrustLondonUK
| | - Neill D. Duncan
- Hammersmith HospitalImperial College Healthcare NHS TrustLondonUK
| | - Frederick W. K. Tam
- Hammersmith HospitalImperial College Healthcare NHS TrustLondonUK,Centre for Inflammatory Disease, Department of MedicineImperial College LondonLondonUK
| | - Damien R. Ashby
- Hammersmith HospitalImperial College Healthcare NHS TrustLondonUK
| |
Collapse
|
14
|
Dubovetsky D, Tyler J, Bisceglia S. Assessing impact of an interdisciplinary erythropoiesis‐stimulating agent dosing protocol at an outpatient community hospital hemodialysis unit. JOURNAL OF THE AMERICAN COLLEGE OF CLINICAL PHARMACY 2022. [DOI: 10.1002/jac5.1675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
| | - Joshua Tyler
- Department of Pharmacy Advent Health Orlando Florida USA
| | | |
Collapse
|
15
|
Liao R, Zhou X, Ma D, Tang J, Zhong H. Iron Deficiency is Associated With Platelet Count Elevation in Patients With Dialysis-dependent Chronic Kidney Disease. J Ren Nutr 2022; 32:587-594. [PMID: 35339349 DOI: 10.1053/j.jrn.2021.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/18/2021] [Accepted: 09/05/2021] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Iron deficiency is common in patients with end-stage renal disease (ESRD). Platelet count changes may reflect iron status, but the relationship between platelet count and iron indices is unclear in patients with ESRD. METHODS We conducted a retrospective study in 1,167 patients with ESRD from 2012 to 2017 in West China Hospital. Baseline data were used to analyze the relationship between the platelet count and iron indices. Patients were followed up for 3 years. RESULTS Patients with iron deficiency (both absolute and functional) had a higher platelet count than those without iron deficiency (174 ± 61 × 109/L vs. 153 ± 58 × 109/L, P < .001). Receiver operating characteristic analysis showed a weak predictive power of platelet count on absolute iron deficiency (area under curve 0.620; cutoff value > 137 × 109/L, sensitivity 76%, specificity 43%) and functional iron deficiency (area under curve 0.540; cutoff value > 124 × 109/L, sensitivity 77%, specificity 32%). Platelet count was negatively correlated with ferritin (Spearman's rho [ρ] -0.1547, P < .001), transferrin saturation (ρ = -0.1895, P < .001), and serum iron (ρ = -0.1466, P < .001). The abovementioned correlations remained significant in multivariate regression (β -0.7285, 95% confidence interval [CI] -1.0757 to -0.3814; β -.00347, 95% CI -0.0520 to -0.0174; β -0.0097, 95% CI -0.0159 to -0.0035, respectively). In unadjusted and adjusted Cox regression models, neither baseline platelet count nor relative thrombocytosis was associated with 3-year mortality. CONCLUSION There was a weak but significant platelet count elevation in patients with ESRD and with iron deficiency. KEY WORDS end-stage renal disease; iron deficiency; iron supplement; mortality; platelet count.
Collapse
Affiliation(s)
- Ruoxi Liao
- Department of Nephrology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xueli Zhou
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Dengyan Ma
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jing Tang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hui Zhong
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
16
|
Borreda I, Zukermann R, Epstein D, Marcusohn E. IV Sodium Ferric Gluconate Complex in Patients Hospitalized Due to Acute Decompensated Heart Failure and Iron Deficiency. J Cardiovasc Pharmacol Ther 2022; 27:10742484211055639. [PMID: 34994220 DOI: 10.1177/10742484211055639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background: Patients suffering from heart failure (HF) and iron deficiency (ID) have worse outcomes. Treatment with intra-venous (IV) ferric carboxymaltose has been shown to reduce HF rehospitalizations and to improve functional capacity and symptoms in patients with HF and reduced ejection fraction (HFrEF). However, IV ferric carboxymaltose is significantly more expensive than IV sodium ferric gluconate complex (SFGC) limiting its availability to most HF patients around the globe. Methods: A retrospective analysis comparing patients admitted to internal medicine or cardiology departments between January 2013 to December 2018 due to acute decompensated HF (ADHF) and treated with or without IV SFGC on top of standard medical therapy. Results: During the study period, a total of 1863 patients were hospitalized due to ADHF with either HFrEF or HF with preserved ejection fraction (HFpEF). Among them, 840 patients had laboratory evidence of iron deficiency (absolute or functional) and met the inclusion criteria. One hundred twenty-two of them (14.5%) were treated with IV SFGC during the index hospitalization. Patients treated with IV iron were more likely to have history of ischemic heart disease, atrial fibrillation, and chronic kidney disease. The rate of readmissions due to ADHF was similar between the groups at 30 days, 3 months, and 1 year. Conclusion: High risk patient hospitalized to ADHF and treated with IV SFGC showed comparable ADHF readmission rates, compared to those who did not receive iron supplementation.
Collapse
Affiliation(s)
- Itay Borreda
- Internal Medicine H, 58878Rambam Health Care Campus, Haifa, Israel
| | - Robert Zukermann
- Intermediate Cardiac Care Unit, 58878Department of Cardiology, Rambam Health Care Campus, Haifa, Israel
| | - Danny Epstein
- Critical Care Division, 58878Rambam Health Care Campus, Haifa, Israel
| | - Erez Marcusohn
- 58878Department of Cardiology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
17
|
Park MY, Le Henaff C, Sitara D. Administration of α-Klotho Does Not Rescue Renal Anemia in Mice. Front Pediatr 2022; 10:924915. [PMID: 35813388 PMCID: PMC9259788 DOI: 10.3389/fped.2022.924915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/27/2022] [Indexed: 12/05/2022] Open
Abstract
Renal anemia is a common complication in chronic kidney disease (CKD), associated with decreased production of erythropoietin (EPO) due to loss of kidney function, and subsequent decreased red blood cell (RBC) production. However, many other factors play a critical role in the development of renal anemia, such as iron deficiency, inflammation, and elevated fibroblast growth factor 23 (FGF23) levels. We previously reported that inhibition of FGF23 signaling rescues anemia in mice with CKD. In the present study we sought to investigate whether α-Klotho deficiency present in CKD also contributes to the development of renal anemia. To address this, we administered α-Klotho to mice with CKD induced by an adenine-rich diet. Mice were sacrificed 24 h after α-Klotho injection, and blood and organs were collected immediately post-mortem. Our data show that α-Klotho administration had no beneficial effect in mice with CKD-associated anemia as it did not increase RBC numbers and hemoglobin levels, and it did not stimulate EPO secretion. Moreover, α-Klotho did not improve iron deficiency and inflammation in CKD as it had no effect on iron levels or inflammatory markers. Interestingly, Klotho supplementation significantly reduced the number of erythroid progenitors in the bone marrow and downregulated renal Epo and Hif2α mRNA in mice fed control diet resulting in reduced circulating EPO levels in these mice. In addition, Klotho significantly decreased intestinal absorption of iron in control mice leading to reduced serum iron and transferrin saturation levels. Our findings demonstrate that α-Klotho does not have a direct role in renal anemia and that FGF23 suppresses erythropoiesis in CKD via a Klotho-independent mechanism. However, in physiological conditions α-Klotho appears to have an inhibitory effect on erythropoiesis and iron regulation.
Collapse
Affiliation(s)
- Min Young Park
- Department of Molecular Pathobiology, NYU College of Dentistry, New York, NY, United States
| | - Carole Le Henaff
- Department of Molecular Pathobiology, NYU College of Dentistry, New York, NY, United States
| | - Despina Sitara
- Department of Molecular Pathobiology, NYU College of Dentistry, New York, NY, United States.,Medicine, NYU School of Medicine, New York, NY, United States
| |
Collapse
|
18
|
Majoni SW, Nelson J, Germaine D, Hoppo L, Long S, Divakaran S, Turner B, Graham J, Cherian S, Pawar B, Rathnayake G, Heron B, Maple-Brown L, Batey R, Morris P, Davies J, Fernandes DK, Sundaram M, Abeyaratne A, Wong YHS, Lawton PD, Taylor S, Barzi F, Cass A. INFERR-Iron infusion in haemodialysis study: INtravenous iron polymaltose for First Nations Australian patients with high FERRitin levels on haemodialysis-a protocol for a prospective open-label blinded endpoint randomised controlled trial. Trials 2021; 22:868. [PMID: 34857020 PMCID: PMC8641231 DOI: 10.1186/s13063-021-05854-w] [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] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/20/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The effectiveness of erythropoiesis-stimulating agents, which are the main stay of managing anaemia of chronic kidney disease (CKD), is largely dependent on adequate body iron stores. The iron stores are determined by the levels of serum ferritin concentration and transferrin saturation. These two surrogate markers of iron stores are used to guide iron replacement therapy. Most Aboriginal and/or Torres Islander Australians of the Northern Territory (herein respectfully referred to as First Nations Australians) with end-stage kidney disease have ferritin levels higher than current guideline recommendations for iron therapy. There is no clear evidence to guide safe and effective treatment with iron in these patients. We aim to assess the impact of intravenous iron treatment on all-cause death and hospitalisation with a principal diagnosis of all-cause infection in First Nations patients on haemodialysis with anaemia, high ferritin levels and low transferrin saturation METHODS: In a prospective open-label blinded endpoint randomised controlled trial, a total of 576 participants on maintenance haemodialysis with high ferritin (> 700 μg/L and ≤ 2000 μg/L) and low transferrin saturation (< 40%) from all the 7 renal units across the Northern Territory of Australia will be randomised 1:1 to receive intravenous iron polymaltose 400 mg once monthly (200 mg during 2 consecutive haemodialysis sessions) (Arm A) or no IV iron treatment (standard treatment) (Arm B). Rescue therapy will be administered when the ferritin levels fall below 700 μg/L or when clinically indicated. The primary outcome will be the differences between the two study arms in the risk of hospitalisation with all-cause infection or death. An economic analysis and several secondary and tertiary outcomes analyses will also be performed. DISCUSSION The INFERR clinical trial will address significant uncertainty on the safety and efficacy of iron therapy in First Nations Australians with CKD with hyperferritinaemia and evidence of iron deficiency. This will hopefully lead to the development of evidence-based guidelines. It will also provide the opportunity to explore the causes of hyperferritinaemia in First Nations Australians from the Northern Territory. TRIAL REGISTRATION This trial is registered with The Australian New Zealand Clinical Trials Registry (ANZCTR): ACTRN12620000705987 . Registered 29 June 2020.
Collapse
Affiliation(s)
- Sandawana William Majoni
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia.
- Department of Nephrology, Division of Medicine, Royal Darwin Hospital, P.O. Box 41326, Casuarina, Darwin, Northern Territory, Australia.
- Flinders University and Northern Territory Medical Program, Royal Darwin Hospital Campus, Darwin, Northern Territory, Australia.
| | - Jane Nelson
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Darren Germaine
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Libby Hoppo
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Stephanie Long
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Shilpa Divakaran
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Department of Nephrology, Division of Medicine, Royal Darwin Hospital, P.O. Box 41326, Casuarina, Darwin, Northern Territory, Australia
| | - Brandon Turner
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Jessica Graham
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Sajiv Cherian
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Flinders University and Northern Territory Medical Program, Royal Darwin Hospital Campus, Darwin, Northern Territory, Australia
- Department of Nephrology, Division of Medicine, Alice Springs Hospital, Alice Springs, Northern Territory, Australia
| | - Basant Pawar
- Department of Nephrology, Division of Medicine, Alice Springs Hospital, Alice Springs, Northern Territory, Australia
| | - Geetha Rathnayake
- Flinders University and Northern Territory Medical Program, Royal Darwin Hospital Campus, Darwin, Northern Territory, Australia
- Chemical Pathology-Territory Pathology, Department of Health, Northern Territory Government, Darwin, Northern Territory, Australia
| | - Bianca Heron
- Department of Nephrology, Division of Medicine, Royal Darwin Hospital, P.O. Box 41326, Casuarina, Darwin, Northern Territory, Australia
| | - Louise Maple-Brown
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Department of Endocrinology, Division of Medicine, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Robert Batey
- Department of Nephrology, Division of Medicine, Alice Springs Hospital, Alice Springs, Northern Territory, Australia
- New South Wales Health, St Leonards, NSW, Australia
| | - Peter Morris
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Department of Pediatrics, Division of Women, Children and Youth, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| | - Jane Davies
- Department of Infectious Diseases, Division of Medicine, Royal Darwin Hospital, Darwin, Northern Territory, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - David Kiran Fernandes
- Department of Nephrology, Division of Medicine, Alice Springs Hospital, Alice Springs, Northern Territory, Australia
| | - Madhivanan Sundaram
- Department of Nephrology, Division of Medicine, Royal Darwin Hospital, P.O. Box 41326, Casuarina, Darwin, Northern Territory, Australia
| | - Asanga Abeyaratne
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Department of Nephrology, Division of Medicine, Royal Darwin Hospital, P.O. Box 41326, Casuarina, Darwin, Northern Territory, Australia
- Flinders University and Northern Territory Medical Program, Royal Darwin Hospital Campus, Darwin, Northern Territory, Australia
| | - Yun Hui Sheryl Wong
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Department of Nephrology, Division of Medicine, Royal Darwin Hospital, P.O. Box 41326, Casuarina, Darwin, Northern Territory, Australia
| | - Paul D Lawton
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- The Central Clinical School, Monash University & Alfred Health, Melbourne, Australia
| | - Sean Taylor
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Department of Nephrology, Division of Medicine, Royal Darwin Hospital, P.O. Box 41326, Casuarina, Darwin, Northern Territory, Australia
| | - Federica Barzi
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- UQ Poche Centre for Indigenous Health, The University of Queensland, St Lucia, Queensland, 4067, Australia
| | - Alan Cass
- Division of Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| |
Collapse
|
19
|
Shah AA, Donovan K, Seeley C, Dickson EA, Palmer AJR, Doree C, Brunskill S, Reid J, Acheson AG, Sugavanam A, Litton E, Stanworth SJ. Risk of Infection Associated With Administration of Intravenous Iron: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e2133935. [PMID: 34767026 PMCID: PMC8590171 DOI: 10.1001/jamanetworkopen.2021.33935] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/13/2021] [Indexed: 12/22/2022] Open
Abstract
Importance Intravenous iron is recommended by many clinical guidelines based largely on its effectiveness in reducing anemia. However, the association with important safety outcomes, such as infection, remains uncertain. Objective To examine the risk of infection associated with intravenous iron compared with oral iron or no iron. Data Sources Medline, Embase, and Cochrane Central Register of Controlled Trials (CENTRAL) were searched for randomized clinical trials (RCTs) from 1966 to January 31, 2021. Ongoing trials were sought from ClinicalTrials.gov, CENTRAL, and the World Health Organization International Clinical Trials Search Registry Platform. Study Selection Pairs of reviewers identified RCTs that compared intravenous iron with oral iron or no iron across all patient populations, excluding healthy volunteers. Nonrandomized studies published since January 1, 2007, were also included. A total of 312 full-text articles were assessed for eligibility. Data Extraction and Synthesis Data extraction and risk of bias assessments were performed according to the Preferred Reporting Items of Systematic Reviews and Meta-analyses (PRISMA) and Cochrane recommendations, and the quality of evidence was assessed using the GRADE (Grades of Recommendation, Assessment, Development, and Evaluation) approach. Two reviewers extracted data independently. A random-effects model was used to synthesize data from RCTs. A narrative synthesis was performed to characterize the reporting of infection. Main Outcomes and Measures The primary outcome was risk of infection. Secondary outcomes included mortality, hospital length of stay, and changes in hemoglobin and red blood cell transfusion requirements. Measures of association were reported as risk ratios (RRs) or mean differences. Results A total of 154 RCTs (32 920 participants) were included in the main analysis. Intravenous iron was associated with an increased risk of infection when compared with oral iron or no iron (RR, 1.17; 95% CI, 1.04-1.31; I2 = 37%; moderate certainty of evidence). Intravenous iron also was associated with an increase in hemoglobin (mean difference, 0.57 g/dL; 95% CI, 0.50-0.64 g/dL; I2 = 94%) and a reduction in the risk of requiring a red blood cell transfusion (RR, 0.93; 95% CI, 0.76-0.89; I2 = 15%) when compared with oral iron or no iron. There was no evidence of an effect on mortality or hospital length of stay. Conclusions and Relevance In this large systematic review and meta-analysis, intravenous iron was associated with an increased risk of infection. Well-designed studies, using standardized definitions of infection, are required to understand the balance between this risk and the potential benefits.
Collapse
Affiliation(s)
- Akshay A. Shah
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Biomedical Research Centre Haematology Theme, Oxford, United Kingdom
- Adult Intensive Care Unit, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Killian Donovan
- Adult Intensive Care Unit, Oxford University Hospitals National Health Service (NHS) Foundation Trust, Oxford, United Kingdom
| | - Claire Seeley
- Department of Anaesthesia, Royal Berkshire Hospitals NHS Foundation Trust, Reading, United Kingdom
| | - Edward A. Dickson
- National Institute for Health Research Biomedical Research Centre in Gastrointestinal and Liver Diseases, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
- Department of Colorectal Surgery, Nottingham University Hospitals NHS Trust, University of Nottingham, Nottingham, United Kingdom
| | - Antony J. R. Palmer
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Carolyn Doree
- Systematic Review Initiative, NHS Blood & Transplant, Oxford, United Kingdom
| | - Susan Brunskill
- Systematic Review Initiative, NHS Blood & Transplant, Oxford, United Kingdom
| | - Jack Reid
- Department of Anaesthesia, Brighton and Sussex University Hospitals NHS Trust, Brighton, United Kingdom
| | - Austin G. Acheson
- National Institute for Health Research Biomedical Research Centre in Gastrointestinal and Liver Diseases, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
- Department of Colorectal Surgery, Nottingham University Hospitals NHS Trust, University of Nottingham, Nottingham, United Kingdom
| | - Anita Sugavanam
- Department of Anaesthesia, Brighton and Sussex University Hospitals NHS Trust, Brighton, United Kingdom
| | - Edward Litton
- Intensive Care Unit, Fiona Stanley Hospital, Perth, Australia
| | - Simon J. Stanworth
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research Biomedical Research Centre Haematology Theme, Oxford, United Kingdom
- Systematic Review Initiative, NHS Blood & Transplant, Oxford, United Kingdom
- Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
20
|
Fletcher A, Forbes A, Svenson N, Wayne Thomas D. Guideline for the laboratory diagnosis of iron deficiency in adults (excluding pregnancy) and children. Br J Haematol 2021; 196:523-529. [PMID: 34693519 DOI: 10.1111/bjh.17900] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 12/22/2022]
Affiliation(s)
- Andrew Fletcher
- Hull University Teaching Hospitals NHS Trust, Castle Hill Hospital, Cottingham, UK
| | - Adam Forbes
- Royal Cornwall Hospital NHS Trust, Truro, UK
| | - Nicola Svenson
- Hull University Teaching Hospitals NHS Trust, Hull Royal Infirmary, Hull, UK
| | | | | |
Collapse
|
21
|
Gutiérrez OM. Treatment of Iron Deficiency Anemia in CKD and End-Stage Kidney Disease. Kidney Int Rep 2021; 6:2261-2269. [PMID: 34514189 PMCID: PMC8418942 DOI: 10.1016/j.ekir.2021.05.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/04/2021] [Accepted: 05/17/2021] [Indexed: 01/27/2023] Open
Abstract
Iron deficiency is common in individuals with chronic kidney disease and plays a major role in the development of anemia. Oral and intravenous iron agents are both available to replete iron in patients with chronic kidney disease diagnosed with iron deficiency. The choice of which agent to use is most often dictated by goals of therapy, tolerability, convenience, and response to prior therapy. Diminished absorption of iron in the gastrointestinal tract and a high incidence of gastrointestinal adverse effects can reduce the efficacy of oral iron agents, necessitating the use of i.v. iron formulations to treat iron deficiency anemia, particularly in patients requiring kidney replacement therapy. Newer oral agents may help to overcome these limitations and help treat iron deficiency in those not requiring kidney replacement therapy. Recent studies have provided new evidence that more aggressive repletion of iron in patients with chronic kidney disease requiring kidney replacement therapy may provide benefits with respect to anemia management and hard clinical outcomes such as cardiovascular disease and survival.
Collapse
Affiliation(s)
- Orlando M. Gutiérrez
- Division of Nephrology, Department of Medicine and Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
22
|
Long-acting erythropoiesis-stimulating agent (ESA) induces physiological erythropoiesis via improvement of iron availability. Int Urol Nephrol 2021; 54:1079-1084. [PMID: 34390438 DOI: 10.1007/s11255-021-02965-w] [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: 03/14/2021] [Accepted: 07/25/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE Previous studies reported that the long-acting erythropoiesis-stimulating agent (ESA) significantly suppresses the expression of hepcidin, which regulates iron availability. In this study, we compared the iron availability for erythropoiesis between short and long-acting ESA over a long period. METHODS We enrolled 69 hemodialysis patients in this study. All patients were treated with short-acting ESA (epoetin-α or epoetin-β) for the first 30 months. Then, all patients switched to long-acting ESA (continuous erythropoietin receptor activator-methoxy polyethylene glycol-epoetin beta) for the next 30 months. We measured their blood levels of Hb, ferritin, iron, total iron-binding capacity, intact-parathyroid hormone, calcium, phosphate, albumin, and highly sensitive CRP level. RESULTS There was no significant change in the dose of short or long-acting ESA during the study period. Compared with the short-acting ESA period, the mean hemoglobin (Hb) and transferrin saturation levels were significantly increased in the long-acting ESA period (from 10.3 ± 0.2 to 10.6 ± 0.3 g/dL). On the other hand, the mean serum ferritin level (from 72 ± 22.2 to 56.3 ± 14 ng/mL) and the dose of IV iron (from 108 ± 63 to 53 ± 27 mg/month) were significantly decreased in the long-acting ESA period. CONCLUSION In this study, we found that anemia treatment with long-acting ESA attenuated the iron utilization for erythropoiesis and maintained target Hb levels without requiring a higher dose of IV iron or ESA.
Collapse
|
23
|
Coyne DW. Proactive High-Dose IV Iron Is Preferred Therapy in ESKD Patients: PRO. KIDNEY360 2021; 3:208-210. [PMID: 35373137 PMCID: PMC8967649 DOI: 10.34067/kid.0002442021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/23/2021] [Indexed: 01/10/2023]
Affiliation(s)
- Daniel W. Coyne
- Division of Nephrology, School of Medicine, Washington University, St. Louis, Missouri
| |
Collapse
|
24
|
Kawai Y, Toya Y, Wakui H, Fujikawa T, Ueda E, Azushima K, Kinguchi S, Mitsuhashi H, Kawano T, Kuji T, Yamaguchi S, Ohnishi T, Tamura K. Potential effective treatment of shortening continuous erythropoietin receptor activator treatment interval combined with iron supplementation in hemodialysis patients. J Pharmacol Sci 2021; 147:118-125. [PMID: 34294362 DOI: 10.1016/j.jphs.2021.05.010] [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: 02/22/2021] [Revised: 04/28/2021] [Accepted: 05/19/2021] [Indexed: 11/29/2022] Open
Abstract
Our previous randomized controlled trial comparing the total dose of weekly versus biweekly continuous erythropoietin receptor activator (CERA) therapy to maintain optimal hemoglobin (Hb) levels showed no significant differences between the two therapies. This post-hoc analysis assessed whether the total dose of weekly versus biweekly CERA therapy to maintain Hb levels among HD patients differed among groups with or without iron supplementation. Of 107 patients, 40 received intravenous iron supplementation due to iron deficiency (iron group) and 67 did not (non-iron group). In the iron group, the weekly therapy tended to require a lower total CERA dose compared with the biweekly therapy (274 ± 274 vs 381 ± 223 μg/12 weeks, P = 0.051). Changes in circulating hepcidin levels, a negative regulator of intestinal iron uptake, after 2 weeks of CERA treatment were significantly lower in the weekly therapy compared with the biweekly therapy (-4.2 ± 6.3 vs 11.1 ± 7.3 ng/mL, P = 0.015). In the non-iron group, there were no significant differences in total CERA dose or changes in hepcidin levels between the two therapies. Shortening the CERA treatment interval combined with iron supplementation may lead to the more efficient treatment of HD patients with iron deficiency.
Collapse
Affiliation(s)
- Yuki Kawai
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Yoshiyuki Toya
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Hiromichi Wakui
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| | - Tetsuya Fujikawa
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan; Center for Health Service Sciences, Yokohama National University, 79-8 Tokiwadai, Hodogaya-ku, Yokohama, 240-8501, Japan
| | - Eiko Ueda
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Kengo Azushima
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Sho Kinguchi
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Hiroshi Mitsuhashi
- Kohsaikai Kamioooka Jinsei Clinic, 1-10-1 Kamioookanishi, Konan-ku, Yokohama, 233-0002, Japan
| | - Tomoyuki Kawano
- Kohsaikai Bunkojin Clinic, 356-3 Yatsucho, Kanazawa-ku, Yokohama, 236-0016, Japan
| | - Tadashi Kuji
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan; Yokodai Central Clinic, 3-9-31 Yokodai, Isogo-ku, Yokohama, 235-0045, Japan
| | - Satoshi Yamaguchi
- Kohsaikai Yokohama Jinsei Hospital, 3-1-28 Konan, Konan-ku, Yokohama, 233-0003, Japan
| | - Toshimasa Ohnishi
- Kohsaikai Kamioooka Jinsei Clinic, 1-10-1 Kamioookanishi, Konan-ku, Yokohama, 233-0002, Japan
| | - Kouichi Tamura
- Department of Medical Science and Cardiorenal Medicine, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| |
Collapse
|
25
|
Savković M, Simić-Ogrizović S, Dopsaj V. Assessment of positive iron balance in end-stage renal disease: Could hepcidin-25 be useful? Int J Lab Hematol 2021; 43:1159-1167. [PMID: 33835732 DOI: 10.1111/ijlh.13539] [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: 01/23/2021] [Revised: 03/18/2021] [Accepted: 03/23/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION The aim of our study was to examine the relationship of hepcidin-25 with red blood cell and reticulocyte indices and to evaluate the diagnostic properties of hepcidin-25 in the assessment of positive iron balance in end-stage renal disease (ESRD) patients. METHODS Eighty anemic ESRD patients (hemoglobin < 110 g/L) were classified as having iron deficiency (ID, N = 20), iron sufficiency (IS, N = 29), and positive iron balance (PB, N = 31) using the conventional biomarkers for iron status evaluation. Hepcidin-25 was determined by a chemiluminescent direct ELISA. RESULTS Hepcidin-25 was significantly negatively correlated with the proportion of hypochromic erythrocytes (%HYPO) (P = .034) and immature reticulocyte fraction (P = .010) in ID and with the absolute reticulocyte concentration in ID (P = .048) and PB (P = .040). In multivariate models, hepcidin-25 was independently negatively associated with the mean reticulocyte hemoglobin content (CHr; β = -0.493, P = .004) and red blood cell size factor (RSf) (β = -0.334, P = .036) only in the PB group. The best hepcidin-25 value to exclude PB was 66.13 µg/L, showing a sensitivity of 61.3%, a specificity of 75.5%, and an AUC of 0.808. CONCLUSION Our results suggest that hepcidin-25 levels are independently negatively associated with the iron demand for the most recent erythropoiesis only in PB. Hepcidin-25 performed acceptable in discriminating anemic ESRD patients with positive iron balance and may prove to be a useful additional tool in the evaluation of iron status.
Collapse
Affiliation(s)
- Miljan Savković
- Center for Medical Biochemistry, Clinical Center of Serbia, Belgrade, Serbia
| | - Sanja Simić-Ogrizović
- General Hospital Medigroup, Belgrade, Serbia.,Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Violeta Dopsaj
- Center for Medical Biochemistry, Clinical Center of Serbia, Belgrade, Serbia.,Department of Medical Biochemistry, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| |
Collapse
|
26
|
Del Vecchio L, Ekart R, Ferro CJ, Malyszko J, Mark PB, Ortiz A, Sarafidis P, Valdivielso JM, Mallamaci F. Intravenous iron therapy and the cardiovascular system: risks and benefits. Clin Kidney J 2021; 14:1067-1076. [PMID: 34188903 PMCID: PMC8223589 DOI: 10.1093/ckj/sfaa212] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Anaemia is a common complication of chronic kidney disease (CKD). In this setting, iron deficiency is frequent because of the combination of increased iron needs to sustain erythropoiesis with increased iron losses. Over the years, evidence has accumulated on the involvement of iron in influencing pulmonary vascular resistance, endothelial function, atherosclerosis progression and infection risk. For decades, iron therapy has been the mainstay of therapy for renal anaemia together with erythropoiesis-stimulating agents (ESAs). Despite its long-standing use, grey areas still surround the use of iron therapy in CKD. In particular, the right balance between either iron repletion with adequate therapy and the avoidance of iron overload and its possible negative effects is still a matter of debate. This is particularly true in patients having functional iron deficiency. The recent Proactive IV Iron Therapy in Haemodialysis Patients trial supports the use of intravenous (IV) iron therapy until a ferritin upper limit of 700 ng/mL is reached in haemodialysis patients on ESA therapy, with short dialysis vintage and minimal signs of inflammation. IV iron therapy has also been proven to be effective in the setting of heart failure (HF), where it improves exercise capacity and quality of life and possibly reduces the risk of HF hospitalizations and cardiovascular deaths. In this review we discuss the risks of functional iron deficiency and the possible benefits and risks of iron therapy for the cardiovascular system in the light of old and new evidence.
Collapse
Affiliation(s)
- Lucia Del Vecchio
- Department of Nephrology and Dialysis, Sant’Anna Hospital, ASST Lariana, Como, Italy
| | - Robert Ekart
- Department of Dialysis, Clinic for Internal Medicine, University Clinical Center Maribor, Maribor, Slovenia
| | - Charles J Ferro
- Renal Unit, University Hospitals Birmingham and Institute of Cardiovascular Science, University of Birmingham, Birmingham, UK
| | - Jolanta Malyszko
- Department of Nephrology, Dialysis and Internal Disease, Medical University of Warsaw, Warsaw, Poland
| | - Patrick B Mark
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
| | - Pantelis Sarafidis
- Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Jose M Valdivielso
- Vascular and Renal Translational Research Group and UDETMA, Institut de Recerca Biomèdica de Lleida, Lleida, Spain
| | - Francesca Mallamaci
- CNR-IFC Clinical Epidemiology of Renal Diseases and Hypertension, Reggio Calabria, Italy
| | | |
Collapse
|
27
|
Majoni SW, Lawton PD, Rathnayake G, Barzi F, Hughes JT, Cass A. Narrative Review of Hyperferritinemia, Iron Deficiency, and the Challenges of Managing Anemia in Aboriginal and Torres Strait Islander Australians With CKD. Kidney Int Rep 2021; 6:501-512. [PMID: 33615076 PMCID: PMC7879094 DOI: 10.1016/j.ekir.2020.10.035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022] Open
Abstract
Aboriginal and Torres Strait Islander Australians (Indigenous Australians) suffer some of the highest rates of chronic kidney disease (CKD) in the world. Among Indigenous Australians in remote areas of the Northern Territory, prevalence rates for renal replacement therapy (RRT) are up to 30 times higher than national prevalence. Anemia among patients with CKD is a common complication. Iron deficiency is one of the major causes. Iron deficiency is also one of the key causes of poor response to the mainstay of anemia therapy with erythropoiesis-stimulating agents (ESAs). Therefore, the effective management of anemia in people with CKD is largely dependent on effective identification and correction of iron deficiency. The current identification of iron deficiency in routine clinical practice is dependent on 2 surrogate markers of iron status: serum ferritin concentration and transferrin saturation (TSAT). However, questions exist regarding the use of serum ferritin concentration in people with CKD because it is an acute-phase reactant that can be raised in the context of acute and chronic inflammation. Serum ferritin concentration among Indigenous Australians receiving RRT is often markedly elevated and falls outside reference ranges within most national and international guidelines for iron therapy for people with CKD. This review explores published data on the challenges of managing anemia in Indigenous people with CKD and the need for future research on the efficacy and safety of treatment of anemia of CKD in patients with high ferritin and evidence iron deficiency.
Collapse
Affiliation(s)
- Sandawana William Majoni
- Department of Nephrology, Division of Medicine, Royal Darwin Hospital, Darwin, Northern Territory, Australia
- Flinders University and Northern Territory Medical Program, Royal Darwin Hospital Campus, Darwin, Northern Territory, Australia
- Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Northern Territory, Australia
| | - Paul D. Lawton
- Department of Nephrology, Division of Medicine, Royal Darwin Hospital, Darwin, Northern Territory, Australia
- Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Northern Territory, Australia
| | - Geetha Rathnayake
- Flinders University and Northern Territory Medical Program, Royal Darwin Hospital Campus, Darwin, Northern Territory, Australia
- Chemical Pathology–Territory Pathology, Department of Health, Northern Territory Government, Northern Territory, Australia
| | - Federica Barzi
- Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Northern Territory, Australia
| | - Jaquelyne T. Hughes
- Department of Nephrology, Division of Medicine, Royal Darwin Hospital, Darwin, Northern Territory, Australia
- Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Northern Territory, Australia
| | - Alan Cass
- Wellbeing and Preventable Chronic Diseases, Menzies School of Health Research, Charles Darwin University, Northern Territory, Australia
| |
Collapse
|
28
|
Lee KH, Ho Y, Tarng DC. Iron Therapy in Chronic Kidney Disease: Days of Future Past. Int J Mol Sci 2021; 22:1008. [PMID: 33498292 PMCID: PMC7863960 DOI: 10.3390/ijms22031008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Anemia affects millions of patients with chronic kidney disease (CKD) and prompt iron supplementation can lead to reductions in the required dose of erythropoiesis-stimulating agents, thereby reducing medical costs. Oral and intravenous (IV) traditional iron preparations are considered far from ideal, primarily due to gastrointestinal intolerability and the potential risk of infusion reactions, respectively. Fortunately, the emergence of novel iron replacement therapies has engendered a paradigm shift in the treatment of iron deficiency anemia in patients with CKD. For example, oral ferric citrate is an efficacious and safe phosphate binder that increases iron stores to maintain hemoglobin levels. Additional benefits include reductions in fibroblast growth factor 23 levels and the activation of 1,25 dihydroxyvitamin D. The new-generation IV iron preparations ferumoxytol, iron isomaltoside 1000, and ferric carboxymaltose are characterized by a reduced risk of infusion reactions and are clinically well tolerated as a rapid high-dose infusion. In patients undergoing hemodialysis (HD), ferric pyrophosphate citrate (FPC) administered through dialysate enables the replacement of ongoing uremic and HD-related iron loss. FPC transports iron directly to transferrin, bypassing the reticuloendothelial system and avoiding iron sequestration. Moreover, this paper summarizes recent advancements of hypoxia-inducible factor prolyl hydroxylase inhibitors and future perspectives in renal anemia management.
Collapse
Affiliation(s)
- Kuo-Hua Lee
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (K.-H.L.); (Y.H.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11217, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), Hsinchu 300, Taiwan
| | - Yang Ho
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (K.-H.L.); (Y.H.)
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (K.-H.L.); (Y.H.)
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11217, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDSB), Hsinchu 300, Taiwan
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu 300, Taiwan
- Department and Institute of Physiology, National Yang-Ming University, Taipei 11217, Taiwan
| |
Collapse
|
29
|
Savković M, Simić-Ogrizović S, Dopsaj V. Factors associated with hepcidin-25 levels in maintenance hemodialysis patients. Ther Apher Dial 2021; 25:565-574. [PMID: 33403761 DOI: 10.1111/1744-9987.13617] [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: 08/09/2020] [Revised: 11/25/2020] [Accepted: 12/22/2020] [Indexed: 12/01/2022]
Abstract
This study aimed to investigate the factors that are independently associated with hepcidin-25 and its relationship with doses of erythropoiesis-stimulating agents (ESAs) and intravenous iron in stable maintenance hemodialysis patients (smHD) stratified by ESAs administration. In 103 adult smHD (ESAs therapy (N = 64) and ESAs-free (N = 39)), median values of biologically active hepcidin-25 (chemiluminescent direct ELISA assay) and ferritin levels were significantly higher whereas red blood cell count, hemoglobin, and hematocrit values were lower in ESAs therapy compared to ESAs-free group (P < .001, for all). Our results suggest that ESAs-independent smHD exhibit supposedly normal hepcidin-25 levels and preserved iron homeostasis, with a lower degree of anemia. The results of our multivariable model indicate that hepcidin-25 levels are independently and positively associated with iron stores and inflammation, and inversely with active erythropoiesis, regardless of ESAs administration. Maintenance ESAs and the intravenous iron dose were not related to hepcidin-25 levels.
Collapse
Affiliation(s)
- Miljan Savković
- Center of Medical Biochemistry, Clinical Center of Serbia, Belgrade, Serbia
| | - Sanja Simić-Ogrizović
- General Hospital Medigroup, Belgrade, Serbia.,Faculty of Medicine, University of Banja Luka, Banja Luka, Bosnia and Herzegovina
| | - Violeta Dopsaj
- Center of Medical Biochemistry, Clinical Center of Serbia, Belgrade, Serbia.,Department of Medical Biochemistry, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| |
Collapse
|
30
|
Pergola PE, Devalaraja M, Fishbane S, Chonchol M, Mathur VS, Smith MT, Lo L, Herzog K, Kakkar R, Davidson MH. Ziltivekimab for Treatment of Anemia of Inflammation in Patients on Hemodialysis: Results from a Phase 1/2 Multicenter, Randomized, Double-Blind, Placebo-Controlled Trial. J Am Soc Nephrol 2021; 32:211-222. [PMID: 33272965 PMCID: PMC7894678 DOI: 10.1681/asn.2020050595] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/14/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Patients with CKD who are on hemodialysis are hyporesponsive to erythropoiesis-stimulating agents (ESAs) because of anemia of inflammation. Interleukin-6 (IL-6) induced hepcidin expression is a key mediator of such inflammation. METHODS This phase 1/2, placebo-controlled trial assessed effects of ziltivekimab, a novel anti-IL-6 ligand antibody, in patients on hemodialysis with rs855791, a single nucleotide polymorphism of the TMPRSS6 gene that is hypothesized to heighten susceptibility to IL-6-mediated inflammatory effects. After a screening period documenting stable ESA and iron dosing, we randomized 61 patients with elevated IL-6 (≥4 pg/ml) to receive placebo or ziltivekimab (doses of 2, 6, or 20 mg), administered intravenously every 2 weeks for 12 weeks during hemodialysis. ESA dose adjustments were allowed after 4 weeks. We analyzed safety and effects on inflammation, iron metabolism, serum albumin, and anti-drug antibodies. RESULTS No patient experienced dose-limiting toxicity. Four patients (two each in the 6- and 20-mg cohorts) died of a treatment-emergent adverse event. Compared with patients receiving placebo, those receiving ziltivekimab experienced significantly greater reductions of high-sensitivity C-reactive protein, serum amyloid A, and fibrinogen from baseline to end of treatment. Median ESA usage decreased by 15,000, 15,000, or 33,000 IU/wk per patient in the 2-, 6-, and 20-mg ziltivekimab cohorts, respectively, compared with no change in the placebo group. We also noted significant dose responses for decreased ESA resistance index and increased serum iron, total iron binding capacity, transferrin saturation, and serum albumin. CONCLUSIONS Ziltivekimab significantly improved markers of inflammation, reduced ESA requirements, and increased serum albumin in patients on hemodialysis with inflammation and hyporesponsiveness to ESA therapy. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER Study to Assess the Safety, Pharmacokinetics, and Pharmacodynamics of Multiple Doses of COR-001, NCT02868229.
Collapse
Affiliation(s)
| | | | - Steven Fishbane
- Division of Nephrology, Department of Medicine, Donald and Barbara Zucker School of Medicine, Great Neck, New York
| | - Michel Chonchol
- Division of Renal Medicine Disease and Hypertension, University of Colorado, Aurora, Colorado
| | | | | | - Larry Lo
- Corvidia Therapeutics, Inc., Waltham, Massachusetts
| | - Kurt Herzog
- Corvidia Therapeutics, Inc., Waltham, Massachusetts
| | - Rahul Kakkar
- Pandion Therapeutics, Inc., Waltham, Massachusetts
| | | |
Collapse
|
31
|
Kassianides X, Hazara AM, Bhandari S. Improving the safety of intravenous iron treatments for patients with chronic kidney disease. Expert Opin Drug Saf 2020; 20:23-35. [PMID: 33203251 DOI: 10.1080/14740338.2021.1853098] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Iron-deficiency anemia in chronic kidney disease (CKD) is common and has prognostic, financial, and quality of life implications. Intravenous (IV) iron is a key intervention for optimal management, however, ongoing safety concerns exist. Area covered: The potential side effects associated with IV iron use are addressed as we review the most recent studies. Hypersensitivity reactions and true anaphylaxis are indeed rare with a greater understanding of the nature of labile iron and 'Fishbane' reactions. Hypophosphatemia appears commoner with certain IV iron preparations, however its significance in CKD requires exploration. The long-standing questions regarding oxidative stress and the potential susceptibility to infections and worsening cardiovascular morbidity are discussed. Iron overload secondary to repeat IV iron infusions is plausible, however, a number of guidelines limit and strictly guide prescription. Expert opinion: The past decade has improved our understanding of IV iron administration safety in patients with CKD. Third generation IV iron compounds have minimized hypersensitivity reactions while allowing high doses to be administered safely and rapidly in non-dialysis-dependent CKD patients. However, differences in safety profiles such as hypophosphatemia require further study and therapy should be tailored to the individual. Clinicians should feel confident in using IV iron therapy.
Collapse
Affiliation(s)
- Xenophon Kassianides
- Academic Renal Medicine, Hull University Teaching Hospitals NHS Trust , Kingston upon Hull, UK
| | - Adil Mohammad Hazara
- Academic Renal Medicine, Hull University Teaching Hospitals NHS Trust , Kingston upon Hull, UK
| | - Sunil Bhandari
- Academic Renal Medicine, Hull University Teaching Hospitals NHS Trust , Kingston upon Hull, UK
| |
Collapse
|
32
|
Karaboyas A, Morgenstern H, Fleischer NL, Schaubel DE, Robinson BM. Replicating Randomized Trial Results with Observational Data Using the Parametric g-Formula: An Application to Intravenous Iron Treatment in Hemodialysis Patients. Clin Epidemiol 2020; 12:1249-1260. [PMID: 33204166 PMCID: PMC7667704 DOI: 10.2147/clep.s283321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/27/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Reproducibility of clinical and epidemiologic research is important to generalize findings and has increasingly been scrutinized. A recently published randomized trial, PIVOTAL, evaluated high vs low intravenous iron dosing strategies to manage anemia in hemodialysis patients in the UK. Our objective was to assess the reproducibility of the PIVOTAL trial findings using data from a well-established cohort study, the Dialysis Outcomes and Practice Patterns Study (DOPPS). METHODS To overcome the absence of randomization in the DOPPS, we applied the parametric g-formula, an extension of standardization to longitudinal data. We estimated the effect of a proactive high-dose vs reactive low-dose iron supplementation strategy on all-cause mortality (primary outcome), hemoglobin, two measures of iron concentration (ferritin and TSAT), and erythropoiesis-stimulating agent dose over 12 months of follow-up in 6325 DOPPS patients. RESULTS Comparing high- vs low-iron dose strategies, the 1-year mortality risk difference was 0.020 (95% CI: 0.008, 0.031) and risk ratio was 1.20 (95% CI: 1.07, 1.33), compared with null 1-year findings in the PIVOTAL trial. Differences in secondary outcomes were directionally consistent but of lesser magnitude than in the PIVOTAL trial. CONCLUSION Our findings are somewhat consistent with the recent PIVOTAL trial, with discrepancies potentially attributable to model misspecification and differences between the two study populations. In addition to the importance of our results to nephrologists and hence hemodialysis patients, our analysis illustrates the utility of the parametric g-formula for generalizing results and comparing complex and dynamic treatment strategies using observational data.
Collapse
Affiliation(s)
- Angelo Karaboyas
- Arbor Research Collaborative for Health, Ann Arbor, MI, USA
- University of Michigan, Department of Epidemiology, Ann Arbor, MI, USA
| | - Hal Morgenstern
- University of Michigan, Departments of Epidemiology and Environmental Health Sciences, School of Public Health, and Department of Urology, Medical School, Ann Arbor, MI, USA
| | - Nancy L Fleischer
- University of Michigan, Department of Epidemiology, Ann Arbor, MI, USA
| | - Douglas E Schaubel
- University of Michigan, Department of Biostatistics, Ann Arbor, MI, USA
- University of Pennsylvania, Department of Biostatistics, Epidemiology and Informatics, Philadelphia, PA, USA
| | - Bruce M Robinson
- Arbor Research Collaborative for Health, Ann Arbor, MI, USA
- University of Michigan, Department of Internal Medicine, Ann Arbor, MI, USA
| |
Collapse
|
33
|
Evans M, Bower H, Cockburn E, Jacobson SH, Barany P, Carrero JJ. Contemporary management of anaemia, erythropoietin resistance and cardiovascular risk in patients with advanced chronic kidney disease: a nationwide analysis. Clin Kidney J 2020; 13:821-827. [PMID: 33123358 PMCID: PMC7577763 DOI: 10.1093/ckj/sfaa054] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 03/23/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Optimal management of chronic kidney disease (CKD) anaemia remains controversial and few studies have evaluated real-world management of anaemia in advanced CKD in the context of guideline recommendations. METHODS We performed an observational study from the Swedish Renal Registry evaluating the epidemiology and treatment patterns of anaemia across Stages 3b-5 in non-dialysis (ND) and dialysis-dependent (DD) CKD patients during 2015. Logistic regression and Cox models explored the associations between anaemia treatments, inflammation, erythropoietin resistance index (ERI) and subsequent 1-year risk of major adverse cardiovascular events (MACEs). RESULTS Data from 14 415 (ND, 11 370; DD, 3045) patients were included. Anaemia occurred in 60% of ND and 93% of DD patients. DD patients used more erythropoiesis-stimulating agents (ESAs; 82% versus 24%) and iron (62% versus 21%) than ND patients. All weekly ESA doses were converted to a weight-adjusted weekly epoetin equivalent dose. The prescribed ESA doses were low to moderate [median 48.2 IU/kg/week (ND), 78.6 IU/kg/week (DD)]. Among ESA-treated patients, 6-21% had haemoglobin (Hb) >13 g/dL and 2-6% had Hb <9 g/dL. Inflammation (C-reactive protein >5 mg/L) was highly prevalent and associated with ERI and higher ESA doses. Higher (>88 IU/kg/week) versus lower (<44 IU/kg/week) ESA doses were associated with a higher risk of MACEs [{ND hazard ratio [HR] 1.36 [95% confidence interval (CI) 1.00-1.86]; DD HR 1.60 [95% CI 1.24-2.06]}. There was no association between iron use and inflammation or MACEs. CONCLUSIONS Anaemia remains highly prevalent in advanced CKD. Patients with anaemia received moderate ESA doses with a relatively low prevalence of iron use. Higher doses of ESA were associated with inflammation and a higher risk of MACE.
Collapse
Affiliation(s)
- Marie Evans
- Department of Renal Medicine, Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Hannah Bower
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Elinor Cockburn
- Medical Affairs, Nordic Operations, Astellas Pharma A/S, Kastrup, Denmark
| | - Stefan H Jacobson
- Department of Clinical Sciences, Division of Nephrology, Danderyd University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Peter Barany
- Department of Renal Medicine, Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Pediatric Nephrology, Karolinska University Hospital, Stockholm, Sweden
| | - Juan-Jesus Carrero
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Punj S, Ghafourian K, Ardehali H. Iron deficiency and supplementation in heart failure and chronic kidney disease. Mol Aspects Med 2020; 75:100873. [PMID: 32753256 DOI: 10.1016/j.mam.2020.100873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/26/2020] [Accepted: 07/02/2020] [Indexed: 12/16/2022]
Abstract
Iron is a key element for normal cellular function and plays a role in many cellular processes including mitochondrial respiration. The role of iron deficiency (ID) in heart failure (HF) has been a subject of debate amid increasing advocacy for intravenous (IV) supplementation. Both the definition and the approach to treatment of ID in HF have been adapted from the experience in patients with chronic kidney disease (CKD). In this review, we highlight the differences in regulatory mechanisms as well as pathophysiology of ID in CKD and HF population both at the systemic and cellular levels. We will review the major clinical trials in HF patients that have shown symptomatic benefit from IV iron supplementation but without effect on clinical outcomes. Intravenous iron loading bypasses the mechanisms that tightly regulate iron uptake and can potentially cause myocardial and endothelial damage by releasing reactive oxygen species. By contrast, newer oral iron preparations do not have similar toxicity concerns and might have a role in heart failure.
Collapse
Affiliation(s)
- Shweta Punj
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Hossein Ardehali
- Department of Medicine, Northwestern University, Chicago, IL, USA; Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
35
|
Susantitaphong P, Siribumrungwong M, Takkavatakarn K, Chongthanakorn K, Lieusuwan S, Katavetin P, Tiranathanagul K, Lekhyananda S, Tungsanga K, Vanichakarn S, Eiam-Ong S, Praditpornsilpa K. Effect of Maintenance Intravenous Iron Treatment on Erythropoietin Dose in Chronic Hemodialysis Patients: A Multicenter Randomized Controlled Trial. Can J Kidney Health Dis 2020; 7:2054358120933397. [PMID: 32612843 PMCID: PMC7307402 DOI: 10.1177/2054358120933397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 04/07/2020] [Indexed: 11/17/2022] Open
Abstract
Background: There is no consensus on intravenous (IV) iron supplement dose, schedule, and
serum ferritin target in functional iron deficiency anemia to maintain
optimum target levels of iron stores by several guidelines. Objective: To examine the effect of IV iron supplementation to different targets of
serum ferritin on erythropoietin dose and inflammatory markers in chronic
hemodialysis (HD) patients with functional iron deficiency anemia. Design: A multicenter, randomized, open-label study. Setting: In a developing country, Thailand. Patients: Chronic HD patients with functional iron deficiency anemia. Measurements: Erythropoietin resistance index, high-sensitivity C-reactive protein, and
fibroblast growth factor 23. Methods: Two hundred adult chronic HD patients with transferrin saturation less than
30% and serum ferritin of 200 to 400 ng/mL were randomized 1:1 to maintain
serum ferritin 200 to 400 ng/mL (low-serum ferritin group, N = 100) or 600
to 700 ng/mL (high-serum ferritin group, N = 100). During a 6-week titration
period, participants randomized to the high-serum ferritin group initially
received 600 mg IV iron (100 mg every week), while the participants in the
low-serum ferritin group did not receive IV iron. During the 6-month
follow-up period, the dose of IV iron was adjusted by protocol. Results: The mean dose of IV iron was 108.3 ± 28.2 mg/month in the low-serum ferritin
group and 192.3 ± 36.2 mg/month in the high-serum ferritin group. The mean
serum ferritin was 367.0 ± 224.9 ng/mL in the low ferritin group and 619.6 ±
265.2 ng/mL in the high ferritin group. The erythropoietin resistance index
was significantly decreased in the high-serum ferritin group compared to the
low-serum ferritin group after receiving IV iron in the 6-week titration
period (mean difference: −113.43 ± 189.14 vs 41.08 ± 207.38 unit/week/g/dL;
P < .001) and 3-month follow-up period (mean
differences: −88.88 ± 234.43 vs −10.48 ± 217.75 unit/week/g/dL;
P = .02). Limitations: Short follow-up period. Conclusion: Maintaining a serum ferritin level of 600 to 700 ng/mL by IV iron
administration of approximately 200 mg per month as a maintenance protocol
can decrease erythropoietin dose requirements in chronic HD patients with
functional iron deficiency anemia. Trials registration: The study was registered with the Thai Clinical Trials Registry
TCTR20180903003.
Collapse
Affiliation(s)
- Paweena Susantitaphong
- Division of Nephrology, Department of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok, Thailand.,Research Unit for Metabolic Bone Disease in CKD Patients, Faculty of Medicine, Chulalongkorn University, Thailand
| | - Monchai Siribumrungwong
- Nephrology Unit, Department of Medicine, Lerdsin Hospital, College of Medicine, Rangsit University, Bangkok, Thailand
| | - Kullaya Takkavatakarn
- Division of Nephrology, Department of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | | | | | - Pisut Katavetin
- Division of Nephrology, Department of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Khajohn Tiranathanagul
- Division of Nephrology, Department of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | | | - Kriang Tungsanga
- Division of Nephrology, Department of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | | | - Somchai Eiam-Ong
- Division of Nephrology, Department of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Kearkiat Praditpornsilpa
- Division of Nephrology, Department of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| |
Collapse
|
36
|
Lobo B, Abdel-Rahman E, Brown D, Dunn L, Bowman B. A recurrent neural network approach to predicting hemoglobin trajectories in patients with End-Stage Renal Disease. Artif Intell Med 2020; 104:101823. [PMID: 32499002 DOI: 10.1016/j.artmed.2020.101823] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/30/2019] [Accepted: 02/17/2020] [Indexed: 12/20/2022]
Abstract
The most severe form of kidney disease, End-Stage Renal Disease (ESRD) is treated with various forms of dialysis - artificial blood cleansing. Dialysis patients suffer many health burdens including high mortality and hospitalization rates, and symptomatic anemia: a low red blood cell count as indicated by a low hemoglobin (Hgb) level. ESRD-induced anemia is treated, with variable patient response, by erythropoiesis stimulating agents (ESAs): expensive injectable medications typically administered during dialysis sessions. The dosing protocol is typically a population level protocol based on original clinical trials, the use of which often results in Hgb cycling. This cycling phenomenon occurs primarily due to the mismatch in the time between dosing decisions and the time it takes for the effects of a dosing change to be fully realized. In this paper we develop a recurrent neural network approach that uses historic data together with future ESA and iron dosing data to predict the 1, 2, and 3 month Hgb levels of patients with ESRD-induced anemia. The results of extensive experimentation indicate that this approach generates predictions that are clinically relevant: the mean absolute error of the predictions is comparable to estimates of the intra-individual variability of the laboratory test for Hgb.
Collapse
Affiliation(s)
- Benjamin Lobo
- Department of Systems & Information Engineering, University of Virginia, Charlottesville, VA 22904, United States
| | - Emaad Abdel-Rahman
- Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA 22908, United States
| | - Donald Brown
- Department of Systems & Information Engineering, University of Virginia, Charlottesville, VA 22904, United States
| | - Lori Dunn
- Medical Center - Pharmacy, University of Virginia, Charlottesville, VA 22908, United States
| | - Brendan Bowman
- Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA 22908, United States
| |
Collapse
|
37
|
Low-Dose Maintenance Intravenous Iron Therapy Can Prevent Anemia in Children with End-Stage Renal Disease Undergoing Chronic Hemodialysis. Int J Nephrol 2020; 2020:3067453. [PMID: 32566294 PMCID: PMC7284959 DOI: 10.1155/2020/3067453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 04/16/2020] [Indexed: 01/16/2023] Open
Abstract
Iron deficiency anemia is common in children with end-stage renal disease (ESRD) on long-term hemodialysis receiving erythropoiesis-stimulating agents. One approach to maintain the iron profile and hemoglobin levels is maintenance therapy with regular low doses of intravenous (IV) iron after initial iron repletion therapy; however, evidence for the benefits of this approach is lacking. This study evaluated the effect of IV iron maintenance therapy on anemia in children on regular hemodialysis. This retrospective cohort study included 41 pediatric ESRD patients with normal hemoglobin and iron status who underwent regular hemodialysis at the Pediatric Dialysis Unit of Cipto Mangunkusumo Hospital, Indonesia, between January 2015 and April 2019. Among these, 21 received IV iron maintenance therapy with two doses of 2 mg/kg of IV iron sucrose every 2 weeks (the treatment group) and 20 did not (the comparison group). Changes in hemoglobin and transferrin saturation were assessed after 6 weeks of observation and compared between the two groups. There was a significant reduction in the mean hemoglobin level compared with the baseline level in the comparison group (21 g/L; 95% CI, 9.3–33 g/L; p=0.001) but not in the treatment group (0.7 g/L; 95% CI, −6.6–8 g/L; p=0.84). The risk of anemia was lower in the treatment group (relative risk = 0.42; 95% CI, 0.22–0.79; p=0.003). Although majority of the patients had high baseline ferritin level, this study indicates that in our setting, ferritin may not be a reliable parameter to review the iron status, as it can be affected by chronic inflammation. Hence, the decision to start IV iron maintenance therapy in patients with hyperferritinemia should consider the patient's clinical condition and morbidity. To conclude, the coadministration of IV iron maintenance therapy is beneficial for maintaining hemoglobin levels and preventing anemia in children with ESRD who are undergoing regular hemodialysis, have achieved the target hemoglobin levels, and have normal iron status.
Collapse
|
38
|
The Impact of Iron Supplementation for Treating Anemia in Patients with Chronic Kidney Disease: Results from Pairwise and Network Meta-Analyses of Randomized Controlled Trials. Pharmaceuticals (Basel) 2020; 13:ph13050085. [PMID: 32365757 PMCID: PMC7281268 DOI: 10.3390/ph13050085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
After relative erythropoietin deficiency, iron deficiency is the second most important contributing factor for anemia in chronic kidney disease (CKD) patients. Iron supplementation is a crucial part of the treatment of anemia in CKD patients, and intravenous (IV) iron supplementation is considered to be superior to per os (PO) iron supplementation. The differences between the available formulations are poorly characterized. This report presents results from pairwise and network meta-analyses carried out after a comprehensive search in sources of published and unpublished studies, according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) recommendations (International prospective register of systematic reviews PROSPERO reference ID: CRD42020148155). Meta-analytic calculations were performed for the outcome of non-response to iron supplementation (i.e., hemoglobin (Hgb) increase of <0.5–1.0 g/dL, or initiation/intensification of erythropoiesis-stimulating agent (ESA) therapy, or increase/change of iron supplement, or requirements of blood transfusion). A total of 34 randomized controlled trials (RCT) were identified, providing numerical data for analyses covering 93.7% (n = 10.097) of the total study population. At the network level, iron supplementation seems to have a more protective effect against the outcome of non-response before the start of dialysis than once dialysis is initiated, and some preparations seem to be more potent (e.g., ferumoxytol, ferric carboxymaltose), compared to the rest of iron supplements assessed (surface under the cumulative ranking area (SUCRA) > 0.8). This study provides parameters for adequately following-up patients requiring iron supplementation, by presenting the most performing preparations, and, indirectly, by making it possible to identify good responders among all patients treated with these medicines.
Collapse
|
39
|
Coyne DW, Fishbane S. The Value of Intravenous Iron: Beyond the Cave of Speculation. J Am Soc Nephrol 2020; 31:896-897. [PMID: 32253272 DOI: 10.1681/asn.2019121340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Daniel W Coyne
- Division of Nephrology, School of Medicine, Washington University, St. Louis, Missouri
| | - Steven Fishbane
- Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, New York
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW We will examine the current and future options in management of anemia in dialysis patients focusing on recent trials in iron supplementation and alternatives to erythropoietin-stimulating agents (ESAs). RECENT FINDINGS We review the literature on Erythropoietin (EPO)-stimulating agents, focusing on the risk benefits of various options available. We review the recent practice changing trial in iron supplementation in dialysis patients with chronic kidney disease and movements in the research on alternatives to EPO-stimulating agents primarily hypoxia-inducible factor prolyl hydroxylase inhibitors (HIF-PHIs). SUMMARY ESAs constitute the mainstay of treatment of anemia in dialysis and evidence does not support the preference of any one type over the other. But concerns exist about the cardiovascular safety of supra-physiological ESA levels. Iron supplementation has been shown to be a well tolerated method to decrease ESA doses while maintaining hemoglobin levels and recent evidence should result in a revisiting of the guidelines for iron supplementation. HIF-PHIs are potentially safe alternatives to ESAs that correct and maintain hemoglobin while maintaining physiological levels of erythropoietin. Ongoing phase III trials for these drugs will likely answer questions of long-term safety regarding these drugs.
Collapse
|
41
|
Miles LF, Coulson TG, Larsen T, Burbury KL, Story DA, Bellomo R. Associations between preoperative inflammatory hyperferritinaemia and outcomes after major abdominal surgery. Br J Anaesth 2020; 124:e195-e197. [PMID: 32000977 DOI: 10.1016/j.bja.2020.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/26/2019] [Accepted: 01/01/2020] [Indexed: 12/01/2022] Open
|
42
|
Ganz T, Aronoff GR, Gaillard CAJM, Goodnough LT, Macdougall IC, Mayer G, Porto G, Winkelmayer WC, Wish JB. Iron Administration, Infection, and Anemia Management in CKD: Untangling the Effects of Intravenous Iron Therapy on Immunity and Infection Risk. Kidney Med 2020; 2:341-353. [PMID: 32734254 PMCID: PMC7380433 DOI: 10.1016/j.xkme.2020.01.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are at increased risk for infection, attributable to immune dysfunction, increased exposure to infectious agents, loss of cutaneous barriers, comorbid conditions, and treatment-related factors (eg, hemodialysis and immunosuppressant therapy). Because iron plays a vital role in pathogen reproduction and host immunity, it is biologically plausible that intravenous iron therapy and/or iron deficiency influence infection risk in CKD. Available data from preclinical experiments, observational studies, and randomized controlled trials are summarized to explore the interplay between intravenous iron and infection risk among patients with CKD, particularly those receiving maintenance hemodialysis. The current evidence base, including data from a recent randomized controlled trial, suggests that proactive judicious use of intravenous iron (in a manner that minimizes the accumulation of non-transferrin-bound iron) beneficially replaces iron stores while avoiding a clinically relevant effect on infection risk. In the absence of an urgent clinical need, intravenous iron therapy should be avoided in patients with active infection. Although serum ferritin concentration and transferrin saturation can help guide clinical decision making about intravenous iron therapy, definition of an optimal iron status and its precise determination in individual patients remain clinically challenging in CKD and warrant additional study.
Collapse
Affiliation(s)
- Tomas Ganz
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA
| | | | | | - Lawrence T Goodnough
- Department of Pathology, Stanford University, Stanford, CA.,Department of Medicine (Hematology), Stanford University, Stanford, CA
| | - Iain C Macdougall
- Department of Renal Medicine, King's College Hospital, London, United Kingdom
| | - Gert Mayer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | - Graça Porto
- Pathology and Molecular Immunology Department, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,i3S, Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal
| | - Wolfgang C Winkelmayer
- Section of Nephrology and Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, TX
| | - Jay B Wish
- Division of Nephrology, Indiana University Health, Indianapolis, IN
| |
Collapse
|
43
|
Batchelor EK, Kapitsinou P, Pergola PE, Kovesdy CP, Jalal DI. Iron Deficiency in Chronic Kidney Disease: Updates on Pathophysiology, Diagnosis, and Treatment. J Am Soc Nephrol 2020; 31:456-468. [PMID: 32041774 PMCID: PMC7062209 DOI: 10.1681/asn.2019020213] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Anemia is a complication that affects a majority of individuals with advanced CKD. Although relative deficiency of erythropoietin production is the major driver of anemia in CKD, iron deficiency stands out among the mechanisms contributing to the impaired erythropoiesis in the setting of reduced kidney function. Iron deficiency plays a significant role in anemia in CKD. This may be due to a true paucity of iron stores (absolute iron deficiency) or a relative (functional) deficiency which prevents the use of available iron stores. Several risk factors contribute to absolute and functional iron deficiency in CKD, including blood losses, impaired iron absorption, and chronic inflammation. The traditional biomarkers used for the diagnosis of iron-deficiency anemia (IDA) in patients with CKD have limitations, leading to persistent challenges in the detection and monitoring of IDA in these patients. Here, we review the pathophysiology and available diagnostic tests for IDA in CKD, we discuss the literature that has informed the current practice guidelines for the treatment of IDA in CKD, and we summarize the available oral and intravenous (IV) iron formulations for the treatment of IDA in CKD. Two important issues are addressed, including the potential risks of a more liberal approach to iron supplementation as well as the potential risks and benefits of IV versus oral iron supplementation in patients with CKD.
Collapse
Affiliation(s)
| | - Pinelopi Kapitsinou
- Feinberg Cardiovascular and Renal Research Institute and
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Pablo E Pergola
- Renal Associates PA, Division of Nephrology, University of Texas Health Science Center at San Antonio, San Antonio, Texas; and
| | - Csaba P Kovesdy
- Division of Nephrology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Diana I Jalal
- Division of Nephrology, University of Iowa Hospitals and Clinics, Iowa City, Iowa;
| |
Collapse
|
44
|
Affiliation(s)
- Jay B. Wish
- Division of Nephrology Case Western Reserve University Cleveland, Ohio, USA
| |
Collapse
|
45
|
Salim SA, Cheungpasitporn W, Elmaraezy A, Jawafi O, Rahman M, Aeddula NR, Tirupathi R, Fülöp T. Infectious complications and mortality associated with the use of IV iron therapy: a systematic review and meta-analysis. Int Urol Nephrol 2019; 51:1855-1865. [PMID: 31485910 DOI: 10.1007/s11255-019-02273-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 08/29/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Parental iron is used to optimize hemoglobin and enhance erythropoiesis in end-stage renal disease along with erythropoietin-stimulating agents. Safety of iron has been debated extensively and there is no definite evidence whether parenteral iron increases the risk of infections and mortality. We performed this meta-analysis to evaluate the incidence of infectious complications, hospitalizations and mortality with use of parenteral iron. METHODS Medical electronic databases [PubMed, EMBASE, Scopus, Web of Science, and cochrane central register for controlled clinical trials (CENTRAL)] were queried for studies that investigated the association between intravenous iron administration and infection in hemodialysis patients. 24 studies (8 Randomized control trials (RCTs) and 16 observational studies) were considered for qualitative and quantitative analysis. RESULTS All-cause mortality Data from 6 RCTs show that high-dose IV iron conferred 17% less all-cause mortality compared to controls; however, this outcome was not statistically significant (OR = 0.83, CI [0.7, 1.01], p = 0.07). Nine observational studies were pooled under the random effects model due to significant heterogeneity (I2 = 83%, p < 0.001). The overall HR showed increased risk of all-cause mortality in the high-dose group but was statistically non-significant (HR = 1.1, CI [1, 1.22], p = 0.06). Infections Four RCTs with no heterogeneity among their data (I2 = 0%, p = 0.61). Under the fixed effect model, there was no difference in the infection rate between high-dose iron and control group (OR = 0.97, CI [0.82, 1.16], p = 0.77); eight observational studies with significant heterogeneity and utilizing random effects model. Summary HR showed increased yet non-significant risk of infection in the high-dose group (HR = 1.13, CI [0.99, 1.28], p = 0.07) Hospitalization 1 RCT and six observational studies provided data for the rate of all-cause hospitalization. There was marked heterogeneity among observational studies. RCT showed no significant difference between high-dose iron and controls in the rate of hospitalization (OR = 1.03, CI [0.87, 1.23], p = 0.71). Summary HR for observational data showed increased rate of hospitalization in the high-dose group; however, this effect was not statistically significant (HR = 1.11, CI [0.99, 1.24], p = 0.07). Cardiovascular events One RCT compared the rate of adverse cardiovascular events between high-dose and low-dose iron. No significant difference was observed between the two groups (22.3% vs 25.6%, p = 0.12). Six heterogeneous observational studies (I2 = 65%, p < 0.001) reported on the rate of cardiovascular events. No significant difference was observed between high-dose iron and controls (HR = 1.18, CI [0.89, 1.57], p = 0.24). CONCLUSION High-dose parenteral iron does not seem to be associated with higher risk of infection, all-cause mortality, increased hospitalization or increased cardiovascular events on analysis of RCTs. Observational studies show increased risk for all-cause mortality, infections and hospitalizations that were not statistically significant and were associated with significant heterogeneity.
Collapse
Affiliation(s)
- Sohail Abdul Salim
- Department of Internal Medicine, Division of Nephrology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216-4505, USA.
| | - Wisit Cheungpasitporn
- Department of Internal Medicine, Division of Nephrology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS, 39216-4505, USA
| | | | - Omar Jawafi
- Computational and Data-Enabled Science, Jackson State University, Jackson, MS, USA
| | - Md Rahman
- Computational and Data-Enabled Science, Jackson State University, Jackson, MS, USA
| | | | | | - Tibor Fülöp
- Division of Nephrology, Department of Internal Medicine, Medical University of South Carolina, Charleston, SC, USA.,Raph H. Johnson VA Medical Center, Charleston, SC, USA
| |
Collapse
|
46
|
Locham S, Mathlouthi A, Dakour-Aridi H, Nejim B, Malas MB. Association between Severe Anemia and Outcomes of Hemodialysis Vascular Access. Ann Vasc Surg 2019; 62:295-303. [PMID: 31449946 DOI: 10.1016/j.avsg.2019.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND The vast majority of patients undergoing hemodialysis (HD) are anemic. The severity of anemia in these patients may influence the postoperative outcomes and the durability of vascular access. Thus, the purpose of this study is to assess the association between anemia and adverse outcomes in patients undergoing HD access placement (arteriovenous grafts and fistula). METHODS Patients with chronic kidney disease stages IV and V recorded in the Vascular Quality Initiative Hemodialysis database between 2011 and 2017 were included. Patients were divided into 3 study groups based on preoperative hemoglobin (Hgb) levels: normal/mild anemia (Hgb: females ≥10 g/dL, males ≥12 g/dL), moderate anemia (Hgb: females: 7-9.9 g/dL, males: 9-11.9 g/dL), and severe anemia (Hgb: females<7 g/dL, males<9 g/dL). Multivariable logistic and Cox regression analyses were implemented to evaluate the association between anemia and 30-day mortality and primary patency (PP) at 1 year. RESULTS A total of 28,000 patients undergoing HD access surgery were identified (normal/mild [42%], moderate [49%], and severe [9%] anemia). Postoperative bleeding (2.1% vs. 2.2% vs. 2.2%) and 30-day outcomes including swelling (0.4% vs. 0.5% vs. 0.7%) and wound infection (0.4% vs. 0.3% vs. 0.1%) were similar in mild/normal, moderate, and severe anemia groups, respectively (All P > 0.05). However, 30-day mortality was significantly higher in patients with severe anemia compared with normal/mild and moderate anemia (2.1% vs. 1.1% and 1.1%, P < 0.001). After adjusting for potential confounders, severe anemia was associated with 90% higher risk of 30-day mortality (odds ratio [95% confidence interval]: 1.90 [1.20-3.00], P = 0.006) and 17% increase in PP loss at 1 year (adjusted hazard ratio [95% confidence interval]: 1.17 [1.02-1.35], P = 0.01) compared with the normal/mild anemia group. However, no significant difference was seen between normal/mild and moderate anemia. CONCLUSIONS In this large study of patients undergoing HD access placement, severe anemia was associated with 90% increased risk of 30-day mortality and 17% increased risk of loss of PP compared with those with normal/mild anemia. Management of severe anemia before surgery might be indicated to reduce operative mortality and improve the durability of HD access.
Collapse
Affiliation(s)
- Satinderjit Locham
- Department of Vascular and Endovascular Surgery, University of California San Diego, La Jolla, CA
| | - Asma Mathlouthi
- Department of Vascular and Endovascular Surgery, University of California San Diego, La Jolla, CA
| | - Hanaa Dakour-Aridi
- Department of Vascular and Endovascular Surgery, University of California San Diego, La Jolla, CA
| | - Besma Nejim
- Hershey PennState College of Medicine, Hershey, PA
| | - Mahmoud B Malas
- Department of Vascular and Endovascular Surgery, University of California San Diego, La Jolla, CA.
| |
Collapse
|
47
|
Rostoker G, Vaziri ND. Risk of iron overload with chronic indiscriminate use of intravenous iron products in ESRD and IBD populations. Heliyon 2019; 5:e02045. [PMID: 31338466 PMCID: PMC6627982 DOI: 10.1016/j.heliyon.2019.e02045] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 03/14/2019] [Accepted: 07/03/2019] [Indexed: 01/19/2023] Open
Abstract
The routine use of recombinant erythropoiesis-stimulating agents (ESA) over the past three decades has enabled the partial correction of anaemia in most patients with end-stage renal disease (ESRD). Since ESA use frequently leads to iron deficiency, almost all ESA-treated haemodialysis patients worldwide receive intravenous iron (IV) to ensure sufficient available iron during ESA therapy. Patients with inflammatory bowel disease (IBD) are also often treated with IV iron preparations, as anaemia is common in IBD. Over the past few years, liver magnetic resonance imaging (MRI) has become the gold standard method for non-invasive diagnosis and follow-up of iron overload diseases. Studies using MRI to quantify liver iron concentration in ESRD have shown a link between high infused iron dose and risk of haemosiderosis in dialysis patients. In September 2017, the Pharmacovigilance Committee (PRAC) of the European Medicines Agency (EMA) considered convergent publications over the last few years on iatrogenic haemosiderosis in dialysis patients and requested that companies holding marketing authorization for iron products should investigate the risk of iron overload, particularly in patients with end-stage renal disease on dialysis and, by analogy, patients with IBD. We present a narrative review of data supporting the views and decision of the EMA, and then give our expert opinion on this controversial field of anaemia therapeutics.
Collapse
Affiliation(s)
- Guy Rostoker
- Division of Nephrology and Dialysis, Hôpital Privé Claude Galien, Ramsay-Générale de Santé, Quincy-sous-Sénart, France
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, University of California, Irvine, USA
| |
Collapse
|
48
|
Karaboyas A, Morgenstern H, Waechter S, Fleischer NL, Vanholder R, Jacobson SH, Sood MM, Schaubel DE, Inaba M, Pisoni RL, Robinson BM. Low hemoglobin at hemodialysis initiation: an international study of anemia management and mortality in the early dialysis period. Clin Kidney J 2019; 13:425-433. [PMID: 32699623 PMCID: PMC7367115 DOI: 10.1093/ckj/sfz065] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 04/29/2019] [Indexed: 02/07/2023] Open
Abstract
Background Anemia at hemodialysis (HD) initiation is common. Correcting low hemoglobin (Hgb) before HD initiation may improve survival by avoiding potential harms of chronic anemia, high doses of erythropoiesis-stimulating agents (ESAs) and intravenous (IV) iron in the early HD period, and/or rapid Hgb rise. Methods We included 4604 incident HD patients from 21 countries in the Dialysis Outcomes and Practice Patterns Study Phases 4–5 (2009–15). Because low Hgb at HD start may reflect comorbidity or ESA hyporesponse, we restricted our analysis to the 80% of patients who achieved Hgb ≥10 g/dL 91–120 days after HD start (Month 4). Results About 53% of these patients had Hgb <10 g/dL in Month 1 (<30 days after HD start); they were younger with a similar comorbidity profile (versus Hgb ≥10 g/dL). Month 1 Hgb was associated with first-year HD mortality (adjusted hazard ratio for 1 g/dL higher Hgb was 0.89; 95% confidence interval: 0.81–0.97), despite minimal differences in Month 4 Hgb. Patients with lower Hgb in Month 1 received higher doses of ESA, but not IV iron, over the first 3 months of HD. Results were consistent when excluding catheter users or adjusting for IV iron and ESA dose over the first 3 months. Conclusions Even among patients with Hgb ≥10 g/dL 3 months later, anemia at HD initiation was common and associated with elevated mortality. A more proactive approach to anemia management in advanced chronic kidney disease (CKD) may thus improve survival on HD, though long-term prospective studies of non-dialysis CKD patients are needed.
Collapse
Affiliation(s)
- Angelo Karaboyas
- Arbor Research Collaborative for Health, Ann Arbor, MI, USA.,Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Hal Morgenstern
- Department of Epidemiology and Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA.,Department of Urology, Medical School, University of Michigan, Ann Arbor, MI, USA
| | | | - Nancy L Fleischer
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | | | - Stefan H Jacobson
- Karolinska Institutet, Danderyd University Hospital, Stockholm, Sweden
| | - Manish M Sood
- Department of Medicine, School of Epidemiology and Public Health, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Canada
| | | | - Masaaki Inaba
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | | | - Bruce M Robinson
- Arbor Research Collaborative for Health, Ann Arbor, MI, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
49
|
Agarwal AK, Yee J. Hepcidin. Adv Chronic Kidney Dis 2019; 26:298-305. [PMID: 31477260 DOI: 10.1053/j.ackd.2019.04.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/20/2022]
Abstract
Dysregulation of metabolism and utilization of iron can lead to the development and maintenance of anemia of CKD. Anemia is prevalent among patients with CKD. The markers of iron sufficiency or availability of iron are far from perfect which results in inaccurate diagnosis and treatment of anemia with poor outcomes. Hepcidin, a 25 amino acid peptide produced by the hepatocytes, has emerged as the key regulator of uptake and release of iron in the tissues to maintain a steady supply of iron to erythron and other tissues while avoiding higher levels of iron that could be detrimental to the organs. Hepcidin itself is regulated by the supply of iron, the need for erythropoiesis, and the state of inflammation. Alterations in hepcidin levels are associated with restricted erythropoiesis, anemia, and iron overload. Discovery of hepcidin and elucidation of its mechanism of action and consequences of its upregulation and suppression have unraveled important insight into many hematologic disorders including anemia of CKD. This knowledge has also unlocked unique opportunities to modulate hepcidin via agonists and antagonists of hepcidin and its feedback pathways to treat clinical conditions. Many such agents are being developed and have potential therapeutic utility in future.
Collapse
|
50
|
Kshirsagar AV, Li X. Long-Term Risks of Intravenous Iron in End-Stage Renal Disease Patients. Adv Chronic Kidney Dis 2019; 26:292-297. [PMID: 31477259 DOI: 10.1053/j.ackd.2019.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/15/2019] [Accepted: 05/15/2019] [Indexed: 01/07/2023]
Abstract
Patients with end-stage renal disease on dialysis commonly receive intravenous iron to treat anemia along with erythropoiesis-stimulating agents. While studies of intravenous iron have demonstrated efficacy in raising hemoglobin, the quantity of administered intravenous iron has raised concerns about iron overload leading to long-term toxicities. The goal of this review is to understand recent trends in intravenous iron use, potential mechanisms of iron toxicity, and to evaluate the available evidence in the literature for potential long-term cardiovascular and infectious complications. We include findings from the recently published landmark clinical trial of intravenous iron for patients receiving hemodialysis to contextualize treatment recommendations.
Collapse
|