1
|
Auerbach M, Wolf M. Intravenous Iron Associated Hypophosphatemia: Much More Than a Laboratory Curiosity. Am J Hematol 2025; 100:752-754. [PMID: 39821908 DOI: 10.1002/ajh.27599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/19/2025]
Affiliation(s)
| | - Myles Wolf
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
2
|
Babah OA, Beňová L, Hanson C, Abioye AI, Larsson EC, Afolabi BB. Screening and treatment practices for iron deficiency in anaemic pregnant women: A cross-sectional survey of healthcare workers in Nigeria. PLoS One 2024; 19:e0310912. [PMID: 39570864 PMCID: PMC11581334 DOI: 10.1371/journal.pone.0310912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/08/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Iron deficiency anaemia in pregnancy is a significant contributor to maternal and perinatal morbidity and mortality globally. Despite international and national guidelines for its screening and treatment, knowledge and prescription practices of healthcare providers vary. AIM To determine maternal healthcare workers' screening and treatment practices for iron deficiency in anaemic pregnancy women in two states in Nigeria. METHODOLOGY This cross-sectional study sampled maternal healthcare workers from 84 randomly selected public health facilities in Lagos and Kano States. Data on methods of diagnosis and prescription practices for iron deficiency anaemia were collected using a self-administered questionnaire. Means and percentages were reported using probability weights, and a comparison of practices of anaemia treatment between doctors and nurses/midwives was done using Chi-square test or Fishers exact. RESULTS Of the 467 maternal healthcare workers surveyed (232 from Lagos, 235 from Kano), 40.0% were doctors, 54.0% nurses or midwives and 6.0% community health extension workers. In the sample, 27.6% always and 58.7% sometimes screened anaemic pregnant women for iron deficiency; among these, 84.7% screened using complete blood count. Oral iron for treatment of iron deficiency anaemia was prescribed by 96.9%. Intravenous iron for treatment was prescribed by 30.2%, but by only by 18.6% as first-line drug (as iron dextran by 69.3% and as iron sucrose by 31.5% of intravenous iron prescribers). Commonest reasons for low usage of intravenous iron were cost and need for venepuncture. Fifty-three percent of maternal healthcare workers' prescribed iron supplements for anaemia during concomitant infection, with the prescription practice similar among doctors versus nurse/midwives (p = 0.074). CONCLUSION We found suboptimal levels of screening for iron deficiency among anaemic pregnant women. Iron deficiency anaemia in pregnancy is almost exclusively treated with oral iron by maternal healthcare workers in the two Nigerian states, similarly between doctors and nurses/midwives. Further research into potential reasons for low screening for iron deficiency and low use of intravenous iron are needed.
Collapse
Affiliation(s)
- Ochuwa Adiketu Babah
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
- Faculty of Clinical Sciences, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
- Department of Obstetrics and Gynaecology, Lagos University Teaching Hospital, Idi-Araba, Lagos, Nigeria
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | - Lenka Beňová
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| | - Claudia Hanson
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Ajibola Ibraheem Abioye
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Elin C. Larsson
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Bosede Bukola Afolabi
- Faculty of Clinical Sciences, College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
- Department of Obstetrics and Gynaecology, Lagos University Teaching Hospital, Idi-Araba, Lagos, Nigeria
- Centre for Clinical Trials and Implementation Science (CCTRIS), College of Medicine, University of Lagos, Idi-Araba, Lagos, Nigeria
| |
Collapse
|
3
|
Van Doren L, Steinheiser M, Boykin K, Taylor KJ, Menendez M, Auerbach M. Expert consensus guidelines: Intravenous iron uses, formulations, administration, and management of reactions. Am J Hematol 2024; 99:1338-1348. [PMID: 38282557 DOI: 10.1002/ajh.27220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/01/2024] [Accepted: 01/08/2024] [Indexed: 01/30/2024]
Abstract
Intravenous iron has become an essential component for the treatment of iron deficiency and iron deficiency anemia. Individuals administering Intravenous iron should have knowledge in intravenous iron administration, including a pre-infusion assessment to evaluate infusion reaction risks, pre- and post-infusion monitoring, identification of and management of infusion reactions, accurate documentation of these reactions, laboratory monitoring and recognition and management of treatment-emergent hypophosphatemia. This comprehensive consensus provides step-by-step guidance and tools for practitioners to promote safe delivery of intravenous iron, recognition, and management of infusion reactions and treatment-emergent hypophosphatemia.
Collapse
Affiliation(s)
- Layla Van Doren
- Section of Hematology, Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Kristen Boykin
- Florida Cancer Specialists and Research Institute, Florida, USA
| | | | - Monica Menendez
- FEM Iron Infusion Centers by Heme Onc Call, Miami, Florida, USA
| | - Michael Auerbach
- Department of Medicine, Georgetown University School of Medicine, Washington, District of Columbia, USA
| |
Collapse
|
4
|
Pellitteri G, Versace S, Merlino G, Nilo A, Gigli GL, Valente M. A comprehensive update on the ADMET considerations for α2δ calcium channel ligand medications for treating restless legs syndrome. Expert Opin Drug Metab Toxicol 2024; 20:133-142. [PMID: 38482850 DOI: 10.1080/17425255.2024.2329738] [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: 01/18/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
INTRODUCTION Restless legs syndrome/Willis-Ekbom disease (RLS/WED) is a sleep-related sensory-motor disorder associated with poor sleep quality and impaired daily functioning. In patients affected by chronic RLS/WED, a pharmacological therapy is recommended. International guidelines suggest to start the treatment with a α2δ calcium channel ligand in most cases, unless contraindicated. AREAS COVERED The present review is based on an extensive Internet and PubMed search from 1986 to 2024. Our purpose is to describe the absorption, distribution, metabolism, and toxicology (ADMET) of the α2δ ligands, with common consideration for the therapeutic class, specificities of different compounds, efficacy, and safety in relation to other treatment options. EXPERT OPINION α2δ ligands are quite similar in their ADMET profiles, sharing most of the pharmacokinetics and potential adverse effects. However, we highlight the linear kinetic of gabapentin enacarbil and pregabalin, differently from gabapentin. α2δ ligands are safe and effective for the treatment of RLS/WED. Additional benefits can be obtained in comorbid insomnia, chronic pain syndromes, history of impulse control disorder, and comorbid anxiety. The use of α2δ ligands is associated with poor risk of augmentation. We still need new long-term safe and effective treatments, which could be developed along with our knowledge of RLS/WED pathophysiology.
Collapse
Affiliation(s)
- Gaia Pellitteri
- Clinical Neurology Unit, Udine University Hospital, Udine, Italy
| | - Salvatore Versace
- Clinical Neurology Unit, Udine University Hospital, Udine, Italy
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Giovanni Merlino
- Clinical Neurology Unit, Udine University Hospital, Udine, Italy
| | - Annacarmen Nilo
- Clinical Neurology Unit, Udine University Hospital, Udine, Italy
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Gian Luigi Gigli
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Mariarosaria Valente
- Clinical Neurology Unit, Udine University Hospital, Udine, Italy
- Department of Medicine (DMED), University of Udine, Udine, Italy
| |
Collapse
|
5
|
Van Doren L, Auerbach M. IV iron formulations and use in adults. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:622-629. [PMID: 38066930 PMCID: PMC10727060 DOI: 10.1182/hematology.2023000495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Intravenous iron has become a major component of the therapeutic armamentarium for iron deficiency and iron deficiency anemia. The earliest formulations were associated with unacceptable toxicity. Newer formulations, with complex carbohydrate cores that bind elemental iron more tightly, allow the administration of full therapeutic doses in 15 to 60 minutes. Nonetheless, a folklore of danger, fueled by earlier formulations no longer available, continues to foment caution. Complement-mediated minor infusion reactions, referred to as complement activation-related pseudo-allergy, resolve without therapy. Inappropriate intervention with vasopressors and H1 blockers converts these minor reactions into hemodynamically significant adverse events. Four new formulations, low-molecular-weight iron dextran, ferumoxytol, ferric carboxymaltose, and ferric derisomaltose, all approved for the treatment of iron deficiency in a host of conditions, are now widely used with an excellent safety profile. Herein, the administration, safety, indications, and management of infusion reactions are discussed. Treatment-emergent hypophosphatemia, a newly recognized side effect for some formulations, is also reviewed. Based on the preponderance of published evidence, intravenous iron should be moved up-front for the treatment of iron deficiency and iron deficiency anemia in those conditions in which oral iron is suboptimal.
Collapse
Affiliation(s)
- Layla Van Doren
- Division of Hematology, Yale School of Medicine, New Haven, CT
| | - Michael Auerbach
- Division of Hematology, Georgetown School of Medicine, Baltimore, MD
| |
Collapse
|
6
|
Maas LA, Krishna M, Parian AM. Ironing It All Out: A Comprehensive Review of Iron Deficiency Anemia in Inflammatory Bowel Disease Patients. Dig Dis Sci 2023; 68:357-369. [PMID: 35930123 DOI: 10.1007/s10620-022-07599-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/10/2022] [Indexed: 12/09/2022]
Abstract
Iron deficiency anemia affects approximately 45% of patients with inflammatory bowel disease (IBD), negatively impacts the quality of life in this patient population, and significantly burdens our healthcare system. The pathogenesis of iron deficiency in IBD patients is multifactorial, including intestinal bleeding, malabsorption, and inadequate oral intake. Regular screening and diagnosis in these patients are imperative, and often patients have mixed iron deficiency anemia and anemia of chronic disease, especially in those with active inflammation. Iron may be replenished either orally or intravenously. While oral iron is safe, affordable, and easy to administer, patients often suffer from intolerable gastrointestinal side effects, and particularly in IBD patients, oral iron may increase inflammation and contribute to flares. Therefore, although it is substantially underused, intravenous (IV) iron is considered first-line treatment for patients with active disease, severe anemia, oral iron intolerance, and erythropoietin requirements. Several IV iron formulations are available, and iron sucrose and ferric carboxymaltose are the most frequently used and well studied in patients with IBD. However, iron isomaltoside could potentially become a popular choice among providers given its safety, efficacy, and convenience. Overall, screening, diagnosis, and treatment of iron deficiency anemia are important in patients with IBD. Individual patient characteristics, risks, and benefits, and advantages and disadvantages, should be considered when determining the best route and formulation for iron repletion.
Collapse
|
7
|
Boots JMM, Quax RAM. High-Dose Intravenous Iron with Either Ferric Carboxymaltose or Ferric Derisomaltose: A Benefit-Risk Assessment. Drug Saf 2022; 45:1019-1036. [PMID: 36068430 PMCID: PMC9492608 DOI: 10.1007/s40264-022-01216-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2022] [Indexed: 11/22/2022]
Abstract
The intravenous iron formulations ferric carboxymaltose (FCM) and ferric derisomaltose (FDI) offer the possibility of administering a large amount of iron in one infusion. This results in faster correction of anemia and the formulations being better tolerated than oral iron formulations. This triad of logistic advantages, improved patient convenience, and fast correction of anemia explains the fact that intravenous iron formulations nowadays are frequently prescribed worldwide in the treatment of iron deficiency anemia. However, these formulations may result in hypophosphatemia by inducing a strong increase in active fibroblast growth factor-23 (FGF-23), a hormone that stimulates renal phosphate excretion. This effect is much more pronounced with FCM than with FDI, and therefore the risk of developing hypophosphatemia is remarkably higher with FCM than with FDI. Repeated use of FCM may result in severe osteomalacia, which is characterized by bone pain, Looser zones (pseudofractures), and low-trauma fractures. Intravenous iron preparations are also associated with other adverse effects, of which hypersensitivity reactions are the most important and are usually the result of a non-allergic complement activation on nanoparticles of free labile iron-Complement Activation-Related Pseudo-Allergy (CARPA). The risk on these hypersensitivity reactions can be reduced by choosing a slow infusion rate. Severe hypersensitivity reactions were reported in < 1% of prospective trials and the incidence seems comparable between the two formulations. A practical guideline has been developed based on baseline serum phosphate concentrations and predisposing risk factors, derived from published cases and risk factor analyses from trials, in order to establish the safe use of these formulations.
Collapse
Affiliation(s)
- Johannes M M Boots
- Department of Internal Medicine, Maasstad Hospital, Maasstadweg 21, 3079 DZ, Rotterdam, The Netherlands.
| | - Rogier A M Quax
- Department of Internal Medicine, Maasstad Hospital, Maasstadweg 21, 3079 DZ, Rotterdam, The Netherlands
| |
Collapse
|
8
|
Spagnuolo R, Abenavoli L, Larussa T, Iannelli C, Pellicano R, Fagoonee S, Doldo P, Luzza F. Safety profile of intravenous iron in inflammatory bowel disease: an up-to-date overview. Minerva Gastroenterol (Torino) 2022; 68:111-118. [PMID: 33267572 DOI: 10.23736/s2724-5985.20.02819-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Up to 30-70% of patients may experience mild and moderate side effects during iron therapy and this is often associated with a poor adherence to therapy. Anemia is frequent in patients with active inflammatory bowel disease (IBD), due to both iron deficiency and chronic inflammation, therefore iron supplementation is frequently needed. Considering that gastrointestinal disorders are the most common side effects with oral iron, in IBD patients intravenous administration must be preferred. Although intravenous iron supplementation remains the most effective therapy of IBD-associated iron deficiency anemia, the perception of risk related to intravenous administration by clinicians could limit this successful strategy. In this narrative review we provided an up to date on the safety of the different iron formulations for intravenous administration, by reporting the most recent studies in IBD patients.
Collapse
Affiliation(s)
- Rocco Spagnuolo
- Department of Clinical and Experimental Medicine, "Magna Graecia" University, Catanzaro, Italy
| | - Ludovico Abenavoli
- Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy -
| | - Tiziana Larussa
- Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Chiara Iannelli
- Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Rinaldo Pellicano
- Unit of Gastroenterology, Molinette Hospital, Città della Salute e della Scienza, Turin, Italy
| | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging, National Research Council, Molecular Biotechnology Center, Turin, Italy
| | - Patrizia Doldo
- Department of Clinical and Experimental Medicine, "Magna Graecia" University, Catanzaro, Italy
| | - Francesco Luzza
- Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| |
Collapse
|
9
|
Arastu AH, Elstrott BK, Martens KL, Cohen JL, Oakes MH, Rub ZT, Aslan JJ, DeLoughery TG, Shatzel J. Analysis of Adverse Events and Intravenous Iron Infusion Formulations in Adults With and Without Prior Infusion Reactions. JAMA Netw Open 2022; 5:e224488. [PMID: 35353168 PMCID: PMC8968468 DOI: 10.1001/jamanetworkopen.2022.4488] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IMPORTANCE Although iron deficiency is common, it remains unclear which iron repletion strategy is associated with the lowest rate of infusion-related adverse events, and how patients with history of infusion reaction should be managed. OBJECTIVE To evaluate rates of infusion reactions among 4 commonly used intravenous iron repletion strategies and determine how readministration was managed in patients with history of reaction. DESIGN, SETTING, AND PARTICIPANTS This cohort study included all patients receiving intravenous iron infusion from January 1, 2015, to September 7, 2021, at 6 centers in Portland, Oregon. Participants included a total of 12 237 patients with iron deficiency, not restricted by etiology. Statistical analysis was performed from September to October 2021. EXPOSURES Type of intravenous iron formulation and concurrent administration of diphenhydramine, epinephrine, famotidine, and/or hydrocortisone, used as surrogate maker of infusion reaction. MAIN OUTCOMES AND MEASURES Incidence of adverse events, including severe events requiring epinephrine, stratified by type of iron formulation, and in patients who received premedication or with history of infusion-related reaction receiving subsequent doses. RESULTS Among 35 737 unique iron infusions (12 237 patients [9480 (77.5%) women; 717 (5.9%) Black; 10 250 (83.7%) White; mean (SD) age of 51 (20) years]), comprising 22 309 iron sucrose doses, 9067 iron dextran total doses (1771 preceded by test dose, 56 test doses alone), 3147 ferumoxytol doses, and 1214 ferric carboxymaltose doses, incidence of adverse events was 3.9% (n = 1389; 95% CI, 3.7%-4.1%). Rate of infusion events differed among iron formulations: 4.3% (n = 970; 95% CI, 4.1%-4.6%) iron sucrose, 3.8% (n = 345, 95% CI: 3.4%-4.2%) iron dextran (test and full doses or test dose alone), 1.8% (n = 57; 95% CI, 1.4%-2.3%) ferumoxytol, and 1.4% (n = 17, 95% CI, 0.8%-2.3%) ferric carboxymaltose (P < .001). Severe adverse events were exceedingly rare with only 2 documented epinephrine administrations, both associated with iron dextran. Incidence of adverse events among those who received premedication was 23-fold higher compared with those who did not (38.6% vs 1.7%, χ21 = 7324.8; P < .001). Among 873 patients with history of infusion reaction who underwent readministration, the majority received the same formulation, which was associated with significantly higher reaction rate particularly if premedication was administered (68% [95% CI, 64%-72%] vs 32% [95% CI, 26%-41%], respectively), compared with those who received an alternate formulation (21% [95% CI, 11%-35%] vs 5% [95% CI, 2%-12%], respectively) (P < .001). CONCLUSIONS AND RELEVANCE These data, and the preponderance of published evidence, suggest that intravenous iron is generally well tolerated with exceedingly low risk of severe reaction, use of premedication and test doses are unnecessary, and that optimal prevention and management of infusion-related reactions warrant further study.
Collapse
Affiliation(s)
- Asad H. Arastu
- Department of Medicine, Oregon Health & Science University, Portland
| | | | - Kylee L. Martens
- Department of Hematology-Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Jonathan L. Cohen
- Department of Pharmacy, Oregon Health & Science University, Portland
| | - Michael H. Oakes
- Department of Medicine, Oregon Health & Science University, Portland
| | - Zhoe T. Rub
- Department of Medicine, Chicago Medical School at Rosalind Franklin University of Medicine and Science, Chicago, Illinois
| | - Joseph J. Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland
| | - Thomas G. DeLoughery
- Department of Hematology-Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland
| | - Joseph Shatzel
- Department of Hematology-Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland
- Department of Biomedical Engineering, Oregon Health & Science University, Portland
| |
Collapse
|
10
|
Fortuny J, von Gersdorff G, Lassalle R, Linder M, Overbeek J, Reinold J, Toft G, Timmer A, Dress J, Blin P, Droz-Perroteau C, Ehrenstein V, Franzoni C, Herings R, Kollhorst B, Moore N, Odsbu I, Perez-Gutthann S, Schink T, Rascher K, Rasouliyan L, Rothman KJ, Saigi-Morgui N, Schaller M, Smits E, Forstner M, Bénichou J, Bircher AJ, Garbe E, Rampton DS, Gutierrez L. Use of intravenous iron and risk of anaphylaxis: A multinational observational post-authorisation safety study in Europe. Pharmacoepidemiol Drug Saf 2021; 30:1447-1457. [PMID: 34181291 PMCID: PMC8457074 DOI: 10.1002/pds.5319] [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: 04/20/2021] [Revised: 06/18/2021] [Accepted: 06/24/2021] [Indexed: 11/11/2022]
Abstract
PURPOSE This post-authorisation safety study estimated the risk of anaphylaxis in patients receiving intravenous (IV) iron in Europe, with interest in iron dextran and iron non-dextrans. Studies conducted in the United States have reported risk of anaphylaxis to IV iron ranging from 2.0 to 6.8 per 10 000 first treatments. METHODS Cohort study of IV iron new users, captured mostly through pharmacy ambulatory dispensing, from populations covered by health and administrative data sources in five European countries from 1999 to 2017. Anaphylaxis events were identified through an algorithm that used parenteral penicillin as a positive control. RESULTS A total of 304 210 patients with a first IV iron treatment (6367 iron dextran), among whom 13-16 anaphylaxis cases were identified and reported as a range to comply with data protection regulations. The pooled unadjusted incidence proportion (IP) ranged from 0.4 (95% confidence interval [CI], 0.2-0.9) to 0.5 (95% CI, 0.3-1.0) per 10 000 first treatments. No events were identified at first dextran treatments. There were 231 294 first penicillin treatments with 30 potential cases of anaphylaxis (IP = 1.2; 95% CI, 0.8-1.7 per 10 000 treatments). CONCLUSION We found an IP of anaphylaxis from 0.4 to 0.5 per 10 000 first IV iron treatments. The study captured only a fraction of IV iron treatments administered in hospitals, where most first treatments are likely to happen. Due to this limitation, the study could not exclude a differential risk of anaphylaxis between iron dextran and iron non-dextrans. The IP of anaphylaxis in users of penicillin was consistent with incidences reported in the literature.
Collapse
Affiliation(s)
- Joan Fortuny
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Barcelona, Spain
| | - Gero von Gersdorff
- Department of Internal Medicine-QiN-group, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Régis Lassalle
- Bordeaux PharmacoEpi, INSERM CIC1401, University of Bordeaux, Bordeaux, France
| | - Marie Linder
- Centre for Pharmacoepidemiology, Karolinska Institutet, Stockholm, Sweden
| | - Jetty Overbeek
- Department Research, PHARMO Institute for Drug Outcomes Research, Utrecht, The Netherlands
| | - Jonas Reinold
- Clinical Epidemiology Group, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
| | - Gunnar Toft
- Department of Clinical Epidemiology, Aarhus University, Aarhus, Denmark
| | - Antje Timmer
- Epidemiology and Biometry Group, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Jochen Dress
- Research Data Center (DaTraV), Federal Institute for Drugs and Medical Devices-BfArM, Bonn, Germany
| | - Patrick Blin
- Bordeaux PharmacoEpi, INSERM CIC1401, University of Bordeaux, Bordeaux, France
| | | | - Vera Ehrenstein
- Department of Clinical Epidemiology, Aarhus University, Aarhus, Denmark
| | - Carla Franzoni
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Barcelona, Spain
| | - Ron Herings
- Department Research, PHARMO Institute for Drug Outcomes Research, Utrecht, The Netherlands.,Amsterdam UMC, Vrije Universiteit Amsterdam, Epidemiology and Biostatistics, Amsterdam, The Netherlands
| | - Bianca Kollhorst
- Clinical Epidemiology Group, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
| | - Nicholas Moore
- Bordeaux PharmacoEpi, INSERM CIC1401, University of Bordeaux, Bordeaux, France
| | - Ingvild Odsbu
- Centre for Pharmacoepidemiology, Karolinska Institutet, Stockholm, Sweden
| | | | - Tania Schink
- Clinical Epidemiology Group, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
| | - Katherine Rascher
- Department of Internal Medicine-QiN-group, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Lawrence Rasouliyan
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Barcelona, Spain
| | - Kenneth J Rothman
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Waltham, Massachusetts, USA
| | - Nuria Saigi-Morgui
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Barcelona, Spain
| | - Mathias Schaller
- Department of Internal Medicine-QiN-group, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Elisabeth Smits
- Department Research, PHARMO Institute for Drug Outcomes Research, Utrecht, The Netherlands
| | | | | | - Jacques Bénichou
- Department of Biostatistics, Rouen University Hospital and Inserm U 1018, University of Rouen, Rouen, France
| | - Andreas J Bircher
- Allergy Unit, Dermatology Clinic University Hospital Basel, Basel, Switzerland.,Faculty of Biomedicine, Università della Svizzera italiana, Lugano, Switzerland
| | - Edeltraut Garbe
- Clinical Epidemiology Group, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
| | - David S Rampton
- Department of Gastroenterology, Royal London Hospital, London, UK
| | - Lia Gutierrez
- Pharmacoepidemiology and Risk Management, RTI Health Solutions, Barcelona, Spain
| |
Collapse
|
11
|
Auerbach M, Henry D, DeLoughery TG. Intravenous ferric derisomaltose for the treatment of iron deficiency anemia. Am J Hematol 2021; 96:727-734. [PMID: 33580972 PMCID: PMC8248147 DOI: 10.1002/ajh.26124] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 01/09/2023]
Abstract
Intravenous (IV) iron is the therapy of choice when oral iron is ineffective or poorly tolerated, yet use has been limited by fears of hypersensitivity reactions (HSRs). Newer formulations that bind iron more tightly and release it more slowly have made the risk of serious or severe HSRs very low. One such formulation, ferric derisomaltose, has been approved in the United States for delivery of 1000 mg iron in a single IV infusion. Ferric derisomaltose rapidly repletes iron parameters with low rates of serious or severe HSRs. Single-infusion iron repletion offers convenience, eliminates adherence concerns, and reduces healthcare resource utilization.
Collapse
Affiliation(s)
- Michael Auerbach
- Department of Medicine Georgetown University School of Medicine Washington District of Columbia
| | - David Henry
- Department of Medicine, Division of Hematology and Oncology Pennsylvania Hospital Philadelphia Pennsylvania
| | - Thomas G. DeLoughery
- Department of Hematology and Medical Oncology, Knight Cancer Center Oregon Health & Sciences University Oregon Portland
| |
Collapse
|
12
|
Gupta S, Panchal P, Gilotra K, Wilfred AM, Hou W, Siegal D, Whitlock RP, Belley-Cote EP. Intravenous iron therapy for patients with preoperative iron deficiency or anaemia undergoing cardiac surgery reduces blood transfusions: a systematic review and meta-analysis. Interact Cardiovasc Thorac Surg 2021; 31:141-151. [PMID: 32642775 DOI: 10.1093/icvts/ivaa094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/14/2020] [Accepted: 05/03/2020] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES The benefits of preoperative intravenous (IV) iron treatment in cardiac surgery patients with preoperative anaemia or iron deficiency have not been well-established. We performed a systematic review and meta-analysis to determine the effects of treating preoperative anaemia or iron deficiency with IV iron in adult cardiac surgery patients. METHODS We searched Cochrane Central Register of Controlled Trials, Medical Literature Analysis and Retrieval Systems Online and Excerpta Medica Database for randomized controlled trials (RCTs) and observational studies comparing IV iron to oral iron or no iron. We performed title and abstract, full-text screening, data extraction and risk of bias assessment independently and in duplicate. We pooled data using a random effects model and evaluated the overall quality of evidence. RESULTS We identified 4 RCTs and 7 observational studies. Pooled data from observational studies suggested a benefit of IV iron compared to no iron on mortality [relative risk 0.39, 95% confidence interval (CI) 0.23-0.65; P < 0.001, very low quality], units transfused per patient (mean difference -1.22, 95% CI -1.85 to -0.60; P < 0.001, very low quality), renal injury (relative risk 0.50, 95% CI 0.36-0.69; P < 0.001, very low quality) and hospital length of stay (mean difference -4.24 days, 95% CI -6.86 to -1.63; P = 0.001, very low quality). Pooled data from RCTs demonstrated a reduction in the number of patients transfused with IV iron compared to oral or no iron (relative risk 0.81, 95% CI 0.70-0.94; P = 0.005, moderate quality). The pooled estimates of effect from RCTs for mortality, hospital length of stay, units transfused per patient and renal injury were consistent in direction with observational studies. CONCLUSIONS This meta-analysis suggests that IV iron improves postoperative morbidity in adult cardiac surgery patients with preoperative anaemia or iron deficiency. A large, rigorous, placebo-controlled, double-blinded, multicentre trial is needed to clarify the role of IV iron in this patient population. CLINICAL TRIAL REGISTRATION International prospective register of systematic reviews ID Number CRD42019122844.
Collapse
Affiliation(s)
- Saurabh Gupta
- Department of Surgery, McMaster University, Hamilton, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Puru Panchal
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Kevin Gilotra
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Ann Mary Wilfred
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Winston Hou
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Deborah Siegal
- Department of Medicine, McMaster University, Hamilton, ON, Canada.,Population Health Research Institute, Hamilton, ON, Canada
| | - Richard P Whitlock
- Department of Surgery, McMaster University, Hamilton, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada.,Population Health Research Institute, Hamilton, ON, Canada
| | - Emilie P Belley-Cote
- Department of Medicine, McMaster University, Hamilton, ON, Canada.,Population Health Research Institute, Hamilton, ON, Canada
| |
Collapse
|
13
|
Rund D. Intravenous iron: do we adequately understand the short- and long-term risks in clinical practice? Br J Haematol 2020; 193:466-480. [PMID: 33216989 DOI: 10.1111/bjh.17202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/28/2020] [Indexed: 12/31/2022]
Abstract
Intravenous (IV) iron as a therapeutic agent is often administered but not always fully understood. The benefits of IV iron are well proven in many fields, particularly in nephrology. IV iron is beneficial not only for true iron deficiency but also for iron-restricted anaemia (functional iron deficiency). Yet, the literature on intravenous iron has many inconsistencies regarding its adverse effects. Over the last several years, newer forms of iron have been developed, leading to the more regular use of iron and in larger doses. This review will summarize some of the older and newer literature regarding the differences among iron products, including the mechanisms and frequency of their adverse events (AEs). The pathway and frequency of an underrecognized adverse event (hypophosphataemia) will be discussed. Recent insights on infection risk and iron handling by macrophages are examined. Potential but presently unproven risks of iron overload due to IV iron are discussed. The impact of these on the risk:benefit ratio and dosing of intravenous iron are considered in different clinical settings, including pregnancy and cancer. IV iron is an essential component of the therapy of anaemia and understanding these issues will enable more informed treatment decisions and knowledgeable use of these drugs.
Collapse
Affiliation(s)
- Deborah Rund
- Hebrew University-Hadassah Medical Organization, Ein Kerem, Jerusalem, Israel
| |
Collapse
|
14
|
Guinn NR, Cooter ML, Maisonave Y, Grimsley A, Cellura C, Grotegut CA, Lamon A, Aronson S. How do I develop a process to effectively treat parturients with iron deficiency anemia? Transfusion 2020; 60:2476-2481. [DOI: 10.1111/trf.15930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/19/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Nicole R. Guinn
- Department of Anesthesiology Duke University Medical Center Durham North Carolina USA
| | - Mary L. Cooter
- Department of Anesthesiology Duke University Medical Center Durham North Carolina USA
| | - Yasmin Maisonave
- Department of Anesthesiology Duke University Medical Center Durham North Carolina USA
| | - Aime Grimsley
- Department of Anesthesiology Duke University Medical Center Durham North Carolina USA
| | - Cindy Cellura
- Department of Anesthesiology Duke University Medical Center Durham North Carolina USA
| | - Chad A. Grotegut
- Department of Obstetrics and Gynecology Duke University Medical Center Durham North Carolina USA
| | - Agnes Lamon
- Department of Anesthesiology Penn Medicine Princeton Health Plainsboro New Jersey USA
| | - Solomon Aronson
- Department of Anesthesiology Duke University Medical Center Durham North Carolina USA
| | | |
Collapse
|
15
|
Sbrana A, Antonuzzo A, Brunello A, Petrelli F, Pronzato P, Tralongo A, Turrini M, Zoratto F, Danova M. Management of anemia in patients with cancer: 2019 Italian Association of Medical Oncology (AIOM) guidelines. TUMORI JOURNAL 2020; 106:337-345. [PMID: 32482149 DOI: 10.1177/0300891620921942] [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] [Indexed: 11/17/2022]
Abstract
This article summarizes the latest Italian Medical Oncology Association (AIOM) guidelines on the management of cancer-related and chemotherapy-related anemia with a particular attention to the use of erythropoiesis-stimulating agents and iron supplementation.
Collapse
Affiliation(s)
- Andrea Sbrana
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.,Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Andrea Antonuzzo
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | | | - Fausto Petrelli
- Medical Oncology Unit, ASST Bergamo Ovest, Treviglio, Bergamo, Italy
| | - Paolo Pronzato
- Medical Oncology Unit, AOU San Martino IRCCS IST, Genova, Italy
| | - Antonino Tralongo
- Medical Oncology Unit, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | - Marianna Turrini
- Medical Oncology Unit, Ospedale del Valdarno, Santa Maria La Gruccia, Azienda USL Toscana Sud-Est, Montevarchi, Toscana, Italy
| | - Federica Zoratto
- Medical Oncology Unit, Ospedale S. Maria Goretti, Latina, Lazio, Italy
| | - Marco Danova
- Internal Medicine and Medical Oncology, ASST Pavia, Pavia, Lombardia, Italy
| |
Collapse
|
16
|
Mattiello V, Schmugge M, Hengartner H, von der Weid N, Renella R. Diagnosis and management of iron deficiency in children with or without anemia: consensus recommendations of the SPOG Pediatric Hematology Working Group. Eur J Pediatr 2020; 179:527-545. [PMID: 32020331 DOI: 10.1007/s00431-020-03597-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/22/2020] [Accepted: 01/27/2020] [Indexed: 11/26/2022]
Abstract
Iron deficiency is the most prevalent nutritional deficiency affecting children and adolescents worldwide. A consistent body of epidemiological data demonstrates an increased incidence of iron deficiency at three timepoints: in the neonatal period, in preschool children, and in adolescents, where it particularly affects females.Conclusion: This narrative review focuses on the most suggestive symptoms of iron deficiency in childhood, describes the diagnostic procedures in situations with or without anemia, and provides Swiss expert-based management recommendations for the pediatric context.What is Known:• Iron deficiency (ID) is one of the most common challenges faced by pediatricians.• Significant progress in the diagnosis and therapy of ID has been made over the last decade.What is New:• Our expert panel provides ID management recommendations based on the best available evidence.• They include strategies for ID diagnosis and therapy, both oral and intravenous.
Collapse
Affiliation(s)
- Veneranda Mattiello
- Department "Woman-Mother-Child and Adolescent", Pediatric Hematology-Oncology Unit, Division of Pediatrics, University Hospital of Geneva, Geneva, Switzerland
| | - Markus Schmugge
- Division of Pediatric Hematology, University Children's Hospital of Zurich, Zurich, Switzerland
| | - Heinz Hengartner
- Pediatric Hematology-Oncology Unit, Children's Hospital of Sankt Gallen, Sankt Gallen, Switzerland
| | - Nicolas von der Weid
- Pediatric Hematology-Oncology Department, University Children's Hospital and University of Basel, Basel, Switzerland
| | - Raffaele Renella
- Department "Woman-Mother-Child", Pediatric Hematology-Oncology Unit, Division of Pediatrics, Lausanne University Hospital and University of Lausanne, Vaudois, BH11, Rue du Bugnon 46, 1011, Lausanne, Switzerland.
| |
Collapse
|
17
|
Pollock RF, Biggar P. Indirect methods of comparison of the safety of ferric derisomaltose, iron sucrose and ferric carboxymaltose in the treatment of iron deficiency anemia. Expert Rev Hematol 2020; 13:187-195. [PMID: 31928094 DOI: 10.1080/17474086.2020.1709437] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Objectives: The benefits of intravenous (IV) iron greatly outweigh the risks, but IV iron formulations carry a small risk of hypersensitivity reactions (HSRs). The objective was to use standardized Medical Dictionary for Regulatory Activities queries (SMQs) to compare the safety of ferric derisomaltose/iron isomaltoside 1000 (FDI), iron sucrose (IS), and ferric carboxymaltose (FCM) using prospective trial data.Methods: Prospective trials reporting the incidence of SMQ-coded serious or severe HSRs were identified in the literature. Four SMQs were used: narrow hypersensitivity terms (A), and broad terms pertaining to potential respiratory HSRs (B), skin HSRs (C), and cardiovascular HSRs (D). Bayesian inference, naïve pooling, and adjusted indirect approaches were employed to compare HSR incidence.Results: Twenty one prospective trials including over 8,000 patients receiving FDI, FCM or IS were retrieved. Odds ratios of any serious or severe HSR (all groups) with FDI relative to FCM were 0.41, 0.39, and 0.45 according to the Bayesian, naïve and adjusted approaches, respectively.Conclusions: The risk of serious or severe HSRs was lower with FDI relative to FCM and IS. Using data from prospective trials including over 8,000 patients coded using a well-defined standard (SMQs) enabled a robust comparison of HSR incidence between the iron formulations.
Collapse
Affiliation(s)
- Richard F Pollock
- Health Economics and Outcomes Research, Covalence Research Ltd, London, UK
| | - Patrick Biggar
- Department of Nephrology, Klinikum Coburg GmbH, Coburg, Germany
| |
Collapse
|
18
|
Ng O, Keeler BD, Mishra A, Simpson JA, Neal K, Al‐Hassi HO, Brookes MJ, Acheson AG. Iron therapy for preoperative anaemia. Cochrane Database Syst Rev 2019; 12:CD011588. [PMID: 31811820 PMCID: PMC6899074 DOI: 10.1002/14651858.cd011588.pub3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Preoperative anaemia is common and occurs in 5% to 76% of patients preoperatively. It is associated with an increased risk of perioperative allogeneic blood transfusion, longer hospital stay, and increased morbidity and mortality. Iron deficiency is one of the most common causes of anaemia. Oral and intravenous iron therapy can be used to treat anaemia. Parenteral iron preparations have been shown to be more effective in conditions such as inflammatory bowel disease, chronic heart failure and postpartum haemorrhage due to rapid correction of iron stores. A limited number of studies has investigated iron therapy for the treatment of preoperative anaemia. The aim of this Cochrane Review is to summarise the evidence for iron supplementation, both enteral and parenteral, for the management of preoperative anaemia. OBJECTIVES To evaluate the effects of preoperative iron therapy (enteral or parenteral) in reducing the need for allogeneic blood transfusions in anaemic patients undergoing surgery. SEARCH METHODS We ran the search on 30 July 2018. We searched the Cochrane Injuries Group's Specialised Register, Cochrane Central Register of Controlled Trials (CENTRAL, the Cochrane Library), Ovid MEDLINE(R), Ovid MEDLINE(R) In-Process & Other Non-Indexed Citations, Ovid MEDLINE(R) Daily and Ovid OLDMEDLINE(R), Embase Classic and Embase (Ovid), CINAHL Plus (EBSCO), PubMed, and clinical trials registries, and we screened reference lists. We ran a top-up search on 28 November 2019; one study is now awaiting classification. SELECTION CRITERIA We included all randomised controlled trials (RCTs) that compared preoperative iron monotherapy to placebo, no treatment, standard care or another form of iron therapy for anaemic adults undergoing surgery. We defined anaemia as haemoglobin values less than 13 g/dL for males and 12 g/dL for non-pregnant females. DATA COLLECTION AND ANALYSIS Two review authors collected data and a third review author checked all collected data. Data were collected on the proportion of patients who receive a blood transfusion, the amount of blood transfused per patient (units), quality of life, ferritin levels and haemoglobin levels, measured as continuous variables at the following predetermined time points: pretreatment (baseline), preoperatively but postintervention, and postoperatively. We performed statistical analysis using the Cochrane software, Review Manager 5. We summarised outcome data in tables and forest plots. We used the GRADE approach to describe the quality of the body of evidence. MAIN RESULTS Six RCTs, with a total of 372 participants, evaluated preoperative iron therapy to correct anaemia before planned surgery. Four studies compared iron therapy (either oral (one study) or intravenous (three studies)) with no treatment, placebo or usual care, and two studies compared intravenous iron therapy with oral iron therapy. Iron therapy was delivered over a range of periods that varied from 48 hours to three weeks prior to surgery. The 372 participants in our analysis fall far short of the 819 required - as calculated by our information size calculation - to detect a 30% reduction in blood transfusions. Five trials, involving 310 people, reported the proportion of participants who received allogeneic blood transfusions. Meta-analysis of iron therapy versus placebo or standard care showed no difference in the proportion of participants who received a blood transfusion (risk ratio (RR) 1.21, 95% confidence interval (CI) 0.87 to 1.70; 4 studies, 200 participants; moderate-quality evidence). Only one study that compared oral versus intravenous iron therapy measured this outcome, and reported no difference in risk of transfusion between groups. There was no difference between the iron therapy and placebo/standard care groups for haemoglobin level preoperatively at the end of the intervention (mean difference (MD) 0.63 g/dL, 95% CI -0.07 to 1.34; 2 studies, 83 participants; low-quality evidence). However, intravenous iron therapy produced an increase in preoperative postintervention haemoglobin levels compared with oral iron (MD 1.23 g/dL, 95% CI 0.80 to 1.65; 2 studies, 172 participants; low-quality evidence). Ferritin levels were increased by intravenous iron, both when compared to standard care ((MD 149.00, 95% CI 25.84 to 272.16; 1 study, 63 participants; low-quality evidence) or to oral iron (MD 395.03 ng/mL, 95% CI 227.72 to 562.35; 2 studies, 151 participants; low-quality evidence). Not all studies measured quality of life, short-term mortality or postoperative morbidity. Some measured the outcomes, but did not report the data, and the studies which did report the data were underpowered. Therefore, uncertainty remains regarding these outcomes. The inclusion of new research in the future is very likely to change these results. AUTHORS' CONCLUSIONS The use of iron therapy for preoperative anaemia does not show a clinically significant reduction in the proportion of trial participants who received an allogeneic blood transfusion compared to no iron therapy. Results for intravenous iron are consistent with a greater increase in haemoglobin and ferritin when compared to oral iron, but do not provide reliable evidence. These conclusions are drawn from six studies, three of which included very small numbers of participants. Further, well-designed, adequately powered, RCTs are required to determine the true effectiveness of iron therapy for preoperative anaemia. Two studies are currently in progress, and will include 1500 randomised participants.
Collapse
Affiliation(s)
- Oliver Ng
- University of NottinghamNottingham Digestive Diseases Centre and Biomedical Research UnitDerby RdNottinghamUKNG7 2UH
| | - Barrie D Keeler
- University of NottinghamNottingham Digestive Diseases Centre and Biomedical Research UnitDerby RdNottinghamUKNG7 2UH
| | - Amitabh Mishra
- University of NottinghamNottingham Digestive Diseases Centre and Biomedical Research UnitDerby RdNottinghamUKNG7 2UH
| | - J A Simpson
- University of NottinghamNottingham Digestive Diseases Centre and Biomedical Research UnitDerby RdNottinghamUKNG7 2UH
| | - Keith Neal
- University of NottinghamDepartment of Epidemiology and Public Health188 Alfreton RoadLittle EatonDerbyUKDE21 5AB
| | - Hafid Omar Al‐Hassi
- University of WolverhamptonOffice (MA112b), Wulfruna StreetWolverhamptonUKWV1 1LY
| | | | - Austin G Acheson
- University of NottinghamNottingham Digestive Diseases Centre and Biomedical Research UnitDerby RdNottinghamUKNG7 2UH
| | | |
Collapse
|
19
|
Yan L, Zhao F, Wang J, Zu Y, Gu Z, Zhao Y. A Safe-by-Design Strategy towards Safer Nanomaterials in Nanomedicines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1805391. [PMID: 30701603 DOI: 10.1002/adma.201805391] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 09/13/2018] [Indexed: 05/25/2023]
Abstract
The marriage of nanotechnology and medicine offers new opportunities to fight against human diseases. Benefiting from their unique optical, thermal, magnetic, or redox properties, a wide range of nanomaterials have shown potential in applications such as diagnosis, drug delivery, or tissue repair and regeneration. Despite the considerable success achieved over the past decades, the newly emerging nanomedicines still suffer from an incomplete understanding of their safety risks, and of the relationships between their physicochemical characteristics and safety profiles. Herein, the most important categories of nanomaterials with clinical potential and their toxicological mechanisms are summarized, and then, based on this available information, an overview of the principles in developing safe-by-design nanomaterials for medical applications and of the recent progress in this field is provided. These principles may serve as a starting point to guide the development of more effective safe-by-design strategies and to help identify the major knowledge and skill gaps.
Collapse
Affiliation(s)
- Liang Yan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Nanoscience National Center for Nanoscience and Technology of China, Beijing, 100190, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Feng Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Nanoscience National Center for Nanoscience and Technology of China, Beijing, 100190, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Nanoscience National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Yan Zu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Nanoscience National Center for Nanoscience and Technology of China, Beijing, 100190, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Nanoscience National Center for Nanoscience and Technology of China, Beijing, 100190, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Nanoscience National Center for Nanoscience and Technology of China, Beijing, 100190, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China, Beijing, 100190, China
| |
Collapse
|
20
|
Lim W, Afif W, Knowles S, Lim G, Lin Y, Mothersill C, Nistor I, Rehman F, Song C, Xenodemetropoulos T. Canadian expert consensus: management of hypersensitivity reactions to intravenous iron in adults. Vox Sang 2019; 114:363-373. [PMID: 30937914 PMCID: PMC6850285 DOI: 10.1111/vox.12773] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/31/2019] [Accepted: 02/25/2019] [Indexed: 12/11/2022]
Abstract
Background and Objectives Rare but potentially life‐threatening hypersensitivity reactions can occur during the administration of intravenous iron. To provide guidance to healthcare professionals caring for adults receiving intravenous iron, a panel of 10 Canadian clinical experts developed a practical algorithm for the identification and management of hypersensitivity reactions to intravenous iron. Materials and methods A systematic search of PubMed to February 2018 was performed. Articles related to hypersensitivity reactions were selected for review. The algorithm was developed during a 1‐day live meeting based on the literature review and clinical expertise where evidence was lacking. The algorithm was then refined through an iterative process involving a web‐based platform and virtual meetings. Results The algorithm provides guidance to healthcare professionals in preparing for and administering IV iron, as well as recognizing and managing hypersensitivity reactions to intravenous iron. Considerations for re‐challenging patients who have experienced prior reactions are provided. Conclusion Healthcare professionals who are involved in the care of patients receiving intravenous iron should be trained to anticipate, recognize and manage hypersensitivity reactions to intravenous iron to optimize patient care.
Collapse
Affiliation(s)
- Wendy Lim
- Department of Medicine, Division of Hematology and Thromboembolism, St. Joseph's Healthcare Hamilton, McMaster University, Hamilton, ON, Canada
| | - Waqqas Afif
- Department of Medicine, Division of Gastroenterology and Hepatology, Montreal General Hospital, McGill University, Montreal, QC, Canada
| | - Sandra Knowles
- Department of Pharmacy, Division of Clinical Pharmacology and Toxicology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Gloria Lim
- Department of Medicine, Division of Hematology and Oncology, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Yulia Lin
- Department of Laboratory Medicine and Molecular Diagnostics, Division of Clinical Pathology, Sunnybrook Health Sciences Centre, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Charmaine Mothersill
- Department of Medicine, Division of Hematology and Oncology, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Irina Nistor
- Department of Gastroenterology and Hepatology, Halton Healthcare Services, Oakville, ON, Canada.,Queen's University, Kingston, ON, Canada
| | - Faisal Rehman
- Department of Medicine, Division of Nephrology, University Hospital, London Health Sciences Centre, Western University, London, ON, Canada
| | - Christine Song
- Department of Medicine, Division of Allergy and Immunology, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Ted Xenodemetropoulos
- Department of Medicine, Division of Gastroenterology, McMaster University Medical Centre, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
21
|
Abstract
INTRODUCTION Ferumoxytol is a superparamagnetic molecule originally developed as a contrast agent for magnetic resonance imaging. Elemental iron is contained within the carbohydrate core and is released slowly after infusion allowing a large dose of iron to be administered in a short period of time. Ferumoxytol, originally approved for iron deficiency in chronic kidney disease, received a broad label for any cause of iron deficiency after oral iron intolerance or in those circumstances when oral iron is ineffective or harmful. Areas covered: The chemistry, pharmacology and pharmacokinetics of ferumoxytol were reviewed. Retrospective, observational, and prospective phase II and III trials were reviewed. When appropriate, comparative safety and efficacy parameters were reported. Differentiation between minor infusion reactions and more severe hypersensitivity reactions that may lead to anaphylaxis is described. Expert commentary: Ferumoxytol is a safe and effective iron formulation providing a means of iron repletion in persons with iron deficiency with or without anemia. Relative to iron sucrose, ferric gluconate, and iron dextran and similar to ferric carboxymaltose and iron isomaltoside, ferumoxytol yields relatively low quantities of labile free iron. Hypersensitivity and anaphylaxis is extremely rare. Hypophosphatemia with ferumoxytol's administration is extremely rare. Optimal strategies for application of ferumoxytol-enhanced imaging and full replacement dosing in a single setting remain to be determined.
Collapse
Affiliation(s)
- Michael Auerbach
- a Auerbach Hematology and Oncology , Georgetown University School of Medicine , Baltimore , Maryland , USA
| | - Glenn M Chertow
- b Division of Nephrology , Stanford University School of Medicine , Stanford , California , USA
| | - Mitchell Rosner
- c Department of Medicine , University of Virigina Health System , Charlottesville , Virignia , USA
| |
Collapse
|
22
|
Niepel D, Klag T, Malek NP, Wehkamp J. Practical guidance for the management of iron deficiency in patients with inflammatory bowel disease. Therap Adv Gastroenterol 2018; 11:1756284818769074. [PMID: 29760784 PMCID: PMC5946590 DOI: 10.1177/1756284818769074] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 03/11/2018] [Indexed: 02/04/2023] Open
Abstract
Iron deficiency or iron deficiency anemia (IDA) are some of the most common systemic complications of inflammatory bowel diseases (IBD). Symptoms such as fatigue, reduced ability to concentrate and reduced exercise tolerance can mimic common symptoms of IBD and can therefore easily be overseen. Furthermore, clinicians tend to see mild to moderate anemia as an inevitable accompaniment of IBD that is sufficiently explained by the underlying disease and does not require further workup. But in contrast to these clinical routines, current guidelines recommend that any degree of anemia in patients with IBD should be further evaluated and treated. Multiple studies have shown that anemia is a main factor for decreased quality of life (QoL) in patients with IBD. Correction of anemia, however, can significantly improve the QoL of patients with IBD. It is therefore recommended that every patient with IBD is regularly screened for iron deficiency and anemia. If detected, appropriate workup and treatment should be initiated. Over the last years, a number of new diagnostic tools and treatment options have been developed. Multiple studies have demonstrated the safety of newer formulations of intravenous iron in patients with IBD and have compared oral and intravenous iron in various situations. Treatment recommendations have changed and new evidence-based guidelines were developed. However, to date these guidelines are still not widely implemented in clinical practice. The aim of this review is to draw attention to the need for treatment for every level of anemia in patients with IBD and to provide some practical guidance for screening, diagnostics, treatment and follow up of IDA in patients with IBD following current international guidelines.
Collapse
Affiliation(s)
- Dorothea Niepel
- Department of Internal Medicine I (Gastroenterology, Hepatology, Infectious Diseases), University Hospital Tübingen, Tübingen, Germany
| | - Thomas Klag
- Department of Internal Medicine I (Gastroenterology, Hepatology, Infectious Diseases), University Hospital Tübingen, Tübingen, Germany
| | - Nisar P. Malek
- Department of Internal Medicine I (Gastroenterology, Hepatology, Infectious Diseases), University Hospital Tübingen, Tübingen, Germany
| | | |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Iron deficiency anaemia (IDA) is common and associated with fatigue, reduced quality of life and poorer clinical outcomes. Treatment with oral iron is often inadequate and international guidelines recommend intravenous (i.v.) iron as the preferred option for the treatment of IDA in certain clinical situations. In this review, we assess the safety of using i.v. iron with a particular focus on patients with chronic kidney disease. RECENT FINDINGS Recent publications have raised safety concerns regarding the incidence of serious reactions accompanying i.v. infusion, as well as the subsequent risk of infections and cardiovascular events. Methodological flaws influence the interpretation of these data that lack evidence from the use of modern irons. The latter have been investigated in several randomized control trials. SUMMARY There is a need for better understanding and definition of the nature of i.v. iron reactions, as many are nonserious infusion reactions rather than true anaphylaxis. Retrospective identification of anaphylaxis is difficult and we suggest the importance of reanalysing data using fatalities or standardized terms as outcome measures. With the exception of high molecular weight iron dextran, serious or life-threatening reactions are rare with the use of i.v. irons, and they can be used safely for the treatment of IDA.
Collapse
|
24
|
Abstract
Iron deficiency is common in patients on chronic dialysis, and most require iron-replacement therapy. In addition to absolute iron deficiency, many patients have functional iron deficiency as shown by a suboptimal response to the use of erythropoietin-stimulating agents. Both absolute and functional iron-deficiency anemia have been shown to respond to intravenous (IV) iron replacement. Although parenteral iron is an efficacious method and superior to standard doses of oral iron in patients on hemodialysis, there are ongoing safety concerns about repeated exposure potentially enhancing infection risk and cardiovascular disease. Each IV iron product is composed of an iron core with a carbohydrate shell. The avidity of iron binding and the type of carbohydrate shell play roles in the safe maximal dose and the frequency and severity of acute infusion reactions. All IV iron products are taken up into the reticuloendothelial system where the shell is metabolized and the iron is stored within tissue ferritin or exported to circulating transferrin. IV iron can be given as large intermittent doses (loading therapy) or in smaller doses at frequent intervals (maintenance dosing regimen). Limited trial data and observational data suggest that a maintenance dosing regimen is more efficacious and possibly safer than loading therapy. There is no consensus regarding the preferred method of iron repletion in patients on peritoneal dialysis, although small studies comparing oral and parenteral iron regimens in these patients have shown the latter to be more efficacious. Use of IV iron in virtually all hemodialysis and many peritoneal dialysis patients remains the standard of care.
Collapse
|
25
|
Wu M, Sun D, Tyner K, Jiang W, Rouse R. Comparative Evaluation of U.S. Brand and Generic Intravenous Sodium Ferric Gluconate Complex in Sucrose Injection: In Vitro Cellular Uptake. NANOMATERIALS (BASEL, SWITZERLAND) 2017; 7:E451. [PMID: 29244729 PMCID: PMC5746941 DOI: 10.3390/nano7120451] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/06/2017] [Accepted: 12/11/2017] [Indexed: 12/20/2022]
Abstract
Iron deficiency anemia is a common clinical consequence for people who suffer from chronic kidney disease, especially those requiring dialysis. Intravenous (IV) iron therapy is a widely accepted safe and efficacious treatment for iron deficiency anemia. Numerous IV iron drugs have been approved by U.S. Food and Drug Administration (FDA), including a single generic product, sodium ferric gluconate complex in sucrose. In this study, we compared the cellular iron uptake profiles of the brand (Ferrlecit®) and generic sodium ferric gluconate (SFG) products. We used a colorimetric assay to examine the amount of iron uptake by three human macrophage cell lines. This is the first published study to provide a parallel evaluation of the cellular uptake of a brand and a generic IV iron drug in a mononuclear phagocyte system. The results showed no difference in iron uptake across all cell lines, tested doses, and time points. The matching iron uptake profiles of Ferrlecit® and its generic product support the FDA's present position detailed in the draft guidance on development of SFG complex products that bioequivalence can be based on qualitative (Q1) and quantitative (Q2) formulation sameness, similar physiochemical characterization, and pharmacokinetic bioequivalence studies.
Collapse
Affiliation(s)
- Min Wu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA.
| | - Dajun Sun
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA.
| | - Katherine Tyner
- Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA.
| | - Wenlei Jiang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA.
| | - Rodney Rouse
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA.
| |
Collapse
|
26
|
Roger SD. Practical considerations for iron therapy in the management of anaemia in patients with chronic kidney disease. Clin Kidney J 2017; 10:i9-i15. [PMID: 29225818 PMCID: PMC5716159 DOI: 10.1093/ckj/sfx100] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/02/2017] [Indexed: 01/24/2023] Open
Abstract
Clinical practice guidelines provide both local and global recommendations for the use of iron therapy in the management of anaemia in patients with chronic kidney disease. However, physicians must interpret and adapt these guidelines to meet the specific needs of their individual patients. The recommendations must also be considered in the context of findings from more recently published clinical trials and observational studies.
Collapse
Affiliation(s)
- Simon D Roger
- Department of Renal Medicine, Gosford Hospital, Gosford, NSW, Australia
| |
Collapse
|
27
|
Allen RP, Picchietti DL, Auerbach M, Cho YW, Connor JR, Earley CJ, Garcia-Borreguero D, Kotagal S, Manconi M, Ondo W, Ulfberg J, Winkelman JW. Evidence-based and consensus clinical practice guidelines for the iron treatment of restless legs syndrome/Willis-Ekbom disease in adults and children: an IRLSSG task force report. Sleep Med 2017; 41:27-44. [PMID: 29425576 DOI: 10.1016/j.sleep.2017.11.1126] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 11/09/2017] [Accepted: 11/13/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND Brain iron deficiency has been implicated in the pathophysiology of RLS, and current RLS treatment guidelines recommend iron treatment when peripheral iron levels are low. In order to assess the evidence on the oral and intravenous (IV) iron treatment of RLS and periodic limb movement disorder (PLMD) in adults and children, the International Restless Legs Syndrome Study Group (IRLSSG) formed a task force to review these studies and provide evidence-based and consensus guidelines for the iron treatment of RLS in adults, and RLS and PLMD in children. METHODS A literature search was performed to identify papers appearing in MEDLINE from its inception to July 2016. The following inclusion criteria were used: human research on the treatment of RLS or periodic limb movements (PLM) with iron, sample size of at least five, and published in English. Two task force members independently evaluated each paper and classified the quality of evidence provided. RESULTS A total of 299 papers were identified, of these 31 papers met the inclusion criteria. Four studies in adults were given a Class I rating (one for IV iron sucrose, and three for IV ferric carboxymaltose); only Class IV studies have evaluated iron treatment in children. Ferric carboxymaltose (1000 mg) is effective for treating moderate to severe RLS in those with serum ferritin <300 μg/l and could be used as first-line treatment for RLS in adults. Oral iron (65 mg elemental iron) is possibly effective for treating RLS in those with serum ferritin ≤75 μg/l. There is insufficient evidence to make conclusions on the efficacy of oral iron or IV iron in children. CONCLUSIONS Consensus recommendations based on clinical practice are presented, including when to use oral iron or IV iron, and recommendations on repeated iron treatments. New iron treatment algorithms, based on evidence and consensus opinion have been developed.
Collapse
Affiliation(s)
- Richard P Allen
- Department of Neurology, Johns Hopkins University, Hopkins Bayview Medical Center, Baltimore, MD, USA.
| | - Daniel L Picchietti
- University of Illinois College of Medicine at Urbana-Champaign and Carle Foundation Hospital, Urbana, IL, USA
| | - Michael Auerbach
- Department of Medicine, Georgetown University, Washington DC, USA
| | - Yong Won Cho
- Department of Neurology, Dongsan Medical Center, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - James R Connor
- Department of Neurosurgery, Penn State Hershey Medical Center, Hershey PA, USA
| | - Christopher J Earley
- Department of Neurology, Johns Hopkins University, Hopkins Bayview Medical Center, Baltimore, MD, USA
| | | | - Suresh Kotagal
- Department of Neurology and the Center for Sleep Medicine, Mayo Clinic, Rochester, MN, USA
| | - Mauro Manconi
- Sleep and Epilepsy Center, Neurocenter of Southern Switzerland, Civic Hospital of Lugano, Lugano, Switzerland
| | - William Ondo
- Methodist Neurological Institute, Weill Cornell Medical School Houston, TX, USA
| | - Jan Ulfberg
- Sleep Disorders Department, Capio Health Center, Örebro, Sweden
| | - John W Winkelman
- Departments of Psychiatry and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
28
|
Astier A, Barton Pai A, Bissig M, Crommelin DJA, Flühmann B, Hecq JD, Knoeff J, Lipp HP, Morell-Baladrón A, Mühlebach S. How to select a nanosimilar. Ann N Y Acad Sci 2017; 1407:50-62. [PMID: 28715605 DOI: 10.1111/nyas.13382] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/10/2017] [Accepted: 04/20/2017] [Indexed: 12/24/2022]
Abstract
Nanomedicines in the class of nonbiological complex drugs (NBCDs) are becoming increasingly available. Up to 23 nanomedicines have been approved, and approximately 50 are in clinical development. Meanwhile, the first nanosimilars have entered the market through the generic approval pathway, but clinical differences have been observed. Many healthcare professionals may be unaware of this issue and must be informed of these clinically relevant variances. This article provides a tool for rational decision making for the inclusion of nanomedicines into the hospital formulary, including defined criteria for evaluation of substitutability or interchangeability. The tool was generated by conducting a roundtable with an international panel of experts and follows the same thought process that was developed and published earlier for the selection of biologicals/biosimilars. In addition to the existing criteria for biosimilars, a set of seven criteria was identified that specifically apply to nanosimilars. These include (1) particle size and size distribution, (2) particle surface characteristics, (3) fraction of uncaptured bioactive moiety, (4) stability on storage, (5) bioactive moiety uptake and (6) distribution, and (7) stability for ready-to-use preparation. Pharmacists should utilize their pharmaceutical expertise to use the appropriate criteria to evaluate the comparability of the drug to decide on interchangeability or substitutability.
Collapse
Affiliation(s)
- Alain Astier
- Department of Pharmacy, Henri Mondor University Hospitals, Créteil, France
| | - Amy Barton Pai
- Department of Clinical Pharmacy, University of Michigan, Ann Arbor, Michigan
| | - Marco Bissig
- Hospital Pharmacy, Ospedale Regionale di Lugano, Lugano, Switzerland
| | - Daan J A Crommelin
- Deptartment of Pharmaceutical Sciences, Utrecht University, the Netherlands
| | | | - Jean-Daniel Hecq
- Hospital Pharmacy, University Hospital of Mont-Godinne, Yvoir, Belgium
| | - Josefien Knoeff
- Vifor Pharma Ltd., Glattbrugg, Switzerland.,Faculty of Sciences, Vrije Universiteit Amsterdam, the Netherlands
| | | | | | - Stefan Mühlebach
- Vifor Pharma Ltd., Glattbrugg, Switzerland.,Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
29
|
Lee D, Barsky D, Hughes R, Driggers J, Poupard N, Mascarenhas M. Evaluation of the safety of iron dextran with parenteral nutrition in the paediatric inpatient setting. Nutr Diet 2017; 74:471-475. [PMID: 28758365 DOI: 10.1111/1747-0080.12355] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/04/2017] [Accepted: 03/12/2017] [Indexed: 11/30/2022]
Abstract
AIM Practitioners often avoid administering iron dextran in parenteral nutrition (PN) for hospitalised children because of the concern for anaphylaxis. The primary aim of the present study was to determine the risk of anaphylaxis associated with exposure to PN containing iron dextran in the inpatient setting. METHODS Charts were reviewed for all children admitted to The Children's Hospital of Philadelphia from January 1, 2011 to December 30, 2013 who received PN containing low molecular weight (LMW) iron dextran. Subject characteristics, primary diagnoses and PN orders were evaluated. The pharmacy adverse events database was queried for adverse drug reactions. RESULTS Over three years, 89 subjects received PN containing a maintenance dose of LMW iron dextran with a total of 2774 days of exposure. Subjects ranged from two months to 21 years of age and received between 1 and 196 days of PN containing iron dextran. The mean dose of iron dextran in children decreased as the weight category increased from <5 kg (0.21 ± 0.05 mg/kg/day) to ≥40 kg (1.9 ± 0.5 mg/day; P-value for trend <0.005). No anaphylactic reactions occurred in any subjects. CONCLUSIONS PN containing a maintenance dose of LMW iron dextran can be safely administered to hospitalised children, and further studies are need to evaluate the potential to prevent iron deficiency anaemia and the need for additional IV iron infusions.
Collapse
Affiliation(s)
- Dale Lee
- Division of Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, Washington, USA
| | - Diane Barsky
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Rachel Hughes
- Department of Pediatrics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA
| | - Julia Driggers
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nicole Poupard
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Maria Mascarenhas
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
30
|
Iyoke CA, Emegoakor FC, Ezugwu EC, Lawani LO, Ajah LO, Madu JA, Ezegwui HU, Ezugwu FO. Effect of treatment with single total-dose intravenous iron versus daily oral iron(III)-hydroxide polymaltose on moderate puerperal iron-deficiency anemia. Ther Clin Risk Manag 2017; 13:647-653. [PMID: 28761350 PMCID: PMC5493201 DOI: 10.2147/tcrm.s112227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background Iron-deficiency anemia is the most common nutritional cause of anemia in pregnancy and is often responsible for puerperal anemia. Puerperal anemia can impair postpartum maternal and neonatal well-being. Objective To determine the effect of treatment of moderate puerperal iron-deficiency anemia using a single intravenous total-dose iron dextran versus daily single dose oral iron(III)-hydroxide polymaltose. Methodology A randomized controlled study in which postpartum women with moderate iron-deficiency anemia were randomized into treatment with either a single total-dose intravenous iron dextran or with daily single doses of oral iron(III)-hydroxide polymaltose tablets for 6 weeks. Effects on hemoglobin concentration using either method were compared at 6 weeks postpartum. Analysis was per protocol using SPSS version 17 for windows. P-values ≤0.05 were considered significant. Results Two hundred eighty-four women were recruited for the study: 142 women received single total dose intravenous infusion of iron dextran while 142 received daily oral iron(III)-hydroxide polymaltose tablets. Approximately 84.0% (237/282) completed the study and were analyzed including 81% (115/142) of those randomized to injectable iron therapy compared to 85.9% (122/142) of those randomized to oral treatment. The proportions of women who had attained hemoglobin concentration of at least 10 g/dL by the 6 weeks postpartum visit did not differ significantly between cases and controls (95.7% vs 94.3%; P=0.73). Similarly, the mean increases in hemoglobin following either therapeutic route were comparable (1.03±0.56 g/dL for intravenous iron and 0.97±0.46 g/dL for the oral group; P=0.42). Conclusion Single total-dose intravenous iron for treatment of puerperal iron-deficiency anemia was as effective as daily single doses of ferric iron tablets. For puerperal patients with iron-deficiency anemia in whom compliance with and tolerability of oral iron are not certain, a single total-dose intravenous iron can be safely offered.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hyginus Uzo Ezegwui
- Department of Obstetrics and Gynaecology, University of Nigeria, Enugu Campus
| | - Frank Okechukwu Ezugwu
- Department of Obstetrics and Gynaecology, College of Medicine, Enugu State University, Enugu, Nigeria
| |
Collapse
|
31
|
Auerbach M, Macdougall I. The available intravenous iron formulations: History, efficacy, and toxicology. Hemodial Int 2017; 21 Suppl 1:S83-S92. [DOI: 10.1111/hdi.12560] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Michael Auerbach
- Auerbach Hematology and Oncology, Private Practice; Rosedale Maryland USA
- Georgetown University School of Medicine; Washington, District of Columbia USA
| | | |
Collapse
|
32
|
In search of alternatives to dopaminergic ligands for the treatment of restless legs syndrome: iron, glutamate, and adenosine. Sleep Med 2017; 31:86-92. [DOI: 10.1016/j.sleep.2016.08.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 08/19/2016] [Accepted: 08/20/2016] [Indexed: 11/21/2022]
|
33
|
Abstract
Given the high comorbidity in patients on hemodialysis and the complexity of the dialysis treatment, it is remarkable how rarely a life-threatening complication occurs during dialysis. The low rate of dialysis emergencies can be attributed to numerous safety features in modern dialysis machines; meticulous treatment and testing of the dialysate solution to prevent exposure to trace elements, toxins, and pathogens; adherence to detailed treatment protocols; and extensive training of dialysis staff to handle medical emergencies. Most hemodialysis emergencies can be attributed to human error. A smaller number are due to rare idiosyncratic reactions. In this review, we highlight major emergencies that may occur during hemodialysis treatments, describe their pathogenesis, offer measures to minimize them, and provide specific interventions to prevent catastrophic consequences on the rare occasions when such emergencies arise. These emergencies include dialysis disequilibrium syndrome, venous air embolism, hemolysis, venous needle dislodgement, vascular access hemorrhage, major allergic reactions to the dialyzer or treatment medications, and disruption or contamination of the dialysis water system. Finally, we describe root cause analysis after a dialysis emergency has occurred to prevent a future recurrence.
Collapse
Affiliation(s)
- Manish Saha
- Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
| | | |
Collapse
|
34
|
Wetmore JB, Weinhandl ED, Zhou J, Gilbertson DT. Relative Incidence of Acute Adverse Events with Ferumoxytol Compared to Other Intravenous Iron Compounds: A Matched Cohort Study. PLoS One 2017; 12:e0171098. [PMID: 28135334 PMCID: PMC5279762 DOI: 10.1371/journal.pone.0171098] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/16/2017] [Indexed: 01/23/2023] Open
Abstract
Concerns persist about adverse reactions to intravenous (IV) iron. We aimed to determine the relative safety of ferumoxytol compared to other IV iron compounds. This retrospective cohort study with propensity-score matching for patients and drug doses used the Medicare 20% random sample to identify patients (1) without chronic kidney disease (non-CKD) and (2) with non-dialysis-dependent chronic kidney disease (NDD-CKD) who received a first dose of IV iron in 2010-2012. Exposures were ferumoxytol, iron sucrose, sodium ferric gluconate, or iron dextran. Outcomes were hypersensitivity symptoms, anaphylaxis, emergency department (ED) encounters, hospitalizations, and death after acute IV iron exposure. In the primary analysis for reactions on the day of or following exposure, there was no difference in hypersensitivity symptoms (hazard ratio 1.04, 95% confidence interval 0.94-1.16) or hypotension (0.83, 0.52-1.34) between 4289 non-CKD ferumoxytol users and an equal number of users of other compounds; results were similar for 7358 NDD-CKD patients and an equal number of controls. All-cause ED encounters or hospitalizations were less common in both the non-CKD (0.56, 0.45-0.70) and NDD-CKD ferumoxytol-treated patients (0.83, 0.71-0.95). Fewer than 10 deaths occurred in both the non-CKD and NDD-CKD ferumoxytol users and in matched controls; the hazard for death did not differ significantly between ferumoxytol users and controls in the non-CKD patients (2.00, 0.33-11.97) or in the NDD-CKD patients (0.25, 0.04-1.52). Multiple sensitivity analyses showed similar results. Ferumoxytol did not appear to be associated with more adverse reactions than other compounds for the treatment of iron-deficiency anemia in both non-CKD and NDD-CKD patients.
Collapse
Affiliation(s)
- James B. Wetmore
- Chronic Disease Research Group, Minneapolis Medical Research Foundation, Minneapolis, Minnesota, United States of America
- Division of Nephrology, Hennepin County Medical Center, Minneapolis, Minnesota, United States of America
| | - Eric D. Weinhandl
- Chronic Disease Research Group, Minneapolis Medical Research Foundation, Minneapolis, Minnesota, United States of America
| | - Jincheng Zhou
- Chronic Disease Research Group, Minneapolis Medical Research Foundation, Minneapolis, Minnesota, United States of America
| | - David T. Gilbertson
- Chronic Disease Research Group, Minneapolis Medical Research Foundation, Minneapolis, Minnesota, United States of America
| |
Collapse
|
35
|
Knoeff J, Flühmann B, Mühlebach S. Medication practice in hospitals: are nanosimilars evaluated and substituted correctly? Eur J Hosp Pharm 2017; 25:79-84. [PMID: 29599970 PMCID: PMC5868288 DOI: 10.1136/ejhpharm-2016-001059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/26/2016] [Accepted: 12/20/2016] [Indexed: 11/04/2022] Open
Abstract
Introduction This study investigates the drug selection and dispensing behaviour of hospital pharmacists of intravenous iron products including iron sucrose and iron sucrose similar, with special emphasis on substitution and interchangeability in France and Spain. Iron-carbohydrate complex drugs represent different available intravenous iron drugs and are part of the non-biological complex drug (NBCD) class, an expanding drug class with up to 30 brands available in intravenous pharmacotherapy and over 50 in clinical development. Follow-on versions of iron sucrose have appeared in some markets such as France and Spain, which were authorised by the generic approval pathway. However, differences in clinical efficacy and safety of iron sucrose similars compared with the reference originator drug Venofer have been observed, putting a question mark on their equivalence as assessed for authorisation and consequently their substitutability and interchangeability. Method 70 French and 70 Spanish hospital pharmacists were surveyed via an online questionnaire on their formulary selection and dispensing behaviour of intravenous iron medicines. Results There is little awareness about the characteristics of this class of drugs and the reported differences in safety and efficacy between iron sucrose and iron sucrose similars. In approximately 85% of cases the intravenous iron is chosen according to the hospital formulary. In 30% (France) and 34% (Spain) of cases an iron sucrose similar was dispensed because the formulary requires dispensing an alternative lower cost drug when available. In 26% (France) and 52% (Spain) of cases the physician is not informed on such a medication change using a similar product. Conclusions Evaluation of NBCD similars for substitution and interchange by hospital pharmacists is rarely based on scientific and clinical criteria but rather on cost aspects only, which does not ensure safe, efficacious and cost-effective use of such drugs.
Collapse
Affiliation(s)
- Josefien Knoeff
- Vifor Pharma Ltd., Glattbrugg, Switzerland.,Faculty of Sciences, Vrije Universiteit Amsterdam, Amsterdam, Noord-Holland, The Netherlands
| | | | - Stefan Mühlebach
- Vifor Pharma Ltd., Glattbrugg, Switzerland.,Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
36
|
Alan Brookhart M, Li X, Kshirsagar AV. What are the Considerations in Balancing Benefits and Risks in Iron Treatment?: Balancing Benefits and Safety with Intravenous Iron Treatment. Semin Dial 2016; 30:25-27. [PMID: 27766683 DOI: 10.1111/sdi.12550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- M Alan Brookhart
- Department of Epidemiology, UNC Gillings School of Global Public Health, UNC Chapel Hill, Chapel Hill, North Carolina.,Cecil G. Sheps Center for Health Services Research, University of North Carolina, Chapel Hill, North Carolina
| | - Xiaojuan Li
- Department of Epidemiology, UNC Gillings School of Global Public Health, UNC Chapel Hill, Chapel Hill, North Carolina.,UNC School of Medicine, University of North Carolina Kidney Center, Chapel Hill, North Carolina
| | - Abhijit V Kshirsagar
- UNC School of Medicine, University of North Carolina Kidney Center, Chapel Hill, North Carolina
| |
Collapse
|
37
|
Wong L, Smith S, Gilstrop M, Derman R, Auerbach S, London N, Lenowitz S, Bahrain H, McClintock J, Auerbach M. Safety and efficacy of rapid (1,000 mg in 1 hr) intravenous iron dextran for treatment of maternal iron deficient anemia of pregnancy. Am J Hematol 2016; 91:590-3. [PMID: 26971581 DOI: 10.1002/ajh.24361] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 03/05/2016] [Accepted: 03/07/2016] [Indexed: 11/09/2022]
Abstract
Maternal iron deficiency anemia (IDA) is associated with risk of adverse perinatal outcomes. Oral iron is recommended to reverse anemia, but has gastrointestinal toxicity and frequent non-adherence. Intravenous (IV) iron is reserved for intolerance of, or unresponsiveness to, oral therapy, malabsorption, and severe anemia (1% with hemoglobin [Hgb] levels <7 g/dL). With rare (<100 per one million) adverse events (AEs) ability to infuse a sufficient dose of low molecular weight iron dextran (LMWID) over 60 min, LMWID is an attractive option. This study demonstrated safety and efficacy of rapid IV infusion of 1,000 mg LMWID to gravidas with moderate to severe IDA. An observational treatment study of 1,000 mg LMWID administered over 1 hr for IDA in 189 consecutive, unselected second and third trimester gravidas after oral iron failure was conducted. All received a test dose of 25 mg LMWID and were monitored for AEs during the 60-min infusion. No premedication was administered unless more than one drug allergy or asthma was present in which case IV methylprednisolone was administered. All were followed through pregnancy and delivery. Monitored parameters included Hgb, mean corpuscular volume, serum ferritin, and percent transferrin saturation. About 189 subjects received 1,000 mg LMWID. No serious AEs occurred. About 2% experienced transient infusion reactions. Hgb improved by 1-1.9 g/dL in 82% and ≥2 g/dL in 24%. Second trimester treatment was not associated with greater Hgb improvement than third trimester treatment. Anemia resolved in 95%. Administration of a single large dose of IV LMWID was effective, safe, and convenient. Am. J. Hematol. 91:590-593, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lee Wong
- Department of Obstetrics and Gynecology; MedStar Franklin Square Medical Center; Baltimore Maryland
| | - Samuel Smith
- Department of Obstetrics and Gynecology; MedStar Franklin Square Medical Center; Baltimore Maryland
| | - Marisa Gilstrop
- Department of Obstetrics and Gynecology; Christiana Care Health System; Newark Delaware
| | - Richard Derman
- Department of Obstetrics and Gynecology; Christiana Care Health System; Newark Delaware
| | - Sarah Auerbach
- West Virginia School of Osteopathic Medicine; Lewisburg West Virginia
| | - Nicola London
- Department of Obstetrics and Gynecology; MedStar Franklin Square Medical Center; Baltimore Maryland
| | - Steven Lenowitz
- Department of Obstetrics and Gynecology; MedStar Franklin Square Medical Center; Baltimore Maryland
| | | | - Jessica McClintock
- Department of Obstetrics and Gynecology; MedStar Franklin Square Medical Center; Baltimore Maryland
| | - Michael Auerbach
- Department of Medicine; Georgetown University School of Medicine; Washington DC
| |
Collapse
|
38
|
DeLoughery TG, Auerbach M. Is low-molecular weight iron dextran really the most risky iron?--Unconvincing data from an unconvincing study. Am J Hematol 2016; 91:451-2. [PMID: 26873795 DOI: 10.1002/ajh.24326] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 02/03/2016] [Indexed: 01/17/2023]
Affiliation(s)
- Thomas G. DeLoughery
- Division of Hematology/Medical Oncology, Department of Medicine; Oregon Health Sciences University; Portland Oregon
- Division of Laboratory Medicine, Department of Pathology; Oregon Health Sciences University; Portland Oregon
| | - Michael Auerbach
- Georgetown University; Washington District of Columbia and Private practice, Baltimore, Maryland
| |
Collapse
|
39
|
Mhaskar R, Wao H, Miladinovic B, Kumar A, Djulbegovic B. The role of iron in the management of chemotherapy-induced anemia in cancer patients receiving erythropoiesis-stimulating agents. Cochrane Database Syst Rev 2016; 2:CD009624. [PMID: 26845108 PMCID: PMC8765740 DOI: 10.1002/14651858.cd009624.pub2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Erythropoiesis-stimulating agents (ESAs) are commonly used to treat chemotherapy-induced anemia (CIA). However, about half of patients do not benefit. OBJECTIVES To evaluate the benefits and harms related to the use of iron as a supplement to ESA and iron alone compared with ESA alone in the management of CIA. SEARCH METHODS We searched for relevant trials from the Cochrane Central Register of Controlled Trials (CENTRAL) (issue 1 January 2016), MEDLINE (1950 to February 2016), and www.clinicaltrials.gov without using any language limits. SELECTION CRITERIA All randomized controlled trials (RCTs) comparing 'iron plus ESA' or 'iron alone' versus 'ESA alone' in people with CIA were eligible for inclusion. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included eight RCTs (12 comparisons) comparing ESA plus iron versus ESA alone enrolling 2087 participants. We did not find any trial comparing iron alone versus ESAs alone in people with CIA. None of the included RCTs reported overall survival. There was a beneficial effect of iron supplementation to ESAs compared with ESAs alone on hematopoietic response (risk ratio (RR) 1.17, 95% confidence interval (CI) 1.09 to 1.26; P < 0.0001; 1712 participants; 11 comparisons; high-quality evidence). Assuming a baseline risk of 35% to 80% for hematopoietic response without iron supplementation, between seven and 16 patients should be treated to achieve hematopoietic response in one patient. In subgroup analyses, RCTs that used intravenous (IV) iron favored ESAs and iron (RR 1.20 (95% CI 1.10 to 1.31); P < 0.00001; 1321 participants; eight comparisons), whereas we found no evidence for a difference in hematopoietic response in RCTs using oral iron (RR 1.04 (95% CI 0.87 to 1.24); P = 0.68; 391 participants; three comparisons). There was no evidence for a difference between the subgroups of IV and oral iron (P = 0.16). There was no evidence for a difference between the subgroups of types of iron (P = 0.31) and types of ESAs (P = 0.16) for hematopoietic response.The iron supplementation to ESAs might be beneficial as fewer participants treated with iron supplementation required red blood cell (RBC) transfusions compared to the number of participants treated with ESAs alone (RR 0.74 (95% CI 0.60 to 0.92); P = 0.007; 1719 participants; 11 comparisons; moderate-quality evidence). Assuming a baseline risk of 7% to 40% for RBC transfusion without iron supplementation, between 10 and 57 patients should be treated to avoid RBC transfusion in one patient.We found no evidence for a difference in the median time to hematopoietic response with addition of iron to ESAs (hazard ratio (HR) 0.93 (95% CI 0.67 to 1.28); P = 0.65; 1042 participants; seven comparisons; low-quality evidence). In subgroup analyses, RCTs in which dextran (HR 0.95 (95% CI 0.36 to 2.52); P = 0.92; 340 participants; three comparisons), sucrose iron (HR 1.15 (95% CI 0.60 to 2.21); P = 0.67; 102 participants; one comparison) and sulfate iron (HR 1.24 (95% CI 0.99 to 1.56); P = 0.06; 55 participants; one comparison) were used showed no evidence for difference between iron supplementation versus ESAs alone compared with RCTs in which gluconate (HR 0.78 (95% CI 0.65 to 0.94); P = 0.01; 464 participants; two comparisons) was used for median time to hematopoietic response (P = 0.02). There was no evidence for a difference between the subgroups of route of iron administration (P = 0.13) and types of ESAs (P = 0.46) for median time to hematopoietic response.Our results indicated that there could be improvement in the hemoglobin (Hb) levels with addition of iron to ESAs (mean difference (MD) 0.48 (95% CI 0.10 to 0.86); P = 0.01; 827 participants; seven comparisons; low-quality evidence). In RCTs in which IV iron was used there was evidence for a difference (MD 0.84 (95% CI 0.21 to 1.46); P = 0.009; 436 participants; four comparisons) compared with oral iron (MD 0.07 (95% CI -0.19 to 0.34); P = 0.59; 391 participants; three comparisons) for mean change in Hb level (P = 0.03). RCTs in which dextran (MD 1.55 (95% CI 0.62 to 2.47); P = 0.001; 102 participants; two comparisons) was used showed evidence for a difference with iron supplementation versus ESAs alone compared with RCTs in which gluconate (MD 0.54 (95% CI -0.15 to 1.22); P = 0.12; 334 participants; two comparisons) and sulfate iron (MD 0.07 (95% CI -0.19 to 0.34); P = 0.59; 391 participants; three comparisons) were used for mean change in Hb level (P = 0.007). RCTs in which epoetin was used showed evidence for a difference with iron supplementation versus ESAs alone (MD 0.77 (95% CI 0.25 to 1.29); P = 0.004; 337 participants; five comparisons) compared with darbepoetin use (MD 0.10 (95% CI -0.13 to 0.33); P = 0.38; 490 participants; two comparisons) for mean change in Hb level (P = 0.02).We found no evidence for a difference in quality of life with addition of iron to ESAs (standardized mean difference 0.01 (95% CI -0.10 to 0.12); P = 0.88; 1124 participants; three RCTs; high-quality evidence).We found no evidence for a difference in risk of grade III-IV thromboembolic events (RR 0.95 (95% CI 0.54 to 1.65); P = 0.85; 783 participants; three RCTs; moderate-quality evidence). The incidence of treatment-related mortality (TRM) was 0% (997 participants; four comparisons; high-quality evidence).Other common adverse events included vomiting, asthenia, and leukopenia, and were similar in both arms.Overall the risk of bias across outcomes was high to low. Since the included RCTs had shorter follow-up duration (up to 20 weeks), the long-term effects of iron supplementation are unknown. Our main reasons for downgrading the quality of evidence were inconsistency across the included studies and imprecision of results. AUTHORS' CONCLUSIONS Our systematic review shows that addition of iron to ESAs offers superior hematopoietic response, reduces the risk of RBC transfusions, and improves Hb levels, and appears to be well tolerated. None of the included RCTs reported overall survival. We found no evidence for a difference in quality of life with iron supplementation.
Collapse
Affiliation(s)
- Rahul Mhaskar
- University of South FloridaCenter for Evidence Based Medicine and Health Outcomes ResearchTampaFloridaUSA
| | - Hesborn Wao
- University of South FloridaCenter for Evidence Based Medicine and Health Outcomes ResearchTampaFloridaUSA
| | - Branko Miladinovic
- University of South FloridaCenter for Evidence Based Medicine and Health Outcomes ResearchTampaFloridaUSA
| | - Ambuj Kumar
- University of South FloridaCenter for Evidence Based Medicine and Health Outcomes ResearchTampaFloridaUSA
| | - Benjamin Djulbegovic
- Moffitt Cancer Center, Division of Oncologic Sciences, University of South FloridaDepartment of Blood and Marrow TransplantationTampaFloridaUSA
| | | |
Collapse
|
40
|
Del Vecchio L, Longhi S, Locatelli F. Safety concerns about intravenous iron therapy in patients with chronic kidney disease. Clin Kidney J 2016; 9:260-7. [PMID: 26985378 PMCID: PMC4792617 DOI: 10.1093/ckj/sfv142] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 11/20/2015] [Indexed: 01/12/2023] Open
Abstract
Anaemia in chronic kidney disease (CKD) is managed primarily with erythropoiesis-stimulating agents (ESAs) and iron therapy. Following concerns around ESA therapy, intravenous (IV) iron is being administered more and more worldwide. However, it is still unclear whether this approach is safe at very high doses or in the presence of very high ferritin levels. Some observational studies have shown a relationship between either high ferritin level or high iron dose and increased risk of death, cardiovascular events, hospitalization or infection. Others have not been able to confirm these findings. However, they suffer from indication biases. On the other hand, the majority of randomized clinical trials have only a very short follow-up (and thus drug exposure) and are inadequate to assess the mortality risk. None of them have tested the role of different iron doses on hard end points. With the lack of clear evidence coming from well-designed and large-scale studies, several data suggest that excessive iron therapy may be toxic in several aspects, ranging from iron overload to tissue damage from labile iron. A number of experimental and clinical data suggest that either excessive iron therapy or iron overload may be a possible culprit of atherogenesis. The process seems to be mediated by oxidative stress. Iron therapy should also be used cautiously in the presence of active infections, since iron is essential for bacterial growth. Recently, the European Medicines Agency officially raised concerns about rare hypersensitivity reactions following IV iron administration. The balance has been in favour of benefits. In several European countries, this has created a lot of confusion and somewhat slowed the run towards excessive use. Altogether, IV iron remains a mainstay of anaemia treatment in CKD patients. However, in our opinion, its excessive use should be avoided, especially in patients with high ferritin levels and when ESA agents are not contraindicated.
Collapse
Affiliation(s)
- Lucia Del Vecchio
- Department of Nephrology and Dialysis , A. Manzoni Hospital , Lecco , Italy
| | - Selena Longhi
- Department of Nephrology and Dialysis , A. Manzoni Hospital , Lecco , Italy
| | | |
Collapse
|
41
|
Ng O, Keeler BD, Mishra A, Simpson A, Neal K, Brookes MJ, Acheson AG. Iron therapy for pre-operative anaemia. Cochrane Database Syst Rev 2015:CD011588. [PMID: 26694949 DOI: 10.1002/14651858.cd011588.pub2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Pre-operative anaemia is common and occurs in up to 76% of patients. It is associated with increased peri-operative allogeneic blood transfusions, longer hospital lengths of stay and increased morbidity and mortality. Iron deficiency is one of the most common causes of this anaemia. Oral iron therapy has traditionally been used to treat anaemia but newer, safer parenteral iron preparations have been shown to be more effective in other conditions such as inflammatory bowel disease, chronic heart failure and post-partum haemorrhage. A limited number of studies look at iron therapy for the treatment of pre-operative anaemia. The aim of this Cochrane review is to summarise the evidence for use of iron supplementation, both enteral and parenteral, for the management of pre-operative anaemia. OBJECTIVES The objective of this review is to evaluate the effects of pre-operative iron therapy (enteral or parenteral) in reducing the need for allogeneic blood transfusions in anaemic patients undergoing surgery. SEARCH METHODS We ran the search on 25 March 2015. We searched the Cochrane Injuries Group's Specialised Register, Cochrane Central Register of Controlled Trials (CENTRAL, The Cochrane Library), Ovid MEDLINE(R), Ovid MEDLINE(R) In-Process & Other Non-Indexed Citations, Ovid MEDLINE(R) Daily and Ovid OLDMEDLINE(R), EMBASE Classic and EMBASE (Ovid), CINAHL Plus (EBSCO), PubMed, clinical trials registries, conference abstracts, and we screened reference lists. SELECTION CRITERIA We included all randomised controlled trials (RCTs) which compared pre-operative iron monotherapy to placebo, no treatment, standard of care or another form of iron therapy for anaemic adults undergoing surgery. Anaemia was defined by haemoglobin values less than 13 g/dL for males and 12 g/dL for non-pregnant females. DATA COLLECTION AND ANALYSIS Data were collected by two authors on the proportion of patients who receive a blood transfusion, amount of blood transfused per patient (units) and haemoglobin measured as continuous variables at pre-determined time-points: pre-treatment, pre-operatively but post-treatment, and post-operatively. Statistical analysis was performed using the Cochrane statistical software, Review Manager 2014. Outcome data were summarised in tables and a forest plot. MAIN RESULTS Three prospective randomised controlled studies evaluated pre-operative iron therapy to correct anaemia (two in colorectal and one in gynaecological surgery) and included 114 patients in total. One compared oral iron versus standard care (Lidder 2007); one intravenous iron versus control (Edwards 2009); and one study compared oral versus intravenous iron (Kim 2009). Both colorectal trials reported the primary outcome (proportion of patients who received allogeneic blood transfusions) and meta-analysis showed a reduction in blood transfusions with the administration of iron therapy, but the reduction was not statistically significant (risk ratio (RR) 0.56, 95% confidence interval (CI) 0.27 to 1.18). All studies reported haemoglobin change but data for the anaemic patients were only available for two studies (Edwards 2009 and Kim 2009). Edwards 2009 showed no difference in haemoglobin at the end of treatment pre-operatively. The intravenous versus oral iron study showed an increase in haemoglobin with intravenous iron at the end of treatment pre-operatively (MD 1.90 g/dL, 95% CI 1.16 to 2.64; participants = 56), but the results are at high risk of bias because participants with less than 80% compliance with therapy were excluded from the analysis and compliance was lower in the oral iron group due to the side-effects of treatment (Kim 2009).None of the studies reported quality of life, short- or long-term mortality or post-operative morbidity. AUTHORS' CONCLUSIONS The use of iron therapy for pre-operative anaemia does not show a statistically significant reduction in the proportion of patients who received an allogeneic blood transfusion compared to no iron therapy. However, the 38 patients in our analysis falls far short of the 819 patients our information size calculation recommended to detect a 30% reduction in blood transfusions. Intravenous iron may be more effective than oral iron at increasing haemoglobin. However, all these conclusions are drawn from only three small randomised controlled studies. Further well designed, adequately powered randomised controlled studies are required to determine the true effectiveness of iron therapy for pre-operative anaemia.
Collapse
Affiliation(s)
- Oliver Ng
- Nottingham Digestive Disease Centre, E Floor West Block, Derby Rd, Nottingham, UK, NG7 2UH
| | | | | | | | | | | | | |
Collapse
|
42
|
Szebeni J, Fishbane S, Hedenus M, Howaldt S, Locatelli F, Patni S, Rampton D, Weiss G, Folkersen J. Hypersensitivity to intravenous iron: classification, terminology, mechanisms and management. Br J Pharmacol 2015; 172:5025-36. [PMID: 26265306 DOI: 10.1111/bph.13268] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 07/20/2015] [Accepted: 07/23/2015] [Indexed: 12/19/2022] Open
Abstract
Intravenous (IV) iron therapy is widely used in iron deficiency anaemias when oral iron is not tolerated or ineffective. Administration of IV-iron is considered a safe procedure, but severe hypersensitivity reactions (HSRs) can occur at a very low frequency. Recently, new guidelines have been published by the European Medicines Agency with the intention of making IV-iron therapy safer; however, the current protocols are still non-specific, non-evidence-based empirical measures which neglect the fact that the majority of IV-iron reactions are not IgE-mediated anaphylactic reactions. The field would benefit from new specific and effective methods for the prevention and treatment of these HSRs, and the main goal of this review was to highlight a possible new approach based on the assumption that IV-iron reactions represent complement activation-related pseudo-allergy (CARPA), at least in part. The review compares the features of IV-iron reactions to those of immune and non-immune HSRs caused by a variety of other infused drugs and thus make indirect inferences on IV-iron reactions. The process of comparison highlights many unresolved issues in allergy research, such as the unsettled terminology, multiple redundant classifications and a lack of validated animal models and lege artis clinical studies. Facts and arguments are listed in support of the involvement of CARPA in IV-iron reactions, and the review addresses the mechanism of low reactogenic administration protocols (LRPs) based on slow infusion. It is suggested that consideration of CARPA and the use of LRPs might lead to useful new additions to the management of high-risk IV-iron patients.
Collapse
Affiliation(s)
- J Szebeni
- Nanomedicine Research and Education Center, Semmelweis University, Budapest, Hungary.,Department of Nanobiotechnology and Regenerative Medicine, Miskolc University, Miskolc, Hungary
| | - S Fishbane
- Hofstra North Shore-LIJ School of Medicine, New York, NY, USA
| | - M Hedenus
- Department for Internal Medicine, Sundsvalls Hospital, Sundsvall, Sweden
| | - S Howaldt
- Research Institute for IBD, HaFCED GmbH & Co. KG, Hamburg, Germany
| | - F Locatelli
- Department of Nephrology, Alessandro Manzoni Hospital, Lecco, Italy
| | - S Patni
- Birmingham Heartlands Hospital, Birmingham, UK
| | - D Rampton
- Barts and the London School of Medicine and Dentistry, London, UK
| | - G Weiss
- Department for Internal Medicine VI, Medical University of Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
43
|
Affiliation(s)
- Yuanzeng Min
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Joseph M Caster
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Michael J Eblan
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
44
|
Ng O, Keeler B, Mishra A, Simpson A, Neal K, Brookes MJ, Acheson AG. Iron therapy for pre-operative anaemia. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2015. [DOI: 10.1002/14651858.cd011588] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
45
|
Bircher AJ, Auerbach M. Hypersensitivity from intravenous iron products. Immunol Allergy Clin North Am 2015; 34:707-23, x-xi. [PMID: 25017687 DOI: 10.1016/j.iac.2014.04.013] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In the last several years, intravenous therapy with iron products has been more widely used. Although it has been a standard procedure in dialysis-associated anemia since the early 1990s, its use is expanding to a host of conditions associated with iron deficiency, especially young women with heavy uterine bleeding and pregnancy. Free iron is associated with unacceptable high toxicity inducing severe, hemodynamically significant symptoms. Subsequently, formulations that contain the iron as an iron carbohydrate nanoparticle have been designed. With newer formulations, including low-molecular-weight iron dextran, iron sucrose, ferric gluconate, ferumoxytol, iron isomaltoside, and ferric carboxymaltose, serious adverse events are rare.
Collapse
Affiliation(s)
- Andreas J Bircher
- Allergy Unit, Dermatology Clinic, University Hospital Basel, Petersgraben 4, Basel 4031, Switzerland.
| | - Michael Auerbach
- Georgetown University School of Medicine, 3900 Reservoir road northwest, Washington, DC 20007, USA; Hematology and Oncology, Private Practice, King Avenue #308, Baltimore, MD 21237, USA
| |
Collapse
|
46
|
|
47
|
Auerbach M, Macdougall IC. Safety of intravenous iron formulations: facts and folklore. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2014; 12:296-300. [PMID: 25074787 PMCID: PMC4111808 DOI: 10.2450/2014.0094-14] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 06/03/2014] [Indexed: 11/21/2022]
Affiliation(s)
- Michael Auerbach
- School of Medicine Georgetown University, Washington DC, United States of America
| | - Iain C. Macdougall
- Department of Nephrology, King’s College Hospital, London, United Kingdom
| |
Collapse
|
48
|
Gilreath JA, Stenehjem DD, Rodgers GM. Diagnosis and treatment of cancer-related anemia. Am J Hematol 2014; 89:203-12. [PMID: 24532336 DOI: 10.1002/ajh.23628] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 10/25/2013] [Accepted: 10/31/2013] [Indexed: 01/04/2023]
Abstract
Cancer-related anemia (CRA) is due to multiple etiologies, including chemotherapy-induced myelosuppression, blood loss, functional iron deficiency, erythropoietin deficiency due to renal disease, marrow involvement with tumor as well as other factors. The most common treatment options for CRA include iron therapy, erythropoietic-stimulating agents (ESAs), and red cell transfusion. Safety concerns as well as restrictions and reimbursement issues surrounding ESA therapy for CRA have resulted in suboptimal treatment. Similarly, many clinicians are not familiar or comfortable using intravenous iron products to treat functional iron deficiency associated with CRA. This article summarizes our approach to treating CRA and discusses commonly encountered clinical scenarios for which current clinical guidelines do not apply.
Collapse
Affiliation(s)
- Jeffrey A. Gilreath
- Department of Pharmacy; Huntsman Cancer Institute; University of Utah Hospitals and Clinics; Salt Lake City Utah
| | - David D. Stenehjem
- Department of Pharmacy; Huntsman Cancer Institute; University of Utah Hospitals and Clinics; Salt Lake City Utah
- Department of Pharmacotherapy; Pharmacotherapy Outcomes Research Center, College of Pharmacy, University of Utah; Salt Lake City Utah
| | - George M. Rodgers
- Department of Medicine; University of Utah Hospitals and Clinics; Salt Lake City Utah
- Department of Pathology; University of Utah Hospitals and Clinics; Salt Lake City Utah
| |
Collapse
|
49
|
Vadhan-Raj S, Strauss W, Ford D, Bernard K, Boccia R, Li J, Allen LF. Efficacy and safety of IV ferumoxytol for adults with iron deficiency anemia previously unresponsive to or unable to tolerate oral iron. Am J Hematol 2014; 89:7-12. [PMID: 23983177 PMCID: PMC4223995 DOI: 10.1002/ajh.23582] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 08/15/2013] [Accepted: 08/21/2013] [Indexed: 12/16/2022]
Abstract
Although oral iron is the initial treatment approach for iron deficiency anemia (IDA), some patients fail to respond to or cannot tolerate oral iron. This double-blind safety and efficacy study of the intravenous (IV) iron, ferumoxytol, randomized patients with a history of unsatisfactory oral iron therapy, or in whom oral iron could not be used, to ferumoxytol (n = 609) or placebo (n = 203). The proportion of patients achieving the primary endpoint (hemoglobin increase ≥2.0 g/dL at Week 5) was 81.1% with ferumoxytol versus 5.5% with placebo (P < 0.0001). The mean increase in hemoglobin from Baseline to Week 5, a secondary endpoint (also the alternative preplanned primary efficacy endpoint for other health authorities), was 2.7 versus 0.1 g/dL (P < 0.0001). Achievement of a hemoglobin ≥12 g/dL, time to a hemoglobin increase ≥2.0 g/dL, and improvement in the Functional Assessment of Chronic Illness Therapy Fatigue score also significantly favored ferumoxytol over placebo at Week 5 (P < 0.0001). Ferumoxytol treatment-emergent adverse events were mainly mild to moderate. Ferumoxytol was effective and well tolerated in patients with IDA of any underlying cause in whom oral iron was ineffective or could not be used. This trial was registered at www.clinicaltrials.gov as #NCT01114139.
Collapse
Affiliation(s)
| | | | - David Ford
- Toronto Digestive Disease Associates, Inc.Vaughan, Ontario, Canada
| | | | - Ralph Boccia
- The Center for Cancer and Blood DisordersBethesda, Maryland
| | - Joe Li
- AMAG Pharmaceuticals, Inc.Lexington, Massachusetts
| | - Lee F Allen
- AMAG Pharmaceuticals, Inc.Lexington, Massachusetts
| |
Collapse
|
50
|
Plummer ES, Crary SE, McCavit TL, Buchanan GR. Intravenous low molecular weight iron dextran in children with iron deficiency anemia unresponsive to oral iron. Pediatr Blood Cancer 2013; 60:1747-52. [PMID: 23832487 DOI: 10.1002/pbc.24676] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 06/07/2013] [Indexed: 01/24/2023]
Abstract
BACKGROUND Iron deficiency anemia (IDA) in children is usually treated with oral iron, yet many respond poorly. Intravenous low molecular weight iron dextran (LMWID) offers the opportunity of employing a single outpatient infusion to correct the anemia and reduce the overall burden of treatment, but its use in children has been limited due to concerns of serious adverse effects. In this study we report our initial experience using LMWID in children with iron deficiency in whom oral iron was ineffective. METHODS We performed a case series of LMWID treatment of children with IDA of diverse etiologies who were poorly responsive to oral iron therapy with the aim of measuring its efficacy and adverse effects. LMWID was administered as a total dose infusion over 60 minutes in the outpatient setting. RESULTS Thirty-one patients age 11 months to 18 years received intravenous LMWID, and 24 were evaluable for hematologic response. Median hemoglobin increments were respectively 3.5, 1.9, and 1.8 g/dl in patients with IDA due to poor nutrition (n = 11), chronic blood loss (n = 13), and miscellaneous causes (n = 7). Two thirds of evaluable patients had a complete hematologic response. Nine of the patients (29%) had mild non-specific adverse effects upon initiation of the LMWID infusion. CONCLUSIONS LMWID as a total dose infusion was well tolerated and effective in a heterogeneous group of children and adolescents with IDA who were refractory to oral iron therapy. Transient reactions were common but not serious.
Collapse
Affiliation(s)
- Ellen S Plummer
- Division of Hematology-Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas; Children's Medical Center, Dallas, Texas
| | | | | | | |
Collapse
|