1
|
White F, Polakowski N, Merlington E, Leimanis M, Dudick B, Parker J, Jousma A, Loji A, Hanson J, Arney M, Boville B. Intravenous Sodium Ferric Gluconate Complex for Hospitalized Pediatric Patients with Iron Deficiency Anemia. CHILDREN (BASEL, SWITZERLAND) 2025; 12:189. [PMID: 40003291 PMCID: PMC11854077 DOI: 10.3390/children12020189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Background/Objectives: Iron deficiency anemia is common in the pediatric population. Red blood cell transfusions, a common acute treatment, pose well-recognized risks including lung injury, circulatory overload, and immune dysfunction. Intravenous iron, specifically sodium ferric gluconate complex (SFGC), is a potential alternative, however investigation on its use in hospitalized children is lacking. This study aims to describe the physiologic response via change in hematologic values to cumulative dose of SFGC, investigate the effect of cumulative dosing on the amount of RBC transfusions received, and comment on its safety. Methods: This is a retrospective investigation of pediatric patients with iron deficiency who received SFGC during their admission to the Helen DeVos Children's Hospital between 2016 and 2018 (N = 85). Results: A total of 258 doses of intravenous SFGC were provided to 85 patients. The average pre-treatment serum hemoglobin was 8.73 ± 1.33, and 7 days post-treatment this increased to 10.41 ± 1.43. Mean corpuscular volume, ferritin, serum ion, total iron binding capacity, reticulocyte percentage, and reticulocyte hemoglobin all increased 7 days post-treatment, as would be suspected, but without any statistically significant difference between hematologic outcomes and cumulative dose of SFGC. Our study did not reveal any correlation between the cumulative dose of SFGC administered and the amount of RBC transfusions received. Only one adverse event was recorded. Conclusions: Our results complement the trend of increased use and emerging evidence of favorable safety profiles of IV iron in the pediatric population. This descriptive investigation revealed that administering higher cumulative doses of SFGC provided no further benefits in terms of hematologic response or RBC transfusion administration.
Collapse
Affiliation(s)
- Felicia White
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (F.W.); (N.P.); (E.M.); (M.L.); (A.L.); (J.H.); (M.A.)
- Department of Pediatrics, Corewell Health Helen DeVos Children’s Hospital, Grand Rapids, MI 49503, USA
| | - Noelle Polakowski
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (F.W.); (N.P.); (E.M.); (M.L.); (A.L.); (J.H.); (M.A.)
| | - Elena Merlington
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (F.W.); (N.P.); (E.M.); (M.L.); (A.L.); (J.H.); (M.A.)
| | - Mara Leimanis
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (F.W.); (N.P.); (E.M.); (M.L.); (A.L.); (J.H.); (M.A.)
- Division of Pediatric Critical Care & Sedation, Corewell Health Helen DeVos Children’s Hospital, Grand Rapids, MI 49503, USA
| | - Brooke Dudick
- Department of Bioinformatics, Corewell Health Health DeVos Children’s Hospital, Grand Rapids, MI 49503, USA; (B.D.); (J.P.)
| | - Jessica Parker
- Department of Bioinformatics, Corewell Health Health DeVos Children’s Hospital, Grand Rapids, MI 49503, USA; (B.D.); (J.P.)
| | - Ashley Jousma
- Department of Quality, Safety, & Experience, Corewell Health Helen DeVos Children’s Hospital, Grand Rapids, MI 49503, USA;
| | - Alexander Loji
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (F.W.); (N.P.); (E.M.); (M.L.); (A.L.); (J.H.); (M.A.)
| | - Jeffrey Hanson
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (F.W.); (N.P.); (E.M.); (M.L.); (A.L.); (J.H.); (M.A.)
| | - Monica Arney
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (F.W.); (N.P.); (E.M.); (M.L.); (A.L.); (J.H.); (M.A.)
| | - Brian Boville
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA; (F.W.); (N.P.); (E.M.); (M.L.); (A.L.); (J.H.); (M.A.)
- Division of Pediatric Critical Care & Sedation, Corewell Health Helen DeVos Children’s Hospital, Grand Rapids, MI 49503, USA
| |
Collapse
|
2
|
Leister J, Norby FL, Boucher AA. Time to Resolution of Severe Anemia in Young Children With Iron Deficiency. J Pediatr Hematol Oncol 2025; 47:e38-e43. [PMID: 39641630 DOI: 10.1097/mph.0000000000002979] [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] [Received: 06/01/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Iron deficiency anemia in children remains a pervasive problem. Prolonged iron deficiency anemia (IDA) is linked to adverse neurodevelopmental outcomes, but the duration of severe IDA (hemoglobin <7 g/dL) in children is poorly studied, particularly for racial/ethnic minority groups. We reviewed the electronic medical records of 92 children (1 to 5 y old) with IDA within a major metropolitan health care system. Duration of anemia, hemoglobin at diagnosis and nadir, age, sex, race/ethnicity, and the use of intravenous interventions were analyzed by Kaplan-Meier curves, Cox regression, and logistic regression. The majority of the included children were Asian (72.8%). Anemia resolution was documented in 68% of cases, and 47% of cases received intravenous intervention. Iron repletion was only confirmed in 37% of cases (n=34), leaving many children with unclear resolution at risk for recurrence. Caucasian children had anemia resolution faster than Black or Asian children, and the latter groups were also less likely to reach hemoglobin normalization. Children with intravenous interventions were more likely to have documented resolution than those with only oral treatment. Those receiving intravenous interventions were more likely to be followed through anemia resolution, although treatment standardization was lacking, and confirmation of iron storage repletion was rarely checked. Future studies should emphasize the importance of ensuring iron storage replacement and potentially utilize time-to-anemia resolution data to determine optimal hemoglobin values for intravenous iron as a first-line intervention.
Collapse
Affiliation(s)
| | - Faye L Norby
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Alexander A Boucher
- Department of Pediatrics
- Department of Medicine, University of Minnesota Medical School
| |
Collapse
|
3
|
Goobie SM, Faraoni D. Perioperative paediatric patient blood management: a narrative review. Br J Anaesth 2025; 134:168-179. [PMID: 39455307 DOI: 10.1016/j.bja.2024.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 10/28/2024] Open
Abstract
Patient blood management (PBM) encompasses implementing multimodal evidence-based strategies to screen, diagnose, and properly treat anaemia and coagulopathies using goal-directed therapy while minimising bleeding. The aim of PBM is to improve clinical care and patient outcomes while managing patients with potential or ongoing critical anaemia, clinically significant bleeding, and coagulopathies. The focus of PBM is patient-centred rather than transfusion-centred. Multimodal PBM strategies are now recommended by international organisations, including the World Health Organization, as a new standard of care and a proven means to safely and effectively manage anaemia and blood loss while minimising unnecessary blood transfusion. Compared with adult PBM, paediatric PBM is currently not routinely accepted as a standard of care. This is partly because of the paucity of robust data on paediatric patient PBM. Managing paediatric bleeding and blood product transfusion presents unique challenges. Neonates, infants, children, and adolescents each have specific considerations based on age, weight, physiology, and pharmacology. This narrative review covers the latest updates for PBM in paediatric surgical populations including the benefits and principles of paediatric PBM, current expert consensus guidelines, and important universal multimodal therapeutic strategies emphasising clinical management of the anaemic, bleeding, or coagulopathic paediatric patient in the perioperative period. Practical paediatric rules for PBM in the perioperative period are highlighted, with review of specific PBM strategies including treatment of preoperative anaemia, restrictive transfusion thresholds, antifibrinolytic agents, cell salvage, standardised transfusion algorithms, and goal-directed therapy based on point-of-care and viscoelastic testing.
Collapse
Affiliation(s)
- Susan M Goobie
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - David Faraoni
- Arthur S. Keats Division of Pediatric Cardiovascular Anesthesia, Department of Anesthesiology, Perioperative and Pain Medicine, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
4
|
Chaber R, Helwich E, Lauterbach R, Mastalerz-Migas A, Matysiak M, Peregud-Pogorzelski J, Styczyński J, Szczepański T, Jackowska T. Diagnosis and Treatment of Iron Deficiency and Iron Deficiency Anemia in Children and Adolescents: Recommendations of the Polish Pediatric Society, the Polish Society of Pediatric Oncology and Hematology, the Polish Society of Neonatology, and the Polish Society of Family Medicine. Nutrients 2024; 16:3623. [PMID: 39519457 PMCID: PMC11547346 DOI: 10.3390/nu16213623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives. Iron deficiency is one of the most common nutritional deficiencies worldwide and is the leading cause of anemia in the pediatric population (microcytic, hypochromic anemia due to iron deficiency). Moreover, untreated iron deficiency can lead to various systemic consequences and can disrupt the child's development. Methods/Results. Therefore, a team of experts from the Polish Pediatric Society, the Polish Society of Pediatric Oncology and Hematology, the Polish Neonatology Society, and the Polish Society of Family Medicine, based on a review of the current literature, their own clinical experience, and critical discussion, has developed updated guidelines for the diagnosis, prevention, and treatment of iron deficiency in children from birth to 18 years of age. These recommendations apply to the general population and do not take into account the specifics of individual conditions and diseases.
Collapse
Affiliation(s)
- Radosław Chaber
- Department of Pediatrics, Institute of Medical Sciences, University of Rzeszow, 35-310 Rzeszow, Poland
- Clinic of Pediatric Oncology and Hematology, State Hospital 2, 35-301 Rzeszow, Poland
| | - Ewa Helwich
- Department of Neonatology and Neonatal Intensive Care, Institute of Mother and Child, 04-370 Warsaw, Poland;
| | - Ryszard Lauterbach
- Clinic of Neonatology, Department of Gynecology and Obstetrics, Jagiellonian University Hospital, 31-501 Cracow, Poland;
| | | | - Michał Matysiak
- Department of Oncology, Children’s Hematology, Clinical Transplantology and Pediatrics, University Clinical Center, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Jarosław Peregud-Pogorzelski
- Department of Pediatrics, Oncology and Pediatric Immunology, Pomeranian Medical University, 70-204 Szczecin, Poland;
| | - Jan Styczyński
- Department of Pediatric Haematology and Oncology, Collegium Medicum, Nicolaus Copernicus University Torun, Jurasz University Hospital 1, 85-094 Bydgoszcz, Poland;
| | - Tomasz Szczepański
- Department of Pediatric Haematology and Oncology, Medical University of Silesia, 41-800 Katowice, Poland;
| | - Teresa Jackowska
- Department of Pediatrics, Centre of Postgraduate Medical Education, 01-809 Warsaw, Poland;
| |
Collapse
|
5
|
Al-Zubeidi D, Davis MB, Rahhal R. Prevention of complications for hospitalized patients receiving parenteral nutrition: A narrative review. Nutr Clin Pract 2024; 39:1037-1053. [PMID: 39152093 DOI: 10.1002/ncp.11201] [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: 05/03/2024] [Revised: 07/17/2024] [Accepted: 07/21/2024] [Indexed: 08/19/2024] Open
Abstract
Hospitalized patients may benefit from parenteral nutrition to address their compromised nutrition status attributed to limited oral/enteral intake and increased nutrient/energy requirement during acute illness. Parenteral nutrition, however, can be associated with many complications that can negatively impact patient outcomes. In this review, we focus on potential metabolic and catheter-related complications associated with parenteral nutrition use. We report on potential risk factors for such complications and highlight strategies for prevention and early recognition. To optimize outcomes, key findings include the creation and implementation of evidence-based protocols with proven efficacy. For each hospital unit delivering parenteral nutrition to patients, tracking compliance with established protocols and patient outcomes is crucial for ongoing improvement through identification of gaps, proper reeducation and training, and ongoing refinement of care protocols. Establishment of specialized inpatient nutrition support teams should be considered.
Collapse
Affiliation(s)
- Dina Al-Zubeidi
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Mary Beth Davis
- College of Nursing, University of Iowa Children's Hospital, Iowa City, Iowa, USA
| | - Riad Rahhal
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
6
|
Roganovic J. Parenteral iron therapy in children with iron deficiency anemia. World J Clin Cases 2024; 12:2138-2142. [PMID: 38808346 PMCID: PMC11129126 DOI: 10.12998/wjcc.v12.i13.2138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/10/2024] [Accepted: 04/07/2024] [Indexed: 04/25/2024] Open
Abstract
Iron deficiency anemia (IDA) continues to be a global public health problem. Oral iron is the universally accepted first-line therapy, and most children have a prompt and favorable response to oral formulations. In subsets of children who fail to respond due to intolerance, poor adherence, or inadequate intestinal absorption, parenteral iron is indicated. Despite numerous studies in adults with IDA of diverse etiologies, pediatric studies on parenteral iron use are very limited. Although mostly retrospective and small, these studies have documented the efficacy and safety profile of intravenous iron formulations. In this editorial the author comments on the most important published data and underscores the need to seriously consider parenteral iron use in children unresponsive to oral therapy.
Collapse
Affiliation(s)
- Jelena Roganovic
- Department of Pediatric Hematology and Oncology, Children’s Hospital Zagreb, 10000 Zagreb, Croatia
- Faculty of Biotechnology and Drug Development, University of Rijeka, Rijeka 51000, Croatia
| |
Collapse
|
7
|
Cohen CT, Powers JM. Nutritional Strategies for Managing Iron Deficiency in Adolescents: Approaches to a Challenging but Common Problem. Adv Nutr 2024; 15:100215. [PMID: 38556251 PMCID: PMC11070695 DOI: 10.1016/j.advnut.2024.100215] [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: 01/26/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024] Open
Abstract
Iron deficiency (ID) is a common and challenging problem in adolescence. In order to prevent, recognize, and treat ID in this age range, it is critical to understand the recommended daily intake of iron in relation to an adolescent's activity, dietary habits, and basal iron losses. Adolescents following vegetarian or vegan diets exclusively rely on plant-based, nonheme iron, which has decreased bioavailability compared with heme iron and requires increased total iron intake. Individuals with disordered eating habits, excessive menstrual blood loss, and certain chronic health conditions (including inflammatory bowel disease and heart failure) are at high risk of ID and the development of symptomatic iron deficiency anemia (IDA). Adolescent athletes and those with sleep and movement disorders may also be more sensitive to changes in iron status. Iron deficiency is typically treated with oral iron supplementation. To maximize iron absorption, oral iron should be administered no more than once daily, ideally in the morning, while avoiding foods and drinks that inhibit iron absorption. Oral iron therapy should be provided for ≥3 mo in the setting of ID to reach a ferritin of 20 ng/mL before discontinuation. Intravenous iron is being increasingly used in this population and has demonstrated efficacy and safety in adolescents. It should be considered in those with persistent ID despite a course of oral iron, severe and/or symptomatic IDA, and chronic inflammatory conditions characterized by decreased gastrointestinal iron absorption.
Collapse
Affiliation(s)
- Clay T Cohen
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Houston, TX, United States
| | - Jacquelyn M Powers
- Division of Hematology/Oncology, Department of Pediatrics, Baylor College of Medicine and Texas Children's Cancer and Hematology Center, Texas Children's Hospital, Houston, TX, United States.
| |
Collapse
|
8
|
Butragueño-Laiseca L, de la Mata Navazo S, Sánchez Galindo AC, Santiago Lozano MJ. Intravenous iron for critically ill children. Comparison of three dose regimens. Pediatr Blood Cancer 2024; 71:e30734. [PMID: 37880937 DOI: 10.1002/pbc.30734] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 10/01/2023] [Accepted: 10/09/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND Anemia is extremely common among patients admitted to pediatric intensive care. Alternative treatments to transfusions such as intravenous iron must be considered. There are no published data for a prospective intravenous (IV) iron study focused in the critically ill children. The objective is to examine the safety and efficacy of intravenous iron sucrose infusion to manage anemia in pediatric critical care. A secondary objective is to examine the effect of different dose regimens of iron sucrose (3, 5, and 7 mg/kg dose). PROCEDURE Prospective investigation of intravenous iron sucrose utilization at a tertiary pediatric intensive care unit between October 2017 and November 2022. RESULTS In all 115 patients received a total of 616 infusions of IV iron. Transferrin saturation index (TSI) was the most common altered iron deficiency biomarker (91.8%). After IV iron treatment, hemoglobin showed a significant increase within a 30-day follow-up (9.2 vs. 11.6 g/dL, p < .001). There was also a significant improvement in TSI and serum iron (p < .001). Iron deficit replacement was higher in the 7 mg/kg dose group (94%) compared to 85.9% in the 5 mg/kg regimen and 77.5% in the lower dose group (p = .008), requiring less doses and a shorter time. Very few mild adverse reactions were reported (1.3% of infusions), with no differences between groups. The most frequent adverse effect was gastrointestinal in three cases. There were no anaphylaxis-like or other serious/life-threatening adverse effects. CONCLUSIONS This is the first study to evaluate intravenous iron therapy in pediatric critical care, providing preliminary evidence of safety and efficacy of IV iron sucrose. The 7 mg/kg dose regimen showed higher iron deficit replacement in a shorter time, which could be beneficial in critically ill children.
Collapse
Affiliation(s)
- Laura Butragueño-Laiseca
- Pediatric Intensive Care Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatrics Department, Universidad Complutense de Madrid, Madrid, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Development Origin Network (RICORS), Instituto de Salud Carlos III, Madrid, Spain
| | - Sara de la Mata Navazo
- Pediatric Intensive Care Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatrics Department, Universidad Complutense de Madrid, Madrid, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Development Origin Network (RICORS), Instituto de Salud Carlos III, Madrid, Spain
| | - Amelia Caridad Sánchez Galindo
- Pediatric Intensive Care Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatrics Department, Universidad Complutense de Madrid, Madrid, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Development Origin Network (RICORS), Instituto de Salud Carlos III, Madrid, Spain
| | - María José Santiago Lozano
- Pediatric Intensive Care Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatrics Department, Universidad Complutense de Madrid, Madrid, Spain
- Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Development Origin Network (RICORS), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
9
|
Korczowski B, Farrell C, Falone M, Blackman N, Rodgers T. Safety, pharmacokinetics, and pharmacodynamics of intravenous ferric carboxymaltose in children with iron deficiency anemia. Pediatr Res 2023; 94:1547-1554. [PMID: 37208431 PMCID: PMC10589089 DOI: 10.1038/s41390-023-02644-9] [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] [Received: 10/11/2022] [Revised: 04/12/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023]
Abstract
BACKGROUND Iron deficiency is the primary cause of anemia in children. Intravenous (IV) iron formulations circumvent malabsorption and rapidly restore hemoglobin. METHODS This Phase 2, non-randomized, multicenter study characterized the safety profile and determined appropriate dosing of ferric carboxymaltose (FCM) in children with iron deficiency anemia. Patients aged 1-17 years with hemoglobin <11 g/dL and transferrin saturation <20% received single IV doses of undiluted FCM 7.5 mg/kg (n = 16) or 15 mg/kg (n = 19). RESULTS The most common drug-related treatment-emergent adverse event was urticaria (in three recipients of FCM 15 mg/kg). Systemic exposure to iron increased in a dose-proportional manner with approximate doubling of mean baseline-corrected maximum serum iron concentration (157 µg/mL with FCM 7.5 mg/kg; and 310 µg/mL with FCM 15 mg/kg) and area under the serum concentration-time curve (1901 and 4851 h·µg/mL, respectively). Baseline hemoglobin was 9.2 and 9.5 g/dL in the FCM 7.5 and 15 mg/kg groups, respectively, with mean maximum changes in hemoglobin of 2.2 and 3.0 g/dL, respectively. CONCLUSIONS In conclusion, FCM was well tolerated by pediatric patients. Improvements in hemoglobin were greater with the higher dose, supporting use of the FCM 15 mg/kg dose in pediatric patients (Clinicaltrials.gov NCT02410213). IMPACT This study provided information on the pharmacokinetics and safety of intravenous ferric carboxymaltose for treatment of iron deficiency anemia in children and adolescents. In children aged 1-17 years with iron deficiency anemia, single intravenous doses of ferric carboxymaltose 7.5 or 15 mg/kg increased systemic exposure to iron in a dose-proportional manner, with clinically meaningful increases in hemoglobin. The most common drug-related treatment-emergent adverse event was urticaria. The findings suggest that iron deficiency anemia in children can be corrected with a single intravenous dose of ferric carboxymaltose and support use of a 15 mg/kg dose.
Collapse
Affiliation(s)
- Bartosz Korczowski
- Department of Pediatrics, Institute of Medical Sciences, Medical College, University of Rzeszów, Rzeszów, Poland.
| | | | | | | | | |
Collapse
|
10
|
Abstract
Medical child abuse (MCA), formerly called Munchausen syndrome by proxy (MSP or MSBP), occurs when a caregiver, usually the mother, falsifies or exaggerates symptoms resulting in harm to a child through inappropriate medical care. MCA is underrecognized, underreported, and results in significant morbidity and mortality. Pediatrics subspecialists should consider MCA when unusual disease presentation [THAT] do not respond to traditional treatments. This article reviews the more common diagnoses encountered in MCA cases by specialty.
Collapse
Affiliation(s)
- Melissa K Egge
- Pediatrics, Stanford Medicine Children's Health - Lucile Packard, 700 Welch Road, Suite 300G, MC 6583, Palo Alto, CA 94304, USA.
| |
Collapse
|
11
|
Intravenous Iron Therapy for Children With Iron Deficiency Anemia. J Pediatr Hematol Oncol 2023; 45:e56-e59. [PMID: 36161971 DOI: 10.1097/mph.0000000000002550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/11/2022] [Indexed: 02/03/2023]
Abstract
Iron deficiency anemia in children is a public health problem. Although oral iron treatment is the first choice, common side effects and compliance problems can cause the treatment to be interrupted. This study retrospectively evaluated children treated with intravenous (IV) iron sucrose or ferric carboxymaltose (FCM) and compared the treatment processes and efficacy. The demographic characteristics and treatment details of the 44 children with iron deficiency anemia were retrospectively evaluated. Iron sucrose was administered to 25 patients and FCM was administered to 19 patients. The IV iron infusion was applied to 64% of the patients because of unresponsiveness to oral treatment, 25% of the patients because of compliance problems, and 11% of the patients because of severe anemia. IV iron therapy increased hemoglobin, mean corpuscular volume, mean corpuscular hemoglobin, red-cell distribution width, and serum ferritin levels and decreased platelet count. The mean number of infusions per patient in the FCM group was lower, and the total treatment time was shorter. In conclusion, IV iron sucrose or FCM can be used in children with nonadherence to oral therapy and severe anemia in addition to specific indications.
Collapse
|
12
|
Abbas M, Hayirli Z, Drakesmith H, Andrews SC, Lewis MC. Effects of iron deficiency and iron supplementation at the host-microbiota interface: Could a piglet model unravel complexities of the underlying mechanisms? Front Nutr 2022; 9:927754. [PMID: 36267902 PMCID: PMC9577221 DOI: 10.3389/fnut.2022.927754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/05/2022] [Indexed: 01/14/2023] Open
Abstract
Iron deficiency is the most prevalent human micronutrient deficiency, disrupting the physiological development of millions of infants and children. Oral iron supplementation is used to address iron-deficiency anemia and reduce associated stunting but can promote infection risk since restriction of iron availability serves as an innate immune mechanism against invading pathogens. Raised iron availability is associated with an increase in enteric pathogens, especially Enterobacteriaceae species, accompanied by reductions in beneficial bacteria such as Bifidobacteria and lactobacilli and may skew the pattern of gut microbiota development. Since the gut microbiota is the primary driver of immune development, deviations from normal patterns of bacterial succession in early life can have long-term implications for immune functionality. There is a paucity of knowledge regarding how both iron deficiency and luminal iron availability affect gut microbiota development, or the subsequent impact on immunity, which are likely to be contributors to the increased risk of infection. Piglets are naturally iron deficient. This is largely due to their low iron endowments at birth (primarily due to large litter sizes), and their rapid growth combined with the low iron levels in sow milk. Thus, piglets consistently become iron deficient within days of birth which rapidly progresses to anemia in the absence of iron supplementation. Moreover, like humans, pigs are omnivorous and share many characteristics of human gut physiology, microbiota and immunity. In addition, their precocial nature permits early maternal separation, individual housing, and tight control of nutritional intake. Here, we highlight the advantages of piglets as valuable and highly relevant models for human infants in promoting understanding of how early iron status impacts physiological development. We also indicate how piglets offer potential to unravel the complexities of microbiota-immune responses during iron deficiency and in response to iron supplementation, and the link between these and increased risk of infectious disease.
Collapse
Affiliation(s)
- Munawar Abbas
- Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Zeynep Hayirli
- Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Simon C. Andrews
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Marie C. Lewis
- Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
13
|
Safety of Ferric Carboxymaltose in Children: Report of a Case Series from Greece and Review of the Literature. Paediatr Drugs 2022; 24:137-146. [PMID: 35083718 DOI: 10.1007/s40272-022-00491-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Parenteral iron is generally considered safe in adults, and severe adverse events are extremely rare. Ferric carboxymaltose (FCM), a third-generation parenteral iron product, is not licensed for pediatric use. OBJECTIVE The aim of this study was to present our data on the safety of FCM in children with iron deficiency (ID) and/or iron deficiency anemia (IDA) and to investigate through a systematic literature review articles reporting on the safety of FCM use in children with ID/IDA. PATIENTS AND METHODS Safety data regarding children treated with FCM for ID/IDA from four pediatric departments in Greece over a 26-month period are presented. Additionally, a literature search was performed in PubMed, Scopus, and Google Scholar on December 4, 2021 for articles reporting on the use of FCM in children with ID/IDA. Review articles, guidelines, case reports/case series, and reports on the use of FCM for conditions other than ID/IDA were excluded. Identified articles were screened for all reported adverse events (AE) that were graded according to the Common Terminology Criteria for Adverse Events, version 5.0. RESULTS In our cohort, 37 children with ID/IDA received 41 FCM infusions. All infusions were tolerated well. In addition, 11 articles reporting 1231 infusions of FCM in 866 children were identified in the literature. Among them, 52 (6%) children developed AE that were graded as mild or moderate (grades I-III). CONCLUSIONS Our patient cohort and this literature review provide further evidence for the good safety profile of FCM in children, although well-designed prospective clinical trials with appropriate safety endpoints are still required.
Collapse
|
14
|
Garcia-Ortega P, Jimenez-Lozano I, Cruz Á, Polo AF, Lopez M, Ariceta G. Safety and effectiveness of ferric carboxymaltose intravenous therapy in pediatric patients with chronic kidney disease. Front Pediatr 2022; 10:967233. [PMID: 36275063 PMCID: PMC9582777 DOI: 10.3389/fped.2022.967233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Iron-deficiency anemia is the most common reason for worsening of the anemia characteristically seen in chronic kidney disease (CKD). Ferric carboxymaltose (FCM) is a macromolecular hydroxide ferric carbohydrate complex that allows high-dose iron to be administered parenterally for gradual, controlled release. The aim of this study was to retrospectively evaluate the safety and effectiveness of FCM treatment in pediatric patients with CKD non-dependent of hemodialysis, seen at a tertiary hospital. Data were collected on demographics, dosage, infusion time, laboratory results, and tolerability of the medicinal product. A total of 79 patients (40.5% girls) were included; the median age [25th percentile (P25) to 75th percentile (P75)] was 9 years (5-13). Laboratory results at 15-45 days post-infusion revealed a median increase of 1.4 g/dL (0.9-1.9) in hemoglobin, 224 μg/L (136-378.5) in ferritin, 37 μg/dL (17.5-71) in serum iron, and 18% (9.3-27.8) in transferrin saturation. All patients tolerated FCM infusions well, and no serious hypersensitivity reactions or anaphylactic reactions were observed. Only one adverse event was identified: drug extravasation at the end of the infusion in a 16-year-old patient. These data provide further evidence for the use of FCM as a safe and effective therapeutic option in pediatric patients with CKD, based on the low incidence of adverse effects, minor intervention required, and anemia improvement based on laboratory results.
Collapse
Affiliation(s)
| | | | - Álejandro Cruz
- Department of Pediatric Nephrology, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | - Mercedes Lopez
- Department of Pediatric Nephrology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Gema Ariceta
- Department of Pediatric Nephrology, Vall d'Hebron University Hospital, Barcelona, Spain.,Department of Pediatrics, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Organizational Strategies for the Management of Intravenous Iron Therapy in Non-Hospitalized Settings: A Safe Opportunity to Implement Patient Blood Management in Italy. Healthcare (Basel) 2021; 9:healthcare9091222. [PMID: 34574994 PMCID: PMC8467602 DOI: 10.3390/healthcare9091222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 01/25/2023] Open
Abstract
This article analyzes the recommendations issued by the Emilia Romagna region in July 2020 on “Organizational strategies for the safe management of intravenous iron therapy in patients in non-hospitalized settings”. The objective of these recommendations is to set up safe intravenous iron administration sites outside the hospital environment across the national territory. The document facilitates the organization of methods for intravenous iron infusion that are safe for the patient and correct from a medico-legal perspective. In addition, it opens the way for the widespread use of iron infusion in the field, providing benefits to patient quality of life. This program prevents unnecessary transfusions, reduces costs, prevents overcrowding in hospitals in the event of a pandemic, and enables patient treatment in the field, thus, saving on the use of personnel.
Collapse
|
16
|
Boucher AA, Bedel A, Jones S, Lenahan SF, Geer R, McGann PT. A retrospective study of the safety and efficacy of low molecular weight iron dextran for children with iron deficiency anemia. Pediatr Blood Cancer 2021; 68:e29024. [PMID: 33769677 DOI: 10.1002/pbc.29024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Iron deficiency anemia (IDA) affects millions of children worldwide. Oral iron replacement is effective but often poorly tolerated. Intravenous iron has been demonstrated to have utility in all ages, but pediatric use remains limited. Low molecular weight iron dextran (LMWID) has a dosing range capable of replacing iron deficits in a single infusion and has been evaluated in small pediatric cohorts, but additional safety and efficacy data are limited. Here, we evaluate the safety and efficacy of LMWID in association with an electronic medical record (EMR)-based effort to optimize dosing. PROCEDURE A retrospective IRB-approved investigation of LMWID utilization at a tertiary pediatric hospital between January 1, 2016 and March 31, 2020 was undertaken to evaluate the therapeutic efficacy and frequency/severity of infusion-related adverse event (AE) in children and adolescents receiving LMWID. Patient demographics and LMWID dosing characteristics were collected, and primary outcome measures included laboratory response and the incidence/severity of any infusion-related events. The utilization of an EMR-based nomogram for LMWID dosing was also evaluated. RESULTS A total of 254 infusions for 191 patients were included (ages 0.7-20.9 years), most with IDA. LMWID replaced at least 75% of the estimated iron deficit in a single infusion for 76% of patients. The mean hemoglobin and ferritin increases were 2.1 g/dl and >100 ng/ml, respectively. Infusion-related AEs were rare, occurring in only 12/254 (4.7%) of infusions and 67% during the test dose; each rapidly resolved without long-term sequelae. No AEs occurred in those <10 years of age. Premedication use markedly decreased with nomogram use without a change in AE rate. CONCLUSIONS In a large institutional cohort, LMWID was well tolerated in children and adolescents, with most patients having their total iron deficits relieved in a single infusion. These data support expanded use of LMWID in the management of pediatric iron deficiency.
Collapse
Affiliation(s)
- Alexander A Boucher
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Ashley Bedel
- Division of Pharmacy, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Sommer Jones
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Stephanie F Lenahan
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Rebecca Geer
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Patrick T McGann
- Division of Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
17
|
Hoyos AB, Vasquez-Hoyos P. Transfusion prevention using erythropoietin, parenteral sucrose iron, and fewer phlebotomies in infants born at ≤30 weeks gestation at a high altitude center: a 10-year experience. J Perinatol 2021; 41:1403-1411. [PMID: 33568772 DOI: 10.1038/s41372-021-00945-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/11/2020] [Accepted: 01/19/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Red blood cell transfusions in infants born at ≤30 weeks gestation are frequent. Erythropoietin therapy reduces transfusions. An increase in hematocrit is an adaptive response at high altitudes but a guaranteed source of iron is necessary for adequate erythropoiesis. METHODS A retrospective cohort study was done to compare red blood cell transfusion practices of the 2019 EpicLatino (EPIC) Latin America network database with a single unit at 2650 m above sea level (LOCAL). The data from LOCAL for three time periods were compared over 10 years based on changes in erythropoietin dose and fewer phlebotomies. The number of cases that received transfusions and the total number of transfusions required were compared. Adjustments were made for known risk factors using a multivariate regression analysis. RESULTS Two hundred and twenty-one cases in LOCAL and 382 cases from EPIC were included. Overall basic demographic characteristics were similar. In EPIC a significantly higher rate of infection (28% vs. 15%) and outborn (10% vs. 1%) was found, but less necrotizing enterocolitis (9% vs. 15%) and use of prenatal steroids (62% vs. 93%) than LOCAL (p < 0.05). EPIC patients received more transfusions (2.6 ± 3 vs. 0.6 ± 1 times) than LOCAL (p < 0.001) and received them significantly more frequently (61% vs. 25%). Within the LOCAL time periods, no statistically significant differences were found other than the need for transfusions (1st 32%, 2nd 28%, 3rd 9%, p = 0.005) and the average number of transfusions (1st 0.8 ± 1.6, 2nd 0.7 ± 1.3, 3rd 0.1 ± 0.3, p = 0.004). These differences remained significant after multivariate regression analysis and adjusting for risk variables. CONCLUSION The combination of erythropoietin, parenteral sucrose iron, fewer phlebotomies during the first 72 h, and delayed umbilical cord clamping seem to reduce red blood cell transfusion needs. This can be extremely important in high altitude units where higher hematocrit is desirable but may also be valuable at sea level.
Collapse
Affiliation(s)
- Angela B Hoyos
- Division of Neonatology, Clínica del Country, Bogota, DC, Colombia. .,Universidad El Bosque, Bogota, Colombia.
| | - Pablo Vasquez-Hoyos
- Sociedad de Cirugía Hospital de San José, Bogota, Colombia.,Department of Pediatrics, Universidad Nacional de Colombia, Bogota, Colombia.,Department of Pediatrics, Fundación Universitaria de Ciencias de la Salud, Bogota, Colombia
| |
Collapse
|
18
|
Kirk SE, Scheurer ME, Bernhardt MB, Mahoney DH, Powers JM. Phosphorus levels in children treated with intravenous ferric carboxymaltose. Am J Hematol 2021; 96:E215-E218. [PMID: 33735470 DOI: 10.1002/ajh.26165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 01/20/2023]
Affiliation(s)
- Susan E. Kirk
- Department of Pediatrics, Baylor College of Medicine Texas Children's Cancer and Hematology Centers Houston Texas
| | - Michael E. Scheurer
- Department of Pediatrics, Baylor College of Medicine Texas Children's Cancer and Hematology Centers Houston Texas
| | - Melanie Brooke Bernhardt
- Department of Pediatrics, Baylor College of Medicine Texas Children's Cancer and Hematology Centers Houston Texas
| | - Donald H. Mahoney
- Department of Pediatrics, Baylor College of Medicine Texas Children's Cancer and Hematology Centers Houston Texas
| | - Jacquelyn M. Powers
- Department of Pediatrics, Baylor College of Medicine Texas Children's Cancer and Hematology Centers Houston Texas
| |
Collapse
|
19
|
Affiliation(s)
- Schuyler Tong
- Department of Pediatrics and Pediatric Hematology Oncology, University of California at San Francisco Benioff Children's Hospital Oakland, Oakland, CA
| | - Elliott Vichinsky
- Department of Pediatrics and Pediatric Hematology Oncology, University of California at San Francisco Benioff Children's Hospital Oakland, Oakland, CA.,Pediatric Hematology Oncology, University of California, San Francisco, CA
| |
Collapse
|
20
|
Numan S, Kaluza K. Systematic review of guidelines for the diagnosis and treatment of iron deficiency anemia using intravenous iron across multiple indications. Curr Med Res Opin 2020; 36:1769-1782. [PMID: 32936683 DOI: 10.1080/03007995.2020.1824898] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To explore current recommendations for intravenous (IV) iron use in clinical guidelines for iron deficiency anemia (IDA) across different therapeutic areas and identify recommendations, if any, for the treatment of IDA. METHODS A literature search was conducted in Medline, EMBASE, BIOSIS, Cochrane Collaboration, and on websites of relevant professional associations. Searches were limited to English publications. 1292 citations were identified, 219 papers were assessed, and 35 guidelines were identified for inclusion. RESULTS The guidelines covered a variety of geographies: United States (US; n = 10); Europe (n = 11); "Rest-of-World" (n = 9); and "Other" organizations (n = 5). These covered a variety of specialties. Guidelines defined iron deficiency and IDA generally by serum ferritin and transferrin saturation levels. One-fifth of the reviewed guidelines (7 of 35) included no mention or recommendation regarding parenteral iron's utility in the management of IDA. Fifteen guidelines recommended using parenteral iron in the management of IDA. Fewer US guidelines included recommendations around IV iron than in Europe or the rest of the world. Approximately 60% of the guidelines have not been updated in ≥5 years and consequently do not reflect current evidence on the safety and efficacy of IV iron. CONCLUSIONS While national and international guidelines for management of IDA exist, many are outdated and do not reflect current evidence including, but not limited to, parenteral iron use. Urgent consideration should be given to updating and clarifying management guidelines for IDA using the latest treatment modalities and options, particularly in the US.
Collapse
Affiliation(s)
- Syed Numan
- Medical Affairs, American Regent, Norristown, PA, USA
| | | |
Collapse
|
21
|
Ozsahin H, Schaeppi M, Bernimoulin M, Allard M, Guidard C, van den Ouweland F. Intravenous ferric carboxymaltose for iron deficiency anemia or iron deficiency without anemia after poor response to oral iron treatment: Benefits and risks in a cohort of 144 children and adolescents. Pediatr Blood Cancer 2020; 67:e28614. [PMID: 32729200 DOI: 10.1002/pbc.28614] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/21/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE The objective of this single-center observational study was to determine the clinical and hematologic responses to intravenous ferric carboxymaltose (FCM) in a cohort of pediatric patients with poor response to oral iron therapy. The occurrence of adverse events was systematically recorded for up to 96 hours after infusion. STUDY DESIGN A retrospective cohort of 144 consecutive patients aged 18 months to < 18 years with iron deficiency anemia (IDA) or iron deficiency (ID) without anemia was investigated. All patients had failed oral iron therapy. The assessments before and after FCM treatment followed a predefined protocol. RESULTS One hundred of 117 (85 %) of patients with complete data achieved the target ferritin level ≥ 30 µg/L after a single FCM dose. Of 77 patients with IDA and complete data, 38 (49%) showed a complete hematological response within 6-12 weeks; a complete or partial response was achieved by 83%. Clinical symptoms improved in 85% of all patients. In 92% of patients (n = 133 /144), FCM infusion was uneventful. During the 96-hour follow-up, five patients reported potentially related symptoms. No serious adverse events occurred. CONCLUSION The study confirms the safety and efficacy of FCM in children (aged 18 months and older) and adolescents unresponsive to oral therapy, in real-world experience. Single-dose FCM treatment was followed by clinical improvement with advantages of safety, compliance, and lower cost compared with previous generation parenteral iron preparations that had to be administered in fractionated sessions.
Collapse
Affiliation(s)
- Hulya Ozsahin
- Centre de Pédiatrie, Clinique des Grangettes, Chêne-Bougeries, Switzerland
| | - Michela Schaeppi
- Centre de Pédiatrie, Clinique des Grangettes, Chêne-Bougeries, Switzerland
| | - Michael Bernimoulin
- Department of Oncology and Hematology, Hôpital de La Tour, Meyrin, Switzerland.,Dianalabs Medical Laboratory, Geneva, Switzerland
| | - Magali Allard
- Centre de Pédiatrie, Clinique des Grangettes, Chêne-Bougeries, Switzerland
| | - Clémence Guidard
- Centre de Pédiatrie, Clinique des Grangettes, Chêne-Bougeries, Switzerland
| | | |
Collapse
|
22
|
Lee B, Bae MI, Eum D, Ntungi AM, Jun B, Min KT. Bleeding properties according to surgical sites during pediatric craniotomy: a retrospective study comparing the two stages of epilepsy surgery. Anesth Pain Med (Seoul) 2020; 15:283-290. [PMID: 33329826 PMCID: PMC7713839 DOI: 10.17085/apm.20010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/08/2020] [Accepted: 07/08/2020] [Indexed: 11/26/2022] Open
Abstract
Background During pediatric epilepsy surgery, due to low circulating blood volume, intraoperative bleeding can result in significant hemodynamic instability, thereby requiring meticulous hemodynamic and transfusion strategies. Knowing the source of bleeding during the procedure would allow medical staff to better prepare the perioperative protocols for these patients. We compared intraoperative bleeding between the first (involving skin to meninges) and second (involving brain parenchyma) stages of epilepsy surgery to investigate the differences between various anatomical sites. Methods We reviewed the electronic medical records of 102 pediatric patients < 14 years old who underwent two-stage epilepsy surgeries during January 2012–December 2016. Invasive subdural grids were placed via craniotomy during Stage 1 and the epileptogenic zone was removed during Stage 2 of the surgery. We compared the volume of intraoperative bleeding between these two surgeries and identified variables associated with bleeding using multivariate regression analysis. Results Both surgeries resulted in similar intraoperative bleeding (24 vs. 26 ml/kg, P = 0.835), but Stage 2 required greater volumes of blood transfusion than Stage 1 (18.4 vs. 14.8 ml/kg, P = 0.011). Massive bleeding was associated with patients < 7 years of age in Stage 1 and weighing < 18 kg in Stage 2. Conclusions The volume of intraoperative bleeding was similar between the two stages of pediatric epilepsy surgery and was large enough to require blood transfusions. Thus, blood loss during pediatric epilepsy surgery occurred at both anatomic sites. This indicates the necessity of early preparation for blood transfusion in both stages of pediatric epilepsy surgery.
Collapse
Affiliation(s)
- Bora Lee
- Department of Anesthesiology and Pain Medicine, Severance Hospital and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Myung Il Bae
- Department of Anesthesiology and Pain Medicine, Severance Hospital and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Darhae Eum
- Department of Anesthesiology and Pain Medicine, Severance Hospital and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Abel Mussa Ntungi
- Department of Anesthesiology, Muhimbili University of Health and Allied Sciences, Dar Es Salaam, United Republic of Tanzania
| | - Byongnam Jun
- Department of Anesthesiology and Pain Medicine, Severance Hospital and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Kyeong Tae Min
- Department of Anesthesiology and Pain Medicine, Severance Hospital and Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Iron Deficiency Anemia in Children Residing in High and Low-Income Countries: Risk Factors, Prevention, Diagnosis and Therapy. Mediterr J Hematol Infect Dis 2020; 12:e2020041. [PMID: 32670519 PMCID: PMC7340216 DOI: 10.4084/mjhid.2020.041] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022] Open
Abstract
Iron deficiency and iron-deficiency anemia (IDA) affects approximately two billion people worldwide, and most of them reside in low- and middle-income countries. In these nations, additional causes of anemia include parasitic infections like malaria, other nutritional deficiencies, chronic diseases, hemoglobinopathies, and lead poisoning. Maternal anemia in resource-poor nations is associated with low birth weight, increased perinatal mortality, and decreased work productivity. Maintaining a normal iron balance in these settings is challenging, as iron-rich foods with good bioavailability are of animal origin and either expensive and/or available in short supply. Apart from infrequent consumption of meat, inadequate vitamin C intake, and diets rich in inhibitors of iron absorption are additional important risk factors for IDA in low-income countries. In-home iron fortification of complementary foods with micronutrient powders has been shown to effectively reduce the risk of iron deficiency and IDA in infants and young children in developing countries but is associated with unfavorable changes in gut flora and induction of intestinal inflammation that may lead to diarrhea and hospitalization. In developed countries, iron deficiency is the only frequent micronutrient deficiency. In the industrialized world, IDA is more common in infants beyond the sixth month of life, in adolescent females with heavy menstrual bleeding, in women of childbearing age and older people. Other special at-risk populations for IDA in developed countries are regular blood donors, endurance athletes, and vegetarians. Several medicinal ferrous or ferric oral iron products exist, and their use is not associated with harmful effects on the overall incidence of infectious illnesses in sideropenic and/or anemic subjects. However, further research is needed to clarify the risks and benefits of supplemental iron for children exposed to parasitic infections in low-income countries, and for children genetically predisposed to iron overload.
Collapse
|
24
|
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
|
25
|
A Review of Intravenous Iron Replacement Medications for Nurse Practitioners. J Nurse Pract 2020. [DOI: 10.1016/j.nurpra.2019.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
26
|
Iron Deficiency Is Associated with Adverse Outcomes in Pediatric Heart Failure. J Pediatr 2020; 216:58-66.e1. [PMID: 31610927 DOI: 10.1016/j.jpeds.2019.08.060] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/14/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate the prevalence of iron deficiency and its association with outcomes in children with heart failure. STUDY DESIGN A single-center retrospective cohort study of patients with heart failure aged 1-21 years from July 2012 to June 2017 with available serum iron studies was performed. Subjects were analyzed in 2 groups: biventricular systolic heart failure (BiV) and single-ventricle congenital heart disease with systolic heart failure (SV). Iron deficiency was defined as ≥2 of the following: serum iron <50 μg/dL, serum ferritin <20 ng/mL, transferrin >300 ng/mL, or transferrin saturation <15%. The primary outcome was a composite adverse event (CAE) of ventricular assist device implantation, heart transplantation, or death, at 3 and 6 months from time of iron studies. RESULTS Of the 107 subjects (77 BiV, 30 SV) included in the study, 56% were iron deficient. Demographics, etiology of heart failure, and chronicity of heart failure symptoms were not associated with iron deficiency. On multivariable analysis, in group BiV, iron deficiency was associated with CAE at 3 months (79% iron deficiency in CAE group vs 37% iron deficiency in non-CAE, P = .001, OR 7, 95% CI 2-21) and 6 months (76% iron deficiency in CAE vs 35% iron deficiency in non-CAE, P = .002, OR 7, 95% CI 2-24). In group SV, iron deficiency was associated with CAE at 6 months (79% iron deficiency in CAE vs 29% iron deficiency in non-CAE, P = .014, OR 8, 95% CI 2-32). CONCLUSIONS Iron deficiency was present in 56% of the pediatric patients with heart failure who were evaluated with iron studies. Iron deficiency was associated with greater risk of ventricular assist device implantation, heart transplantation, or death.
Collapse
|
27
|
Powers JM, Buchanan GR. Disorders of Iron Metabolism: New Diagnostic and Treatment Approaches to Iron Deficiency. Hematol Oncol Clin North Am 2019; 33:393-408. [PMID: 31030809 DOI: 10.1016/j.hoc.2019.01.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Iron deficiency anemia is the leading cause of anemia worldwide and affects many young children and adolescent girls in the United States. Its signs and symptoms are subtle despite significant clinical effects. Iron deficiency anemia is diagnosed clinically by the presence of risk factors and microcytic anemia. Improvement following a trial of oral iron therapy is confirmative. An array of iron laboratory tests is available with variable indications. Clinical trial and iron absorption data support a shift to lower-dose oral iron therapy. Intravenous iron should be considered in children who fail oral iron or who have more complex disorders.
Collapse
Affiliation(s)
- Jacquelyn M Powers
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Texas Children's Cancer and Hematology Centers, 6701 Fannin Street, Suite 1580, Houston, TX 77030, USA.
| | - George R Buchanan
- Pediatric Hematology-Oncology, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, H3.104A, Dallas, TX 75390-9063, USA
| |
Collapse
|
28
|
Powers JM, O'Brien SH. How I approach iron deficiency with and without anemia. Pediatr Blood Cancer 2019; 66:e27544. [PMID: 30393941 DOI: 10.1002/pbc.27544] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022]
Abstract
Iron deficiency anemia remains a common referral to the pediatric hematology-oncology subspecialist. Improved understanding of iron homeostasis, including the effects of the regulatory hormone hepcidin, recent adult and pediatric clinical trial data, as well as the availability of safer formulations of intravenous iron, have resulted in additional considerations when making treatment recommendations in such patients. Young children and adolescent females remain the most commonly affected groups, but children with complex medical or chronic inflammatory conditions including comorbid gastrointestinal disorders also require special consideration.
Collapse
Affiliation(s)
- Jacquelyn M Powers
- Department of Pediatrics, Section of Hematology/Oncology, Baylor College of Medicine, Houston, Texas.,Texas Children's Hospital, Houston, Texas
| | - Sarah H O'Brien
- Division of Hematology/Oncology, Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
29
|
Abstract
BACKGROUND Ferumoxytol, an "off-label" contrast agent, allows for better cardiac MRI quality as compared with gadolinium-based contrast agents. However, hypotension has been reported with the use of ferumoxytol for indications other than cardiac MRI. The purpose of our investigation was to evaluate the safety of ferumoxytol in children undergoing general anaesthesia for cardiac MRI. METHODS Medical records of children undergoing general anaesthesia for cardiac MRI were reviewed. Baseline demographic and medical characteristics, as well as imaging and anaesthetic duration and technique, were collected. The incidence of hypotension or other adverse events', need for vasoactive support, or airway intervention throughout the anaesthetic, was recorded. RESULTS A total of 95 patients were identified, 61 received ferumoxytol and 34 received gadolinium. There were no significant differences between groups with respect to age, weight, or baseline blood pressure. The incidence of low blood pressure - systolic or mean - after contrast administration did not differ between groups, and there was no difference in sustained hypotension or use of vasopressors between groups. One patient who received ferumoxytol had possible anaphylaxis. The image acquisition time (45 versus 68 min, p=0.002) and anaesthesia duration (100 versus 132 min, p=0.02) were shorter in the ferumoxytol group. CONCLUSION Transient low blood pressure was common in children undergoing cardiac MRI with anaesthesia, but the incidence of hypotension did not differ between ferumoxytol and gadolinium groups. The use of ferumoxytol was associated with significantly shorter scan time and anaesthesia duration, as well as a decreased need for airway intervention.
Collapse
|
30
|
Boucher AA, Pfeiffer A, Bedel A, Young J, McGann PT. Utilization trends and safety of intravenous iron replacement in pediatric specialty care: A large retrospective cohort study. Pediatr Blood Cancer 2018; 65:e26995. [PMID: 29369486 DOI: 10.1002/pbc.26995] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/19/2017] [Accepted: 01/02/2018] [Indexed: 01/20/2023]
Abstract
BACKGROUND Iron deficiency is a common and clinically diverse hematologic disorder in childhood for which oral iron is often an infeasible or ineffective treatment option. Intravenous (IV) iron can be an efficient and highly successful means of iron replacement but its use has not been well-characterized on a large scale in pediatrics. PROCEDURE All IV iron doses administered to patients for iron replacement therapy at a tertiary pediatric hospital from January 2010 through October 2016 were evaluated. Analyses included patient demographics, underlying medical conditions, and detailed information for each dose. Individual chart review was performed to identify infusion-related reactions. Nephrology patients as well as those patients 21 years or older at the time of the first infusion were excluded. RESULTS A total of 1,088 doses of IV iron administered to 194 patients met inclusion criteria. A wide variety of specialties prescribed IV iron, with gastroenterology, hematology, and hospital medicine being the highest users in this cohort. A majority of patients (68%) required multiple infusions and dosing was highly variable, ranging from 1.3-1,030 mg per infusion. Premedication use was infrequent (10.3% of doses) and no severe infusion-associated reactions occurred. CONCLUSIONS IV iron is commonly prescribed by certain pediatric specialties but there is little standardization in the indications, formulations, or dosing. These data suggest that IV iron should be considered a safe alternative for iron deficiency treatment in pediatrics when oral iron is either unsuccessful or contraindicated.
Collapse
Affiliation(s)
- Alexander A Boucher
- Division of Hematology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Amanda Pfeiffer
- Division of Hematology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ashley Bedel
- Division of Pharmacy, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jennifer Young
- Division of Pharmacy, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Patrick T McGann
- Division of Hematology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
31
|
Mantadakis E. Intravenous iron: Safe and underutilized in children. Pediatr Blood Cancer 2018; 65:e27016. [PMID: 29493089 DOI: 10.1002/pbc.27016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Elpis Mantadakis
- Department of Pediatrics, Faculty of Medicine, University General Hospital of Evros, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
32
|
Miele E, Shamir R, Aloi M, Assa A, Braegger C, Bronsky J, de Ridder L, Escher JC, Hojsak I, Kolaček S, Koletzko S, Levine A, Lionetti P, Martinelli M, Ruemmele F, Russell RK, Boneh RS, van Limbergen J, Veereman G, Staiano A. Nutrition in Pediatric Inflammatory Bowel Disease: A Position Paper on Behalf of the Porto Inflammatory Bowel Disease Group of the European Society of Pediatric Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol Nutr 2018; 66:687-708. [PMID: 29570147 DOI: 10.1097/mpg.0000000000001896] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS A growing body of evidence supports the need for detailed attention to nutrition and diet in children with inflammatory bowel disease (IBD). We aimed to define the steps in instituting dietary or nutritional management in light of the current evidence and to offer a useful and practical guide to physicians and dieticians involved in the care of pediatric IBD patients. METHODS A group of 20 experts in pediatric IBD participated in an iterative consensus process including 2 face-to-face meetings, following an open call to Nutrition Committee of the European Society of Pediatric Gastroenterology, Hepatology and Nutrition Porto, IBD Interest, and Nutrition Committee. A list of 41 predefined questions was addressed by working subgroups based on a systematic review of the literature. RESULTS A total of 53 formal recommendations and 47 practice points were endorsed with a consensus rate of at least 80% on the following topics: nutritional assessment; macronutrients needs; trace elements, minerals, and vitamins; nutrition as a primary therapy of pediatric IBD; probiotics and prebiotics; specific dietary restrictions; and dietary compounds and the risk of IBD. CONCLUSIONS This position paper represents a useful guide to help the clinicians in the management of nutrition issues in children with IBD.
Collapse
Affiliation(s)
- Erasmo Miele
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II," Naples, Italy
| | - Raanan Shamir
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marina Aloi
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Rome, Italy
| | - Amit Assa
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Christian Braegger
- Division of Gastroenterology and Nutrition, University Children's Hospital Zurich, Zurich, Switzerland
| | - Jiri Bronsky
- Gastroenterology and Nutrition Unit, Department of Paediatrics, 2nd Faculty of Medicine, Charles, University and Motol University Hospital, Prague, Czech Republic
| | - Lissy de Ridder
- Department of Paediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Johanna C Escher
- Department of Paediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Iva Hojsak
- Department of Gastroenterology, Children Hospital Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sanja Kolaček
- Department of Gastroenterology, Children Hospital Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sibylle Koletzko
- Dr. von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Arie Levine
- Wolfson Medical Center, Sackler School of Medicine, Tel-Aviv, Israel
| | - Paolo Lionetti
- Meyer Children Hospital, University of Florence, Florence, Italy
| | - Massimo Martinelli
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II," Naples, Italy
| | - Frank Ruemmele
- Université Sorbonne Paris Cité, Université Paris Descartes, and Assistance publique-hôpitaux de Paris, Hôpital Necker-Enfants malades, Service de gastroentérologie pédiatrique, Paris, France
| | - Richard K Russell
- Department of Paediatric Gastroenterology, The Royal Hospital for Children, Glasgow, Scotland
| | | | - Johan van Limbergen
- Department of Pediatrics, Division of Pediatric Gastroenterology & Nutrition, IWK Health Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gigi Veereman
- Department of Paediatric Gastroenterology and Nutrition, University Hospital Brussels, Free University Brussels, Brussels, Belgium
| | - Annamaria Staiano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II," Naples, Italy
| |
Collapse
|
33
|
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
|
34
|
Tan MLN, Windscheif PM, Thornton G, Gaynor E, Rodrigues A, Howarth L. Retrospective review of effectiveness and safety of intravenous ferric carboxymaltose given to children with iron deficiency anaemia in one UK tertiary centre. Eur J Pediatr 2017; 176:1419-1423. [PMID: 28844092 DOI: 10.1007/s00431-017-2995-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 08/06/2017] [Accepted: 08/10/2017] [Indexed: 11/27/2022]
Abstract
UNLABELLED In the paediatric population, ferric carboxymaltose (FCM) is only licenced for use in children older than 14 years, and the data in younger children remains scarce. We retrospectively reviewed data of all paediatric patients less than 14 years old who had received FCM infusion from August 2011 to June 2015 at the John Radcliffe Hospital (Oxford University Hospitals), UK. The patient demographics, significant medical history, FCM dose, and blood investigations (pre-FCM and post-FCM) were reviewed. Of the 51 children, 41 had inflammatory bowel disease. There were 24 girls and 27 boys, aged 1 to 13 years, mean (SD) weight 28.4 (13.6) kg. Fifteen patients received at least one more course of FCM up to 35 months later. The time interval between pre-FCM and post-FCM investigations was 1 to 8 months. An improved, median (range) rise in blood indices following one FCM infusion was haemoglobin 2.7 (- 2.4 to 7) g/dL, serum iron 6.6 (- 0.6 to 21.1) μmol/L, and transferrin saturation 14 (- 14 to 38)%. No adverse outcomes were documented. CONCLUSIONS FCM was effective in increasing the key blood indices with no adverse outcomes in children less than 14 years of age, with a range of different conditions, majority with gastrointestinal disorders such as IBD. What is Known: • Ferric carboxymaltose (FCM) given via the intravenous (IV) route has been used widely in adults for the treatment of iron deficiency anaemia. • Sparse data exists on FCM use in paediatric population, including young children What is New: • FCM infusion should be considered as a means of iron administration in the paediatric population less than 14 years of age • No adverse outcomes were recorded following FCM in a young paediatric population (less than 14 years of age); the majority of whom had gastrointestinal disorders.
Collapse
Affiliation(s)
- Michelle Li Nien Tan
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Paediatrics 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, 119228, Singapore.
| | | | - Graham Thornton
- Department of Paediatric Gastroenterology, Oxford University Hospitals Trust, Oxford, UK
| | - Ed Gaynor
- Department of Paediatric Gastroenterology, Oxford University Hospitals Trust, Oxford, UK
| | - Astor Rodrigues
- Department of Paediatric Gastroenterology, Oxford University Hospitals Trust, Oxford, UK
| | - Lucy Howarth
- Department of Paediatric Gastroenterology, Oxford University Hospitals Trust, Oxford, UK
| |
Collapse
|
35
|
Carvalho FSGD, de Medeiros IA, Antunes H. Prevalence of iron deficiency anemia and iron deficiency in a pediatric population with inflammatory bowel disease. Scand J Gastroenterol 2017. [PMID: 28643539 DOI: 10.1080/00365521.2017.1342137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Iron deficiency is the most common cause of anemia in children with inflammatory bowel disease, although the real prevalence is unknown. Intravenous iron is suggested as the first line treatment. This study aims to determine the prevalence of iron deficiency anemia in children with inflammatory bowel disease followed in a Pediatric Gastroenterology Unit of a tertiary center and to evaluate this unit's experience with intravenous iron. MATERIALS AND METHODS A retrospective cohort study was designed involving children with inflammatory bowel disease followed in that unit between January 2001 and April 2016. Laboratory results were collected at the moment of diagnosis, after one-year follow-up and prior each IV iron administration performed during the study period. Anemia was defined according to World Health Organization criteria and the iron deficiency was defined using recent guidelines. RESULTS Were studied 69 patients 71% had CD and 29% UC. 50.7% were female. Mean patient age at diagnosis was 13.3 years (range 1--17 years). Prevalence of ID and IDA at diagnosis was 76.8% and 43.5%, respectively. After one year follow-up, those values decreased to 68.1% (p = .182) and 21.7% (p = .002), respectively. Hemoglobin significantly increased (p < .001). Intravenous iron was administered to 92.8% of patients. No adverse reactions were reported. CONCLUSIONS Intravenous iron is the first line in the treatment of Iron deficiency anemia in Inflammatory Bowel disease and it is safe and effective. Persistent anemia and iron deficiency are common.
Collapse
Affiliation(s)
| | - Inês Ambrósio de Medeiros
- a Gastroenterology, Hepatology and Nutrition Unit, Pediatrics Department , Hospital de Braga , Braga , Portugal
| | - Henedina Antunes
- a Gastroenterology, Hepatology and Nutrition Unit, Pediatrics Department , Hospital de Braga , Braga , Portugal.,b Life and Health Sciences Research Institute (ICVS), School of Medicine , University of Minho , Braga , Portugal
| |
Collapse
|
36
|
Abstract
Iron deficiency anemia (IDA) is the most common nutritional deficiency in children. Most children with IDA are treated with oral iron preparations. However, intravenous (IV) iron is an alternative for children with severe IDA who have difficulty in adhering to or absorbing oral iron. We sought to describe the safety and effectiveness of IV iron sucrose for treatment of IDA in children. Pharmacy records of children who received IV iron sucrose at a children's hospital between 2004 and 2014 were reviewed. Laboratory markers of anemia and iron studies were obtained and preinfusion and postinfusion values were compared. Records were also reviewed for adverse reactions. A total of 142 patients received IV iron sucrose over 10 years. The mean age was 11 years, 9 months. One patient of 142 developed cough and wheezing during the infusion. No other adverse events were found. IV iron sucrose resulted in a statistically significant and clinically meaningful increase in hemoglobin, mean corpuscular volume, serum iron, ferritin, and % iron saturation, with a corresponding decrease in total iron binding capacity. The use of IV iron sucrose in pediatric patients with IDA is safe and leads to a moderate increase in hemoglobin and substantial improvement in iron studies.
Collapse
|
37
|
Karadeniz C, Özdemir R, Demirol M, Katipoğlu N, Yozgat Y, Meşe T, Ünal N. Low Iron Stores in Otherwise Healthy Children Affect Electrocardiographic Markers of Important Cardiac Events. Pediatr Cardiol 2017; 38:909-914. [PMID: 28271153 DOI: 10.1007/s00246-017-1596-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 02/23/2017] [Indexed: 11/28/2022]
Abstract
Both an excess of iron and iron deficiency (ID) may lead to significant cardiac problems. Parameters that represent ventricular repolarization heterogeneity, like QT dispersion (QTd), corrected QT dispersion (QTcd), the interval between the peak and the end of the T wave (Tp-e), and Tp-e dispersion, have not been evaluated in otherwise healthy children with low iron levels before. Here we assessed the effects of low iron storage on P wave dispersion (PWd), QTd, Tp-e intervals, and Tp-e dispersion in otherwise healthy children. We prospectively reviewed 283 patients who were referred to pediatric cardiology department for cardiac evaluation due to murmurs and who were found to have no structural heart disease. The patients were divided into three groups according to their ferritin levels: Group 1: ferritin <15 ng/mL (n = 58); Group 2: ferritin 15-25 ng/mL (n = 80); Group 3: ferritin >25 ng/mL (n = 145). P wave duration (PW), QT and Tp-e intervals, and PW, QT, corrected QT (QTc), and Tp-e dispersions were significantly higher in patients whose ferritin level was <15 ng/mL. A negative correlation was found between ferritin level and QT and QTc intervals, and QT, QTc, and Tp-e dispersions. Our results showed that a low serum ferritin level is associated with changes in some ECG parameters such as prolonged PWd, Tp-e interval, QT, QTc, and Tp-e dispersions in otherwise healthy children, and studies of other populations indicated that these parameters may predict arrhythmias in selected patients. These patients may be considered at some risk of developing arrhythmias. Therefore, careful evaluation of these ECG parameters is necessary in otherwise healthy children with low iron stores.
Collapse
Affiliation(s)
- Cem Karadeniz
- Department of Pediatric Cardiology, Izmir Dr. Behcet Uz Children's Hospital, 1374 St. No: 11 Alsancak, Izmir, Turkey.
| | - Rahmi Özdemir
- Department of Pediatric Cardiology, Izmir Dr. Behcet Uz Children's Hospital, 1374 St. No: 11 Alsancak, Izmir, Turkey
| | - Mustafa Demirol
- Department of Pediatric Cardiology, Izmir Dr. Behcet Uz Children's Hospital, 1374 St. No: 11 Alsancak, Izmir, Turkey
| | - Nagehan Katipoğlu
- Department of Pediatrics, Izmir Dr. Behcet Uz Children's Hospital, Izmir, Turkey
| | - Yılmaz Yozgat
- Department of Pediatric Cardiology, Izmir Dr. Behcet Uz Children's Hospital, 1374 St. No: 11 Alsancak, Izmir, Turkey
| | - Timur Meşe
- Department of Pediatric Cardiology, Izmir Dr. Behcet Uz Children's Hospital, 1374 St. No: 11 Alsancak, Izmir, Turkey
| | - Nurettin Ünal
- Department of Pediatric Cardiology, Izmir Dr. Behcet Uz Children's Hospital, 1374 St. No: 11 Alsancak, Izmir, Turkey
| |
Collapse
|
38
|
Mantadakis E, Roganovic J. Safety and efficacy of ferric carboxymaltose in children and adolescents with iron deficiency anemia. J Pediatr 2017; 184:241. [PMID: 28196681 DOI: 10.1016/j.jpeds.2017.01.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/13/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Elpis Mantadakis
- Department of Pediatrics Pediatric Hematology/Oncology Unit University General Hospital of Evros Alexandroupolis, Thrace, Greece
| | - Jelena Roganovic
- Department of Pediatrics Division of Hematology and Oncology Clinical Hospital Centre Rijeka Rijeka, Croatia
| |
Collapse
|
39
|
MacQueen BC, Baer VL, Scott DM, Ling CY, O’Brien EA, Boyer C, Henry E, Fleming RE, Christensen RD. Iron Supplements for Infants at Risk for Iron Deficiency. Glob Pediatr Health 2017; 4:2333794X17703836. [PMID: 28491927 PMCID: PMC5405879 DOI: 10.1177/2333794x17703836] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 03/08/2017] [Indexed: 11/30/2022] Open
Abstract
Professional societies have published recommendations for iron dosing of preterm neonates, but differences exist between guidelines. To help develop standardized guidelines, we performed a 10-year analysis of iron dosing in groups at risk for iron deficiency: IDM (infants of diabetic mothers), SGA (small for gestational age), and VLBW premature neonates (very low birth weight, <1500 g). We analyzed iron dosing after red cell transfusions and erythropoiesis-stimulating agents (ESA). Of IDM, 11.8% received iron in the hospital; 9.8% of SGA and 27.1% of VLBW neonates received iron. Twenty percent of those who received iron had it started by day 14; 63% by 1 month. Supplemental iron was stopped after red cell transfusions in 73% of neonates receiving iron. An ESA was administered to 1677, of which 33% received iron within 3 days. This marked variation indicates that a consistent approach is needed, and using this report and a literature review, we standardized our iron-dosing guidelines.
Collapse
Affiliation(s)
| | | | - Danielle M. Scott
- Intermountain Healthcare, Salt Lake City, UT, USA
- Intermountain Medical Center, Murray, UT, USA
| | - Con Yee Ling
- University of Utah, Salt Lake City, UT, USA
- Intermountain Healthcare, Salt Lake City, UT, USA
| | - Elizabeth A. O’Brien
- University of Utah, Salt Lake City, UT, USA
- Intermountain Healthcare, Salt Lake City, UT, USA
| | | | - Erick Henry
- Intermountain Healthcare, Salt Lake City, UT, USA
- Institute for Healthcare Delivery Research, Salt Lake City, UT, USA
| | | | - Robert D. Christensen
- University of Utah, Salt Lake City, UT, USA
- Intermountain Healthcare, Salt Lake City, UT, USA
| |
Collapse
|
40
|
Hassan N, Boville B, Reischmann D, Ndika A, Sterken D, Kovey K. Intravenous Ferumoxytol in Pediatric Patients With Iron Deficiency Anemia. Ann Pharmacother 2017. [DOI: 10.1177/1060028017699429] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background: Iron deficiency anemia (IDA) is common in children. Limited data exist on the efficacy and safety of ferumoxytol in children. Objective: To assess the efficacy of 10 mg/kg dose given over 15-60 minutes in correcting IDA and report any adverse drug reactions (ADRs). Methods: We conducted a retrospective review of all patients who received ferumoxytol infusions for the management of IDA by the Pediatric Blood Management Program between October 2010 and March 2015. Results: A total of 110 infusions were given to 54 patients. Compared with baseline preinfusion hemoglobin (Hb; 9.2 ± 1.9 g/dL), a significant rise was seen at 1 week and 4 weeks postinfusion (11.5 ± 1.5 and 11.8 ± 1.7 g/dL, respectively, P < 0.001). Also, a significant rise in serum ferritin at 1 week and 4 weeks postinfusion was seen (51 ± 71 vs 192 ± 148 and 89 ± 135 ng/mL, P < 0.001 and <0.035, respectively). Patients who concomitantly received erythropoietin had a significantly larger Hb rise from baseline than those who did not at 4 weeks (2.7 ± 2.2 vs 1.6 ± 1.1 g/dL, P < 0.017). ADRs included pruritus (n = 1), urticaria (n = 1), and multisymptom episodes (n = 3) that included shortness of breath, chest tightness, back pain, and epigastric cramping that responded to therapy with IV diphenhydramine and methylprednisolone. Conclusion: Ferumoxytol was effective in treating IDA in our small study. Slow infusion rate and close monitoring allowed early detection of the infrequent ADRs.
Collapse
Affiliation(s)
- Nabil Hassan
- Children’s Hospital of Illinois at OSF St Frances, Peoria, IL, USA
| | - Brian Boville
- Helen Devos Children’s Hospital, Grand Rapids, MI, USA
| | | | - Akunne Ndika
- Grand Rapids Medical Education Partners, MI, USA
| | - David Sterken
- Helen Devos Children’s Hospital, Grand Rapids, MI, USA
| | - Karen Kovey
- Parkview Regional Medical Center, Fort Wayne, IN, USA
| |
Collapse
|
41
|
Powers JM, Shamoun M, McCavit TL, Adix L, Buchanan GR. Intravenous Ferric Carboxymaltose in Children with Iron Deficiency Anemia Who Respond Poorly to Oral Iron. J Pediatr 2017; 180:212-216. [PMID: 27776750 DOI: 10.1016/j.jpeds.2016.09.053] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/28/2016] [Accepted: 09/19/2016] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To assess the benefits and risks of intravenous (IV) ferric carboxymaltose (FCM) in children with iron deficiency anemia (IDA). STUDY DESIGN In a retrospective cohort study of patients seen at our center, we identified all FCM infusions in children with IDA over a 12-month period through a query of pharmacy records. Clinical data, including hematologic response and adverse effects, were extracted from the electronic medical record. RESULTS A total of 116 IV FCM infusions were administered to 72 patients with IDA refractory to oral iron treatment (median age, 13.7 years; range, 9 months to 18 years). Median preinfusion and postinfusion hemoglobin values were 9.1 g/dL and 12.3 g/dL, respectively (at 4-12 weeks after the initial infusion; n = 53). Sixty-five patients (84%) experienced no adverse effects. Minor transient complications were encountered during or immediately after 7 infusions. CONCLUSION FCM administered as a short IV infusion without a test dose proved to be safe and highly effective in a small yet diverse population of infants, children, and adolescents with IDA refractory to oral iron therapy.
Collapse
Affiliation(s)
- Jacquelyn M Powers
- Division of Hematology/Oncology, Baylor College of Medicine, Houston, TX; Department of Pediatrics, Baylor College of Medicine, Houston, TX; Texas Children's Hospital, Houston, TX.
| | - Mark Shamoun
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX; Children's Health, Dallas, TX
| | - Timothy L McCavit
- Division of Hematology/Oncology, Cook Children's Hospital, Fort Worth, TX; Department of Pediatrics, Cook Children's Hospital, Fort Worth, TX
| | | | - George R Buchanan
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX; Children's Health, Dallas, TX; Division of Hematology/Oncology, The University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
42
|
Affiliation(s)
- Jelena Roganovic
- Division of Hematology and Oncology, Department of Pediatrics, Clinical Hospital Centre Rijeka, Rijeka, Istarska 43, HR, 51000, Rijeka, Croatia.
| |
Collapse
|