scholarly journals Iron Supplementation: What's New?

Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. SCI-43-SCI-43
Author(s):  
Iain C. Macdougall

Abstract Supplemental orally-administered iron has been available for centuries, but parenteral administration of iron as a therapeutic agent dates from the 1930s. The original agents were iron salts, and their administration to man was associated with severe and unacceptable side-effects even with doses of 4-8 mg. The modern-day practice of IV iron supplementation was transformed in the 1940s with the introduction of iron incorporated into a carbohydrate shell, to allow slow release in the circulation and rapid binding to plasma transferrin. The older iron-carbohydrate complexes included iron dextran, iron sucrose, and iron gluconate, with different iron release kinetics. Iron dextran had significant toxicity with possible anaphylactic reactions. Other iron preparations can also induce hypersensitivity reactions, although recent evidence suggests that these are not immunoglobulin-mediated, but may be due to free iron in circulation or mediated via complement, the so-called CARPA reactions (complement activation-related pseudo-allergy). Newer oral iron preparations include ferric citrate which is much more tolerable than ferrous sulphate, while newer IV iron compounds include ferric carboxymaltose, iron isomaltoside 1000 , and ferumoxytol. Iron supplementation has traditionally been used to ameliorate iron deficiency anaemia, and in the early 1990s, IV iron was found to be mandatory in dialysis patients to support erythropoiesis under stimulation with recombinant human erythropoietin. Recent data also suggest that IV iron may have benefits in chronic heart failure, independent of any effect on haemoglobin or red cell production, possibly by enhancing cardiac myocyte mitochondrial function. Although most RCTs suggest better efficacy with IV iron versus oral iron, there are some concerns regarding the safety of parenteral iron, not only in relation to hypersensitivity reactions, but also exacerbation of oxidative stress or infections. Several ongoing RCTs are seeking to provide further evidence in this regard, e.g. the PIVOTAL trial. Newer strategies for replenishing iron stores include HIF prolyl hydroxylase inhibitors (HIF PHIs), antagonists of the hepcidin pathway, or dialysate iron administration as ferric pyrophosphate citrate. HIF PHIs (such as roxadustat and vadaddustat) have been shown to reduce hepcidin, probably indirectly via stimulation of erythropoiesis, as well as by stimulating a number of iron-regulatory genes, and allowing greater amounts of iron to be absorbed orally. Many molecules designed to interfere with hepcidin activity are in varying stages of clinical development, including monoclonal antibodies against hepcidin or its signalling pathway, spiegelmers, anticalins, and antisense oligonucleotides. Administering ferric pyrophosphate citrate (Triferric™) across dialysis has recently become clinical reality in the US, although this is not available in Europe. Thus, strategies to supplement iron have developed considerably over the last two decades, with several novel approaches transitioning from bench to bedside. Disclosures Macdougall: Astellas: Honoraria, Research Funding; GlaxoSmithKline: Consultancy; Akebia: Consultancy; Vifor Pharma: Consultancy, Honoraria, Research Funding, Speakers Bureau; Pharmacosmos: Honoraria; AMAG: Consultancy, Honoraria, Research Funding; FibroGen: Consultancy, Speakers Bureau.

Animals ◽  
2020 ◽  
Vol 10 (7) ◽  
pp. 1113
Author(s):  
Rafał Mazgaj ◽  
Mateusz Szudzik ◽  
Paweł Lipiński ◽  
Aneta Jończy ◽  
Ewa Smuda ◽  
...  

Background: The similarities between swine and humans in physiological and genomic patterns, as well as significant correlation in size and anatomy, make pigs an useful animal model in nutritional studies during pregnancy. In humans and pigs iron needs exponentially increase during the last trimester of pregnancy, mainly due to increased red blood cell mass. Insufficient iron supply during gestation may be responsible for the occurrence of maternal iron deficiency anemia and decreased iron status in neonates. On the other hand, preventive iron supplementation of non-anemic mothers may be of potential risk due to iron toxicity. Several different regimens of iron supplementation have been applied during pregnancy. The majority of oral iron supplementations routinely applied to pregnant sows provide inorganic, non-heme iron compounds, which exhibit low bioavailability and intestinal side effects. The aim of this study was to check, using pig as an animal model, the effect of sucrosomial ferric pyrophosphate (SFP), a new non-heme iron formulation on maternal and neonate iron and hematological status, placental transport and pregnancy outcome; Methods: Fifteen non-anemic pregnant sows were recruited to the experiment at day 80 of pregnancy and randomized into the non-supplemented group (control; n = 5) and two groups receiving oral iron supplementation—sows given sucrosomial ferric pyrophosphate, 60 mg Fe/day (SFP; n = 5) (SiderAL®, Pisa, Italy) and sows given ferrous sulfate 60 mg Fe/day (Gambit, Kutno, Poland) (FeSO4; n = 5) up to delivery (around day 117). Biological samples were collected from maternal and piglet blood, placenta and piglet tissues. In addition, data on pregnancy outcome were recorded.; Results: Results of our study show that both iron supplements do not alter neither systemic iron homeostasis in pregnant sows nor their hematological status at the end of pregnancy. Moreover, we did not detect any changes of iron content in the milk and colostrum of iron supplemented sows in comparison to controls. Neonatal iron status of piglets from iron supplemented sows was not improved compared with the progeny of control females. No statistically significant differences were found in average piglets weight and number of piglets per litter between animals from experimental groups. The placental expression of iron transporters varied depending on the iron supplement.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3363-3363
Author(s):  
Florian Scotté ◽  
Kamel Laribi ◽  
Christian Gisselbrecht ◽  
Dominique Spaeth ◽  
Emna Kasdaghli ◽  
...  

Abstract Background Recommendations on erythropoiesis-stimulating agents (ESAs) for the management of chemotherapy-induced anemia (CIA) are well established (Schrijvers D et al. Ann Oncol 2010;21[suppl 5]:v244-7). Iron supplementation can further improve treatment response of CIA, particularly in the case of iron deficiency (Pedrazzoli P et al. J Clin Oncol 2008;26:1619-25; Auerbach M et al. J Clin Oncol 2004;22:1301-7), but is under-used. Objective To evaluate the effect of epoetin alfa biosimilar, with or without iron, on CIA in current oncology and hematology practice. Methods SYNERGY was an observational, longitudinal, prospective, multicenter study conducted in France, from a representative, random sample of oncologists and hematologists. Patients of these clinicians were aged ≥18 years with solid tumors, lymphoma and/or myeloma and CIA, eligible for treatment with epoetin alfa biosimilar and followed for 12-16 weeks. A subanalysis describing the treatment response to epoetin alfa biosimilar in patients with/without iron supplementation and their target hemoglobin (Hb) levels is presented here. Results Overall, 2167 patients were enrolled by 195 French oncologists/hematologists during June 2012-December 2014. The analysis included 2076 patients, of whom half were male. At inclusion, mean age ± standard deviation (SD) was 67±12 years and 75.7% (n=1517) of patients had a World Health Organization performance status of 0 or 1. Most patients had not received any blood transfusions (90.0%, n=1867) or ESAs (93.1%, n=1932) in the year before the inclusion visit. A total of 31.6% (n=655) patients received iron supplementation, of whom 58.9% (n=386) received intravenous (IV) iron, 40.5% (n=265) had oral iron and 0.6% (n=4) were prescribed both oral and IV iron. An iron status assessment was more common in patients who were given iron supplementation, while over a third of patients who did not have an iron status were prescribed iron (Table). At follow-up, over 70% of patients had a maximum Hb level above 11 g/dL, regardless of iron status (Table). For patients with and without added iron, the mean change in Hb level was 2.26 g/dL and 2.22 g/dL at maximum during the study and 1.71±1.52 g/dL and 1.59±1.60 g/dL at final visit, respectively. Iron status results were used to define patients as having absolute iron deficiency (coefficient of saturation of transferrin [CST] <20% and ferritin <100 μg/100 mL), functional iron deficiency (CST <20% and ferritin ≥100 µg/100 mL) or no deficiency (CST ≥20%). The majority of patients with no iron supplementation had no deficiency compared with a minority of patients with iron supplementation (Table). Patients with absolute iron deficiency given iron supplementation responded better to epoetin alfa biosimilar (increase of ≥1 g/dL since enrollment or increase of ≥2 g/dL, in the absence of transfusion in the 3 previous weeks) than those not given iron supplementation (74.5% vs 65.5%, p=0.403). Similar results were observed for patients with added IV iron. Table 1.Iron supplementationNo iron supplementationIron status assessed; n (%) No Yesn=655 259 (39.5) 396 (60.5)n=1421 743 (52.3) 678 (47.7)Hb levels (g/dL) at inclusion; mean ± SDn=655 9.57±0.7n=1421 9.62±0.7Maximum Hb value (g/dL) reached during study in patients who completed the study; n (%)n=593n=1280≤9 g/dL13 (2.2)16 (1.3)9-11 g/dL157 (26.5)340 (26.5)≥11 g/dL423 (71.4)924 (72.2)Maximum change in Hb value; mean g/dL ± SD, in patients who completed studyn=593 2.22±1.4n=1280 2.26±1.4Association of epoetin alfa biosimilar use* and iron deficiency; n (%)Absolute iron deficiency49 (62.0)30 (38.0)Functional iron deficiency182 (56.5)140 (43.5)No iron deficiency88 (18.6)384 (81.4)Responder to epoetin alfa biosimilar*; n (%)Absolute iron deficiency35 (74.5; p=0.403)19 (65.5)Functional iron deficiency109 (68.1; p=0.958)78 (67.8)No iron deficiency56 (69.1; p=0.489)251 (73.0)*During the first or second chemotherapy cycle after inclusionHb, hemoglobin; SD, standard deviation Conclusions Overall, these results provide real-life evidence that epoetin alfa biosimilar was effective in treating anemia. Iron supplementation improved the response to epoetin alfa biosimilar in patients with an absolute iron deficiency, suggesting that iron status can be used to optimize treatment of patients with CIA with ESAs and iron supplementation. Disclosures Scotté: Hospira SAS: Research Funding. Laribi:Hospira SAS: Research Funding. Gisselbrecht:Bertram Glass: Research Funding; Chugai Pharmaceutical: Research Funding; Baxter: Research Funding; Roche: Consultancy, Research Funding; Hospira SAS: Research Funding. Spaeth:Hospira SAS: Research Funding. Kasdaghli:Hospira: Employment. Albrand:Hospira: Employment. Leutenegger:GECEM: Employment; Hospira SAS: Research Funding. Ray-Coquard:PharmaMar: Consultancy, Other: Paid instructor; Roche: Consultancy, Other: Paid instructor; Amgen: Consultancy, Other: Paid instructor; Hospira SAS: Research Funding.


2016 ◽  
Vol 45 (1) ◽  
pp. 49-59 ◽  
Author(s):  
Julia Cordelia Hempel ◽  
Felix Poppelaars ◽  
Mariana Gaya da Costa ◽  
Casper F.M. Franssen ◽  
Thomas P.G. de Vlaam ◽  
...  

Background: Intravenous (IV) iron preparations are widely used in the treatment of anemia in patients undergoing hemodialysis (HD). All IV iron preparations carry a risk of causing hypersensitivity reactions. However, the pathophysiological mechanism is poorly understood. We hypothesize that a relevant number of these reactions are mediated by complement activation, resulting in a pseudo-anaphylactic clinical picture known as complement activation-related pseudo allergy (CARPA). Methods: First, the in-vitro complement-activating capacity was determined for 5 commonly used IV iron preparations using functional complement assays for the 3 pathways. Additionally, the preparations were tested in an ex-vivo model using the whole blood of healthy volunteers and HD patients. Lastly, in-vivo complement activation was tested for one preparation in HD patients. Results: In the in-vitro assays, iron dextran, and ferric carboxymaltose caused complement activation, which was only possible under alternative pathway conditions. Iron sucrose may interact with complement proteins, but did not activate complement in-vitro. In the ex-vivo assay, iron dextran significantly induced complement activation in the blood of healthy volunteers and HD patients. Furthermore, in the ex-vivo assay, ferric carboxymaltose and iron sucrose only caused significant complement activation in the blood of HD patients. No in-vitro or ex-vivo complement activation was found for ferumoxytol and iron isomaltoside. IV iron therapy with ferric carboxymaltose in HD patients did not lead to significant in-vivo complement activation. Conclusion: This study provides evidence that iron dextran and ferric carboxymaltose have complement-activating capacities in-vitro, and hypersensitivity reactions to these drugs could be CARPA-mediated.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 14-15
Author(s):  
Melissa T. Maltez ◽  
Johnathan P. Mack

Background: Clinical trials studying the efficacy of intravenous (IV) iron replacement have used a variety of dosing schedules, even for the same iron formulations and there isn't a clear standard for how IV iron replacement should be scheduled. Differences in iron absorption efficiency have been observed with different oral iron supplementation schedules but it is not known if the same is true for IV iron. The objective of this study was to evaluate the association between iron administration schedule and hemoglobin response in patients treated at a tertiary care center. Methods: Data was collected retrospectively using The Ottawa Hospital Data Warehouse (OHDW) capturing all iron infusions given at 3 local institutions between January 2007 and December 2018. Patients that received at least 2 intravenous iron infusions within 180 days of each other were included. A 'course' of iron replacement was defined as consecutive infusions with ≤180 days between doses. Patients transfused red blood cells within 90 days of the last iron infusion in each course were excluded. Patient age, sex, dose and formulation of administered, date of each iron infusion, and laboratory parameters from the period starting 3 months before the first infusion to 6 months after the last infusion were extracted for analysis. Patients were categorized into 4 groups based on the mean time between iron infusions in each course: 1-10 days, 11-20 days, 21-30 days, and &gt;30 days. Achieving a maximum hemoglobin (Hb) 10 g/L or higher than the pre-infusion Hb was defined as a 'good' response. A logistic regression model was used to evaluate the association between interval between infusions and achieving a 'good' Hb response. The model was adjusted for sex, age, presence of chronic kidney disease (CKD), dose of iron per course, number of infusions per course, and number of courses. CKD was defined as having a serum creatinine &gt;177 µmol/L. Iron sucrose, iron gluconate, and iron dextran were available for administration during the study period. Results: A total of 4350 patients were included in the analysis. These patients received a total of 6409 courses of iron replacement, with a median of 2 courses (interquartile range [IQR] 1-3) per patient, and 4 infusions (IQR 3-6) per replacement course. Infusions were given a median of 21.9 days (IQR 12-34) apart in each course, with a range of 1-179 days. Iron sucrose was given in most courses (81.6%), followed by iron gluconate (18.2%) and iron dextran (0.1%). Patient characteristics are summarized in Table 1 and laboratory values prior to the first infusion of each course are summarized in Table 2. The interval between iron infusions was associated with Hb response. Results of the logistic regression are summarized Table 3. Compared with patients receiving infusions every 10-20 days, patients receiving infusions more frequently or less frequently were less likely to achieve a good Hb response. Male sex was associated with increased odds of response, while increasing age, having CKD, and receiving more courses of iron were associated with decreased odds of response. Conclusions: In this single-center retrospective cohort analysis, an association between interval of iron infusions and hematologic response was observed. Patients given iron every 10-20 days more likely to achieve a good response compared with more, or less frequent dosing intervals. Strengths of this study include the large sample size, adjustment for sex, age, presence of CKD, and number of iron infusions and courses given. Transfused patients were excluded, so the hemoglobin response is attributable to iron replacement. The study has several important limitations. The clinical decision-making for the infusion schedule was not known and factors that went into this decision are a possible source of confounding (for example, ongoing bleeding). The suspected cause of iron deficiency was not known. Additionally, concomitant oral iron supplementation and iron infusions or transfusions occurring outside the study institution are unknowns and may influence to outcome. The hypothesis-generating findings suggest that the schedule of iron administration may play an important role in hematologic response to iron infusions. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1552-1552 ◽  
Author(s):  
Christian Lerchenmueller ◽  
Faress Husseini ◽  
Bernd Gaede ◽  
Tony Mossman ◽  
Tamas Suto ◽  
...  

Abstract Background and Aim: The role of IV iron supplementation during treatment with erythropoiesis-stimulating agents (ESAs) in patients with CIA is of increasing interest as a possible means of improving response. This randomized, open-label, multicenter study was designed to evaluate the safety and efficacy of IV iron vs standard practice in CIA patients receiving darbepoetin alfa. Interim efficacy analyses showed a higher response rate for darbepoetin alfa with IV iron compared to darbepoetin alfa with standard iron practice with no difference in the safety profile between the treatment arms (Vanderbroek et al, EHA 2006). Iron parameters are reported here. Methods: Eligible patients were diagnosed with a non-myeloid malignancy and had CIA with a baseline hemoglobin (Hb) value < 11g/dL. All patients received darbepoetin alfa 500 mcg administered Q3W with the SureClick™ prefilled autoinjector. Patients were randomized 1:1 to IV iron 200 mg (single dose Q3W at the same time as darbepoetin alfa or in 2 doses of 100 mg within 3 weeks) or standard practice (oral iron or no iron). Randomization was stratified by tumor type and baseline Hb category (< 10 or ≥10 g/dL). Results: A total of 400 patients were randomized. Mean (SD) age of the study population was 61.4 (11.5) years; range, 20–86. Sixty percent (n=241) of participants were women; 28% (n=114) had lung or gynecological tumors; and 52% (n=208) had a baseline Hb value ≥10 g/dL. In the interim analysis population (n=196), the mean (SD) weekly dose of IV iron was 64.8 (6.6) mg in the IV iron group (n=100). In the standard practice group, 28 of 96 patients (29%) received oral iron and 2 (2%) received IV iron (these patients were analyzed as randomized). Mean (standard error) serum ferritin concentrations and percent transferrin saturation (TSAT) in the 2 groups from baseline (BL) to end of study (EOS) are shown in the figure. Conclusions: The combination of darbepoetin alfa Q3W and IV iron appeared to be associated with a trend toward increased mean serum ferritin levels compared to the standard practice control arm. In contrast, mean TSAT surprisingly appeared to be similar in the 2 groups for most of the study period, perhaps suggesting that TSAT is influenced by other factors. Iron management appears to be an important factor in the response to ESAs and the findings presented here suggest the need for additional exploration into iron uptake and demand in cancer patients treated with darbepoetin alfa. Serum Ferritin Concentration Serum Ferritin Concentration Transferrin Saturation (%) Transferrin Saturation (%)


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3184-3184
Author(s):  
Ellen S. Plummer ◽  
Shelley E. Crary ◽  
George R. Buchanan

Abstract 3184 Background: While iron deficiency anemia (IDA) is among the most common hematologic disorders during childhood, management strategies for patients poorly responsive to oral iron therapy have not been well studied. Children treated for IDA often have a poor response to oral iron due to noncompliance, intolerance of side effects, malabsorption, ongoing blood loss, or a combination of these factors. Alternative treatment approaches are therefore needed. Intravenous (IV) iron, including low molecular weight iron dextran (LMWID), offers the possibility of correcting the anemia and repleting iron stores using a single dose, potentially decreasing the overall burden of treatment. Use of LMWID in children has been limited due to concerns about anaphylaxis associated with high molecular weight iron dextran preparations, even though in adults the risk of anaphylaxis is decreased when alternative IV iron preparations, including LMWID, are employed. In this study we report our initial experience with LMWID in children with IDA. Methods: We performed a retrospective record review of patients receiving IV LMWID for IDA in the Center for Cancer and Blood Disorders at Children's Medical Center Dallas between December 1, 2010 and July 31, 2011. Records were reviewed for age, indication for LMWID, concurrent medical problems, use of premedication, initial and follow-up hemoglobin values, adverse events (AEs), and prior or subsequent receipt of other IV iron preparations. The primary study aim was to characterize the clinical course of patients receiving LMWID to inform a planned prospective cohort study of IV iron in children with IDA poorly responsive to oral iron therapy. Results: A total of 18 patients, age 11 months (mos) to 18 years (yrs), received IV LMWID during the study period. 11 of them (median age 13 yrs) received LMWID for IDA secondary to external blood loss due to menorrhagia (n=3), gastrointestinal disease (n=3), hemophilia (n=2), Von Willebrand disease (n=2), and immune thrombocytopenia (n=1). Five (median age 2 yrs) had IDA due to nutritional deficiency, and two patients had multiple causes for their IDA. 14 patients (77.8%) received their initial LMWID infusion without AEs, and all demonstrated an increase in hemoglobin (mean 3 g/dL) 4 to 7 weeks following infusion. Premedication with diphenhydramine, acetaminophen, hydrocortisone, or a combination of these was given to 6 of the 14 patients (42.8%) at the discretion of the treating physician based on history of atopy. The average dose of LMWID was 600 mg (20.2mg/kg) with a range of 150 mg to 1 gram (6.9 mg/kg to 30.9 mg/kg). 3 of these 14 patients (21.4%) required a subsequent infusion to achieve and maintain a normal hemoglobin due to ongoing blood loss. 6 patients (33.3%) had transient AEs during LMWID infusion including hives (n=3), tachycardia (n=2), chest tightness (n=1), fever (n=2), nausea (n=1), vomiting (n=1), sweating (n=2), and cough (n=1). 2 of them were able to complete the infusion without further sequelae after receiving diphenhydramine or hydrocortisone. Only one of the patients with AEs had received premedication, although on review 3 of the 6 patients with AEs had a concurrent medical problem affecting immune function including asthma and orthotopic liver transplant. No patient required hospital admission or treatment of the AE beyond the day of their clinic visit. 4 of the 6 patients with AEs related to LMWID subsequently received IV iron sucrose infusions without any complications. Conclusions: Among 18 children with IDA receiving LMWID planned as a single dose infusion, treatment was well tolerated and effective in 14 of them and associated with only transient AEs in 6. The latter patients were able to either receive the remainder of the LMWID infusion or an alternative iron preparation without complication. Some patients with ongoing blood loss needed additional infusions, although the majority of children were treated effectively with a single dose. These encouraging results support the need for further study of LMWID in children with IDA unresponsive to oral iron therapy or even as an initial treatment alternative to the oral route. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4552-4552
Author(s):  
Jacquelyn M. Powers ◽  
Mark P. Shamoun ◽  
Timothy L. McCavit ◽  
Leah Adix ◽  
George R. Buchanan

Abstract Background The standard first line therapy for iron deficiency anemia (IDA) is oral iron. Yet, many patients fail to respond to oral iron due to poor adherence and/or adverse effects. Intravenous (IV) iron is an effective means of treating IDA in patients with malabsorption of iron or who are non-adherent and/or experience adverse effects with oral iron. Some IV iron preparations carry an FDA-mandated black box warning and/or require a test dose or prolonged infusion. Ferric carboxymaltose (FCM, Injectafer®) is a relatively new IV iron preparation with demonstrated safety and efficacy in adults with IDA. The manufacturer recommended dosing is 15 mg/kg/dose (maximum 750 mg) x2 doses administered at least 7 days apart, and each individual infusion can be administered over 10 to 15 minutes, without the need for a test dose. Limited data exist on its use in children. Our objective was to assess the hematologic response and adverse effects of IV FCM in a diverse population of infants, children and adolescents with IDA who failed oral iron therapy. Method All children with IDA who received FCM at Children's Health from June 1, 2014 through June 10, 2015 were included. Subjects were identified via search of pharmacy records. All patients received at least one dose of FCM 15 mg/kg (maximum 750 mg) administered as a 15-minute IV infusion (without test dose or pre-medications). Patient characteristics, adverse effects and hematologic response were retrospectively collected from the electronic medical record. Results During the study frame, one hundred twenty-five infusions of FCM were administered to 87 patients (71% female) with a median age of 14 years (range 9 months to 20.8 years). The most common racial/ethnic group was Caucasian/White (Latino) at 45% followed by African American/Black and Caucasian/White (Non-Latino), each at 22%. The primary etiologies were heavy menstrual bleeding (38%), nutritional (24%), and GI bleeding and/or malabsorption (20%) with the remaining 18% representing other/mixed causes of IDA (e.g., inflammatory). The median dose administered during a single infusion was 750 mg (range 132 to 750 mg). No adverse effects were noted during or following the infusion in 77 subjects. Two patients had transient tingling, nausea and/or mild abdominal pain. Five others developed generalized pruritis and/or urticaria and received diphenhydramine and/or hydrocortisone, with prompt resolution. Two adolescents had more clinically significant reactions, 1 with nausea/vomiting post-infusion (likely psychogenic) requiring admission, and 1 with dyspnea 2 minutes into the infusion, requiring its immediate termination and administration of diphenhydramine, hydrocortisone and normal saline with prompt symptom resolution. One patient experienced asymptomatic extravasation during the second infusion which resulted in localized iron-staining of the skin. Median pre-infusion hemoglobin concentration for all patients was 9.1 g/dL (range 3.9 to 13.3 g/dL) (Table). A follow-up measurement was available for 76 patients at a median time of 6 weeks (range 1 to 30 weeks) post-initial infusion with a median hemoglobin increase of 3.3 g/dL (range -1.5 to 9.5 g/dL). Conclusion Intravenous FCM, administered in an outpatient infusion setting as one or two short IV infusions and without need for a test dose, was safe and effective in most children and adolescents with IDA refractory to oral iron therapy. Further clinical data are necessary to more fully characterize the extent of adverse effects in young patients. Prospective studies of IV FCM in children are indicated to assess clinical efficacy, including outcomes such as health related quality of life and fatigue. Table. *Hematologic Response to FCM Pre-Infusion **Post-Infusion Hemoglobin concentration (g/dL) All Etiologies, Pre (n=87), Post (n=76) Heavy menstrual bleeding, Pre (n=33), Post (n=26) Nutritional, Pre (n=21), Post (n=20) - 9.1 (3.9 to 13.3) 9.3 (4.2 to 13.3) 8.8 (4.9 to 12.2) - 12.2 (7.1 to 16) 12.7 (8.8 to 16) 12.2 (10.5 to 13.7) Mean corpuscular volume (fl), Pre (n=87), Post (n=76) 71.6 (49.5 to 97.4) 80.9 (53.3 to 102) Serum ferritin (ng/mL), Pre (n=80), Post (n=60) 5.2 (0.6 to 288.6) 115.7 (2.3 to 679.3) *Median laboratory values are reported. **Follow-up laboratory testing occurred at median time of 6 weeks (range 1 to 30 weeks) post-infusion. Disclosures Powers: Gensavis Pharmaceuticals, LLC: Research Funding. McCavit:Pfizer: Research Funding; Gensavis LLC: Research Funding; Novartis: Speakers Bureau. Adix:Gensavis Pharmaceuticals, LLC: Research Funding. Buchanan:Gensavis Pharmaceuticals, LLC: Research Funding.


2007 ◽  
Vol 27 (2_suppl) ◽  
pp. 255-260
Author(s):  
David W. Johnson

Iron supplementation is required in a preponderance of peritoneal dialysis (PD) patients treated with erythropoietic stimulatory agents (ESAs). Although many authors and clinical practice guidelines recommend primary oral iron supplementation in ESA-treated PD patients, numerous studies have clearly demonstrated that, because of a combination of poor bioavailability of oral iron, gastrointestinal intolerance, and noncompliance, oral iron supplementation is insufficient for maintaining a positive iron balance in these patients over time. Controlled trials have demonstrated that, in iron-deficient and iron-replete PD patients alike, intravenous (IV) iron supplementation results in superior iron stores and hemoglobin levels with fewer side effects than oral iron produces. Careful monitoring of iron stores in patients receiving IV iron supplementation is important in view of conflicting epidemiologic links between IV iron loading and infection and cardiovascular disease. Emerging new iron therapies such as heme iron polypeptide and ferumoxytol may further enhance the tolerability, efficacy, and ease of administration of iron in PD patients.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2334-2334 ◽  
Author(s):  
Michael Auerbach ◽  
Lars L Lykke

Abstract Introduction: Iron deficiency anemia (IDA) is prevalent and associated with reduced quality of life (QoL) and worsened disease outcomes. Oral iron administration remains the front line standard but its use is associated with an unacceptably high incidence of gastrointestinal side effects and low adherence. Further, in many conditions associated with blood loss, iron losses may exceed the capacity for oral iron absorption. Use of intravenous (IV) iron, especially if given in a single dose, may result in better compliance, fewer visits to the medical practitioner, and overall improvement in QoL. To date, no product has been licensed in the USA for single visit complete replacement dosing. To address this issue, this trial compared the safety and efficacy of iron isomaltoside 1000 with the most commonly prescribed IV formulation, iron sucrose, in patients intolerant of, or unresponsive to oral iron or likely to receive a blood transfusion. Methods: This was a randomized, open-label, comparative, multi-center trial conducted in the USA. Patients with IDA were randomized 2:1 to either iron isomaltoside administered as a single dose of 1000 mg infused over 20 min at baseline or iron sucrose administered as 200 mg IV injections according to label and repeated up to 5 times. The primary endpoints were adjudicated serious or severe hypersensitivity events starting on or after the first dose of treatment (if the upper bound of the 95 % CI was <3 %, the safety objective was met) and change in hemoglobin (Hb) from baseline to week 8. Results: A total of 1512 patients were enrolled of whom 26 % had gastrointestinal and 50 % gynecologic blood loss. The mean (standard deviation [SD]) age was 44 (15) years. The mean (SD) cumulative dose of iron was 975 (145) mg and 905 (217) mg in the iron isomaltoside and iron sucrose group, respectively. All required 1 visit for iron correction with iron isomaltoside and 4 to 5 visits for iron sucrose. The frequency of subjects with serious or severe hypersensitivity reactions was 0.3% in the iron isomaltoside group versus 0.4 % in the iron sucrose group (95% CI: 0.88 for iron isomaltoside and 1.45 for iron sucrose), meeting the primary safety endpoint. The treatment groups had statistically similar adverse drug reactions (ADRs), 12.5 % in the iron isomaltoside group and 12.8 % in the iron sucrose group. Only 0.2 % of patients in iron isomaltoside group and 0.4 % in iron sucrose group experienced serious ADRs. There were no related fatalities. The frequency of composite cardiovascular safety endpoint was 0.8 % in the iron isomaltoside group and 1.2 % in the iron sucrose group (p = 0.57). The frequency of hypophosphatemia (s-phosphate <2 mg/dL) was low and similar in the 2 groups (3.9 % in the iron isomaltoside and 2.3 % in the iron sucrose group). No patients had s-phosphate <1 mg/dL. The primary efficacy endpoint of non-inferiority in Hb change from baseline to week 8 was met. Iron isomaltoside lead to a significantly more rapid and increased Hb response in the first 2 weeks. This was reflected in both Hb change from baseline and proportion of responders with Hb increases ≥2 g/dL. Hb increased with least square means of 0.70 and 0.44 g/dL at week 1 (p <0.0001) and 1.48 and 1.19 g/dL at week 2 (p <0.0001) for iron isomaltoside and iron sucrose respectively, and the proportion of responders were 5.3 and 2.5 % at week 1 (p = 0.0077) and 32.6 and 20.8 % at week 2 (p <0.0001) for iron isomaltoside and iron sucrose respectively. Larger improvement in Functional Assessment of Chronic Illness Therapy (FACIT) fatigue scores were observed with iron isomaltoside at week 1 (p = 0.04). A faster and greater response with iron isomaltoside was also observed for s-ferritin and transferrin saturation. Conclusion: Iron isomaltoside 1000 administered as 1000 mg in a single visit resulted in a faster and more pronounced hematological response and improvement in fatigue compared to iron sucrose which requires multiple visits. The safety profile was similar with a low frequency of hypersensitivity reactions, cardiovascular events, and serious ADRs. The frequency of hypophosphatemia was low in both treatment groups and no patients had s-phosphate <1 mg/dL. Disclosures Auerbach: AMAG Pharmaceuticals: Research Funding; Pharmacosmos A/S: Research Funding. Lykke:Pharmacosmos A/S: Employment.


Sign in / Sign up

Export Citation Format

Share Document