scholarly journals Micronized, Microencapsulated Ferric Iron Supplementation in the Form of >Your< Iron Syrup Improves Hemoglobin and Ferritin Levels in Iron-Deficient Children: Double-Blind, Randomized Clinical Study of Efficacy and Safety

Nutrients ◽  
2021 ◽  
Vol 13 (4) ◽  
pp. 1087
Author(s):  
Aida Zečkanović ◽  
Marko Kavčič ◽  
Tomaž Prelog ◽  
Alenka Šmid ◽  
Janez Jazbec

A major problem of oral iron supplementation efficacy in children is its tolerability and compliance. We aimed to determine the safety and efficacy of a novel food supplement >Your< Iron Syrup in the replenishment of iron stores and improvement of hematological parameters in iron-deficient children aged nine months to six years. We randomized 94 healthy children with iron deficiency in a ratio of 3:1 to either receive >Your< Iron Syrup or placebo. A 12-week supplementation with >Your< Iron Syrup resulted in a significant increase in ferritin and hemoglobin levels as compared to placebo (p = 0.04 and p = 0.02). Adverse events were reported with similar frequencies across both study arms. >Your< Iron Syrup represents an effective, well-tolerated, and safe option for the management of nutritional iron deficiency in children.

2017 ◽  
Vol 147 (12) ◽  
pp. 2297-2308 ◽  
Author(s):  
Michael J Wenger ◽  
Laura E Murray-Kolb ◽  
Julie EH Nevins ◽  
Sudha Venkatramanan ◽  
Gregory A Reinhart ◽  
...  

Abstract Background: Iron deficiency and iron deficiency anemia have been shown to have negative effects on aspects of perception, attention, and memory. Objective: The purpose of this investigation was to assess the extent to which increases in dietary iron consumption are related to improvements in behavioral measures of perceptual, attentional, and mnemonic function. Methods: Women were selected from a randomized, double-blind, controlled food-fortification trial involving ad libitum consumption of either a double-fortified salt (DFS) containing 47 mg potassium iodate/kg and 3.3 mg microencapsulated ferrous fumarate/g (1.1 mg elemental Fe/g) or a control iodized salt. Participants' blood iron status (primary outcomes) and cognitive functioning (secondary outcomes) were assessed at baseline and after 10 mo at endline. The study was performed on a tea plantation in the Darjeeling district of India. Participants (n = 126; 66% iron deficient and 49% anemic at baseline) were otherwise healthy women of reproductive age, 18–55 y. Results: Significant improvements were documented for iron status and for perceptual, attentional, and mnemonic function in the DFS group (percentage of variance accounted for: 16.5%) compared with the control group. In addition, the amount of change in perceptual and cognitive performance was significantly (P < 0.05) related to the amount of change in blood iron markers (mean percentage of variance accounted for: 16.0%) and baseline concentrations of blood iron markers (mean percentage of variance accounted for: 25.0%). Overall, there was evidence that the strongest effects of change in iron status were obtained for perceptual and low-level attentional function. Conclusion: DFS produced measurable and significant improvements in the perceptual, attentional, and mnemonic performance of Indian female tea pickers of reproductive age. This trial was registered at clinicaltrials.gov as NCT01032005.


2000 ◽  
pp. 217-223 ◽  
Author(s):  
M Zimmermann ◽  
P Adou ◽  
T Torresani ◽  
C Zeder ◽  
R Hurrell

OBJECTIVE: In developing countries, many children are at high risk for both goiter and iron-deficiency anemia. Because iron deficiency may impair thyroid metabolism, the aim of this study was to determine if iron supplementation improves the response to oral iodine in goitrous, iron-deficient anemic children. DESIGN: A trial of oral iodized oil followed by oral iron supplementation in an area of endemic goiter in the western Ivory Coast. METHODS: Goitrous, iodine-deficient children (aged 6-12 years; n=109) were divided into two groups: Group 1 consisted of goitrous children who were not anemic; Group 2 consisted of goitrous children who were iron-deficient anemic. Both groups were given 200mg oral iodine as iodized oil. Thyroid gland volume using ultrasound, urinary iodine concentration (UI), serum thyroxine (T(4)) and whole blood TSH were measured at baseline, and at 1, 5, 10, 15 and 30 weeks post intervention. Beginning at 30 weeks, the anemic group was given 60mg oral iron as ferrous sulfate four times/week for 12 weeks. At 50 and 65 weeks after oral iodine (8 and 23 weeks after completing iron supplementation), UI, TSH, T(4) and thyroid volume were remeasured. RESULTS: The prevalence of goiter at 30 weeks after oral iodine in Groups 1 and 2 was 12% and 64% respectively. Mean percent change in thyroid volume compared with baseline at 30 weeks in Groups 1 and 2 was -45.1% and -21.8% respectively (P<0.001 between groups). After iron supplementation in Group 2, there was a further decrease in mean thyroid volume from baseline in the anemic children (-34.8% and -38.4% at 50 and 65 weeks) and goiter prevalence fell to 31% and 20% at 50 and 65 weeks. CONCLUSION: Iron supplementation may improve the efficacy of oral iodized oil in goitrous children with iron-deficiency anemia.


2018 ◽  
Vol 11 (3) ◽  
pp. 85 ◽  
Author(s):  
Eka Ginanjar ◽  
Lilik Indrawati ◽  
Iswari Setianingsih ◽  
Djumhana Atmakusumah ◽  
Alida Harahap ◽  
...  

Plasma non-transferrin-bound iron (NTBI) is potentially harmful due to the generation of free radicals that cause tissue damage in vascular and other diseases. Studies in iron-replete and iron-deficient subjects, receiving a single oral test dose of Fe(II)SO4 or NaFe(III)EDTA with water, revealed that FeSO4 was well absorbed when compared with NaFeEDTA, while only the Fe(II) compound showed a remarkable increase of NTBI. As NaFeEDTA is successfully used for food fortification, a double-blind randomized cross-over trial was conducted in 11 healthy women with uncomplicated iron deficiency. All subjects received a placebo, 6.5 mg FeSO4, 65 mg FeSO4, 6.5 mg NaFeEDTA, and 65 mg NaFeEDTA with a traditional Indonesian breakfast in one-week intervals. Blood tests were carried out every 60 min for five hours. NTBI detection was performed using the fluorescein-labeled apotransferrin method. Plasma iron values were highly increased after 65 mg NaFeEDTA, twice as high as after FeSO4. A similar pattern was seen for NTBI. After 6.5 mg of NaFeEDTA and FeSO4, NTBI was hardly detectable. NaFeEDTA was highly effective for the treatment of iron deficiency if given with a meal, inhibiting the formation of nonabsorbable Fe-complexes, while NTBI did not exceed the range of normal values for iron-replete subjects.


BMJ Open ◽  
2020 ◽  
Vol 10 (8) ◽  
pp. e037232
Author(s):  
Jordie AJ Fischer ◽  
Lulu X Pei ◽  
David M Goldfarb ◽  
Arianne Albert ◽  
Rajavel Elango ◽  
...  

IntroductionThe WHO recommends daily oral iron supplementation for 12 weeks in women and adolescents where anaemia prevalence is greater than 40%. However, if iron deficiency is not a major cause of anaemia, then, at best, untargeted iron supplementation is a waste of resources; at worst, it could cause harm. Further, different forms of iron with varying bioavailability may present greater risks of harm.Methods and analysisA 12-week three-arm, double-blind, randomised controlled supplementation trial was conducted in Cambodia to determine if there is potential harm associated with untargeted iron supplementation. We will recruit and randomise 480 non-pregnant women (ages 18–45 years) to receive one of three interventions: 60 mg elemental iron as ferrous sulfate (the standard, commonly used form), 18 mg ferrous bisglycinate (a highly bioavailable iron amino acid chelate) or placebo. We will measure ferritin concentrations (to evaluate non-inferiority between the two forms of iron), as well as markers of potential harm in blood and stool (faecal calprotectin, gut pathogen abundance and DNA damage) at baseline and 12 weeks. Mixed-effects generalised linear models will be used to assess the effect of iron on ferritin concentration and markers of potential harm at 12 weeks.Ethics and disseminationEthical approval was obtained from the University of British Columbia Clinical Research Ethics Board (H18-02610), the Children's and Women's Health Centre of British Columbia Research Ethics Board (H18-02610) and the National Ethics Committee for Health Research in Cambodia (273-NECHR). Findings will be published in peer-reviewed journals, presented to stakeholders and policymakers globally and shared within participants’ communities.Trial registration numberClinicalTrials.gov Registry (NCT04017598).


2020 ◽  
Vol 16 (2) ◽  
pp. 220-227 ◽  
Author(s):  
Siti Helmyati ◽  
Endang Sutriswati Rahayu ◽  
Bernadette Josephine Istiti Kandarina ◽  
Mohammad Juffrie

Background: Iron deficiency may inhibit the height increase and weight gain of children. On the other hand, the supplementation of iron causes gut microbiota imbalance which leads to inflammation and diarrhea. The addition of synbiotic fermented milk is expected to have beneficial effects on iron supplementation. This study aimed to determine the effects of iron supplementation only and its administration with synbiotic fermented milk on iron status, body height and weight, and gut microbiota profile of iron deficient elementary school children. Methods: This research was an experimental study with pre and post test conducted on 59 irondeficient children. Subjects were given iron supplementation in syrups (IS group) or given iron supplementation in syrup with fermented milk (containing synbiotic Lactobacillus plantarum Dad 13 and fructo-oligosaccharide) (ISFM group) for 3 months. The body weight and height, hemoglobin and serum ferritin levels, and total number of Lactobacilli, Enterobacteria, Bifidobacteria, and Escherichia coli were measured at the beginning and the end of the study. Results: The body height in the ISFM group increased significantly than that in IS group after the intervention (1.67 vs. 2.42, p<0.05). The hemoglobin and serum ferritin levels in IS and ISFM groups were improved significantly (p<0.05) although the difference between the two groups was not significant (p>0.05). The results showed no significant difference of gut microbiota profile between the IS and ISFM groups (p>0.05). Conclusion: There is no difference on the iron status, height, weight, and gut microbiota profile of iron-deficient primary school children received iron supplementation only or iron supplementation with synbiotic fermented milk.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2052-2052
Author(s):  
Eldad A. Hod ◽  
Eric H. Ekland ◽  
Shruti Sharma ◽  
Boguslaw S. Wojczyk ◽  
David A. Fidock ◽  
...  

Abstract Abstract 2052 To clarify the interactions between iron status, oral iron supplementation, and bacterial and malarial infections, we examined iron-replete mice and mice with dietary iron deficiency infected with Salmonella typhimurium, Plasmodium yoelii, or both, with and without oral iron administration. These studies were designed to identify potential mechanisms underlying the increased risk of severe illness and death in children in a malaria-endemic region who received routine iron and folic acid supplementation during a randomized, controlled trial in Pemba, Tanzania (Sazawal et al. Lancet 2006;367:133-43). To this end, weanling C57BL/6 female mice were fed an iron-replete or an iron-deficient diet, the latter of which resulted in severe iron deficiency anemia. Groups of mice were then infected by intraperitoneal injection of Salmonella typhimurium strain LT2, Plasmodium yoelii strain 17X parasites, or both. With Salmonella infection alone, iron-deficient mice had a median survival (7.5 days, N=8) approximately half that of iron-replete mice (13 days, N=10, p<0.0001). At death, the mean level of bacteremia was significantly higher in infected iron-deficient mice. In blood cultures performed at death, all iron-deficient mice were bacteremic, but bacteria were detected in only 4 of 10 iron-replete mice. Both iron-deficient and iron-replete Salmonella-infected mice had gross hepatosplenomegaly with hepatitis, distorted hepatic and splenic architecture, massive expansion of the splenic red pulp with inflammatory cells, and Gram-negative bacilli by tissue Gram stain. With P. yoelii infection alone, iron-deficient and iron-replete mice cleared the infection at similar rates (by ~13 days following infection, N=5 in each group) and no deaths due to parasitemia occurred. With Salmonella and P. yoelii co-infection, death was earlier than with Salmonella alone in iron-replete mice (median survival of 10 vs. 13 days; N=10 in each group; p=0.005), but not in iron-deficient mice (median survival of 7 vs. 7.5 days; N=10 and 8, respectively; p=0.8). To examine the effect of short-term oral iron supplementation with Salmonella infection alone, mice received daily iron (ferrous sulfate, 1 mg/kg) by gavage for 4 days before infection with Salmonella, and supplementation continued for a total of 10 days. After gavage, plasma non-transferrin-bound iron (NTBI) appeared at 1–2 hours with a mean peak level of approximately 5 μM. In iron-deficient mice, short-term oral iron supplementation did not fully correct the iron deficiency anemia or replenish iron stores. Oral iron supplementation reduced the median survival of both iron-deficient and iron-replete Salmonella-infected mice by approximately 1 day; the difference was significant only in the iron-replete group (N=5, p<0.05). In summary, these results indicate that iron deficiency decreases the survival of Salmonella-infected mice; the median survival of iron-deficient mice was approximately half that of those that were iron replete. These observations are similar to those in the Pemba sub-study in which iron-deficient children given placebo had a 200% increase in the risk of adverse events relative to iron-replete children. Iron deficiency had no apparent effect on the course of infection with P. yoelii but further studies with more virulent Plasmodium species are needed. Co-infection with Salmonella and Plasmodium significantly increased mortality as compared to single infections, but only in iron-replete mice. Oral iron supplementation of Salmonella-infected mice significantly decreased the median survival, but only of iron-replete animals; however, our study may have had insufficient power to detect an effect on iron-deficient mice. Systematic examination in mice of the effect of iron supplements on the severity of malarial and bacterial infection in iron-replete and iron-deficient states may ultimately help guide the safe and effective use of iron interventions in humans in areas with endemic malaria. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1998 ◽  
Vol 92 (8) ◽  
pp. 2934-2939 ◽  
Author(s):  
Pauli Suominen ◽  
Kari Punnonen ◽  
Allan Rajamäki ◽  
Kerttu Irjala

Despite the established utility of serum transferrin receptor (sTfR), serum ferritin, and the sTfR/log ferritin ratio (TfR-F Index) in the diagnosis of iron deficiency (ID) anemia, the numeric values of these parameters, which are indicative of subclinical ID, remain to be clearly defined. In this study, 65 apparently healthy nonanemic adults (22 men and 43 women) were treated with 3 months of oral iron supplementation to evaluate its effect on parameters reflecting iron status and to determine the prevalence of subclinical iron deficiency in apparently healthy adults. Significant supplementation-induced changes were observed in sTfR, ferritin, and TfR-F Index values in women, whereas in men, none of the studied parameters showed any significant change. Iron-deficient erythropoiesis (IDE) was not observed in men, but was found in 17 women (40%). Although individuals with a compromised iron status may be represented in substantial numbers in conventional reference populations, they can be readily identified using sTfR, ferritin, and TfR-F Index determinations. © 1998 by The American Society of Hematology.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 35-35
Author(s):  
Brian Sandri ◽  
Gabriele Lubach ◽  
Eric Lock ◽  
Pam Kling ◽  
Michael Georgieff ◽  
...  

Background: Pre-anemic iron deficiency (ID) and ID anemia in infancy is associated with long-term effects on brain development. Iron is an essential micronutrient needed for multiple neurodevelopmental processes, including energy production, myelination and neurotransmission. Serum and cerebrospinal fluid (CSF) metabolomic and proteomic analyses can potentially characterize the metabolic and protein signatures of aberrant iron-dependent brain development pathways. A better understanding of the timing and course of the biochemical signatures of ID-induced dysfunction in the heme and intrathecal compartments may help with early diagnosis and treatment of iron deficiency prior to onset of anemia. Objective: To determine proteomic and metabolomic signatures of ID-induced dysfunction in serum and CSF of infant monkeys prior to the onset of iron deficiency anemia (IDA). Methods: Hematological parameters were determined in rhesus monkey infants (Macaca mulatta) at 4 and 6 months of age as they progressed from preanemic ID to IDA (ID group; n=6) or remained iron-sufficient(IS group; n=12). Blood and CSF were collected at 4 and 6 months and metabolically profiled using HPLC/MS with isobaric standards for identification and quantification. Proteomic quantification utilized a custom TMT-labeled workflow with an Orbitrap Fusion Tribrid mass spectrometer. Serum and CSF metabolomic and proteomic profiles of the ID and IS groups were compared, along with a a third group of infants that became IDA earlier at 4 months of age (IDA group; n=3). Results: Evidence of pre-anemic ID was apparent in some infant at 4 months of age with clinical IDA evident at 6 months A (Hgb &lt;10 g/dL and MCV &lt;60 fL). A total of 1,128 CSF proteins and 251 serum proteins were identified and quantified. Over 4,000 metabolite features were observed in serum and CSF across three modalities including reverse phase positive, reverse phase negative and HILIC mode. Pathway analyses provided evidence of increased inflammatory activity, impacts on neural pathways, and bioindicators of acute phase responses in ID and IDA groups. Signs of these metabolic changes were already present in the preanemic ID? period (4 months). Conclusions: Biomarkers indicating indicative of increased inflammatory activity with downstream effects on pathways involved in normal neural function were detectable in both the CSF and serum of ID monkeys prior to the onset of clinical anemia. Brain-specific proteomic and metabolomic profiling of novel analytes in circulation can facilitate earlier diagnosis and treatment of infantile ID to reduce neurodevelopmental risk. Funded by NIH/NICHD Grant HD089989, this abstract is sponsored by ASH member Zohar Sachs MD, PhD Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3722-3722
Author(s):  
S.G. Berrak ◽  
Meryem Angaji ◽  
Emine Turkkan ◽  
Cengiz Canpolat ◽  
Cetin Timur ◽  
...  

Abstract Background: Microbial killing killing of by phagocytic cells by iron containing myeloperoxidase pathway is part of the immune system. We aimed to investigate the effects of iron deficiency anemia (IDA) on oxidative burst response of phagocytic cells and understand if the effect is reversible after iron supplementation. Material and methods: There were 57 IDA patients and 31 healthy children as the control group that were aged between 6 months-12 years with similar demographic status. IDA patients were classified according to the severity of their anemia as follows; group 1 (Hb<8 gr/dl), group 2 (Hb≥8 gr/dl and Hb≤10 gr/dl), and group 3 (Hb>10 gr/dl). Neutrophil and monocyte oxidative burst response of each three groups of IDA and control group were compared by flow cytometry. IDA group were given oral iron supplementation (3 mg/kg-Ferrosanol) therapy. On day 15 of iron supplementation therapy neutrophil and monocyte oxidative burst response of each three groups of IDA were rechecked and compared to the control group. In order to minimize the discrepancies observed in functional studies, the oxidative burst index was calculated as the proportion of oxidative burst response in IDA group to the simultaneous oxidative burst response in the control group. Results: The number of infection episodes observed in the last three months was significantly higher in the IDA group (0–13 episodes) than the control group (1–2 episodes) (p=0.05). Neutrophil (p=0.1) and monocyte (p=0.02) oxidative burst responses in IDA group were found to be less than the control group. On day 15 of iron supplementation therapy, these values were observed to increase to the control group’s values. Evaluation of neutophil and monocyte burst indexes at the diagnosis of IDA revealed that the neutrophil burst indexes in the Hb ≤10 gr/dl group (p=0.006), and monocyte burst indexes in the Hb <8 gr/dl group (p=0.01) were significantly low. The initially observed significant differences in oxidative burst responses disappeared at day 15 of iron supplementation therapy in all hemoglobin groups. Conclusion: Correction of the differences in oxidative burst response after iron supplementation therapy implies the fact that IDA might be the reason for changes in neutophil and monocyte burst indexes.


Sign in / Sign up

Export Citation Format

Share Document