Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.284
Filter
Add more filters








Publication year range
2.
Sci Rep ; 14(1): 15674, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977742

ABSTRACT

Our study aims to validate safety and efficacy of Feroglobin capsule compared with different iron supplementations in adult subjects diagnosed with non-anemic to mild anemic iron deficiency and fatigue. Enrolled 302 participants diagnosed with non-anemic to mild anemic iron deficiency and fatigue. Group A (n = 147) received Feroglobin, Group B (n = 146) received standard of care [Haem Up Gems capsules (Ferrous fumarate) or Fericip tablets (Ferrous ascorbate)]. 293 subjects completed the study with follow-up visits on days 30, 60, and 90. Feroglobin treatment significantly increased hemoglobin levels from mean 12.43 g/dl to 13.24 g/dl in 90 days. Ferritin levels improved significantly by 442.87% compared to the standard care's 256.67%. Fatigue scale scores reduced by 47.51%, and all presenting health complaints resolved completely. Gastrointestinal symptoms observed were similar in both the groups. Both groups exhibited moderate treatment adherence. Quality of life improved in pain and general health domains, exhibiting a good tolerability. Adverse events were unrelated to the investigational products. Feroglobin serves as an efficacious therapeutic alternative for improving hemoglobin, ferritin, and reducing fatigue with low doses compared to standard of care. However, longer-term effects of low-dose require further investigations in different target groups.


Subject(s)
Anemia, Iron-Deficiency , Dietary Supplements , Ferrous Compounds , Hemoglobins , Humans , Female , Male , Adult , Middle Aged , Hemoglobins/analysis , Anemia, Iron-Deficiency/drug therapy , Ferrous Compounds/administration & dosage , Ferrous Compounds/therapeutic use , Quality of Life , Iron/administration & dosage , Iron/therapeutic use , Ferritins/blood , Fatigue/drug therapy , Fatigue/etiology , Treatment Outcome , Aged
3.
BMJ Open ; 14(6): e084033, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862227

ABSTRACT

OBJECTIVE: This systematic review and meta-analysis aimed to comprehensively assess the impact of weekly iron-folic acid supplementation (WIFAS) on the nutrition, health and educational outcomes of children and adolescents in sub-Saharan Africa. DESIGN: A systematic review and meta-analysis was used. DATA SOURCES: Five databases, namely, MEDLINE, Scopus, Web of Science, Cochrane Library and Google Scholar, were systematically searched for relevant articles up to 23 August 2023. ELIGIBILITY CRITERIA: It was focused on randomised controlled trials involving children and adolescents in sub-Saharan Africa, exploring the effects of iron supplementation on various outcomes, such as serum ferritin and haemoglobin levels, anaemia, mental health and school performance. DATA EXTRACTION AND SYNTHESIS: The Joanna Briggs Institute Critical Appraisal tools were used for quality assessment, with two independent reviewers thoroughly evaluating each paper. Using the Cochrane risk of bias tool, we evaluated the certainty of evidence such as the risk of bias, inconsistency, indirectness, imprecision and publication bias. RESULTS: A systematic review of 10 articles revealed that WIFAS significantly increased serum ferritin levels in adolescent girls (Hedge's g=0.53, 95% CI 0.28 to 0.78; heterogeneity I2=41.21%, p<0.001) and haemoglobin levels in school-aged children (Hedge's g=0.37, 95% CI 0.01 to 0.73; heterogeneity I2=91.62%, p<0.001). The analysis further demonstrated a substantial reduction in the risk of anaemia by 20% (risk ratio=0.8, 95% CI 0.69 to 0.93; heterogeneity I2=28.12%, p<0.001). CONCLUSION: WIFAS proved effective in enhancing serum ferritin and haemoglobin concentrations and lowering the risk of anaemia in school-aged children and adolescents compared with a placebo. Similarly, there are not enough studies to examine the effects of WIFAS on school performance. However, information regarding mental health problems, mortality and potential side effects remains insufficient. PROSPERO REGISTRATION NUMBER: CRD42023397898.


Subject(s)
Dietary Supplements , Folic Acid , Iron , Mental Health , Humans , Child , Adolescent , Africa South of the Sahara , Iron/administration & dosage , Iron/therapeutic use , Folic Acid/administration & dosage , Folic Acid/therapeutic use , Ferritins/blood , Anemia, Iron-Deficiency/prevention & control , Hemoglobins/analysis , Randomized Controlled Trials as Topic , Female , Nutritional Status
4.
J Healthc Qual ; 46(4): 220-227, 2024.
Article in English | MEDLINE | ID: mdl-38833574

ABSTRACT

INTRODUCTION: Studies have demonstrated the efficacy of intravenous (IV) iron when administered to patients with congestive heart failure (CHF) and iron deficiency (ID). We aimed to better understand the adherence of treatment for ID among a population with CHF, with particular interest in high-risk groups not often studied due to inadequate recruitment. METHODS: A retrospective chart review at our institution was conducted from January 1, 2012, to July 7, 2021. Analysis included hospitalized patients with CHF and ID and dividing these patients into two time periods based on changes in iron treatment patterns and treatment between sexes. RESULTS: Four thousand eight hundred thirteen patients were included in this study. During the "early era," 7.0% of patients with CHF and ID received IV iron compared with 20.9% of "late-era" patients. Female patients with ID were statistically less likely to receive IV iron when compared with male patients, both unadjusted (0.66, confidence interval [CI] 0.55-0.79, p < .0001) and adjusted (0.72, CI 0.59-0.87, p < .0001) for covariates. CONCLUSION: This study illustrates improved adherence to treatment for ID among hospitalized population with CHF and ID over time but persistent undertreatment remains. Future studies will need to identify the barriers to treating female patients with CHF and ID to reduce these disparities.


Subject(s)
Heart Failure , Humans , Heart Failure/drug therapy , Female , Male , Retrospective Studies , Aged , Middle Aged , Anemia, Iron-Deficiency/drug therapy , Hospitalization/statistics & numerical data , Aged, 80 and over , Iron/therapeutic use , Iron/administration & dosage , Iron Deficiencies , Sex Factors
5.
JAMA Netw Open ; 7(5): e2414305, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38819821

ABSTRACT

Importance: Optimal oral iron supplementation strategy is unclear in patients with iron deficiency anemia (IDA) who have either normal kidney function (NKF) or chronic kidney disease (CKD). Objective: To investigate the association of different oral iron supplementation strategies with the change in hemoglobin and iron indices among patients with IDA with either NKF or CKD. Design, Setting, and Participants: This retrospective cohort study was conducted between 2009 and 2019 at nationwide Veterans Health Administration facilities. Eligible participants included veterans with IDA (defined as hemoglobin <12 g/dL and either iron saturation <20% or ferritin <50 ng/mL) who received their first outpatient prescription of oral iron. Patients were further divided into those with NKF (estimated glomerular filtration rate >60 mL/min/1.73 m2) and CKD (estimated glomerular filtration rate ≥15 mL/min/1.73 m2 and <60 mL/min/1.73 m2). Data analysis was conducted from February to October 2023. Exposures: Patients were classified into 3 groups based on their oral iron dosing schedule: daily (once a day), multiple doses per day (MDD; ≥2 times per day), or alternate-day dose (ADD). Main Outcomes and Measures: The primary outcomes were change of hemoglobin, ferritin, total iron binding capacity (TIBC), and iron saturation (ISAT), which were calculated with linear mixed-effects models. Results: A total of 71 677 veterans with IDA (63 202 male [88.2%] and 8475 female [11.8%]; mean [SD] age, 68.47 [13.09] years), including 47 201 with NKF and 24 476 with CKD, were identifed. In patients with NKF in the daily group, hemoglobin increased from baseline (estimated per-30-day difference [SE], 0.27 [0.00] g/dL; P < .001). In comparison with the daily group, hemoglobin increased more in the MDD group (estimated per-30-day difference [SE], 0.08 [0.03] g/dL; P < .001), but no difference was noted in the ADD group (estimated per-30-day difference [SE], -0.01 [0.01] g/dL; P = .38). Ferritin, ISAT, and TIBC results were similar, except TIBC showed less change in the ADD group compared with the daily group. Patients with CKD showed similar trends but smaller magnitudes in changes. Among patients with NKF, the adjusted mean increase in hemoglobin was 1.03 g/dL (95% CI, 1.01-1.06 g/dL) for those in the daily group, 1.38 g/dL (95% CI, 1.36-1.40 g/dL) for those in the MDD group, and 0.93 g/dL (95% CI, 0.84-1.02 g/dL) for those in the ADD group at 90 days. Among patients with CKD, the adjusted mean increase in hemoglobin was 0.71 g/dL (95% CI, 0.68-0.73 g/dL) for those in the daily group, 0.99 g/dL (95% CI, 0.97-1.01 g/dL) for those in the MDD group, and 0.62 g/dL (95% CI, 0.52-0.73 g/dL) for those in the ADD group at 90 days. Conclusions and Relevance: In this retrospective cohort study of veterans with IDA, there was no significant difference in the improvement of hemoglobin and iron indices between daily and ADD groups, but quickest improvement was observed in the MDD group. These findings suggest that the choice of oral iron therapy should depend on the rapidity of response desired and patient preference due to adverse effects.


Subject(s)
Anemia, Iron-Deficiency , Veterans , Humans , Anemia, Iron-Deficiency/drug therapy , Male , Female , Retrospective Studies , Veterans/statistics & numerical data , Middle Aged , Administration, Oral , United States/epidemiology , Aged , Iron/administration & dosage , Iron/therapeutic use , Renal Insufficiency, Chronic/complications , Hemoglobins/analysis , Glomerular Filtration Rate
6.
PLoS One ; 19(5): e0302964, 2024.
Article in English | MEDLINE | ID: mdl-38776289

ABSTRACT

BACKGROUND: Iron deficiency anemia (IDA) is a prevalent hematological complication associated with gastrointestinal (GI) cancers due to an increased loss of iron and decreased iron absorption. The purpose of this systematic review is to evaluate the use of parenteral iron to treat IDA in patients with GI cancer. METHODS: PubMed, Cochrane, EMBASE, CINHAL and Scopus were searched from January 1, 2010 to September 29, 2023 with no language restrictions. We excluded editorials, case reports, abstracts, conference papers, and poster presentations. Studies were included if they discussed IDA, GI neoplasms, use of iron supplementation (with or without erythropoietin-stimulating agents [ESAs]), defined anemia and had an adult patient population. We assessed the efficacy of parenteral iron in comparison to other iron supplementation methods when treating IDA in patients with GI cancer. The Cochrane Risk of Bias Tool 2 (RoB 2) and the Risk Of Bias In Non-randomized Studies of Interventions (ROBINS-I) assessment tools were used to assess the quality of the included studies. Moreover, the Cochrane Effective Practice and Organization data collection form was used to collect pertinent study information. RESULTS: Our search yielded 3,969 studies across all databases. Twenty-one studies were included (6 randomized control trials; 15 non-randomized studies). Of the 15 studies evaluating hemoglobin (Hb) response, seven studies found an increase in Hb levels when patients were treated with IV iron. The 14 studies evaluating red blood cell (RBC) transfusion rates found conflicting differences in RBC transfusion needs when treated with IV iron. Studies analyzing health related outcomes typically found an increase in quality of life and decreased post-operative complications. DISCUSSION: This review demonstrates improved outcomes of IDA in patients with GI cancer treated with IV iron instead of other iron supplementation methods. Timely diagnosis and appropriate IDA management can greatly improve quality of life in this patient population, especially if myelosuppressive chemotherapy is required.


Subject(s)
Anemia, Iron-Deficiency , Gastrointestinal Neoplasms , Iron , Humans , Anemia, Iron-Deficiency/drug therapy , Gastrointestinal Neoplasms/complications , Gastrointestinal Neoplasms/drug therapy , Iron/administration & dosage , Iron/therapeutic use , Administration, Intravenous
7.
Medicina (Kaunas) ; 60(5)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38792868

ABSTRACT

Background and Objectives: The administration of iron to premature newborns is a common intervention aimed at preventing iron deficiency (ID). However, there is no consensus on the optimal timing and dosage for iron supplementation in this population. This study evaluates the effects and potential adverse outcomes of administering iron on the 7th and 21st days of life in premature infants. Materials and Methods: This research was conducted on 108 premature neonates at the "Louis Turcanu" Children's Emergency Clinical Hospital in Timisoara, Romania. The study population was divided into a control group of 48 newborns who did not receive iron supplementation and an intervention group of 60 newborns who did. The analysis utilized univariate and multivariate regression to examine binary outcomes. Results: The findings indicate that iron supplementation significantly increased the risk of anemia during the premature period at 21 days of life, as demonstrated by both univariate and multivariate regression analyses, with an odds ratio (OR) of 2.40 (95% CI, 1.01-5.68) and an adjusted odds ratio (AOR) of 2.75 (95% CI, 1.06-7.11), respectively. Contrary to expectations, iron supplementation did not significantly alter the risk of abnormal serum ferritin or iron levels at 21 days of life, according to the univariate analysis (p = 0.380 and p = 0.526, respectively). Conclusions: The observed increase in the risk of anemia without a corresponding improvement in the serum ferritin or iron levels suggests the need for further investigation into alternative strategies for iron supplementation in premature newborns.


Subject(s)
Anemia, Iron-Deficiency , Infant, Premature , Iron , Humans , Infant, Newborn , Prospective Studies , Male , Female , Iron/administration & dosage , Iron/therapeutic use , Romania/epidemiology , Anemia, Iron-Deficiency/drug therapy , Cohort Studies , Dietary Supplements , Ferritins/blood
8.
Clin Med (Lond) ; 24(3): 100211, 2024 May.
Article in English | MEDLINE | ID: mdl-38643833

ABSTRACT

Patients with heart failure (HF) and iron deficiency are at increased risk of adverse clinical outcomes. We searched databases for randomised controlled trials that compared IV iron to placebo, in patients with HF with reduced ejection fraction (HFrEF). A total of 7,813 participants, all having HFrEF with 3,998 receiving IV iron therapy, and 3,815 control recipients were included. There was a significant improvement in Kansas City Cardiomyopathy Questionnaire favouring IV iron with MD 7.39, 95% CI [3.55, 11.22], p = 0.0002. Subgroup analysis, based on acute and chronic HF, has displayed a sustained statistical significance. Additionally, a significant increase in the left ventricular ejection fraction % was observed, with MD 3.76, 95% CI [2.32, 5.21], p < 0.00001. A significant improvement in 6-min walk test was noted, with MD 34.87, 95% CI [20.02, 49.72], p < 0.00001. Furthermore, IV iron showed significant improvement in NYHA class, peak VO2, serum ferritin, and haemoglobin levels. Finally, despite the lack of difference in terms of all-cause hospitalisation and HF-related death, IV iron was associated with a significant reduction in HF-related, any cardiovascular reason hospitalisations, and all-cause death; which supports the need for implementation of IV iron as a standard of care in patients with HF and iron deficiency.


Subject(s)
Heart Failure , Iron , Stroke Volume , Humans , Administration, Intravenous , Anemia, Iron-Deficiency/drug therapy , Heart Failure/drug therapy , Heart Failure/physiopathology , Iron/administration & dosage , Iron/therapeutic use , Iron Deficiencies
10.
J Nutr ; 154(6): 1927-1935, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615735

ABSTRACT

BACKGROUND: Anemia may be associated with poor clinical outcomes among people living with human immunodeficiency virus (HIV) (PLHIV) despite highly active antiretroviral therapy (HAART). There are concerns that iron supplementation may be unsafe to prevent and treat anemia among PLHIV. OBJECTIVE: The objective of the study was to evaluate the associations of anemia and iron supplementation with mortality and viral load among PLHIV in Tanzania. METHODS: We analyzed data from a cohort of 70,442 nonpregnant adult PLHIV in Tanzania conducted between 2015 and 2019. Regression models evaluated the relationships between anemia severity and iron supplement use with mortality and unsuppressed HIV-1 viral load among all participants and stratified by whether participants were initiating or continuing HAART. RESULTS: Anemia was associated with an increased risk of mortality and unsuppressed viral load for participants who initiated or continued HAART. Iron supplement use was associated with reduced mortality risk but also had a greater risk of an unsuppressed viral load among participants continuing HAART. There was no association of iron supplement use with mortality, and unsuppressed viral load among PLHIV that were initiating HAART. There was a stronger negative association between iron supplement use and the risk of having an unsuppressed viral load among participants with stage III/IV disease compared with stage I/II disease. CONCLUSIONS: Anemia is associated with increased risk of mortality and unsuppressed viral load, but the benefits and safety of iron supplements appear to differ for those initiating compared with continuing ART as well as by HIV disease severity.


Subject(s)
Anemia , Dietary Supplements , HIV Infections , Iron , Viral Load , Humans , Tanzania/epidemiology , HIV Infections/drug therapy , HIV Infections/mortality , HIV Infections/complications , Male , Female , Adult , Anemia/mortality , Middle Aged , Iron/blood , Iron/administration & dosage , Iron/therapeutic use , Antiretroviral Therapy, Highly Active , Cohort Studies , Young Adult
11.
Indian Pediatr ; 61(7): 621-626, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38655891

ABSTRACT

OBJECTIVE: This study aimed to assess the efficacy of different oral iron preparations prescribed for prevention of iron deficiency anemia in healthy infants. METHODS: This retrospective study enrolled infants aged between 6 and 12 months who were initiated on iron prophylaxis at four months of age. Enrolled children consistently used specific iron preparations (ferrous, ferric or liposomal iron) and had their complete blood counts and serum ferritin levels assessed within the 6-12 month timeframe. Blood values and iron prophylaxis type (ferrous (Fe+2), ferric (Fe+3), liposomal iron) were recorded. Chi-square test was used to compare the hemoglobin and ferritin levels levels between groups. Univariate and multivariate regression analyses assessed the risk of anemia. RESULTS: The study included 371 children (ferrous sulphate - 60, iron hydroxide-polymaltose complex - 137 and liposomal ferric pyrophosphate - 174) with a mean (SD) age 9.1 (1.3) mo. Iron deficiency in different groups were: liposomal iron (46.0%), ferric iron (44.5%), and ferrous iron (5.0%). Mean (SD) serum ferritin levels (µg/L) were higher in the ferrous group [30.1 (10.8)] compared to infants receiving ferric [17.6 (14.50)] and liposomal iron [15.4 (12.1)] (P < 0.001). Mean (SD) hemoglobin levels (g/dL) were significantly higher in the ferrous group [12.4 (0.8)] compared to ferric [11.9 (1.1)] and liposomal iron group [12.0 (1.1)]; P =0.008. Multiple regression analysis showed that ferrous group was associated with a lower risk of iron deficiency [OR (95% CI) 0.04 (0.01-0.15), P < 0.001]. CONCLUSION: Ferrous iron demonstrated superior efficacy compared to ferric and liposomal iron. Further studies are needed to establish alternative iron preprations in children.


Subject(s)
Anemia, Iron-Deficiency , Ferric Compounds , Ferrous Compounds , Liposomes , Humans , Infant , Anemia, Iron-Deficiency/prevention & control , Anemia, Iron-Deficiency/blood , Retrospective Studies , Male , Female , Ferrous Compounds/administration & dosage , Ferrous Compounds/therapeutic use , Ferric Compounds/administration & dosage , Ferric Compounds/therapeutic use , Ferritins/blood , Iron/administration & dosage , Iron/blood , Iron/therapeutic use , Hemoglobins/analysis , Hemoglobins/drug effects
12.
Sci Rep ; 14(1): 7924, 2024 04 04.
Article in English | MEDLINE | ID: mdl-38575644

ABSTRACT

Neonatal hypoxic-ischemic brain injury (HIBI) results in part from excess reactive oxygen species and iron-dependent lipid peroxidation (i.e. ferroptosis). The vitamin D precursor 7-dehydrocholesterol (7-DHC) may inhibit iron-dependent lipid peroxidation. Primary neurons underwent oxygen and glucose deprivation (OGD) injury and treatment with 7-DHC-elevating medications such as cariprazine (CAR) or vehicle. Postnatal day 9 mice underwent sham surgery or carotid artery ligation and hypoxia and received intraperitoneal CAR. In neurons, CAR administration resulted in significantly increased cell survival compared to vehicle controls, whether administered 48 h prior to or 30 min after OGD, and was associated with increased 7-DHC. In the mouse model, malondialdehyde and infarct area significantly increased after HIBI in the vehicle group, which were attenuated by post-treatment with CAR and were negatively correlated with tissue 7-DHC concentrations. Elevating 7-DHC concentrations with CAR was associated with improved cellular and tissue viability after hypoxic-ischemic injury, suggesting a novel therapeutic avenue.


Subject(s)
Dehydrocholesterols , Ferroptosis , Hypoxia-Ischemia, Brain , Animals , Mice , Animals, Newborn , Brain , Hypoxia/complications , Oxygen/therapeutic use , Ischemia/complications , Iron/therapeutic use
14.
COPD ; 21(1): 2322605, 2024 12.
Article in English | MEDLINE | ID: mdl-38591165

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a chronic respiratory condition characterized by persistent inflammation and oxidative stress, which ultimately leads to progressive restriction of airflow. Extensive research findings have cogently suggested that the dysregulation of essential transition metal ions, notably iron, copper, and zinc, stands as a critical nexus in the perpetuation of inflammatory processes and oxidative damage within the lungs of COPD patients. Unraveling the intricate interplay between metal homeostasis, oxidative stress, and inflammatory signaling is of paramount importance in unraveling the intricacies of COPD pathogenesis. This comprehensive review aims to examine the current literature on the sources, regulation, and mechanisms by which metal dyshomeostasis contributes to COPD progression. We specifically focus on iron, copper, and zinc, given their well-characterized roles in orchestrating cytokine production, immune cell function, antioxidant depletion, and matrix remodeling. Despite the limited number of clinical trials investigating metal modulation in COPD, the advent of emerging methodologies tailored to monitor metal fluxes and gauge responses to chelation and supplementation hold great promise in unlocking the potential of metal-based interventions. We conclude that targeted restoration of metal homeostasis represents a promising frontier for ameliorating pathological processes driving COPD progression.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Humans , Copper/therapeutic use , Lung , Oxidative Stress , Iron/therapeutic use , Zinc/therapeutic use
16.
Circ Heart Fail ; 17(4): e011351, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38572652

ABSTRACT

BACKGROUND: Studies have shown an association between iron deficiency (ID) and clinical outcomes in patients with heart failure (HF), irrespective of the presence of ID anemia (IDA). The current study used population-level data from a large, single-payer health care system in Canada to investigate the epidemiology of ID and IDA in patients with acute HF and those with chronic HF, and the iron supplementation practices in these settings. METHODS: All adult patients with HF in Alberta between 2012 and 2019 were identified and categorized as acute or chronic HF. HF subtypes were determined through echocardiography data, and ID (serum ferritin concentration <100 µg/L, or ferritin concentration between 100 and 300 µg/L along with transferrin saturation <20%), and IDA through laboratory data. Broad eligibility for 3 clinical trials (AFFIRM-AHF [Study to Compare Ferric Carboxymaltose With Placebo in Patients With Acute HF and ID], IRONMAN [Intravenous Iron Treatment in Patients With Heart Failure and Iron Deficiency], and HEART-FID [Randomized Placebocontrolled Trial of Ferric Carboxymaltose as Treatment for HF With ID]) was determined. RESULTS: Among the 17 463 patients with acute HF, 38.5% had iron studies tested within 30 days post-index-HF episode (and 34.2% of the 11 320 patients with chronic HF). Among tested patients, 72.6% of the acute HF and 73.9% of the chronic HF were iron-deficient, and 51.4% and 49.0% had IDA, respectively. Iron therapy was provided to 41.8% and 40.5% of patients with IDA and acute or chronic HF, respectively. Of ID patients without anemia, 19.9% and 21.7% were prescribed iron therapy. The most common type of iron therapy was oral (28.1% of patients). Approximately half of the cohort was eligible for each of the AFFIRM-AHF, intravenous iron treatment in patients with HF and ID, and HEART-FID trials. CONCLUSIONS: Current practices for investigating and treating ID in patients with HF do not align with existing guideline recommendations. Considering the gap in care, innovative strategies to optimize iron therapy in patients with HF are required.


Subject(s)
Anemia, Iron-Deficiency , Ferric Compounds , Heart Failure , Iron Deficiencies , Maltose/analogs & derivatives , Adult , Humans , Iron/therapeutic use , Heart Failure/diagnosis , Heart Failure/drug therapy , Heart Failure/epidemiology , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/epidemiology , Ferritins , Dietary Supplements , Alberta/epidemiology
17.
Swiss Med Wkly ; 154: 3635, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38579297

ABSTRACT

BACKGROUND: Iron deficiency without anaemia is a common health problem, especially in young menstruating women. The efficacy of the usually recommended oral iron supplementation is limited due to increased plasma hepcidin concentration, which reduces iron absorption and leads to side effects such as intestinal irritation. This observation raises the question of how low-dose iron therapy may affect plasma hepcidin levels and whether oral iron intake dose-dependently affects plasma hepcidin production. METHODS: Fifteen non-anaemic women with iron deficiency (serum ferritin ≤30 ng/ml) received a single dose of 0, 6, 30, or 60 mg of elemental oral iron as ferrous sulfate on different days. Plasma hepcidin was measured before and seven hours after each dose. RESULTS: Subjects had an average age of 23 (standard deviation = 3.0) years and serum ferritin of 24 ng/ml (interquartile range = 16-27). The highest mean change in plasma hepcidin levels was measured after ingesting 60 mg of iron, increasing from 2.1 ng/ml (interquartile range = 1.6-2.9) to 4.1 ng/ml (interquartile range = 2.5-6.9; p < 0.001). Iron had a significant dose-dependent effect on the absolute change in plasma hepcidin (p = 0.008), where lower iron dose supplementation resulted in lower plasma hepcidin levels. Serum ferritin levels were significantly correlated with fasting plasma hepcidin levels (R2 = 0.504, p = 0.003) and the change in plasma hepcidin concentration after iron intake (R2 = 0.529, p = 0.002). CONCLUSION: We found a dose-dependent effect of iron supplementation on plasma hepcidin levels. Lower iron dosage results in a smaller increase in hepcidin and might thus lead to more efficient intestinal iron absorption and fewer side effects. The effectiveness and side effects of low-dose iron treatment in women with iron deficiency should be further investigated. This study was registered at the Swiss National Clinical Trials Portal (2021-00312) and ClinicalTrials.gov (NCT04735848).


Subject(s)
Hepcidins , Iron , Female , Humans , Anemia, Iron-Deficiency/drug therapy , Dietary Supplements , Ferritins , Hepcidins/drug effects , Hepcidins/metabolism , Iron/pharmacology , Iron/therapeutic use , Iron Deficiencies/drug therapy , Nutritional Status
18.
Am J Hematol ; 99(6): 1077-1083, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38476079

ABSTRACT

Restless legs syndrome (RLS) is a neurological disorder that can have a profound effect on sleep and quality of life. Idiopathic RLS is associated with brain iron insufficiency despite normal peripheral iron stores. There is, however, a five- to six-fold increase in prevalence of RLS in patients with iron deficiency anemia (IDA). Several open-label trials have demonstrated symptomatic improvement in RLS following treatment of IDA using oral or intravenous iron supplementation. To date, there have been no randomized double-blind controlled trials of intravenous iron compared with oral iron for the treatment of RLS patients with IDA. In the current study, oral ferrous sulfate and ferumoxytol were compared for efficacy and speed of response for treatment of RLS occurring in patients with IDA. The planned recruitment for this study was 70 patients with RLS and IDA, to be randomly assigned 1:1 to oral or intravenous iron, using double-blind, double-dummy procedures. At Week 6, the primary outcomes of Clinical Global Impression-Improvement score and change from baseline in the International Restless Legs Syndrome Study Group rating scale score were assessed. Due to challenges, performing the clinical trial during the COVID-19 pandemic, final-week data were found missing for 30 patients. As a result, in order to maintain the prespecified statistical analysis, an additional 30 patients were recruited. Both IV and oral iron were associated with a marked improvement in RLS symptoms, with no statistically significant difference between treatment groups. No serious adverse events were observed in either treatment group.


Subject(s)
Administration, Intravenous , Anemia, Iron-Deficiency , Ferrous Compounds , Restless Legs Syndrome , Humans , Restless Legs Syndrome/drug therapy , Anemia, Iron-Deficiency/drug therapy , Administration, Oral , Double-Blind Method , Male , Female , Pilot Projects , Middle Aged , Ferrous Compounds/administration & dosage , Ferrous Compounds/therapeutic use , Ferrous Compounds/adverse effects , Adult , Aged , Treatment Outcome , Ferrosoferric Oxide/administration & dosage , Ferrosoferric Oxide/therapeutic use , Ferrosoferric Oxide/adverse effects , Iron/administration & dosage , Iron/therapeutic use
19.
Matern Child Nutr ; 20(3): e13636, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38456385

ABSTRACT

In 2013, the World Health Organisation (WHO) updated the recommendations for micronutrient deficiency correction in hospitalised under-5 children with complicated severe acute malnutrition (SAM). This study aimed to describe the micronutrient deficiency correction practices in relation to WHO 2013 recommendations. Data from medical records of under-5 children admitted for SAM management at two hospitals in South Africa and three tertiary hospitals in Ghana were extracted. Micronutrient correction practices were compared to the WHO 2013 recommendations by considering the dosage, timing of micronutrient supplementation (vitamin A, iron and folic acid) and therapeutic feeds administered. In total, 723 medical records were included. Nearly half (48.3%) of the children received at least one of the studied micronutrients as a supplement. Vitamin A was supplemented in 27.4% of the children, while iron and folic acid were supplemented in 9.5% and 34.9%, respectively. Among the children who received vitamin A, 60.1% received the first dose on Day 1 of admission. Also, 46.4% of the iron-supplemented children received iron within the first week of admission. Vitamin A, iron and folic acid were administered within the dose range of 100,000-180,000 IU, 3.1-7.7 mg per kg per day, and 3-5 mg per day, respectively. Additionally, 71.7% of the children reportedly received therapeutic feeds that met WHO recommendations. The micronutrient deficiency correction practices regarding dose and timing differed from the 2013 WHO guidelines. Qualitative studies investigating the reasons for the disparities are recommended.


Subject(s)
Dietary Supplements , Folic Acid , Micronutrients , Severe Acute Malnutrition , World Health Organization , Humans , Infant , Ghana , Micronutrients/administration & dosage , Micronutrients/deficiency , South Africa , Child, Preschool , Female , Severe Acute Malnutrition/diet therapy , Severe Acute Malnutrition/therapy , Folic Acid/administration & dosage , Male , Vitamin A/administration & dosage , Practice Guidelines as Topic , Iron/administration & dosage , Iron/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL