Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
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
2.
Eur J Med Res ; 28(1): 489, 2023 Nov 07.
Article in English | MEDLINE | ID: mdl-37936193

ABSTRACT

BACKGROUND: Roxadustat is an oral hypoxia inducing factor-prolyl hydroxylase inhibitor (HIF-PHI) that regulates iron metabolism in patients with chronic kidney disease (CKD) primarily by reducing hepcidin levels and mobilizing internal iron stores. More data are needed to demonstrate the efficacy of roxadustat in regulating iron metabolism in patients with peritoneal dialysis (PD) compared with erythropoiesis stimulating agents (ESAs). METHODS: This prospective cohort study enrolled PD patients with a mean hemoglobin level of 60-100 g/L. All subjects were randomized into two groups at a ratio of 2:1 the roxadustat group (106 cases), and the ESA group (53 cases). The primary endpoint was the change in the iron biomarker levels and the proportion of patients with absolute iron deficiency and functional iron deficiency. RESULTS: Compared with ESAs, roxadustat significantly decreased hepcidin level (difference, - 20.09 ng/mL; 95% CI, - 30.26 to - 9.92), attenuated the increase in serum soluble transferrin receptor (sTFR) level (difference, - 7.87 nmol/L; 95% CI, - 12.11 to - 3.64), and reduced the proportion of patients with functional iron deficiency (roxadustat, 11.43%; ESA, 33.33%). There was no significant difference in safety of the two groups over the duration of the study. CONCLUSIONS: Compared with ESA group, roxadustat group showed significant differences in all iron biomarker levels except serum ferritin (sFt) and transferrin saturation (TSAT). These results suggest that roxadustat was superior to ESAs as a therapy for iron metabolism in PD patients. TRIAL REGISTRATION: This study completed Chinese Clinical Trial Registration on March 4, 2022 (registration number: ChiCTR2200057231).


Subject(s)
Glycine , Iron Deficiencies , Isoquinolines , Humans , Biomarkers , Glycine/pharmacology , Hematinics , Hepcidins , Iron/metabolism , Iron Deficiencies/drug therapy , Isoquinolines/pharmacology , Peritoneal Dialysis , Prospective Studies
3.
San Salvador; ISSS; oct. 2023.
Non-conventional in Spanish | BRISA/RedTESA | ID: biblio-1537775

ABSTRACT

ÁREA DESCRIPTIVA DEL PROBLEMA DE SALUD: Definición: La anemia y la ferropenia son dos importantes comorbilidades comunes en pacientes con Insuficiencia Cardíaca (IC) y se asocian a un mal estado clínico y a peores resultados a corto y largo plazo; la anemia y la ferropenia son de hecho, medidores de pobre pronóstico en pacientes con IC, corregir estas comorbilidades sería una diana terapéutica atractiva y novedosa para mejorar los resultados. La anemia se asocia de forma independiente con la gravedad de la IC y la mortalidad, y la deficiencia de hierro parece asociarse de forma exclusiva con una menor capacidad de ejercicio. La deficiencia de hierro suele definirse como un nivel de ferritina <100 µg /L o un nivel de 100 a 300 µg/L, si la saturación de transferrina es <20%. Se ha demostrado que la repleción intravenosa de hierro mejora la capacidad de ejercicio y la calidad de vida. Principales manifestaciones clínicas: la identificación de los síntomas es un paso clave en el diagnóstico; estos incluyen aquellos relacionados a la sobrecarga hídrica (disnea, ortopnea, edema, dolor por la congestión hepática, y discomfort abdominal asociado a la distensión por la ascitis) y aquellos secundarios a la reducción del gasto cardíaco (debilidad, fatiga) que se pronuncian más con el ejercicio.(2) En cuanto a la anemia, definida como la reducción de 1 o más componentes de la línea roja celular (concentración de hemoglobina, hematocrito, o conteo de glóbulos rojos) una concentración baja de hemoglobina o bajo hematocrito son los parámetros ampliamente usados para el diagnóstico, con los siguientes rangos. METODOLOGÍA: Se realizó una búsqueda en las principales bases de datos bibliográficas Pubmed Carboximaltosa férrica; Insuficiencia cardiaca; Deficiencia de hierro; Hierro Sacarosa; Anemia; cirugía cardiovascular. Se filtra la búsqueda a Estudios Clínicos fase 111, controlados randomizados, Revisiones Sistemáticas, Meta-análisis, Guías de Práctica Clínica, además se limitó la búsqueda estudios en humanos. También se realiza búsqueda manual en otras bases de datos bibliográficas (Cochrane, NIH, TRIP 0ATABASE), en buscadores genéricos de internet, agencias de evaluación de tecnologías sanitarias y financiadores de salud. Se priorizó la inclusión de revisiones sistemáticas, meta-análisis, estudios clínicos aleatorizados y controlados, guías de práctica clínica, evaluaciones de tecnología sanitaria, evaluaciones económicas y políticas de cobertura de otros sistemas de salud. CONCLUSIONES. Eficacia: Carboximaltosa Férrica es un medicamento propuesto para el tratamiento de pacientes con anemia por déficit de hierro, en quienes no se puede corregir esta afección con hierro oral, o se necesita hacerlo de una manera rápida; La evaluación de este efecto se ha hecho a través de ensayos clínicos controlados y aleatorizados. Estos estudios sugieren que el uso de carboximaltosa férrica puede ser eficaz para corregir la anemia secundaria por déficit de hierro, pero es importante recalcar que los estudios pivotales están hechos comparando su uso con placebo; la utilización se asocia con mejoría en la calidad de vida de estos pacientes y menos tiempo de estancia hospitalaria; sin embargo, estos mismos informes refieren que no hay disminución en la probabilidad de muerte en general para estos pacientes; es muy importante mencionar que a pesar de que la carboximaltosa férrica al momento está siendo muy estudiada para el uso de pacientes con déficit de hierro e insuficiencia cardíaca, los preparados como Hierro Sacarosa también han sido estudiados en esta patología , y faltan más investigaciones donde se comparen estos dos preparados para obtener mayor evidencia sobre el beneficio de usar uno sobre el otro; además, también hay una gran falta de estudios específicos sobre la población preoperatoria que se someterá a cirugía cardiovascular; si bien es cierto se hace un especial énfasis acerca de la importancia de identificar y corregir la anemia preoperatoria, las guías clínicas actualizadas y revisiones de la literatura no especifican a la carboximaltosa férrica como medicamento de primera línea en el manejo de los pacientes sometidos a cirugía cardiovascular. También, es de hacer notar que la mayoría de investigaciones se han realizado en poblaciones específicas que no pueden ser generalizados a otras poblaciones, así como hay heterogeneidad en los parámetros estudiados entre ellos como los valores de hemoglobina en cada estudio siendo un dato importante para esta revisión. Seguridad: De acuerdo a la revisión realizada se puede concluir que la administración de Carboximaltosa férrica es segura; La reacción adversa reportada con mayor frecuencia fue náuseas (que se produce en el 3,2% de los sujetos), seguida por reacciones en el lugar de inyección/perfusión, hipofosfatemia, cefalea, rubefacción, mareos e hipertensión. Las reacciones en el lugar de inyección/perfusión se componen de varias reacciónes adversas que de forma individual son poco frecuentes o raras. La más grave es la reacción anafiláctica (rara); se han notificado muertes con su uso; los estudios también demuestran que ambas terapias presentan reacciones similares, siendo las más comunes los trastornos de hipersensibilidad, gastrointestinales y desequilibrios hidroelectrolíticos. Aunque es cierto que la literatura describe un ligero aumento en la frecuencia de estas reacciones con Hierro Sacarosa, también se menciona que la mayoría de ellas son rápidamente reversibles y no ponen en peligro la vida de los pacientes. Costo: Para el ISSS, el uso de Carboximaltosa Férrica en pacientes con Insuficiencia Cardíaca y anemia podría considerarse una opción rentable a largo plazo. Dado que se requiere una única aplicación y menos personal y espacio hospitalario, el costo de adquisición se puede recuperar a través de la reducción de recursos de salud utilizados y la ocupación de camas hospitalarias. Aunque al comparar el costo de adquisición de este medicamento con Hierro Sacarosa, un medicamento ya disponible en la institución, se observa que, a pesar de que se necesitan dosis mayores de Hierro Sacarosa para lograr el mismo efecto, el costo total de estas dosis no supera la aplicación individual de Carboximaltosa Férrica. Conveniencia: A pesar de lo antes mencionado sobre el beneficio del uso de carboximaltosa de hierro, en pacientes con déficit de hierro e insuficiencia cardíaca; La mayoría de documentos mencionan que el hierro intravenoso podría controlar los niveles de hemoglobina en los pacientes sometidos a cirugía electiva, no está clara la repercusión del hierro intravenoso sobre los resultados (transfusiones sanguíneas, riesgo de infecciones, o supervivencia en general). En la institución se cuenta con Hierro Sacarosa, un preparado endovenoso que también se utiliza para la corrección de la anemia por déficit de hierro; las ventajas que se presentan de la carboximaltosa férrica a este preparado se centran en menor número de dosis y menor estancia hospitalaria, que se traducen en menor uso de recursos y menor tiempo cama hospitalaria; sin embargo, no hay estudios que prueben el beneficio en una rápida corrección de hemoglobina utilizando carboximaltosa férrica sobre hierro sacarosa, por lo que no es posible concluir que sea más conveniente su uso en pacientes preoperatorios que necesiten un rápido restablecimiento de estos valores.


Subject(s)
Humans , Cardiovascular Surgical Procedures , Heart Failure, Diastolic/physiopathology , Ferric Oxide, Saccharated/therapeutic use , Iron Deficiencies/drug therapy , Anemia/drug therapy , Health Evaluation/economics , Efficacy
4.
Biomed Pharmacother ; 167: 115510, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37757490

ABSTRACT

The study evaluated the safety and effectiveness of the generic intravenous (IV) iron treatment (Feriv®), in a Spanish cohort with absolute iron deficiency (ID) (serum ferritin <50 ng/ml, with or without anaemia) (n = 122; 91% women; median age of 44 years [IQR: 33.7-54]). Iron-related biomarkers were measured before treatment (baseline), 2 weeks after beginning the protocol (intermediate control, IC) and between 7 and 10 days after treatment completion (final time-point). Primary efficacy endpoints were ferritin levels ≥ 50 ng/ml, anaemia restoration or an increase in haemoglobin (Hb) of at least one point in patients without baseline anaemia. After treatment, iron-related biomarkers improved, including ferritin, Hb, sideremia, transferrin, transferrin saturation index, soluble transferrin receptor (sTfR), and hepcidin. Baseline ferritin concentration (13.5 ng/ml [IQR: 8-24.2]) increased at the IC and continued rising at the final time-point, reaching a median ferritin of 222 ng/ml and 97.3% of patients ≥ 50 ng/ml. At the final time-point, anaemia prevalence decreased from 26.2% to 5%, while the 34.1% without baseline anaemia showed an increase in Hb of at least one point. Headache was the only drug-adverse event recorded in 2.3% of patients. At a late time-point (27.5 median weeks after ending therapy [IQR: 22-40]), evaluated in a subgroup of 66 patients, 18% had ferritin levels < 50 ng/ml. Multivariate analysis showed that low baseline ferritin and high sTfR/hepcidin ratio tended to be independently associated with ID recurrence. Feriv® is a safe, effective first-line treatment for absolute ID, with improvement of serum ferritin and Hb. ID recurrence was associated with the baseline degree of iron stores depletion, indicated by serum ferritin, and sTfR/hepcidin ratio.


Subject(s)
Ferric Oxide, Saccharated , Iron Deficiencies , Adult , Female , Humans , Male , Middle Aged , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/etiology , Biomarkers/blood , Dietary Supplements , Ferric Oxide, Saccharated/administration & dosage , Ferric Oxide, Saccharated/adverse effects , Ferritins/blood , Hemoglobins/metabolism , Hepcidins/blood , Iron/metabolism , Receptors, Transferrin , Transferrin , Administration, Intravenous , Iron Deficiencies/complications , Iron Deficiencies/drug therapy
5.
N Engl J Med ; 389(11): 975-986, 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37632463

ABSTRACT

BACKGROUND: Ferric carboxymaltose therapy reduces symptoms and improves quality of life in patients who have heart failure with a reduced ejection fraction and iron deficiency. Additional evidence about the effects of ferric carboxymaltose on clinical events is needed. METHODS: In this double-blind, randomized trial, we assigned ambulatory patients with heart failure, a left ventricular ejection fraction of 40% or less, and iron deficiency, in a 1:1 ratio, to receive intravenous ferric carboxymaltose or placebo, in addition to standard therapy for heart failure. Ferric carboxymaltose or placebo was given every 6 months as needed on the basis of iron indexes and hemoglobin levels. The primary outcome was a hierarchical composite of death within 12 months after randomization, hospitalizations for heart failure within 12 months after randomization, or change from baseline to 6 months in the 6-minute walk distance. The significance level was set at 0.01. RESULTS: We enrolled 3065 patients, of whom 1532 were randomly assigned to the ferric carboxymaltose group and 1533 to the placebo group. Death by month 12 occurred in 131 patients (8.6%) in the ferric carboxymaltose group and 158 (10.3%) in the placebo group; a total of 297 and 332 hospitalizations for heart failure, respectively, occurred by month 12; and the mean (±SD) change from baseline to 6 months in the 6-minute walk distance was 8±60 and 4±59 m, respectively (Wilcoxon-Mann-Whitney P = 0.02; unmatched win ratio, 1.10; 99% confidence interval, 0.99 to 1.23). Repeated dosing of ferric carboxymaltose appeared to be safe with an acceptable adverse-event profile in the majority of patients. The number of patients with serious adverse events occurring during the treatment period was similar in the two groups (413 patients [27.0%] in the ferric carboxymaltose group and 401 [26.2%] in the placebo group). CONCLUSIONS: Among ambulatory patients who had heart failure with a reduced ejection fraction and iron deficiency, there was no apparent difference between ferric carboxymaltose and placebo with respect to the hierarchical composite of death, hospitalizations for heart failure, or 6-minute walk distance. (Funded by American Regent, a Daiichi Sankyo Group company; HEART-FID ClinicalTrials.gov number, NCT03037931.).


Subject(s)
Ferric Compounds , Heart Failure , Iron Deficiencies , Humans , Heart Failure/complications , Heart Failure/drug therapy , Heart Failure/physiopathology , Iron Deficiencies/complications , Iron Deficiencies/drug therapy , Quality of Life , Stroke Volume , Ventricular Function, Left , Ferric Compounds/administration & dosage , Ferric Compounds/adverse effects , Ferric Compounds/therapeutic use , Double-Blind Method , Administration, Intravenous , Ambulatory Care
6.
Nephrol Ther ; 18(6S1): 6S25-6S32, 2022 Dec.
Article in French | MEDLINE | ID: mdl-36585122

ABSTRACT

Anemia is a common complication of chronic kidney disease (CKD). The insufficient erythropoietin (EPO) production by the kidneys and iron deficiency are the main causes. Iron supplementation and the administration of recombinant EPO are the main treatment modalities. New iron formulations that can be administered orally, intravenously or directly via the dialysate have recently been developed to improve efficacy and tolerance. Ferric citrate administered orally can effectively corrects anemia in case of iron deficiency and in addition chelate phosphate in the gut lumen. Ferric carboxymaltose allows intravenous administration of larger doses given less frequently. Ferric pyrophosphate citrate administered directly via the dialysate allows the compensation of iron losses during the hemodialysis session. HIF-prolyl-hydroxylase inhibitors are a new therapeutic class of erythropoiesis-stimulating agents. Orally administered, they act by stabilizing the HIF transcription factor involved in the initiation of erythropoietin production by hypoxia. Several clinical studies have recently evaluated these new molecules in comparison with recombinant EPO. In CKD patients not yet on dialysis or undergoing dialysis therapy non-inferiority in correcting anemia has been demonstrated compared with recombinant EPO. The decrease in circulating hepcidin they induce appears greater than that induced by injectable recombinant EPO. Presently available reports on the safety of HIF-prolyl-hydroxylase inhibitors are reassuring but need to be confirmed in longer-term studies of larger size. © 2022 Published by Elsevier Masson SAS on behalf of Société francophone de néphrologie, dialyse et transplantation.


Subject(s)
Anemia , Erythropoietin , Iron Deficiencies , Prolyl-Hydroxylase Inhibitors , Renal Insufficiency, Chronic , Humans , Anemia/drug therapy , Anemia/etiology , Dialysis Solutions , Erythropoietin/therapeutic use , Iron/therapeutic use , Iron Deficiencies/drug therapy , Iron Deficiencies/etiology , Kidney , Prolyl-Hydroxylase Inhibitors/therapeutic use , Renal Insufficiency, Chronic/complications
7.
Immunobiology ; 227(6): 152297, 2022 11.
Article in English | MEDLINE | ID: mdl-36327544

ABSTRACT

BACKGROUND: Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) needs iron to replicate itself. Coronaviruses are able to upregulate Chop/Gadd153 and Arg1 genes, consequently leading to CD8 lymphocytes decrease, degradation of asparagine and decreased nitric oxide (NO), thus impairing immune response and antithrombotic functions. Little is known about regulation of genes involved in iron metabolism in paucisymptomatic patients with COVID-19 disease or in patients with iron deficiency treated with sucrosomial iron. METHODS: Whole blood was taken from the COVID-19 patients and from patients with sideropenic anemia, treated or not (control group) with iron supplementations. Enrolled patients were: affected by COVID19 under sucrosomal iron support (group A), affected by COVID-19 not under oral iron support (group B), iron deficiency not under treatment, not affected by COVID19 (control group). After RNA extraction and complementary DNA (cDNA) synthesis of Arg1, Hepcidin and Chop/Gadd153, gene expression from the 3 groups was measured by qRT-PCR. M2 macrophages were detected by cytofluorimetry using CD163 and CD14 markers. RESULTS: Forty patients with COVID-19 (group A), 20 patients with iron deficiency treated with sucrosomial iron (group B) and 20 patients with iron deficiency not under treatment (control group) were enrolled. In all the patients supported with oral sucrosomial iron, the gene expression of Chop, Arg1 and Hepcidin genes was lower than in sideropenic patients not supported with iron, M1 macrophages polarization and functional iron deficiency was also lower in group A and B, than observed in the control group. CONCLUSIONS: New oral iron formulations, as sucrosomial iron, are able to influence the expression of genes like Chop and Arg1 and to influence M2 macrophage polarization mainly in the early phase of COVID-19 disease.


Subject(s)
COVID-19 , Ferric Compounds , Iron Deficiencies , Iron , Humans , COVID-19/complications , Homeostasis , Iron/metabolism , Iron Deficiencies/complications , Iron Deficiencies/drug therapy , SARS-CoV-2 , Ferric Compounds/therapeutic use , Macrophages
8.
J Cachexia Sarcopenia Muscle ; 13(6): 2637-2649, 2022 12.
Article in English | MEDLINE | ID: mdl-36321348

ABSTRACT

Iron is an essential nutrient for oxygen supply and aerobic metabolism. Iron deficiency impacts cellular respiration and mitochondrial energy metabolism, which can lead to reduced skeletal muscle function and muscle mass, causing sarcopenia. Intravenous iron offers the ability to rapidly correct iron deficiency, but the functional impact on patient mental and physical health is unclear. We assessed the effects of intravenous iron therapy on physical function and quality of life in the treatment of adults with non-anaemic iron deficiency. An update and reanalysis of a previously published Cochrane systematic review was performed to assess randomized controlled trials that compared any intravenous iron preparation with placebo in adults. The primary functional outcome measure was physical performance as defined by the trial authors. Secondary outcome measures included fatigue and quality-of-life scores, and adverse effects at the end of follow-up. Biochemical efficacy was assessed by change in serum ferritin and haemoglobin concentration levels. Twenty-one randomized controlled trials, comprising 3514 participants, were included. Intravenous iron compared with placebo resulted in significantly increased physical function measured by mean peak oxygen consumption (mean difference [MD] 1.77 mL/kg/min, 95% confidence interval [CI] 0.57 to 2.97). An overall improvement in fatigue was seen (standardized MD 0.30, 95% CI -0.52 to -0.09) but no overall difference in quality of life (MD 0.15, 95% CI -0.01 to 0.31). Biochemically, intravenous iron resulted in improved serum ferritin (MD 245.52 µg/L, 95% CI 152.1 to 338.9) and haemoglobin levels (MD 4.65 g/L, 95% CI 2.53 to 6.78). There was a higher risk of developing mild adverse events in the intravenous iron group compared with the placebo group (risk ratio 1.77, 95% CI 1.10 to 2.83); however, no differences were seen in serious adverse events (risk difference 0, 95% CI -0.01 to 0.01). The quality of evidence was rated 'low' and 'very low' for all outcome variables, except for fatigue, mainly due to most studies being judged as having a high risk of bias. In non-anaemic iron-deficient adults, the use of intravenous iron compared with placebo improved physical function and reduced fatigue scores. However, we remain uncertain about the efficacy in this population due to low-quality evidence, and there is a need for further studies to address potential impact on overall quality of life.


Subject(s)
Iron Deficiencies , Iron , Adult , Humans , Fatigue/drug therapy , Fatigue/etiology , Hemoglobins , Iron/therapeutic use , Iron Deficiencies/drug therapy , Quality of Life
9.
Ter Arkh ; 94(4): 572-578, 2022 May 26.
Article in Russian | MEDLINE | ID: mdl-36286810

ABSTRACT

Iron deficiency is frequent in patients with chronic heart failure (CHF) with a prevalence of 50%, and its frequency varies depending on the study groups. The presence of iron deficiency limits erythropoiesis, leading to the development of anemia over time in patients with CHF, regardless of gender, race, and left ventricular ejection fraction (LVEF). Observational studies demonstrate a higher prevalence of iron deficiency in women and in patients with higher NYHA (New York Heart Association) functional class, decreased LVEF, increased brain natriuretic peptide (NT-proBNP), or increased high-sensitivity C-reactive protein. Iron deficiency and anemia in patients with CHF are independently associated with a decreased exercise capacity, hospitalizations for CHF, an increase in overall mortality and mortality from cardiovascular diseases. The clinical significance of iron deficiency requires the need to diagnose iron metabolism in all patients with CHF. Current guidelines for the diagnosis and treatment of CHF indicate the need to determine the level of ferritin and saturation of transferrin in all patients with a suspected diagnosis of heart failure. The use of oral iron therapy in patients with CHF demonstrates its low efficacy in correcting this condition according to the clinical trials. At the same time the use of intravenous iron therapy is safe and improves symptoms, exercise capacity and quality of life in patients with heart failure with reduced ejection fraction and iron deficiency, which has been shown both in international placebo-controlled trials and meta-analyses. The use of iron carboxymaltose should improve CHF symptoms, exercise capacity and quality of life in patients with CHF and LVEF45%. Intravenous iron therapy has also been shown to reduce readmissions for CHF in patients with an LVEF50% who have recently been hospitalized for worsening CHF.


Subject(s)
Iron Deficiencies , Iron , Female , Humans , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/epidemiology , C-Reactive Protein , Chronic Disease , Ferritins/therapeutic use , Heart Failure , Iron/therapeutic use , Iron Deficiencies/drug therapy , Natriuretic Peptide, Brain , Quality of Life , Stroke Volume , Transferrins/therapeutic use , Ventricular Function, Left , Male , Observational Studies as Topic , Controlled Clinical Trials as Topic , Meta-Analysis as Topic
10.
J Ayub Med Coll Abbottabad ; 34(2): 317-320, 2022.
Article in English | MEDLINE | ID: mdl-35576294

ABSTRACT

BACKGROUND: Iron deficiency is the most common nutritional deficiency worldwide. Common causes of IDA in children are excessive consumption of cow's milk and prolonged breast feeding with delayed and poor weaning. Oral iron is the first line of treatment in children with IDA but occasionally there is inadequate response due to poor compliance. The objective of this study is to assess the effectiveness and safety of intravenous iron in children with IDA, poorly compliant to oral iron therapy. Methods: This study consisted of 90 children from 12-60 months with IDA who were not responding adequately to oral iron therapy. Total iron requirement was calculated and given intravenously (IV) in two divided doses over two consecutive days. Participants were followed up at 2 and 4 weeks to assess the rise in haemoglobin level. Any adverse event was also noted. SPSS version 25 was used for statistical analysis. RESULTS: Of the 90 enrolled children the mean age was 23.1±10.7 months, 47 (52.2%) were males and 43(47.8%) were females. The mean ferritin level before IV iron therapy was 3.75±2.53 ng/ml and mean haemoglobin was 5.9±1.3 g/dL. After IV iron therapy the haemoglobin level was raised to 8.38±1.09 g/dl and 9.74±0.88 g/dl at 2 and 4 weeks respectively which was statistically significant (p<0.05). The adverse events were fever in 3 (3.3%) and urticaria in 2 (2.2%) patients. CONCLUSIONS: Intravenous iron therapy is effective and safe to raise the haemoglobin levels in children with IDA who are poorly compliant to oral iron therapy.


Subject(s)
Anemia, Iron-Deficiency , Iron Deficiencies , Iron , Administration, Intravenous , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/etiology , Animals , Child, Preschool , Female , Hemoglobins/analysis , Humans , Infant , Iron/administration & dosage , Iron/therapeutic use , Iron Deficiencies/drug therapy , Male
11.
Nutrients ; 14(3)2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35276810

ABSTRACT

Iron deficiency in pregnancy is a major public health problem that causes maternal complications. The objective of this randomized, controlled trial was to examine the bioavailability, efficacy, and safety of oral ferrous bisglycinate plus folinic acid supplementation in pregnant women with iron deficiency. Subjects (12−16 weeks of gestation, n = 120) were randomly allocated to receive oral iron as ferrous bisglycinate (equiv. iron 24 mg) in supplement form with folinic acid and multivitamins (test group, n = 60) or as ferrous fumarate (equiv. iron 66 mg iron, control group, n = 60) after breakfast daily. Iron absorption was assessed by measuring fasted serum iron levels at 1 and 2 h immediately after supplementation. Hematological biomarkers and iron status were assessed before intervention, and at 3 and 6 months. Side effects were monitored throughout the intervention. A significant increase in serum iron was seen in both groups (p < 0.001) during the bioavailability assessment; however, the test group increases were comparatively higher than the control values at each timepoint (p < 0.001). Similarly, both test and control groups demonstrated a statistically significant increases in hemoglobin (Hb) (p < 0.001), erythrocytes (p < 0.001), reticulocytes (p < 0.001), mean corpuscular volume (MCV) (p < 0.001), mean corpuscular hemoglobin (MCH) (p < 0.001), mean corpuscular hemoglobin concentration (MCHC) (p < 0.001), % transferrin saturation (p < 0.001), and ferritin (p < 0.001) at 3 and 6 months after supplementation. However, in all cases, the test group increases were numerically larger than the control group increases at each timepoint. The test intervention was also associated with significantly fewer reports of nausea, abdominal pain, bloating, constipation, or metallic taste (p < 0.001). In conclusion, ferrous bisglycinate with folinic acid as a multivitamin nutraceutical format is comparable to standard ferrous fumarate for the clinical management of iron deficiency during pregnancy, with comparatively better absorption, tolerability, and efficacy and with a lower elemental iron dosage.


Subject(s)
Anemia, Iron-Deficiency , Ferrous Compounds , Iron Deficiencies , Pregnancy Complications , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/drug therapy , Biomarkers/blood , Female , Ferrous Compounds/therapeutic use , Glycine/therapeutic use , Humans , Iron Deficiencies/blood , Iron Deficiencies/drug therapy , Leucovorin/therapeutic use , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/drug therapy
13.
J Cardiovasc Pharmacol Ther ; 27: 10742484211055639, 2022.
Article in English | MEDLINE | ID: mdl-34994220

ABSTRACT

Background: Patients suffering from heart failure (HF) and iron deficiency (ID) have worse outcomes. Treatment with intra-venous (IV) ferric carboxymaltose has been shown to reduce HF rehospitalizations and to improve functional capacity and symptoms in patients with HF and reduced ejection fraction (HFrEF). However, IV ferric carboxymaltose is significantly more expensive than IV sodium ferric gluconate complex (SFGC) limiting its availability to most HF patients around the globe. Methods: A retrospective analysis comparing patients admitted to internal medicine or cardiology departments between January 2013 to December 2018 due to acute decompensated HF (ADHF) and treated with or without IV SFGC on top of standard medical therapy. Results: During the study period, a total of 1863 patients were hospitalized due to ADHF with either HFrEF or HF with preserved ejection fraction (HFpEF). Among them, 840 patients had laboratory evidence of iron deficiency (absolute or functional) and met the inclusion criteria. One hundred twenty-two of them (14.5%) were treated with IV SFGC during the index hospitalization. Patients treated with IV iron were more likely to have history of ischemic heart disease, atrial fibrillation, and chronic kidney disease. The rate of readmissions due to ADHF was similar between the groups at 30 days, 3 months, and 1 year. Conclusion: High risk patient hospitalized to ADHF and treated with IV SFGC showed comparable ADHF readmission rates, compared to those who did not receive iron supplementation.


Subject(s)
Ferric Compounds/therapeutic use , Heart Failure/drug therapy , Hematinics/therapeutic use , Iron Deficiencies/drug therapy , Patient Readmission/statistics & numerical data , Acute Disease , Administration, Intravenous , Aged , Aged, 80 and over , Female , Ferric Compounds/administration & dosage , Heart Failure/complications , Hematinics/administration & dosage , Humans , Iron Deficiencies/complications , Israel , Male , Retrospective Studies
14.
Theranostics ; 12(2): 796-816, 2022.
Article in English | MEDLINE | ID: mdl-34976214

ABSTRACT

Ferumoxytol is an intravenous iron oxide nanoparticle formulation that has been approved by the U.S. Food and Drug Administration (FDA) for treating anemia in patients with chronic kidney disease. In recent years, ferumoxytol has also been demonstrated to have potential for many additional biomedical applications due to its excellent inherent physical properties, such as superparamagnetism, biocatalytic activity, and immunomodulatory behavior. With good safety and clearance profiles, ferumoxytol has been extensively utilized in both preclinical and clinical studies. Here, we first introduce the medical needs and the value of current iron oxide nanoparticle formulations in the market. We then focus on ferumoxytol nanoparticles and their physicochemical, diagnostic, and therapeutic properties. We include examples describing their use in various biomedical applications, including magnetic resonance imaging (MRI), multimodality imaging, iron deficiency treatment, immunotherapy, microbial biofilm treatment and drug delivery. Finally, we provide a brief conclusion and offer our perspectives on the current limitations and emerging applications of ferumoxytol in biomedicine. Overall, this review provides a comprehensive summary of the developments of ferumoxytol as an agent with diagnostic, therapeutic, and theranostic functionalities.


Subject(s)
Drug Repositioning , Ferrosoferric Oxide/therapeutic use , Metal Nanoparticles/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Drug Approval , Humans , Iron , Iron Deficiencies/drug therapy , Magnetic Resonance Imaging , Metal Nanoparticles/chemistry , Precision Medicine , United States , United States Food and Drug Administration
15.
Exp Mol Pathol ; 124: 104740, 2022 02.
Article in English | MEDLINE | ID: mdl-34998864

ABSTRACT

Aloin, an anthraquinone glycoside, is one of other C-glycosides found in the leaf exudate of Aloe plant. Aloin possesses several biologic activities, including antitumor activity in vitro and in vivo. However, aloin treatment has shown iron deficiency anemia and erythropoiesis in vivo. The present study was undertaken to verify if iron supplementation could alleviate these perturbations, compared to doxorubicin, an anthracycline analog. Oral iron supplementation (20.56 mg elemental Fe/kg bw) to aloin-treated rats normalized red blood corpuscles count, hemoglobin concentration, and serum levels of total iron binding capacity and saturated transferrin, as well as hepatic iron content, hepcidin level, and mRNA expression of ferritin heavy chain (Ferr-H) and transferrin receptor-1 (TfR-1) genes. Although, serum hyperferremia, and leukocytosis were maintained, yet the spleen iron overload was substantially modulated. However, combined aloin and iron treatment increased iron storage levels in the heart and bone marrow, compared to aloin treatment per se. On other hand, oral iron supplementation to rats treated with doxorubicin (15 mg/kg bw) lessened the increase in the spleen iron content concomitantly with hepatic hepcidin level, rebound hepatic iron content to normal level, and by contrast augmented serum levels of iron and transferrin saturation. Also, activated Ferr-H mRNA expression and repressed TfR-1 mRNA expression were recorded, compared to doxorubicin treatment per se. Histopathological examination of the major body iron stores in rats supplemented with iron along with aloin or doxorubicin showed an increase in extramedullary hematopoiesis. In conclusion, iron supplementation restores the disturbances in iron homeostasis and erythropoiesis induced by aloin treatment.


Subject(s)
Anemia, Iron-Deficiency , Dietary Supplements , Emodin/analogs & derivatives , Iron , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/metabolism , Animals , Emodin/adverse effects , Emodin/pharmacology , Erythropoiesis/drug effects , Glycosides/adverse effects , Glycosides/pharmacology , Hepcidins/blood , Hepcidins/drug effects , Iron/metabolism , Iron/therapeutic use , Iron Deficiencies/drug therapy , Iron Deficiencies/metabolism , Liver/metabolism , Rats , Receptors, Transferrin/blood , Receptors, Transferrin/drug effects , Spleen/metabolism
16.
Ann Thorac Surg ; 113(1): 316-323, 2022 01.
Article in English | MEDLINE | ID: mdl-33345781

ABSTRACT

BACKGROUND: Over the last decade, preoperative anemia has become recognized as a clinical condition in need of management. Although the etiology of preoperative anemia can be multifactorial, two thirds of anemic elective surgical patients have iron deficiency anemia. At the same time, one third of nonanemic elective surgical patients are also iron deficient. METHODS: Modified RAND Delphi methodology was used to identify areas of consensus among an expert panel regarding the management of iron deficiency in patients undergoing cardiac surgery. A list of statements was sent to panel members to respond to using a five-point Likert scale. All panel members subsequently attended a face-to-face meeting. The initial survey was presented and discussed, and panel members responded to each statement on the Likert scale again. Based on the second survey, the panel came to a consensus on recommendations. RESULTS: The panel recommended all patients undergoing cardiac surgery be evaluated for iron deficiency, whether or not anemia is present. Evaluation should include iron studies and reticulocyte hemoglobin content. If iron deficiency is present, with or without anemia, patients should receive parenteral iron. Erythropoietin-stimulating agents may be appropriate for some patients. CONCLUSIONS: Consensus of an expert panel resulted in a standardized approach to diagnosing and managing iron deficiency in patients undergoing cardiac surgery.


Subject(s)
Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/drug therapy , Cardiac Surgical Procedures , Heart Diseases/complications , Heart Diseases/surgery , Iron Deficiencies/complications , Iron Deficiencies/drug therapy , Delphi Technique , Humans , Preoperative Period
17.
J Nippon Med Sch ; 89(2): 203-211, 2022 May 12.
Article in English | MEDLINE | ID: mdl-34526463

ABSTRACT

BACKGROUND: Although some studies have described the association between serum ferritin levels and specific disorders in child and adolescent psychiatry, few have focused on mental status per se with low serum ferritin levels in children and adolescents. This study examined the effects of iron administration on psychological status of children and adolescents with reduced serum ferritin concentration. METHODS: This prospective study evaluated 19 participants aged 6-15 years with serum ferritin levels <30 ng/mL who visited a mental health clinic and received oral iron administration for 12 weeks. The participants were assessed using the Clinical Global Impression Severity (CGI-S), Profile of Mood States 2nd Edition Youth-Short (POMS), Center for Epidemiologic Studies Depression Scale (CES-D), and Pittsburgh Sleep Quality Index (PSQI). In addition to serum ferritin, blood biochemical values such as hemoglobin (Hb) and mean corpuscular volume (MCV) were examined. School attendance was recorded. RESULTS: The most prevalent physical symptoms were fatigability and insomnia. The CGI-S, PSQI, and CES-D scores decreased significantly following iron supplementation, whereas the scores of almost all POMS subscales improved significantly at week 12. No participant had hemoglobin levels <12 g/dL. Serum ferritin concentration increased significantly, whereas Hb and MCV remained unchanged. At baseline, 74% of the participants did not attend school regularly; this number improved to varying degrees by week 12. CONCLUSIONS: Serum ferritin levels would be preferable to be measured in children and adolescents with insomnia and/or fatigability regardless of psychiatric diagnoses or gender. Iron supplementation can improve the hypoferritinemia-related psychological symptoms of children and adolescents, such as poor concentration, anxiety, depression, low energy and/or irritability.


Subject(s)
Iron Deficiencies , Iron , Adolescent , Anxiety , Child , Depression , Dietary Supplements , Ferritins , Hemoglobins , Humans , Iron/therapeutic use , Iron Deficiencies/drug therapy , Iron Deficiencies/psychology , Prospective Studies , Sleep Initiation and Maintenance Disorders
18.
Am J Nephrol ; 52(6): 450-466, 2021.
Article in English | MEDLINE | ID: mdl-34280923

ABSTRACT

BACKGROUND: Patients with CKD frequently have anemia that results from iron-restricted erythropoiesis and inflammation. Anemia of CKD is currently managed with iron supplements and erythropoiesis-stimulating agents (ESAs) to promote erythropoiesis and with RBC transfusion in severe cases. Hyporesponse to ESAs, or the need for larger than usual doses to attain a given hemoglobin (Hb) level, is associated with increased morbidity and mortality and presents a pressing clinical challenge, particularly for patients on dialysis. This paper reviews ESA hyporesponse and potential new therapeutic options in the management of anemia of CKD. SUMMARY: The most common causes of ESA hyporesponse include iron deficiency and inflammation, and to a lesser degree, secondary hyperparathyroidism, inadequate dialysis, malnutrition, and concomitant medications. Management of ESA hyporesponse is multipronged and involves treating low level infections, ensuring adequate nutrition, and optimizing iron status and dialysis modality, although some patients can remain refractory. Inflammation directly increases production and secretion of hepcidin, contributes to an impaired response to hypoxia, and suppresses proliferation of erythroid progenitors. Coordination of renal and hepatic erythropoietin (EPO) production and iron metabolism is under the control of hypoxia-inducible factors (HIF), which are in turn regulated by HIF-prolyl hydroxylases (HIF-PHs). HIF-PHs and hepcidin are therefore attractive potential drug targets particularly in patients with ESA hyporesponse. Several oral HIF-PH inhibitors have been evaluated in patients with anemia of CKD and have been shown to increase Hb and reduce hepcidin regardless of inflammation, iron status, or dialysis modality. These sustained effects are achieved through more modest increases in endogenous EPO compared with ESAs. Key Messages: Treatments that address ESA hyporesponse remain a significant unmet clinical need in patients with anemia of CKD. New therapies such as HIF-PH inhibitors have the potential to address fundamental aspects of ESA hyporesponse and provide a new therapeutic option in these patients.


Subject(s)
Anemia/therapy , Enzyme Inhibitors/therapeutic use , Hematinics/therapeutic use , Renal Insufficiency, Chronic/complications , Anemia/etiology , Hemoglobins/metabolism , Hepcidins/antagonists & inhibitors , Humans , Hyperparathyroidism, Secondary/complications , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Infections/complications , Infections/drug therapy , Inflammation/complications , Iron/therapeutic use , Iron Deficiencies/complications , Iron Deficiencies/drug therapy , Nutritional Status , Renal Insufficiency, Chronic/therapy
19.
Blood Cells Mol Dis ; 90: 102575, 2021 09.
Article in English | MEDLINE | ID: mdl-33989937

ABSTRACT

In order to reduce iron deficiency in neonates at-risk for iron deficiency, we implemented a guideline to increase the consistency of early iron supplementation in infants of diabetic mothers, small for gestational age neonates and very low birthweight premature neonates. Three years following implementation we performed a retrospective analysis in order to assess adherence to the guideline and to compare timing of early iron supplementation and reticulocyte-hemoglobin (RET-He) values at one month of life in at-risk infants. Adherence with early iron supplementation guidelines was 73.4% (399/543) with 51% (275/543) having RET-He values obtained at one month. Despite good adherence, 16% (44/275) had RET-He <25 pg (5th percentile for gestational age). No infants receiving red blood cell transfusion (0/20) had RET-He <25 pg vs. 26.1% (40/153) of those treated with darbepoetin (p < 0.001). There was no evidence of increased feeding intolerance (episodes of emesis/day) with early iron supplementation.


Subject(s)
Intensive Care Units, Neonatal , Iron Deficiencies/drug therapy , Iron/administration & dosage , Female , Humans , Infant, Newborn , Iron/adverse effects , Iron Deficiencies/blood , Male , Retrospective Studies
20.
Eur J Clin Invest ; 51(11): e13624, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34043809

ABSTRACT

BACKGROUND: Over the last years, several trials offered new evidence on heart failure (HF) treatment. DESIGN AND RESULTS: For HF with reduced left ventricular ejection fraction, type 2 sodium-glucose cotransporter inhibitors, aside from sacubitril-valsartan, demonstrated extraordinary efficacy in ameliorating patients' prognosis. Some new molecules (eg vericiguat, omecamtiv mecarbil and ferric carboxymaltose) correct iron deficiency and have shown to be capable of furthering reducing the burden of HF hospitalisation. Finally, there is new evidence on the possible therapeutic approaches of HF patients with mid-range or preserved left ventricular ejection fraction. CONCLUSIONS: This review aimed to revise the main novelties in the field of HF therapy and focus on how the daily clinical approach to patient treatment is changing.


Subject(s)
Angiotensin Receptor Antagonists/therapeutic use , Cardiotonic Agents/therapeutic use , Heart Failure/drug therapy , Hospitalization/statistics & numerical data , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Vasodilator Agents/therapeutic use , Aminobutyrates/therapeutic use , Biphenyl Compounds/therapeutic use , Drug Combinations , Ferric Compounds/therapeutic use , Heart Failure/complications , Heart Failure/physiopathology , Heterocyclic Compounds, 2-Ring/therapeutic use , Humans , Iron Deficiencies/complications , Iron Deficiencies/drug therapy , Maltose/analogs & derivatives , Maltose/therapeutic use , Pyrimidines/therapeutic use , Stroke Volume/physiology , Urea/analogs & derivatives , Urea/therapeutic use , Valsartan/therapeutic use , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL
...