Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 645
1.
Am J Hematol ; 99(6): 1031-1039, 2024 Jun.
Article En | MEDLINE | ID: mdl-38429922

Patients with sickle cell disease (SCD) and other anemias who receive blood transfusions are at risk of organ damage due to transfusional iron overload. Deferiprone is an iron chelator with a well-established safety and efficacy profile that is indicated for the treatment of transfusional iron overload. Here, we report safety data from the large-scale, retrospective Ferriprox® Total Care Registry, which involved all patients with SCD taking deferiprone following the 2011 approval of deferiprone in the United States through August 2020. A total of 634 patients who had initiated deferiprone treatment were included. The mean (SD) duration of deferiprone exposure in the registry was 1.6 (1.6) years (range 0 to 9.7 years). In the overall patient population (N = 634), 64.7% (n = 410) of patients reported a total of 1885 adverse events (AEs). In subgroup analyses, 54.6% (n = 71) of pediatric patients and 67.3% (n = 339) of adult patients reported AEs. The most common AEs reported in patients receiving deferiprone were sickle cell crisis (22.7%), nausea (12.1%), vomiting (8.7%), abdominal discomfort (5.4%), and fatigue (5.4%). Neutropenia was reported in four (0.6%) patients and severe neutropenia/agranulocytosis (defined as absolute neutrophil count <0.5 × 109/L) was reported in two (0.3%) patients. Of patients with evaluable data, all cases of neutropenia and severe neutropenia/agranulocytosis resolved with deferiprone discontinuation. Results from the nearly 10 years of real-world data collected in the Ferriprox® Total Care Registry demonstrate that deferiprone is safe and well tolerated in patients with SCD or other anemias who have transfusional iron overload.


Anemia, Sickle Cell , Deferiprone , Iron Chelating Agents , Registries , Humans , Deferiprone/therapeutic use , Deferiprone/adverse effects , Anemia, Sickle Cell/drug therapy , Male , Child , Adult , Female , Adolescent , Iron Chelating Agents/therapeutic use , Iron Chelating Agents/adverse effects , Iron Chelating Agents/administration & dosage , Retrospective Studies , Iron Overload/drug therapy , Iron Overload/etiology , Child, Preschool , Young Adult , Middle Aged , Infant
4.
N Engl J Med ; 387(22): 2045-2055, 2022 12 01.
Article En | MEDLINE | ID: mdl-36449420

BACKGROUND: Iron content is increased in the substantia nigra of persons with Parkinson's disease and may contribute to the pathophysiology of the disorder. Early research suggests that the iron chelator deferiprone can reduce nigrostriatal iron content in persons with Parkinson's disease, but its effects on disease progression are unclear. METHODS: We conducted a multicenter, phase 2, randomized, double-blind trial involving participants with newly diagnosed Parkinson's disease who had never received levodopa. Participants were assigned (in a 1:1 ratio) to receive oral deferiprone at a dose of 15 mg per kilogram of body weight twice daily or matched placebo for 36 weeks. Dopaminergic therapy was withheld unless deemed necessary for symptom control. The primary outcome was the change in the total score on the Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS; range, 0 to 260, with higher scores indicating more severe impairment) at 36 weeks. Secondary and exploratory clinical outcomes at up to 40 weeks included measures of motor and nonmotor disability. Brain iron content measured with the use of magnetic resonance imaging was also an exploratory outcome. RESULTS: A total of 372 participants were enrolled; 186 were assigned to receive deferiprone and 186 to receive placebo. Progression of symptoms led to the initiation of dopaminergic therapy in 22.0% of the participants in the deferiprone group and 2.7% of those in the placebo group. The mean MDS-UPDRS total score at baseline was 34.3 in the deferiprone group and 33.2 in the placebo group and increased (worsened) by 15.6 points and 6.3 points, respectively (difference, 9.3 points; 95% confidence interval, 6.3 to 12.2; P<0.001). Nigrostriatal iron content decreased more in the deferiprone group than in the placebo group. The main serious adverse events with deferiprone were agranulocytosis in 2 participants and neutropenia in 3 participants. CONCLUSIONS: In participants with early Parkinson's disease who had never received levodopa and in whom treatment with dopaminergic medications was not planned, deferiprone was associated with worse scores in measures of parkinsonism than those with placebo over a period of 36 weeks. (Funded by the European Union Horizon 2020 program; FAIRPARK-II ClinicalTrials.gov number, NCT02655315.).


Antiparkinson Agents , Deferiprone , Iron Chelating Agents , Iron , Parkinson Disease , Substantia Nigra , Humans , Deferiprone/administration & dosage , Deferiprone/adverse effects , Deferiprone/pharmacology , Deferiprone/therapeutic use , Iron/analysis , Iron/metabolism , Levodopa/therapeutic use , Neutropenia/chemically induced , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/adverse effects , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Substantia Nigra/chemistry , Substantia Nigra/diagnostic imaging , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Disease Progression , Double-Blind Method , Administration, Oral , Brain/diagnostic imaging , Brain Chemistry , Dopamine Agents/administration & dosage , Dopamine Agents/adverse effects , Dopamine Agents/pharmacology , Dopamine Agents/therapeutic use , Antiparkinson Agents/administration & dosage , Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacology , Antiparkinson Agents/therapeutic use
5.
J Pediatr Hematol Oncol ; 44(1): e26-e34, 2022 01 01.
Article En | MEDLINE | ID: mdl-34986131

BACKGROUND: Studies on the increased body iron load in patients with thalassemia major have thoroughly demonstrated the problems caused by iron overload. In patients who undergo hematopoietic stem cell transplantation (HSCT) as curative therapy, iron overload continues long after transplantation. There are few pediatric studies on chelation therapy in the posttransplant period. In this study, we present the outcomes of our patients who received posttransplant oral chelation therapy. PATIENTS AND METHODS: This retrospective observational study evaluated the outcomes of pediatric patients with thalassemia major who used oral chelation therapy after allogeneic HSCT at the Akdeniz University Pediatric Bone Marrow Unit between January 2008 and October 2019. RESULTS: Deferasirox therapy was initiated in 58 pediatric patients who underwent HSCT for thalassemia. Pretreatment mean serum ferritin was 2166±1038 ng/mL. Treatment was initiated at a mean of 12±6.7 months after transplantation and continued for a mean of 15.7±11.5 months. At treatment discontinuation, the mean serum ferritin was 693±405 ng/mL and the mean reduction was -1472.75±1121.09 ng/mL (P<0.001 vs. posttreatment). Serum ferritin was below 500 ng/mL in 52% of the patients at treatment discontinuation. Manageable side effects such as nausea, vomiting, liver enzyme elevation, and proteinuria were observed in 17% of the patients, while one patient developed ototoxicity. CONCLUSIONS: Deferasirox therapy effectively reduces iron overload in the posttransplant period. Studies evaluating the effects of early treatment on the graft may help to establish guidelines for posttransplant chelation therapy. Clear guidelines are needed regarding when to initiate and discontinue treatment.


Deferasirox/administration & dosage , Hematopoietic Stem Cell Transplantation , Iron Chelating Agents/administration & dosage , Iron Overload/therapy , Thalassemia/therapy , Adolescent , Allografts , Child , Child, Preschool , Deferasirox/adverse effects , Female , Ferritins/blood , Humans , Iron Chelating Agents/adverse effects , Iron Overload/blood , Male , Retrospective Studies , Thalassemia/blood
6.
Biomed Pharmacother ; 145: 112381, 2022 Jan.
Article En | MEDLINE | ID: mdl-34736078

The oxidative status of twenty-three ß-thalassemia/hemoglobin E patients was evaluated after administration of 75 mg/kg deferiprone (GPO-L-ONE®) divided into 3 doses daily for 12 months. Serum ferritin was significantly decreased; the median value at the initial and final assessments was 2842 and 1719 ng/mL, respectively. Progressive improvement with significant changes in antioxidant enzyme activity, including plasma paraoxonase (PON) and platelet-activating factor acetylhydrolase (PAF-AH), and in antioxidant enzymes in red blood cells (glutathione peroxidase (GPx), catalase and superoxide dismutase (SOD)) were observed at 3-6 months of treatment. The levels of total GSH in red blood cells were significantly increased at the end of the study. Improved red blood cell membrane integrity was also demonstrated using the EPR spin labeling technique. Membrane fluidity at the surface and hydrophobic regions of the red blood cell membrane was significantly changed after 12 months of treatment. In addition, a significant increase in hemoglobin content was observed (6.6 ± 0.7 and 7.5 ± 1.3 g/dL at the initial assessment and at 6 months, respectively). Correlations were observed between hemoglobin content, membrane fluidity and antioxidant enzymes in red blood cells. The antioxidant activity of deferiprone may partly be explained by progressive reduction of redox active iron that catalyzes free radical reactions, as demonstrated by the EPR spin trapping technique. In conclusion, iron chelation therapy with deferiprone notably improved the oxidative status in thalassemia, consequently reducing the risk of oxidative-related complications. Furthermore, the improvement in red blood cell quality may improve the anemia situation in patients.


Deferiprone/pharmacology , Iron Chelating Agents/pharmacology , Iron/metabolism , beta-Thalassemia/drug therapy , Adolescent , Adult , Antioxidants/metabolism , Deferiprone/administration & dosage , Erythrocytes/drug effects , Erythrocytes/metabolism , Female , Ferritins/blood , Glutathione Peroxidase/metabolism , Hemoglobin E/metabolism , Humans , Iron Chelating Agents/administration & dosage , Male , Middle Aged , Oxidation-Reduction , Superoxide Dismutase/metabolism , Young Adult
7.
Pathol Int ; 72(2): 83-95, 2022 Feb.
Article En | MEDLINE | ID: mdl-34965001

Asbestos fibers have been used as an industrial and construction material worldwide due to their high durability and low production cost. Commercial usage of asbestos is currently prohibited in Japan; however, the risk of asbestos-induced malignant mesothelioma (MM) remains. According to epidemiological data, the onset of MM is estimated to occur after a latent period of 30-40 years from initial exposure to asbestos fibers; thus, the continuous increase in MM is a concern. To explore the molecular mechanisms of MM using animal models, iron saccharate with iron chelator-induced sarcomatoid mesothelioma (SM) revealed hallmarks of homozygous deletion of Cdkn2a/2b by aCGH and microRNA-199/214 by expression microarray. Oral treatment of iron chelation by deferasirox decreased the rate of high-grade SM. Moreover, phlebotomy delayed MM development in crocidolite-induced MM in rats. In Divalent metal transporter 1 (Dmt1) transgenic mice, MM development was delayed because of low reactive oxygen species (ROS) production. These results indicate the importance of iron and ROS in mesothelial carcinogenesis. The aims of this review focus on the pathogenesis of elongated mineral particles (EMPs), including asbestos fibers and multiwalled carbon nanotubes (MWCNTs) that share similar rod-like shapes in addition to the molecular mechanisms of MM development.


Asbestos/adverse effects , Iron/metabolism , Mesothelioma, Malignant/pathology , Mineral Fibers/adverse effects , Nanotubes, Carbon/adverse effects , Reactive Oxygen Species/metabolism , Animals , Asbestos, Crocidolite/adverse effects , Carcinogenesis , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Deferasirox/administration & dosage , Humans , Iron Chelating Agents/administration & dosage , Mesothelioma, Malignant/chemically induced , Mice , Mice, Transgenic , Oxidative Stress
8.
Food Funct ; 12(21): 10887-10902, 2021 Nov 01.
Article En | MEDLINE | ID: mdl-34643632

Iron deficiency is a global nutritional problem that adversely affects the functional regulation of the immune system. In the process of treatment through iron supplementation, the problem of excessive iron intake often occurs, which increases the level of inflammation in the body. Excessive iron can also lead to an increase in intestinal iron-requiring pathogenic bacteria and an imbalance of intestinal flora. In this study, we aim to explore the effect of Ejiao peptide-iron (EPI) chelates on the intestinal flora and inflammation of ICR mice having iron-deficiency anemia (IDA). The mice were given low, medium, and high doses of EPI and FeSO4 (1.0, 2.0 and 3.0 mg Fe per kg weight, respectively) daily for 4 weeks by intragastric administration. IDA mice showed increased inflammation levels and decreased sIgA secretion, which were restored after intervention with EPI at different doses. Intestinal mucosal ulcers, inflammatory cell infiltration, and oxidative stress in the colon tissue were reduced, and intestinal permeability was improved. Furthermore, 16S rDNA gene sequencing revealed that EPI increased microbial diversity and richness, changing the community structure, therefore, alleviating microbiota dysbiosis caused by IDA (e.g. the proportion of Firmicutes/Bacteroides). Different from the traditional iron supplement FeSO4, when the pathogenic bacteria (e.g. Helicobacter and Erysipelatoclostridium) increase and the beneficial bacteria (e.g. Bifidobacterium and Blautia) decrease at high doses, EPI shows higher safety at a high dose, thereby maintaining a healthier intestinal homeostasis.


Anemia, Iron-Deficiency/drug therapy , Gastrointestinal Microbiome/drug effects , Gelatin/therapeutic use , Inflammation/drug therapy , Intestinal Diseases/drug therapy , Iron Chelating Agents/therapeutic use , Animals , Bacteria/genetics , Biomarkers , Gene Expression Regulation/drug effects , High-Throughput Nucleotide Sequencing , Iron Chelating Agents/administration & dosage , Male , Mice , Mice, Inbred ICR , Oxidative Stress/drug effects , RNA/genetics , RNA/metabolism , RNA, Bacterial , RNA, Ribosomal, 16S
9.
Expert Rev Clin Pharmacol ; 14(11): 1445-1453, 2021 Nov.
Article En | MEDLINE | ID: mdl-34486906

Background and aim: We conducted a systematic review to apprise the efficacy of silymarin in conjunction with standard iron chelators on iron overload for transfusion-dependent ß-thalassemia (TDT) patients.Methods: We searched PubMed, Web of Science, Scopus, Sciencedirect, the Cochrane Library (the Cochrane Database of Systematic Reviews, and the Cochrane Central Register of Controlled Trials (CENTRAL) to 1 May 2020. All randomized controlled trials (RCTs) studies comparing the effect of iron chelators alone versus silymarin plus standard routine treatment on iron burden amid TDT were included in this review. Primary outcomes comprised serum ferritin level (ng/mL), liver iron concentration (LIC Fe/kg dry weight), and total iron binding capacity (TIBC mcg/dL)Results: Combination therapy of silymarin and iron chelators showed a significant improvement in serum ferritin level in TDT patients, compared to nonsilymarin users [eight studies, n = 477]; weighted mean difference (WMD) -1.79, 95% confidence interval [CI] -2.86 to -0.72, I2 96.1%; P = 0.001. Concurrent treatment with silymarin failed to significantly decrease LIC in TDT patients [two studies, n = 106]; WMD 0.74, 95% CI -1.62 to 3.10, I2 96.6%; P = 0.54.Conclusion: There is no evidence of the effectiveness of adding silymarin to standard iron chelators to reduce iron load in TDT.


Iron Chelating Agents/administration & dosage , Silymarin/administration & dosage , beta-Thalassemia/therapy , Blood Transfusion , Drug Therapy, Combination , Ferritins/blood , Humans , Iron Overload/drug therapy , Randomized Controlled Trials as Topic
10.
J Ayub Med Coll Abbottabad ; 33(2): 207-212, 2021.
Article En | MEDLINE | ID: mdl-34137530

BACKGROUND: Thalassemia major is the severe form of ß thalassemia characterized by severe anaemia, hepatosplenomegaly and facioskeletal changes due to increased haemolysis of defective red blood cells. In iron overload states, high levels of iron exceed the iron-carrying capacity of transferrin within the plasma, leading to the formation of nontransferrin-bound iron form. These nontransferrin-bound iron forms can be taken up into cells, including liver, heart, and endocrine cells leading to organ damage. To prevent complications associated with hemosiderosis, iron chelation therapy remains one of the main objectives of clinical management of the patients affected by Thalassemia Major. METHODS: Thirty-seven patients were enrolled using non randomized convenience sampling technique after the written consent from patients. Patients age 2-30 years were enrolled in this study. Serum Ferritin, ALT, Serum Creatinine were checked at the start of the study, 3 months, 6months and then at the end of the study, i.e., at 9 months of the commencement of the study. They were also assessed for other side effects pertaining to oral tolerability of the drug like vomiting, nausea, GI upset, diarrhoea, urinary complaints or any other subjective complaint. RESULTS: Of the 37 patients, 20 were male (54.1%) and 17 were female (45.9%). Mean age of the patients was 10.2 years (Min. 3 years, Max 21 years). The average serum Ferritin at baseline was noted as 3440 which increased after a period of 3 months, 6 months and 9 months with average of 3359, 3677 and 4394 respectively. After the period of 9 months largest 95% confidence interval of serum Ferritin levels was observed in the range of 3420.17 to 5368.63. In our study, 17 patients required alternative chelation (46%). These patients needed IV Deferioxamine because of the rising trend of Serum Ferritin after the study. CONCLUSIONS: From the results of our study, we infer that oral Deferasirox is not an effective iron chelator. If the patients are taking oral deferasirox, their Serum Ferritin should be checked 3 monthlies. The drug is effective only in maintaining Serum Ferritin levels with levels less than 1500ng/ml. Intravenous Deferioxamine still should be preferred over oral iron chelators for effective control of iron overload and its complications.


Deferasirox/therapeutic use , Iron Chelating Agents/therapeutic use , beta-Thalassemia/drug therapy , Administration, Intravenous , Adolescent , Adult , Benzoates/adverse effects , Child , Child, Preschool , Deferasirox/administration & dosage , Deferasirox/adverse effects , Erythrocyte Count , Female , Hepatomegaly , Humans , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/adverse effects , Male , Triazoles/adverse effects , Young Adult
11.
Sci Rep ; 11(1): 12581, 2021 06 15.
Article En | MEDLINE | ID: mdl-34131221

Deferasirox (DFX) is the newest among three different chelators available to treat iron overload in iron-loading anaemias, firstly released as Dispersible Tablets (DT) and more recently replaced by Film-Coated Tablets (FCT). In this retrospective observational study, pharmacokinetics, pharmacodynamics, and safety features of DFX treatment were analyzed in 74 patients that took both formulations subsequently under clinical practice conditions. Bioavailability of DFX FCT compared to DT resulted higher than expected [Cmax: 99.5 (FCT) and 69.7 (DT) µMol/L; AUC: 1278 (FCT) and 846 (DT), P < 0.0001]. DFX FCT was also superior in scalability among doses. After one year of treatment for each formulation, no differences were observed between the treatments in the overall iron overload levels; however, DFX FCT but not DT showed a significant dose-response correlation [Spearman r (dose-serum ferritin variation): - 0.54, P < 0.0001]. Despite being administered at different dosages, the long-term safety profile was not different between formulations: a significant increase in renal impairment risk was observed for both treatments and it was reversible under strict monitoring (P < 0.002). Altogether, these data constitute a comprehensive comparison of DFX formulations in thalassaemia and other iron-loading anaemias, confirming the effectiveness and safety characteristics of DFX and its applicability for treatment tailoring.


Anemia/drug therapy , Deferasirox/administration & dosage , Iron Overload/drug therapy , Thalassemia/drug therapy , Adult , Anemia/blood , Anemia/epidemiology , Anemia/pathology , Chelation Therapy/trends , Deferasirox/pharmacokinetics , Female , Ferritins/blood , Humans , Iron/blood , Iron/metabolism , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/pharmacokinetics , Iron Overload/blood , Iron Overload/epidemiology , Iron Overload/pathology , Male , Middle Aged , Retrospective Studies , Thalassemia/blood , Thalassemia/epidemiology , Thalassemia/pathology
12.
Br J Haematol ; 194(1): 200-210, 2021 07.
Article En | MEDLINE | ID: mdl-33942901

Beta-thalassaemia is an inherited blood disorder characterised by ineffective erythropoiesis and anaemia. Consequently, hepcidin expression is reduced resulting in increased iron absorption and primary iron overload. Hepcidin is under the negative control of transmembrane serine protease 6 (TMPRSS6) via cleavage of haemojuvelin (HJV), a co-receptor for the bone morphogenetic protein (BMP)-mothers against decapentaplegic homologue (SMAD) signalling pathway. Considering the central role of the TMPRSS6/HJV/hepcidin axis in iron homeostasis, the inhibition of TMPRSS6 expression represents a promising therapeutic strategy to increase hepcidin production and ameliorate anaemia and iron overload in ß-thalassaemia. In the present study, we investigated a small interfering RNA (siRNA) conjugate optimised for hepatic targeting of Tmprss6 (SLN124) in ß-thalassaemia mice (Hbbth3/+ ). Two subcutaneous injections of SLN124 (3 mg/kg) were sufficient to normalise hepcidin expression and reduce anaemia. We also observed a significant improvement in erythroid maturation, which was associated with a significant reduction in splenomegaly. Treatment with the iron chelator, deferiprone (DFP), did not impact any of the erythroid parameters. However, the combination of SLN124 with DFP was more effective in reducing hepatic iron overload than either treatment alone. Collectively, we show that the combination therapy can ameliorate several disease symptoms associated with chronic anaemia and iron overload, and therefore represents a promising pharmacological modality for the treatment of ß-thalassaemia and related disorders.


Deferiprone/therapeutic use , Erythropoiesis/drug effects , Hepcidins/biosynthesis , Iron Chelating Agents/therapeutic use , Iron Overload/prevention & control , Membrane Proteins/antagonists & inhibitors , RNA, Small Interfering/therapeutic use , beta-Thalassemia/drug therapy , Acetylgalactosamine/administration & dosage , Animals , Deferiprone/administration & dosage , Disease Models, Animal , Drug Therapy, Combination , Female , Gene Expression Profiling , Hepcidins/genetics , Humans , Iron/blood , Iron Chelating Agents/administration & dosage , Iron Overload/etiology , Liver/metabolism , Magnesium/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , RNA Interference , RNA, Small Interfering/administration & dosage , Reactive Oxygen Species , Serine Endopeptidases/genetics , Spleen/metabolism , Spleen/ultrastructure , Zinc/metabolism , beta-Thalassemia/complications , beta-Thalassemia/metabolism , beta-Thalassemia/physiopathology
13.
Expert Opin Drug Saf ; 20(8): 937-947, 2021 Aug.
Article En | MEDLINE | ID: mdl-33877003

Introduction: Beta-thalassemia is an autosomal recessive hereditary anemia characterized by reduced or absent ß-globin chain synthesis, affecting about 60,000 people peryear. Management for ß-thalassemia major includes regular blood transfusions followed by iron chelating therapy and drug targeting ineffective erythropoiesis.Areas covered: The safety of licensed drugs for the management of ß-thalassemia is reviewed, using evidence from clinical trials and observational research. Such drugs include the iron chelators and the erythrocyte maturation agent luspatercept. The safety of emerging treatment, such as hydroxyurea and thalidomide is also reviewed.Expert opinion: Beta-thalassemia is arare disease, and is not surprising that there are limited studies investigating the safety of drugs used in this disease. Indeed, although observational studies are the main source of drug safety information in areal-world setting, only eleven studies were identified for iron-chelators and none of these estimated the risk of agiven safety outcome. Future work should aim to better leverage existing sources of real-world datato investigate drug safety in thalassemia.


Activin Receptors, Type II/adverse effects , Immunoglobulin Fc Fragments/adverse effects , Iron Chelating Agents/adverse effects , Recombinant Fusion Proteins/adverse effects , beta-Thalassemia/drug therapy , Activin Receptors, Type II/administration & dosage , Hematinics/administration & dosage , Hematinics/adverse effects , Humans , Hydroxyurea/administration & dosage , Hydroxyurea/adverse effects , Immunoglobulin Fc Fragments/administration & dosage , Iron Chelating Agents/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Thalidomide/administration & dosage , Thalidomide/adverse effects , beta-Thalassemia/physiopathology
14.
Am J Clin Nutr ; 114(2): 780-793, 2021 08 02.
Article En | MEDLINE | ID: mdl-33871598

BACKGROUND: The iron-chelating activities of polyphenols raise concern whether there is a risk of iron deficiency or anemia induced by polyphenol supplementation. Results from clinical trials regarding the effects of polyphenol supplementation on iron status and erythropoiesis are inconclusive. OBJECTIVE: We performed a systematic review and meta-analysis of randomized controlled trials to determine the effects of polyphenol supplementation on iron status and erythropoiesis. METHODS: Published articles were searched between May 1988 and 7 December, 2020. Finally, we identified 34 randomized controlled trials. Random-effects meta-analyses were performed to obtain the weighted mean difference of serum iron (SI), transferrin saturation (TS), ferritin, and hemoglobin concentration. Funnel plots and Egger's test were used to determine the risk of bias. The robustness of the effect sizes was examined by sensitivity analysis. RESULTS: Polyphenol supplementation had an inhibitory effect on the SI concentration (-13.72 µg/dL; 95% CI: -20.74, -6.71) and TS (-3.10%; 95% CI: -4.93, -1.27), with no effect on ferritin (-9.34 ng/mL; 95% CI: -28.55, 9.87). Polyphenols increased the hemoglobin concentration (8.53 g/L; 95% CI: 3.33, 13.73). In healthy participants, polyphenol reduced the TS (-3.83%; 95% CI: -7.47, -0.19) and increased the hemoglobin concentration (12.87 g/L; 95% CI: 1.61, 24.14). Similarly, polyphenol reduced the SI concentration (-8.60 µg/dL; 95% CI: -16.10, -1.10) and increased the hemoglobin concentration (8.50 g/L; 95% CI: 0.86, 16.15) in patients with metabolic diseases. In patients with ß-thalassemia, polyphenol decreased the SI concentration (-23.19 µg/dL; 95% CI: -35.84, -10.55), TS (-3.23%; 95% CI: -5.54, -0.91), and ferritin concentration (-223.62 ng/mL; 95% CI: -359.32, -87.91), but had no effect on the hemoglobin concentration. CONCLUSION: Healthy individuals and patients with metabolic diseases may benefit from the positive impact of polyphenols on erythropoiesis. Patients with ß-thalassemia may benefit from the effect of polyphenols on reducing SI. This trial was registered at PROSPERO (International prospective register of systematic reviews) as CRD42020161983.


Anemia, Iron-Deficiency/chemically induced , Erythropoiesis/drug effects , Iron Chelating Agents/administration & dosage , Iron/metabolism , Polyphenols/administration & dosage , Dietary Supplements , Humans , Iron Chelating Agents/adverse effects , Polyphenols/adverse effects
15.
PLoS One ; 16(4): e0249527, 2021.
Article En | MEDLINE | ID: mdl-33822801

OBJECTIVE: A meta-analysis was conducted to assess the effects of dietary chromium picolinate (CrPic) supplementation on broiler growth performance and to determine whether such effects are regulated by broiler strains, sex, environmental stress, or contextual factors including study area and years. METHODS: Eligible studies were identified by searching the Web of Science, Springer, Elsevier, ScienceDirect, Taylor & Francis Online databases. Weighted average differences with corresponding 95% confidence intervals were computed with a random-effects model. We performed subgroup analysis stratified by study area, published years, broiler strains and sex, and environmental stress. Publication bias was assessed with Egger's test method. A total of 15 studies eligible for inclusion. RESULTS: The results indicated that CrPic supplementation significantly improved broiler growth performance and subgroup analysis confirmed this conclusion. We also found that Ross 308 or male broilers might be more sensitive to CrPic supplementation and showed better growth performance. A model was used to obtain the amount of chromium addition under the optimal growth performance, which suggested that the maximum value of average daily gain (ADG) was reached when chromium addition was 1810 µg/kg. The results of the sensitivity analysis showed low sensitivity and high stability of the meta-analysis. CONCLUSIONS: CrPic supplementation had a positive effect on the growth performance of broilers, and this meta-analysis provides a more accurate value of chromium addition, which may be beneficial for the practice of the broiler industry.


Animal Feed/analysis , Chickens/growth & development , Dietary Supplements , Iron Chelating Agents/administration & dosage , Picolinic Acids/administration & dosage , Animals
16.
Cancer Res ; 81(9): 2289-2303, 2021 05 01.
Article En | MEDLINE | ID: mdl-33685989

Deferoxamine (DFO) represents a widely used iron chelator for the treatment of iron overload. Here we describe the use of mitochondrially targeted deferoxamine (mitoDFO) as a novel approach to preferentially target cancer cells. The agent showed marked cytostatic, cytotoxic, and migrastatic properties in vitro, and it significantly suppressed tumor growth and metastasis in vivo. The underlying molecular mechanisms included (i) impairment of iron-sulfur [Fe-S] cluster/heme biogenesis, leading to destabilization and loss of activity of [Fe-S] cluster/heme containing enzymes, (ii) inhibition of mitochondrial respiration leading to mitochondrial reactive oxygen species production, resulting in dysfunctional mitochondria with markedly reduced supercomplexes, and (iii) fragmentation of the mitochondrial network and induction of mitophagy. Mitochondrial targeting of deferoxamine represents a way to deprive cancer cells of biologically active iron, which is incompatible with their proliferation and invasion, without disrupting systemic iron metabolism. Our findings highlight the importance of mitochondrial iron metabolism for cancer cells and demonstrate repurposing deferoxamine into an effective anticancer drug via mitochondrial targeting. SIGNIFICANCE: These findings show that targeting the iron chelator deferoxamine to mitochondria impairs mitochondrial respiration and biogenesis of [Fe-S] clusters/heme in cancer cells, which suppresses proliferation and migration and induces cell death. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/9/2289/F1.large.jpg.


Carcinogenesis/drug effects , Deferoxamine/administration & dosage , Iron Chelating Agents/administration & dosage , Iron/metabolism , Mitochondria/metabolism , Mitophagy/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Tumor Burden/drug effects , Animals , Cell Death/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Heme/metabolism , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mitochondria/drug effects , Neoplasms/pathology , PC-3 Cells , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
17.
Int J Cancer ; 149(2): 347-357, 2021 07 15.
Article En | MEDLINE | ID: mdl-33662150

Esophageal cancer is a disease showing poor prognosis. Although combination chemotherapy using cisplatin (CDDP) and 5-fluorouracil is standard for unresectable esophageal cancer, the response rate is 35%. Cancer stem cells (CSCs) and inflammation are reportedly responsible for the poor prognosis of esophageal cancer. However, comprehensive analyses have not been conducted and proposals for progress remain lacking. Iron is known to be a key factor in the stemness of CSCs. Our study focused on the therapeutic potential of iron control using iron chelators for CSCs in esophageal cancer. Among 134 immunohistochemically analyzed cases, Nanog expression was high in 98 cases and low in 36 cases. High Nanog expression correlated with low overall and disease-free survivals. The iron chelators deferasirox (DFX) and SP10 suppressed the proliferation and expression of stemness markers in TE8 and OE33 cells. DFX and SP10 did not induce compensatory interleukin (IL)-6 secretion, although CDDP did result in high induction. Moreover, BBI608 and SSZ, as other CSC-targeting drugs, could not suppress the expression of stemness markers. Overall, Nanog expression appears related to poor prognosis in esophageal cancer patients, and inhibition of stemness and compensatory IL-6 secretion by iron chelators may offer a novel therapeutic strategy for esophageal cancer.


Drug Resistance, Neoplasm/drug effects , Esophageal Neoplasms/drug therapy , Gene Expression Profiling/methods , Iron Chelating Agents/administration & dosage , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Iron Chelating Agents/pharmacology , Male , Mice , Nanog Homeobox Protein/drug effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Prognosis , Sequence Analysis, RNA , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
18.
Int J Mol Sci ; 22(2)2021 Jan 16.
Article En | MEDLINE | ID: mdl-33467196

In ß-thalassemia, ineffective erythropoiesis leads to anemia and systemic iron overload. The management of iron overload by chelation therapy is a standard of care. However, iron chelation does not improve the ineffective erythropoiesis. We recently showed that the oral ferroportin inhibitor VIT-2763 ameliorates anemia and erythropoiesis in the Hbbth3/+ mouse model of ß-thalassemia. In this study, we investigated whether concurrent use of the iron chelator deferasirox (DFX) and the ferroportin inhibitor VIT-2763 causes any pharmacodynamic interactions in the Hbbth3/+ mouse model of ß-thalassemia. Mice were treated with VIT-2763 or DFX alone or with the combination of both drugs once daily for three weeks. VIT-2763 alone or in combination with DFX improved anemia and erythropoiesis. VIT-2763 alone decreased serum iron and transferrin saturation (TSAT) but was not able to reduce the liver iron concentration. While DFX alone had no effect on TSAT and erythropoiesis, it significantly reduced the liver iron concentration alone and in the presence of VIT-2763. Our results clearly show that VIT-2763 does not interfere with the iron chelation efficacy of DFX. Furthermore, VIT-2763 retains its beneficial effects on improving ineffective erythropoiesis when combined with DFX in the Hbbth3/+ mouse model. In conclusion, co-administration of the oral ferroportin inhibitor VIT-2763 and the iron chelator DFX is feasible and might offer an opportunity to improve both ineffective erythropoiesis and iron overload in ß-thalassemia.


Benzimidazoles/pharmacology , Erythropoiesis/drug effects , Oxazoles/pharmacology , Pyridines/pharmacology , beta-Thalassemia/drug therapy , Administration, Oral , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Benzimidazoles/therapeutic use , Cation Transport Proteins/antagonists & inhibitors , Cells, Cultured , Deferasirox/administration & dosage , Deferasirox/pharmacology , Deferasirox/therapeutic use , Drug Combinations , Drug Interactions , Female , Iron/blood , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Male , Mice , Mice, Inbred C57BL , Oxazoles/administration & dosage , Oxazoles/adverse effects , Oxazoles/therapeutic use , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/therapeutic use , Transferrin/metabolism
19.
PLoS One ; 16(1): e0238793, 2021.
Article En | MEDLINE | ID: mdl-33481797

There is limited data on iron reduction therapy (IRT) after successful allogeneic haematopoietic stem cell transplantation (aHSCT) for patients with thalassemia major (TM). We present the long term outcome of IRT in 149 patients with TM who underwent aHSCT during January, 2001-December, 2012. The median age was 7 years (range:1-18) and 92 (61.7%) belonged to Pesaro class 3 with a median ferritin at aHSCT of 2480ng/ml (range:866-8921). IRT was reinitiated post-aHSCT at a median of 14 months (range:5-53) post aHSCT with phlebotomy alone in 10 (6.7%) patients or iron chelation alone in 60 (40.3%) patients while 79 (53%) were treated with the combination. Reduction in serum ferritin/month [absolute quantity (ng/ml/month) was as follows: 87 (range:33-195), 130 (range:17-1012) and 147 (range:27.7-1427) in the phlebotomy, chelation and combination therapy groups, respectively (p = 0.038). With a median follow up of 80 months (range:37-182), target ferritin level of <300ng/ml was achieved in 59(40%) while a level <500ng/ml was achieved in 88 patients (59%) in a median duration of 41 months of IRT (range: 3-136). Patients in class III risk category and higher starting serum ferritin levels (>2500ng/ml) were associated with delayed responses to IRT. Our data shows that IRT may be needed for very long periods in ex-thalassaemics to achieve target ferritin levels and should therefore be carefully planned and initiated as soon as possible after aHSCT. A combination of phlebotomy and iron chelators is more effective in reducing iron overload.


Iron Chelating Agents/pharmacology , Iron/metabolism , beta-Thalassemia/drug therapy , Adolescent , Allografts/drug effects , Benzoates/administration & dosage , Child , Child, Preschool , Deferasirox/administration & dosage , Female , Ferritins/analysis , Ferritins/blood , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Infant, Newborn , Iron Chelating Agents/administration & dosage , Iron Overload/etiology , Male , Phlebotomy/methods , Triazoles/administration & dosage
20.
Molecules ; 26(1)2021 Jan 02.
Article En | MEDLINE | ID: mdl-33401708

Iron is an essential element in multiple biochemical pathways in humans and pathogens. As part of the innate immune response in local infection, iron availability is restricted locally in order to reduce overproduction of reactive oxygen species by the host and to attenuate bacterial growth. This physiological regulation represents the rationale for the therapeutic use of iron chelators to support induced iron deprivation and to treat infections. In this review paper we discuss the importance of iron regulation through examples of local infection and the potential of iron chelation in treating infection.


Infections/drug therapy , Iron Chelating Agents/pharmacology , Iron Chelating Agents/therapeutic use , Iron/metabolism , Deferiprone/pharmacology , Deferiprone/therapeutic use , Eye Infections/drug therapy , Homeostasis , Host-Pathogen Interactions/physiology , Humans , Infections/metabolism , Iron Chelating Agents/administration & dosage , Keratitis/drug therapy , Keratitis/microbiology , Siderophores/metabolism , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , Wound Infection/drug therapy , Wound Infection/microbiology
...