1
|
Kwiatkowski JL, Thompson AA, Tricta F, Temin NT, Rozova A, Fradette C, Badawy SM. Real-world evidence: Long-term safety of deferiprone in a large cohort of patients with sickle cell disease enrolled in a registry for up to 10 years. Am J Hematol 2024; 99:1031-1039. [PMID: 38429922 DOI: 10.1002/ajh.27276] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
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.
Collapse
Affiliation(s)
- Janet L Kwiatkowski
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alexis A Thompson
- Division of Hematology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | - Anna Rozova
- Chiesi Canada Corporation, Toronto, Ontario, Canada
| | | | - Sherif M Badawy
- Division of Hematology, Oncology and Stem Cell Transplantation, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
2
|
Wei Z, Yu H, Zhao H, Wei M, Xing H, Pei J, Yang Y, Ren K. Broadening horizons: ferroptosis as a new target for traumatic brain injury. BURNS & TRAUMA 2024; 12:tkad051. [PMID: 38250705 PMCID: PMC10799763 DOI: 10.1093/burnst/tkad051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/24/2023] [Accepted: 10/15/2023] [Indexed: 01/23/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability worldwide, with ~50 million people experiencing TBI each year. Ferroptosis, a form of regulated cell death triggered by iron ion-catalyzed and reactive oxygen species-induced lipid peroxidation, has been identified as a potential contributor to traumatic central nervous system conditions, suggesting its involvement in the pathogenesis of TBI. Alterations in iron metabolism play a crucial role in secondary injury following TBI. This study aimed to explore the role of ferroptosis in TBI, focusing on iron metabolism disorders, lipid metabolism disorders and the regulatory axis of system Xc-/glutathione/glutathione peroxidase 4 in TBI. Additionally, we examined the involvement of ferroptosis in the chronic TBI stage. Based on these findings, we discuss potential therapeutic interventions targeting ferroptosis after TBI. In conclusion, this review provides novel insights into the pathology of TBI and proposes potential therapeutic targets.
Collapse
Affiliation(s)
- Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, China
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, No. 1, Longhu Middle Ring Road, Jinshui District, Zhengzhou, China
| | - Haihan Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou, China
| | - Huijuan Zhao
- Henan International Joint Laboratory of Thrombosis and Hemostasis, College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, No. 1, Longhu Middle Ring Road, Jinshui District, Luoyang, China
| | - Mingze Wei
- The Second Clinical Medical College, Harbin Medical University, No. 263, Kaiyuan Avenue, Luolong District, Harbin, China
| | - Han Xing
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, No. 246, Xuefu Road, Nangang District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou 450052, China
| | - Jinyan Pei
- Quality Management Department, Henan No.3 Provincial People’s Hospital, No. 198, Funiu Road, Zhongyuan District, Henan province, Zhengzhou 450052, China
| | - Yang Yang
- Clinical Systems Biology Research Laboratories, Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, No. 198, Funiu Road, Zhongyuan District, Zhengzhou 450052, China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, No. 246, Xuefu Road, Nangang District, Zhengzhou 450052, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, No. 1, Jianshe East Road, Erqi District, Zhengzhou 450052, China
| |
Collapse
|
3
|
Hurtado J, Sellak H, Joseph G, Lewis CV, Naudin CR, Garcia S, Wodicka JR, Archer DR, Taylor WR. Accelerated atherosclerosis in beta-thalassemia. Am J Physiol Heart Circ Physiol 2023; 325:H1133-H1143. [PMID: 37682237 PMCID: PMC10908407 DOI: 10.1152/ajpheart.00306.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/25/2023] [Accepted: 09/06/2023] [Indexed: 09/09/2023]
Abstract
Children with beta-thalassemia (BT) present with an increase in carotid intima-medial thickness, an early sign suggestive of premature atherosclerosis. However, it is unknown if there is a direct relationship between BT and atherosclerotic disease. To evaluate this, wild-type (WT, littermates) and BT (Hbbth3/+) mice, both male and female, were placed on a 3-mo high-fat diet with low-density lipoprotein receptor suppression via overexpression of proprotein convertase subtilisin/kexin type 9 (PCSK9) gain-of-function mutation (D377Y). Mechanistically, we hypothesize that heme-mediated oxidative stress creates a proatherogenic environment in BT because BT is a hemolytic anemia that has increased free heme and exhausted hemopexin, heme's endogenous scavenger, in the vasculature. We evaluated the effect of hemopexin (HPX) therapy, mediated via an adeno-associated virus, to the progression of atherosclerosis in BT and a phenylhydrazine-induced model of intravascular hemolysis. In addition, we evaluated the effect of deferiprone (DFP)-mediated iron chelation in the progression of atherosclerosis in BT mice. Aortic en face and aortic root lesion area analysis revealed elevated plaque accumulation in both male and female BT mice compared with WT mice. Hemopexin therapy was able to decrease plaque accumulation in both BT mice and mice on our phenylhydrazine (PHZ)-induced model of hemolysis. DFP decreased atherosclerosis in BT mice but did not provide an additive benefit to HPX therapy. Our data demonstrate for the first time that the underlying pathophysiology of BT leads to accelerated atherosclerosis and shows that heme contributes to atherosclerotic plaque development in BT.NEW & NOTEWORTHY This work definitively shows for the first time that beta-thalassemia leads to accelerated atherosclerosis. We demonstrated that intravascular hemolysis is a prominent feature in beta-thalassemia and the resulting increases in free heme are mechanistically relevant. Adeno-associated virus (AAV)-hemopexin therapy led to decreased free heme and atherosclerotic plaque area in both beta-thalassemia and phenylhydrazine-treated mice. Deferiprone-mediated iron chelation led to deceased plaque accumulation in beta-thalassemia mice but provided no additive benefit to hemopexin therapy.
Collapse
Affiliation(s)
- Julian Hurtado
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Hassan Sellak
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Giji Joseph
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Caitlin V Lewis
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Crystal R Naudin
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Sergio Garcia
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - James Robert Wodicka
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - David R Archer
- Aflac Cancer and Blood Disorders Center at Children's Healthcare of Atlanta and the Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States
| | - W Robert Taylor
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
- Cardiology Division, Atlanta Veterans Affairs Medical Center, Atlanta, Georgia, United States
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
4
|
Suzuki R, Iki N. Kinetic aspects of iron(III)-chelation therapy with deferasirox (DFX) revealed by the solvolytic dissociation rate of the Fe(III)-DFX complex estimated with capillary electrophoretic reactor. J Inorg Biochem 2023; 241:112131. [PMID: 36706491 DOI: 10.1016/j.jinorgbio.2023.112131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
Capillary electrophoresis was used to estimate the solvolytic dissociation rate (kd) of metal complexes of deferasirox (DFX, H3L), a drug used to treat iron overload. Inert CoIIIL23- did not dissociate. The estimated kd value for FeIIIL23- was (2.7 ± 0.3) × 10-4 s-1 (298 K, pH 7.4). The kd values of other complexes (AlIIIL23-, NiIIL24-, and MnIIL-) were in the range 10-3-10-4 s-1. In contrast, ZnIIL- and CuIIL- were too labile to allow kd estimation. The fact that the half-life of FeIIIL23- (43.3 min) is shorter than the blood half-life of DFX (8-16 h) implies that the blood concentration of DFX should be high enough to prevent dissociation of FeIIIL23-. The possibility of a safer iron-chelation therapy that avoids excretion of other essential metal ions such as ZnII is discussed, highlighting the importance of selectivity in terms of kinetic stability.
Collapse
Affiliation(s)
- Ryota Suzuki
- Graduate School of Environmental Studies, Tohoku University, 6-6-07 Aramaki-Aoba, Aoba-ku, 980-8579, Sendai, Japan
| | - Nobuhiko Iki
- Graduate School of Environmental Studies, Tohoku University, 6-6-07 Aramaki-Aoba, Aoba-ku, 980-8579, Sendai, Japan.
| |
Collapse
|
5
|
Fares MY, Hegazy MA, El-Sayed GM, Abdelrahman MM, Abdelwahab NS. Quality by design approach for green HPLC method development for simultaneous analysis of two thalassemia drugs in biological fluid with pharmacokinetic study. RSC Adv 2022; 12:13896-13916. [PMID: 35548387 PMCID: PMC9084420 DOI: 10.1039/d2ra00966h] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/21/2022] [Indexed: 01/01/2023] Open
Abstract
This work implements a combined experimental approach of analytical quality-by-design (AQbD) and green analytical chemistry (GAC) to develop an HPLC method for simultaneous determination of the two thalassemia drugs, deferasirox (DFX) and deferiprone (DFP), in biological fluid for the first time. This integration was designed to maximize efficiency and minimize environmental impacts, as well as energy and solvent consumption. To accomplish this goal, an analytical quality-by-design approach was performed, beginning with quality risk assessment and scouting analysis, followed by Placket-Burman design screening for five chromatographic parameters. Critical method parameters were thoroughly recognized and then optimized by using a two levels-three factors custom experimental design to evaluate the optimum conditions that achieved the highest resolution with acceptable peak symmetry within the shortest run time. The desirability function was used to define the optimal chromatographic conditions, and the optimal separation was achieved using an XBridge® HPLC RP-C18 (4.6 × 250 mm, 5 μm) column with ethanol : acidic water at pH 3.0 adjusted by phosphoric acid in the ratio of (70 : 30, v/v) as the mobile phase at a flow rate of 1 mL min-1 with UV detection at 225 nm at a temperature of 25 °C. Linearity was obtained over the concentration range of 0.30-20.00 μg mL-1 and 0.20-20.00 μg mL-1 for DFX and DFP, respectively, using 20.00 μg mL-1 ibuprofen (IBF) as an internal standard. The established method's greenness profile was evaluated and measured using various assessment tools, and the developed method was green. For the validation of the developed method, FDA recommendations were followed, and all the results obtained met the acceptance criteria. The suggested method was successfully used to study the pharmacokinetic parameters of DFX and DFP in rat plasma. Due to the substantial increase in bioavailability of the two iron chelating drugs, the results from this study strongly recommend their co-administration.
Collapse
Affiliation(s)
- Michel Y Fares
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Nahda University Sharq El-Nile 62511 Beni-Suef Egypt
| | - Maha A Hegazy
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt +20-100-548-6038
| | - Ghada M El-Sayed
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt +20-100-548-6038
| | - Maha M Abdelrahman
- Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Beni-Suef University Alshaheed Shehata Ahmad Hegazy St 62514 Beni-Suef Egypt
| | - Nada S Abdelwahab
- Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Beni-Suef University Alshaheed Shehata Ahmad Hegazy St 62514 Beni-Suef Egypt
| |
Collapse
|
6
|
Kwiatkowski JL, Hamdy M, El-Beshlawy A, Ebeid FSE, Badr M, Alshehri A, Kanter J, Inusa B, Adly AAM, Williams S, Kilinc Y, Lee D, Tricta F, Elalfy MS. Deferiprone vs deferoxamine for transfusional iron overload in SCD and other anemias: a randomized, open-label noninferiority study. Blood Adv 2022; 6:1243-1254. [PMID: 34847228 PMCID: PMC8864642 DOI: 10.1182/bloodadvances.2021004938] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 10/14/2021] [Indexed: 11/26/2022] Open
Abstract
Many people with sickle cell disease (SCD) or other anemias require chronic blood transfusions, which often causes iron overload that requires chelation therapy. The iron chelator deferiprone is frequently used in individuals with thalassemia syndromes, but data in patients with SCD are limited. This open-label study assessed the efficacy and safety of deferiprone in patients with SCD or other anemias receiving chronic transfusion therapy. A total of 228 patients (mean age: 16.9 [range, 3-59] years; 46.9% female) were randomized to receive either oral deferiprone (n = 152) or subcutaneous deferoxamine (n = 76). The primary endpoint was change from baseline at 12 months in liver iron concentration (LIC), assessed by R2* magnetic resonance imaging (MRI). The least squares mean (standard error) change in LIC was -4.04 (0.48) mg/g dry weight for deferiprone vs -4.45 (0.57) mg/g dry weight for deferoxamine, with noninferiority of deferiprone to deferoxamine demonstrated by analysis of covariance (least squares mean difference 0.40 [0.56]; 96.01% confidence interval, -0.76 to 1.57). Noninferiority of deferiprone was also shown for both cardiac T2* MRI and serum ferritin. Rates of overall adverse events (AEs), treatment-related AEs, serious AEs, and AEs leading to withdrawal did not differ significantly between the groups. AEs related to deferiprone treatment included abdominal pain (17.1% of patients), vomiting (14.5%), pyrexia (9.2%), increased alanine transferase (9.2%) and aspartate transferase levels (9.2%), neutropenia (2.6%), and agranulocytosis (0.7%). The efficacy and safety profiles of deferiprone were acceptable and consistent with those seen in patients with transfusion-dependent thalassemia. This trial study was registered at www://clinicaltrials.gov as #NCT02041299.
Collapse
Affiliation(s)
- Janet L. Kwiatkowski
- Division of Hematology, The Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA
| | - Mona Hamdy
- Department of Pediatrics, School of Medicine, Cairo University, Cairo, Egypt
| | - Amal El-Beshlawy
- Department of Pediatric Hematology, Pediatric Hospital of Cairo University, Cairo, Egypt
| | - Fatma S. E. Ebeid
- Pediatric Hematology Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohammed Badr
- Department of Pediatrics, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Julie Kanter
- Division of Hematology and Oncology, Department of Medicine, University of Alabama, Birmingham, AL
| | - Baba Inusa
- Paediatric Haematology, Evelina Children's Hospital, Guy’s and St. Thomas NHS Foundation Trust, London, United Kingdom
| | - Amira A. M. Adly
- Pediatric Hematology Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Suzan Williams
- Department of Haematology and Oncology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Yurdanur Kilinc
- Department of Pediatric Hematology, Faculty of Medicine, Cukurova University, Adana, Turkey; and
| | - David Lee
- Hematology/Immunology Program, Chiesi Rare Disease, Toronto, ON, Canada
| | - Fernando Tricta
- Hematology/Immunology Program, Chiesi Rare Disease, Toronto, ON, Canada
| | - Mohsen S. Elalfy
- Pediatric Hematology Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
7
|
Doyle EK, Thornton S, Ghugre NR, Coates TD, Nayak KS, Wood JC. Effects of B 1 + Heterogeneity on Spin Echo-Based Liver Iron Estimates. J Magn Reson Imaging 2021; 55:1419-1425. [PMID: 34555245 PMCID: PMC8940739 DOI: 10.1002/jmri.27928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 09/07/2021] [Accepted: 09/10/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Liver iron concentration (LIC) measured by MRI has become the clinical reference standard for managing iron overload in chronically transfused patients. Transverse relaxivity (R2 or R2 * ) measurements are converted to LIC units using empirically derived calibration curves. HYPOTHESIS That flip angle (FA) error due to B1 + spatial heterogeneity causes significant LIC quantitation error. B1 + scale (b1 , [FAactual /FAspecified ]) variation is a major problem at 3 T which could reduce the accuracy of transverse relaxivity measurements. STUDY TYPE Prospective. POPULATION Forty-seven subjects with chronic transfusional iron overload undergoing clinically indicated LIC assessment. FIELD STRENGTH/SEQUENCE 5 T/3 T dual-repetition time B1 + mapping sequence ASSESSMENT: We quantified the average/standard deviation b1 in the right and left lobes of the liver from B1 + maps acquired at 1.5 T and 3 T. The impact of b1 variation on spin echo LIC estimates was determined using a Monte Carlo model. STATISTICAL TESTS Mean, median, and standard deviation in whole liver and right and left lobes; two-sided t-test between whole-liver b1 means. RESULTS Average b1 within the liver was 99.3% ± 12.3% at 1.5 T versus 69.6% ± 14.6% at 3 T and was independent of iron burden (P < 0.05). Monte Carlo simulations demonstrated that b1 systematically increased R2 estimates at lower LIC (<~25 mg/g at 1.5 T, <~15 mg/g at 3 T) but flattened or even inverted the R2 -LIC relationship at higher LIC (≥~25 mg/g to 1.5 T, ≥~15 mg/g to 3 T); changes in the R2 -LIC relationship were symmetric with respect to over and under excitation and were similar at 1.5 T and 3 T (for the same R2 value). The R2 * -LIC relationship was independent of b1 . CONCLUSION Spin echo R2 measurement of LIC at 3 T is error-prone without correction for b1 errors. The impact of b1 error on current 1.5 T spin echo-based techniques for LIC quantification is large enough to introduce measurable intersubject variability but the in vivo effect size needs a dedicated validation study. LEVEL OF EVIDENCE 1. TECHNICAL EFFICACY STAGE 2.
Collapse
Affiliation(s)
- Eamon K Doyle
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.,Division of Cardiology and Radiology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Samuel Thornton
- Ming Hsieh Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, California, USA
| | - Nilesh R Ghugre
- Schulich Heart Research Program, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada.,Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Thomas D Coates
- Division of Hematology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Krishna S Nayak
- Ming Hsieh Department of Electrical and Computer Engineering, University of Southern California, Los Angeles, California, USA
| | - John C Wood
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.,Division of Cardiology and Radiology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
8
|
Zhang J, Zhao H, Yao G, Qiao P, Li L, Wu S. Therapeutic potential of iron chelators on osteoporosis and their cellular mechanisms. Biomed Pharmacother 2021; 137:111380. [PMID: 33601146 DOI: 10.1016/j.biopha.2021.111380] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/30/2021] [Accepted: 02/08/2021] [Indexed: 12/22/2022] Open
Abstract
Iron is an essential trace element in the metabolism of almost all living organisms. Iron overload can disrupt bone homeostasis by significant inhibition of osteogenic differentiation and stimulation of osteoclastogenesis, consequently leading to osteoporosis. Iron accumulation is also involved in the osteoporosis induced by multiple factors, such as estrogen deficiency, ionizing radiation, and mechanical unloading. Iron chelators are first developed for treating iron overloaded disorders. However, growing evidence suggests that iron chelators can be potentially used for the treatment of bone loss. In this review, we focus on the therapeutic effects of iron chelators on bone loss. Iron chelators have therapeutic effects not only on iron overload induced osteoporosis, but also on osteoporosis induced by estrogen deficiency, ionizing radiation, and mechanical unloading, and in Alzheimer's disease-associated osteoporotic deficits. Iron chelators differently affect the cellular behaviors of bone cells. For osteoblast lineage cells (bone mesenchymal stem cells and osteoblasts), iron chelation stimulates osteogenic differentiation. Conversely, iron chelation significantly inhibits osteoclast differentiation. These different responses may be associated with the different needs of iron during differentiation. Fibroblast growth factor 23, angiogenesis, and antioxidant capability are also involved in the osteoprotective effects of iron chelators.
Collapse
Affiliation(s)
- Jian Zhang
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China.
| | - Hai Zhao
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Gang Yao
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Penghai Qiao
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Longfei Li
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Shuguang Wu
- Institute of Laboratory Animal Science, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
9
|
Neutropenia and Life-threatening Agranulocytosis Among Children With β-Thalassemia Treated With Oral Iron Chelators in a Community With Background of Ethnic Neutropenia. J Pediatr Hematol Oncol 2020; 42:e750-e755. [PMID: 31876777 DOI: 10.1097/mph.0000000000001699] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Neutropenia and agranulocytosis are rare side effects of deferiprone (DFP) in patients treated for iron overload. Unfortunately, no study directly addressed special risks in countries with a background of ethnic neutropenia, such as Oman. AIM The aim of this study was to report the incidence of neutropenia in Omani children with β-thalassemia treated with different iron chelators. METHODS/DESIGN This was a retrospective study that included 179 Omani pediatric patients. For patients who developed neutropenia, demographic data, iron-chelating agent, clinical presentation, management, and outcome were recorded. Detailed clinical, laboratory±radiologic information was added for patients with agranulocytosis. RESULTS Neutropenia was encountered in 43.6% of patients: severe neutropenia in 10 patients, moderate in 29, and mild in 69 patients. Severe and moderate neutropenia had similar incidence among DFP-exposed and DFP-naive patients (P=0.515 and 0.421, respectively), while mild neutropenia was common among DFP-naive patients (P=0.0001). A higher incidence of DFP-related agranulocytosis (4.1%) was noted compared with previous reports, but none had a fatal outcome. CONCLUSIONS In a community with ethnic neutropenia, mild to moderate neutropenia is common. Higher incidence of severe neutropenia and agranulocytosis mandates careful monitoring and rational modification of iron-chelating agents to avoid life-threatening complications.
Collapse
|
10
|
d'Arqom A, G Putri M, Savitri Y, Rahul Alfaidin AM. Vitamin and mineral supplementation for β-thalassemia during COVID-19 pandemic. Future Sci OA 2020; 6:FSO628. [PMID: 33230422 PMCID: PMC7434224 DOI: 10.2144/fsoa-2020-0110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 07/31/2020] [Indexed: 01/22/2023] Open
Abstract
AIM Low levels of immune-related micronutrients have been identified in β-thalassemia samples. Moreover, the excess amount of iron, contributing to oxidative stress in the pathogenesis of the disease, alters the immune system in β-thalassemia, which is important during the COVID-19 pandemic. MATERIALS & METHODS Searches of PUBMED and EMBASE were conducted to identify the level and supplementation of micronutrients in β-thalassemia, published from 2001-May 2020. RESULTS The review found six observational and five interventional studies supporting the importance of supplementing vitamins and minerals among patients with β-thalassemia. CONCLUSION Supplementation of immune-related vitamins and minerals might bring benefits to the immune system, especially in reducing oxidative stress in β-thalassemia.
Collapse
Affiliation(s)
- Annette d'Arqom
- Department of Pharmacology & Therapy, Faculty of Medicine, Universitas Airlangga, Surabaya, 60131, Indonesia
| | - Melvanda G Putri
- Faculty of Medicine, Universitas Airlangga, Surabaya, 60131, Indonesia
| | - Yovani Savitri
- Faculty of Medicine, Universitas Airlangga, Surabaya, 60131, Indonesia
| | | |
Collapse
|
11
|
Rajendrakumar S, Durga ASVS, Nanubolu JB, Balasubramanian S. Two novel polymorphic forms of iron-chelating agent deferiprone. ACTA CRYSTALLOGRAPHICA SECTION C-STRUCTURAL CHEMISTRY 2020; 76:193-200. [PMID: 32022715 DOI: 10.1107/s2053229620000959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/24/2020] [Indexed: 11/11/2022]
Abstract
Thalassemia is a genetic blood disorder requiring life-long blood transfusions. This process often results in iron overload and can be treated by an iron-chelating agent, like deferiprone (3-hydroxy-1,2-dimethylpyridin-4-one), C7H9NO2, in an oral formulation. The first crystal structure of deferiprone, (Ia), was reported in 1988 [Nelson et al. (1988). Can. J. Chem. 66, 123-131]. In the present study, two novel polymorphic forms, (Ib) and (Ic), of deferiprone were identified concomitantly with polymorph (Ia) during the crystallization experiments. Polymorph (Ia) was redetermined at low temperature for comparison of the structural features and lattice energy values with polymorphs (Ib) and (Ic). Polymorph (Ia) crystallized in the orthorhombic space group Pbca, whereas both polymorphs (Ib) and (Ic) crystallized in the monoclinic space group P21/c. The asymmetric units of (Ia) and (Ib) contain one deferiprone molecule, while polymorph (Ic) has three crystallographically independent molecules (A, B and C). All three polymorphs have similar hydrogen-bonding features, such as an R22(10) dimer formed by O-H...O hydrogen bonds, an R43(20) tetramer formed by C-H...O hydrogen bonds and π-π interactions, but the polymorphs differ in their molecular arrangements in the solid state and are classified as packing polymorphs. O-H...O and C-H...O hydrogen bonds lead to the formation of two-dimensional hydrogen-bonded parallel sheets which are interlinked by π-π stacking interactions. In the three-dimensional crystal packing, the deferiprone molecules were aggregated as corrugated sheets in polymorphs (Ia) and (Ic), whereas in polymorph (Ib), they were aggregated as a square-grid network. The characteristic crystalline peaks of polymorphs (Ia), (Ib) and (Ic) were established through powder X-ray diffraction analysis. The Rietveld analysis was also performed to estimate the contribution of the polymorphs to the bulk material.
Collapse
Affiliation(s)
- Satyasree Rajendrakumar
- Department of Pharmaceutical Analysis, Osmania University College of Technology, Hyderabad, Telangana 500 007, India
| | | | - Jagadeesh Babu Nanubolu
- Center for X-ray Crystallography, Analytical Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500 007, India
| | - Sridhar Balasubramanian
- Center for X-ray Crystallography, Analytical Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana 500 007, India
| |
Collapse
|
12
|
Sumneang N, Siri-Angkul N, Kumfu S, Chattipakorn SC, Chattipakorn N. The effects of iron overload on mitochondrial function, mitochondrial dynamics, and ferroptosis in cardiomyocytes. Arch Biochem Biophys 2019; 680:108241. [PMID: 31891670 DOI: 10.1016/j.abb.2019.108241] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/22/2019] [Accepted: 12/25/2019] [Indexed: 02/06/2023]
Abstract
Excessive iron accumulation in the heart can lead to iron overload cardiomyopathy (IOC), the leading cause of death in hemochromatosis patients. Current understanding regarding the mechanism by which iron overload causes a deterioration in cardiac performance, mitochondrial dysfunction, and impaired mitochondrial dynamics remains limited. Ferroptosis, a newly identified form of regulated cell death, has recently been revealed influencing the pathophysiological process of IOC. Nevertheless, the direct effect of cardiac iron overload on ferroptotic cell death is incompletely characterized. This review article comprehensively summarizes and discusses the effects of iron overload on cardiac mitochondrial function, cardiac mitochondrial dynamics, ferroptosis of cardiomyocytes, and left ventricular function in in vitro and in vivo reports. This review also provides relevant consistent and controversial information which can facilitate further mechanistic investigation into iron-induced cardiac dysfunction in the clinical setting in the near future.
Collapse
Affiliation(s)
- Natticha Sumneang
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Natthaphat Siri-Angkul
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sirinart Kumfu
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
13
|
Tran NT, Akkawat B, Morales NP, Rojnuckarin P, Luechapudiporn R. Antiplatelet activity of deferiprone through cyclooxygenase-1 inhibition. Platelets 2019; 31:505-512. [PMID: 31366263 DOI: 10.1080/09537104.2019.1648782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Thalassemia patients are susceptible to both iron overload and thromboembolism. Deferiprone is an iron chelator that shows an antiplatelet activity and thus may alleviate platelet hyperactivation in thalassemia. Therefore, this study aimed to characterize the inhibitory effects and mechanisms of deferiprone on normal human platelets. The results illustrated that deferiprone inhibited platelet aggregation at the iron chelating concentrations (0.08-0.25 mmol/l). Deferiprone inhibited human platelet aggregation stimulated by arachidonic acid and ADP more potently than epinephrine and collagen, with the IC50 of 0.24 mmol/l and 0.25 mmol/l vs. 3.36 mmol/l and 3.73 mmol/l, respectively. Interestingly, deferiprone significantly inhibited COX-1 activity, with the IC50 of 0.33 mmol/l, and slightly increased cAMP level at the high concentration of 4 mmol/l. Moreover, the results from molecular docking showed that deferiprone interacted closely with key residues in the peroxidase active site of COX-1. These results suggested that deferiprone possessed antiplatelet activity mainly through the inhibition of COX-1 activity.
Collapse
Affiliation(s)
- Ngan Thi Tran
- Pharmacology and Toxicology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University , Bangkok, Thailand
| | - Benjaporn Akkawat
- Department of Medicine, Chulalongkorn University , Bangkok, Thailand
| | | | | | - Rataya Luechapudiporn
- Natural Products for Ageing and Chronic Diseases Research Unit, Chulalongkorn University , Bangkok, Thailand.,Department of Pharmacology and Physiology, Faculty ofPharmaceutical Sciences, Chulalongkorn University , Bangkok, Thailand
| |
Collapse
|
14
|
Sarno F, Papulino C, Franci G, Andersen JH, Cautain B, Melardo C, Altucci L, Nebbioso A. 3-Chloro- N'-(2-hydroxybenzylidene) benzohydrazide: An LSD1-Selective Inhibitor and Iron-Chelating Agent for Anticancer Therapy. Front Pharmacol 2018; 9:1006. [PMID: 30245629 PMCID: PMC6137965 DOI: 10.3389/fphar.2018.01006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 08/17/2018] [Indexed: 01/09/2023] Open
Abstract
Despite the discovery and development of novel therapies, cancer is still a leading cause of death worldwide. In order to grow, tumor cells require large quantities of nutrients involved in metabolic processes, and an increase in iron levels is known to contribute to cancer proliferation. Iron plays an important role in the active site of a number of proteins involved in energy metabolism, DNA synthesis and repair, such as ribonucleotide reductase, which induce G0/S phase arrest and exert a marked antineoplastic effect, particularly in leukemia and neuroblastoma. Iron-depletion strategies using iron chelators have been shown to result in cell cycle arrest and apoptosis. Deferoxamine (DFO) was the first FDA-approved drug for the treatment of iron overload pathologies, and has also been recognized as having anticancer properties. The high cost, low permeability and short plasma half-life of DFO led to the development of other iron-chelating drugs. Pyridoxal isonicotinoyl hydrazone (PIH) and its analogs chelate cellular iron by tridentate binding, and inhibit DNA synthesis more robustly than DFO, demonstrating an effective antiproliferative activity. Here, we investigated the biological effects of a PIH derivative, 3-chloro-N′-(2-hydroxybenzylidene)benzohydrazide (CHBH), known to be a lysine-specific histone demethylase 1A inhibitor. We showed that CHBH is able to induce cell proliferation arrest in several human cancer cell lines, including lung, colon, pancreas and breast cancer, at micromolar levels. Our findings indicate that CHBH exerts a dual anticancer action by strongly impairing iron metabolism and modulating chromatin structure and function.
Collapse
Affiliation(s)
- Federica Sarno
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | | | - Gianluigi Franci
- Epi-C srl, Naples, Italy.,Dipartimento di Medicina Sperimentale, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Jeanette H Andersen
- Marbio, The University of Tromsø - The Arctic University of Norway, Tromsø, Norway
| | - Bastien Cautain
- Fundación MEDINA, Centro de Excelencia en Investigación de Medicamentos Innovadores en Andalucía, Granada, Spain
| | - Colombina Melardo
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Lucia Altucci
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Angela Nebbioso
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
15
|
Jaiswal S, Hishikar R, Khandwal O, Agarwal M, Joshi U, Halwai A, Maheshwari B, Sheohare R. Efficacy of Deferasirox as an Oral Iron Chelator in Paediatric Thalassaemia Patients. J Clin Diagn Res 2017; 11:FC01-FC03. [PMID: 28384880 DOI: 10.7860/jcdr/2017/22650.9395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/22/2016] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Thalassaemia Major patients require frequent blood transfusion leading to iron overload. Excessive iron gets deposited in vital organs and leads to dysfunction of the heart, liver, anterior pituitary, pancreas, and joints. Our body has limited mechanism to excrete iron, so patients with iron overload and its complications need safe and effective iron chelation therapy. AIM To assess the efficacy of Deferasirox (DFX) as an iron chelator, with specific reference to reduction in serum ferritin level. MATERIALS AND METHODS This is a prospective; observational study done in 45 multitransfused Thalassaemia Major Children receiving DFX therapy at registered Thalassaemia society Raipur Chhattisgarh. DFX was given in an initial dose of 20 mg/kg/day and according to response increased to a maximum of 40 mg/kg/day. Serum ferritin level was estimated at time of registration and at every three monthly intervals (four times during study period). The primary end point of the study was change in serum ferritin level after 12 months of DFX therapy. RESULTS The mean serum ferritin before DFX therapy of all cases was 3727.02 ng/mL. After 12 months of mean dose of 38 mg/kg/day of DFX, the mean decline in serum ferritin was 1207.11 ng/mL (drop by 32.38%, p-value <0.001). CONCLUSION DFX monotherapy has a good safety profile and effectively chelates total body iron in Thalassaemia major patients.
Collapse
Affiliation(s)
- Shikha Jaiswal
- Assistant Professor, Department of Pharmacology, PT JNM Medical College , Raipur, Chhattisgarh, India
| | - Rajesh Hishikar
- Professor and Head, Department of Pharmacology, PT JNM Medical college , Raipur, Chhattisgarh, India
| | - Onkar Khandwal
- Professor, Department of Paediatrics, PT JNM Medical College , Raipur, Chhattisgarh, India
| | - Manju Agarwal
- Assistant Professor, Department of Pharmacology, PT JNM Medical College , Raipur, Chhattisgarh, India
| | - Usha Joshi
- Associate Professor, Department of Pharmacology, PT JNM Medical College , Raipur, Chhattisgarh, India
| | - Ajay Halwai
- Assistant Professor, Department of Pharmacology, PT JNM Medical College , Raipur, Chhattisgarh, India
| | - Basant Maheshwari
- Associate Professor, Department of Pharmacology, PT JNM Medical College , Raipur, Chhattisgarh, India
| | - Raka Sheohare
- Consultant Diabetologist and Non Invasive Cardiologist, Lifeline Hospital , Raipur, Chhattisgarh, India
| |
Collapse
|
16
|
Gotsbacher MP, Telfer TJ, Witting PK, Double KL, Finkelstein DI, Codd R. Analogues of desferrioxamine B designed to attenuate iron-mediated neurodegeneration: synthesis, characterisation and activity in the MPTP-mouse model of Parkinson's disease. Metallomics 2017; 9:852-864. [DOI: 10.1039/c7mt00039a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
One dual-function (2) and one first-generation (9) conjugate of the Fe(iii) chelator desferrioxamine B (DFOB,1) showed significant rescue of neurons in the MPTP mouse model of Parkinson's disease.
Collapse
Affiliation(s)
- Michael P. Gotsbacher
- School of Medical Sciences (Pharmacology)
- and Bosch Institute
- The University of Sydney
- Australia
| | - Thomas J. Telfer
- School of Medical Sciences (Pharmacology)
- and Bosch Institute
- The University of Sydney
- Australia
| | - Paul K. Witting
- School of Medical Sciences (Pathology)
- and Bosch Institute
- The University of Sydney
- Australia
| | - Kay L. Double
- Brain and Mind Centre
- and School of Medical Sciences (Biomedical Sciences)
- The University of Sydney
- Australia
| | | | - Rachel Codd
- School of Medical Sciences (Pharmacology)
- and Bosch Institute
- The University of Sydney
- Australia
| |
Collapse
|
17
|
Nurchi VM, Crisponi G, Lachowicz JI, Medici S, Peana M, Zoroddu MA. Chemical features of in use and in progress chelators for iron overload. J Trace Elem Med Biol 2016; 38:10-18. [PMID: 27365273 DOI: 10.1016/j.jtemb.2016.05.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/20/2016] [Accepted: 05/30/2016] [Indexed: 01/19/2023]
Abstract
An excessive amount of iron may become extremely toxic both for its ability to generate reactive oxygen species, and for the lack of regulatory mechanisms for iron excretion in humans. Chelation therapy has been introduced in clinical practice in the 1970's to defend thalassemia patients from the effects of iron overload and it has dramatically changed both life expectancy and quality of life. The disadvantages of the drugs in clinical use make the research for new, more suitable iron chelating agents, urgent. This review defines the requirements of an iron chelator, then points out the principal chemical features of the iron chelators in use. Finally, a survey on the last ten years of the literature relative to iron chelators is done, and the most interesting ligands are presented, with particular emphasis to those that reached clinical trials.
Collapse
Affiliation(s)
- Valeria Marina Nurchi
- Department of Chemical and Geological Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato-Cagliari, Italy.
| | - Guido Crisponi
- Department of Chemical and Geological Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato-Cagliari, Italy
| | - Joanna I Lachowicz
- Department of Chemical and Geological Sciences, University of Cagliari, Cittadella Universitaria, 09042 Monserrato-Cagliari, Italy
| | - Serenella Medici
- Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy
| | - Massimiliano Peana
- Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy
| | | |
Collapse
|
18
|
Belen BF, Polat M, Özsevik SN, Soylu E. Frequency of neutropenia among Turkish and Syrian pediatric thalassemia patients under deferiprone monotherapy. Pediatr Hematol Oncol 2016; 33:51-8. [PMID: 26918459 DOI: 10.3109/08880018.2015.1106627] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Weekly monitoring of absolute neutrophil count (ANC) under deferiprone therapy in thalassemia patients is recommended to avoid agranulocytosis adverse event. Actually, this recommendation may not be applicable in clinical setting. Our study aimed to establish incidence of neutropenia under deferiprone (DFP) monotherapy when it was monitored bimonthly due to socioeconomic conditions effecting local and refugee thalassemic patients including Syrian origin (SYR; n = 26) and Turkish origin (TR; n = 26) groups. Patients on DFP were followed up for 12 months. Fifteen neutropenic episodes were seen in 5 patients. All 5 patients (4 from SYR group and 1 from TR group) had splenomegaly and hypersplenism, and neutropenia ceased in 4 patients after splenectomy despite continuation of deferiprone. In the TR group, the frequency of patients who have neutropenia (absolute neutrophil count [ANC] <1500/mm(3)) was 3.8% (n = 1) in the 1st month, no patients in TR group had neutropenia until 10th month when again there was 1 patient with mild neutropenia. In SYR group, the frequency of patients who have neutropenia was 3.8% (n = 1), 7.7% (n = 2), and 11.5% (n = 3) in the 1st, 2nd, and 3rd months, respectively, and was found to be 3.8% (n = 1) between 6 and 12 months. Whether or not DFP therapy should be interrupted in case of mild neutropenia and the frequency of monitoring ANC in real-life conditions should be documented with further studies. Other causes of neutropenia in DFP-treated patients should also be kept in mind.
Collapse
Affiliation(s)
- Burcu Fatma Belen
- a Department of Pediatric Hematology, İzmir Katip Celebi University Tepecik Training and Research Hospital , İzmir , Turkey
| | - Meltem Polat
- b Department of Pediatric Infectious Diseases, Gazi University Medical Faculty , Ankara , Turkey
| | | | - Esma Soylu
- c Department of Pediatrics, Gaziantep Childrens' Hospital , Gaziantep , Turkey
| |
Collapse
|
19
|
Lachowicz JI, Nurchi VM, Crisponi G, Jaraquemada-Pelaez MDG, Ostrowska M, Jezierska J, Gumienna-Kontecka E, Peana M, Zoroddu MA, Choquesillo-Lazarte D, Niclós-Gutiérrez J, González-Pérez JM. Zinc(II) and copper(II) complexes with hydroxypyrone iron chelators. J Inorg Biochem 2015; 151:94-106. [PMID: 26281974 DOI: 10.1016/j.jinorgbio.2015.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 12/20/2022]
Abstract
High stability of the complexes formed at physiological pH is one of the basic requisites that a good iron chelator must possess. At the same time the chelating agent must be selective toward iron, i.e., the stability of iron complexes must be significantly higher than that of the complexes formed with essential metal ions, in order that these last ones do not perturb iron chelation. In the frame of our research on iron chelators we have designed and synthesized a series of tetradentate derivatives of kojic acid, and examined their binding properties toward Fe(3+) and Al(3+). In this paper, for a characterization of the behavior of the proposed iron chelating agents in biological fluids, their complex formation equilibria with copper(II) and zinc(II) ions have been fully characterized together with a speciation study, showing the degree at which the iron chelators interfere with the homeostatic equilibria of these two essential metal ions.
Collapse
Affiliation(s)
- Joanna Izabela Lachowicz
- Department of Chemistry and Geology, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Cagliari, Italy.
| | - Valeria Marina Nurchi
- Department of Chemistry and Geology, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Cagliari, Italy
| | - Guido Crisponi
- Department of Chemistry and Geology, University of Cagliari, Cittadella Universitaria, 09042 Monserrato, Cagliari, Italy
| | | | - Małgorzata Ostrowska
- Faculty of Chemistry, University of Wrocław, Joliot-Curie Street 14, 50-383 Wrocław, Poland
| | - Julia Jezierska
- Faculty of Chemistry, University of Wrocław, Joliot-Curie Street 14, 50-383 Wrocław, Poland
| | | | - Massimiliano Peana
- Department of Chemistry and Pharmacy, University of Sassari, Via Vienna 2, 07100 Sassari, Italy
| | | | - Duane Choquesillo-Lazarte
- Laboratorio de Estudios Cristalográficos, IACT, CSIC-Universidad de Granada, Av. de las Palmeras 4, E-18100 Armilla, Granada, Spain
| | - Juan Niclós-Gutiérrez
- Department of Inorganic Chemistry, Faculty of Pharmacy, Campus Cartuja, University of Granada, E-18071 Granada, Spain
| | - Josefa Maria González-Pérez
- Department of Inorganic Chemistry, Faculty of Pharmacy, Campus Cartuja, University of Granada, E-18071 Granada, Spain
| |
Collapse
|
20
|
Crisponi G, Nurchi VM, Crespo-Alonso M, Sanna G, Zoroddu MA, Alberti G, Biesuz R. A Speciation Study on the Perturbing Effects of Iron Chelators on the Homeostasis of Essential Metal Ions. PLoS One 2015; 10:e0133050. [PMID: 26192307 PMCID: PMC4508027 DOI: 10.1371/journal.pone.0133050] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 06/22/2015] [Indexed: 01/19/2023] Open
Abstract
A number of reports have appeared in literature calling attention to the depletion of essential metal ions during chelation therapy on β-thalassaemia patients. We present a speciation study to determine how the iron chelators used in therapy interfere with the homeostatic equilibria of essential metal ions. This work includes a thorough analysis of the pharmacokinetic properties of the chelating agents currently in clinical use, of the amounts of iron, copper and zinc available in plasma for chelation, and of all the implied complex formation constants. The results of the study show that a significant amount of essential metal ions is complexed whenever the chelating agent concentration exceeds the amount necessary to coordinate all disposable iron--a frequently occurring situation during chelation therapy. On the contrary, copper and zinc do not interfere with iron chelation, except for a possible influence of copper on iron speciation during deferiprone treatment.
Collapse
Affiliation(s)
- Guido Crisponi
- Dipartimento di Scienze Chimiche e Geologiche, University of Cagliari, Cagliari, Italy
| | - Valeria Marina Nurchi
- Dipartimento di Scienze Chimiche e Geologiche, University of Cagliari, Cagliari, Italy
| | - Miriam Crespo-Alonso
- Dipartimento di Scienze Chimiche e Geologiche, University of Cagliari, Cagliari, Italy
| | - Gavino Sanna
- Dipartimento di Chimica e Farmacia, University of Sassari, Sassari, Italy
| | | | | | | |
Collapse
|
21
|
Elalfy MS, Saber MM, Adly AAM, Ismail EA, Tarif M, Ibrahim F, Elalfy OM. Role of vitamin C as an adjuvant therapy to different iron chelators in young β-thalassemia major patients: efficacy and safety in relation to tissue iron overload. Eur J Haematol 2015; 96:318-26. [PMID: 26018112 DOI: 10.1111/ejh.12594] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Vitamin C, as antioxidant, increases the efficacy of deferoxamine (DFO). AIM To investigate the effects of vitamin C as an adjuvant therapy to the three used iron chelators in moderately iron-overloaded young vitamin C-deficient patients with β-thalassemia major (β-TM) in relation to tissue iron overload. METHODS This randomized prospective trial that included 180 β-TM vitamin C-deficient patients were equally divided into three groups (n = 60) and received DFO, deferiprone (DFP), and deferasirox (DFX). Patients in each group were further randomized either to receive vitamin C supplementation (100 mg daily) or not (n = 30). All patients received vitamin C (group A) or no vitamin C (group B) were followed up for 1 yr with assessment of transfusion index, hemoglobin, iron profile, liver iron concentration (LIC) and cardiac magnetic resonance imaging (MRI) T2*. RESULTS Baseline vitamin C was negatively correlated with transfusion index, serum ferritin (SF), and LIC. After vitamin C therapy, transfusion index, serum iron, SF, transferrin saturation (Tsat), and LIC were significantly decreased in group A patients, while hemoglobin and cardiac MRI T2* were elevated compared with baseline levels or those in group B without vitamin C. The same improvement was found among DFO-treated patients post-vitamin C compared with baseline data. DFO-treated patients had the highest hemoglobin with the lowest iron, SF, and Tsat compared with DFP or DFX subgroups. CONCLUSIONS Vitamin C as an adjuvant therapy possibly potentiates the efficacy of DFO more than DFP and DFX in reducing iron burden in the moderately iron-overloaded vitamin C-deficient patients with β-TM, with no adverse events.
Collapse
Affiliation(s)
- Mohsen S Elalfy
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Maha M Saber
- Child Health in Complementary Medicine, National Research Center, Cairo, Egypt
| | | | - Eman A Ismail
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed Tarif
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Fatma Ibrahim
- Child Health in Complementary Medicine, National Research Center, Cairo, Egypt
| | - Omar M Elalfy
- Child Health in Complementary Medicine, National Research Center, Cairo, Egypt
| |
Collapse
|
22
|
Athauda D, Foltynie T. The ongoing pursuit of neuroprotective therapies in Parkinson disease. Nat Rev Neurol 2014; 11:25-40. [PMID: 25447485 DOI: 10.1038/nrneurol.2014.226] [Citation(s) in RCA: 185] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many agents developed for neuroprotective treatment of Parkinson disease (PD) have shown great promise in the laboratory, but none have translated to positive results in patients with PD. Potential neuroprotective drugs, such as ubiquinone, creatine and PYM50028, have failed to show any clinical benefits in recent high-profile clinical trials. This 'failure to translate' is likely to be related primarily to our incomplete understanding of the pathogenic mechanisms underlying PD, and excessive reliance on data from toxin-based animal models to judge which agents should be selected for clinical trials. Restricted resources inevitably mean that difficult compromises must be made in terms of trial design, and reliable estimation of efficacy is further hampered by the absence of validated biomarkers of disease progression. Drug development in PD dementia has been mostly unsuccessful; however, emerging biochemical, genetic and pathological evidence suggests a link between tau and amyloid-β deposition and cognitive decline in PD, potentially opening up new possibilities for therapeutic intervention. This Review discusses the most important 'druggable' disease mechanisms in PD, as well as the most-promising drugs that are being evaluated for their potential efficiency in treatment of motor and cognitive impairments in PD.
Collapse
Affiliation(s)
- Dilan Athauda
- Sobell Department of Motor Neuroscience, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| | - Thomas Foltynie
- Sobell Department of Motor Neuroscience, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
23
|
Dutta S, Lahiri S, Banerjee A, Saha S, Dasgupta D. Association of antitumor antibiotic Mithramycin with Mn2+ and the potential cellular targets of Mithramycin after association with Mn2+. J Biomol Struct Dyn 2014; 33:434-46. [PMID: 24559512 DOI: 10.1080/07391102.2014.887031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Mithramycin (MTR), an aureolic acid group of antitumor antibiotic is used for the treatment of several types of tumors. We have reported here the association of MTR with an essential micronutrient, manganese (Mn(2+)). Spectroscopic methods have been used to characterize and understand the kinetics and mechanism of complex formation between them. MTR forms a single type of complex with Mn(2+) in the mole ratio of 2:1 [MTR: Mn(2+)] via a two step kinetic process. Circular dichroism (CD) spectroscopic study indicates that the complex [(MTR)2 Mn(2+)] has a right-handed twist conformation similar in structure with the complexes reported for Mg(2+) and Zn(2+). This conformation allows binding via minor groove of DNA with (G, C) base preference during the interaction with double-stranded B-DNA. Using absorbance, fluorescence, and CD spectroscopy we have shown that [(MTR)2 Mn(2+)] complex binds to double-stranded DNA with an apparent dissociation constant of 32 μM and binding site size of 0.2 (drug/nucleotide). It binds to chicken liver chromatin with apparent dissociation constant value 298 μM. Presence of histone proteins in chromatin inhibits the accessibility of the complex for chromosomal DNA. We have also shown that MTR binds to Mn(2+) containing metalloenzyme manganese superoxide dismutase from Escherichia coli.
Collapse
Affiliation(s)
- Shreyasi Dutta
- a Biophysics & Structural Genomics Division , Saha Institute of Nuclear Physics , Block-AF, Sector-I, Bidhan Nagar, Kolkata - 700 064 , India
| | | | | | | | | |
Collapse
|
24
|
Abstract
Histochemical and MRI studies have demonstrated that MS (multiple sclerosis) patients have abnormal deposition of iron in both gray and white matter structures. Data is emerging indicating that this iron could partake in pathogenesis by various mechanisms, e.g., promoting the production of reactive oxygen species and enhancing the production of proinflammatory cytokines. Iron chelation therapy could be a viable strategy to block iron-related pathological events or it can confer cellular protection by stabilizing hypoxia inducible factor 1α, a transcription factor that normally responds to hypoxic conditions. Iron chelation has been shown to protect against disease progression and/or limit iron accumulation in some neurological disorders or their experimental models. Data from studies that administered a chelator to animals with experimental autoimmune encephalomyelitis, a model of MS, support the rationale for examining this treatment approach in MS. Preliminary clinical studies have been performed in MS patients using deferoxamine. Although some side effects were observed, the large majority of patients were able to tolerate the arduous administration regimen, i.e., 6-8 h of subcutaneous infusion, and all side effects resolved upon discontinuation of treatment. Importantly, these preliminary studies did not identify a disqualifying event for this experimental approach. More recently developed chelators, deferasirox and deferiprone, are more desirable for possible use in MS given their oral administration, and importantly, deferiprone can cross the blood-brain barrier. However, experiences from other conditions indicate that the potential for adverse events during chelation therapy necessitates close patient monitoring and a carefully considered administration regimen.
Collapse
|
25
|
Sripetchwandee J, Pipatpiboon N, Chattipakorn N, Chattipakorn S. Combined therapy of iron chelator and antioxidant completely restores brain dysfunction induced by iron toxicity. PLoS One 2014; 9:e85115. [PMID: 24400127 PMCID: PMC3882264 DOI: 10.1371/journal.pone.0085115] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 12/02/2013] [Indexed: 12/25/2022] Open
Abstract
Background Excessive iron accumulation leads to iron toxicity in the brain; however the underlying mechanism is unclear. We investigated the effects of iron overload induced by high iron-diet consumption on brain mitochondrial function, brain synaptic plasticity and learning and memory. Iron chelator (deferiprone) and antioxidant (n-acetyl cysteine) effects on iron-overload brains were also studied. Methodology Male Wistar rats were fed either normal diet or high iron-diet consumption for 12 weeks, after which rats in each diet group were treated with vehicle or deferiprone (50 mg/kg) or n-acetyl cysteine (100 mg/kg) or both for another 4 weeks. High iron-diet consumption caused brain iron accumulation, brain mitochondrial dysfunction, impaired brain synaptic plasticity and cognition, blood-brain-barrier breakdown, and brain apoptosis. Although both iron chelator and antioxidant attenuated these deleterious effects, combined therapy provided more robust results. Conclusion In conclusion, this is the first study demonstrating that combined iron chelator and anti-oxidant therapy completely restored brain function impaired by iron overload.
Collapse
Affiliation(s)
- Jirapas Sripetchwandee
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Noppamas Pipatpiboon
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- * E-mail:
| | - Siriporn Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
26
|
Abstract
This article provides a practice-based and concise review of the etiology, diagnosis, and management of acquired aplastic anemia in children. Bone marrow transplantation, immunosuppressive therapy, and supportive care are discussed in detail. The aim is to provide the clinician with a better understanding of the disease and to offer guidelines for the management of children with this uncommon yet serious disorder.
Collapse
Affiliation(s)
- Helge D. Hartung
- Division of Hematology, Department of Pediatrics, Comprehensive Bone Marrow Failure Center, The Children’s Hospital of Philadelphia, 3615 Civic Center Boulevard, ARC 302, Philadelphia, PA 19104, USA
| | - Timothy S. Olson
- Division of Oncology, Department of Pediatrics, Comprehensive Bone Marrow Failure Center, The Children’s Hospital of Philadelphia, 3615 Civic Center Boulevard, ARC 302, Philadelphia, PA 19104, USA
| | - Monica Bessler
- Division of Hematology, Department of Pediatrics, Comprehensive Bone Marrow Failure Center, The Children’s Hospital of Philadelphia, 3615 Civic Center Boulevard, ARC 302, Philadelphia, PA 19104, USA,Division of Hemato-Oncology, Department of Medicine, Hospital of the University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, 1218 Penn Tower, Philadelphia, PA 19104, USA
| |
Collapse
|