Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 489
Filter
1.
Int J Pediatr Otorhinolaryngol ; 177: 111868, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38252990

ABSTRACT

BACKGROUND: Hearing impairment has frequently been described in ß-thalassemia patients with a significant impact on the patients' quality of life. Most studies provided evidence of deferoxamine (DFO) dose-related ototoxicity, however, the data is scarce regarding deferasirox (DFX) as a sole iron chelator. AIM: We aimed to assess the prevalence and risk factors of sensorineural hearing loss (SNHL) and vestibular dysfunction in regularly transfused ß-thalassemia patients who had been treated with DFX film coated tablets. METHODS: We conducted a case control study on 57 transfusion dependent ß-thalassemia patients with a mean age of 15.3 years who received DFX FCT as monotherapy for at least one consecutive year, and 57 healthy age and sex-matching controls. Comprehensive audiological evaluations using pure tone audiometry (PTA) and transient evoked otoacoustic emission (TEOAE) as well as vestibular evaluation using Video-nystagmography (VNG) were done. RESULTS: SNHL was identified in 12 patients (21.1 %) using PTA and a statistically significant difference was detected between controls and patients at 6 KHz and 12 KHz frequencies. A higher incidence of SNHL was detected using TEOAE, 22 patients (43.1 %) failed to pass TEOAE, with a statistically significant decrease in the signal at frequencies 1, 4 KHz bilaterally and at frequencies 1.5, 2 KHz in the right ear compared to controls. Canal paresis was detected in 21 (36.8 %) of thalassemic children using bithermal caloric test with significantly more unilateral weakness than control children (P = 0.008). We found no significant correlation between audio-vestibular dysfunction and age, sex, serum ferritin, frequency of blood transfusion and dose of DFX FCT in thalassemic children. CONCLUSION: We conclude that the incidence of SNHL and vestibular dysfunction was high among transfusion dependent ß-thalassemia patients. Therefore, we recommend performing pre-treatment baseline audio-vestibular assessment and yearly audio-vestibular monitoring to early detect high risk patients and initiate timely management to prevent permanent damage.


Subject(s)
Hearing Loss, Sensorineural , beta-Thalassemia , Child , Humans , Adolescent , beta-Thalassemia/complications , beta-Thalassemia/therapy , Deferasirox/adverse effects , Deferoxamine/adverse effects , Case-Control Studies , Quality of Life , Hearing Loss, Sensorineural/chemically induced , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/epidemiology
2.
Photodiagnosis Photodyn Ther ; 45: 103961, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38163453

ABSTRACT

BACKGROUND: We present a case of bilateral maculopathy associated with deferoxamine mesylate (DFO) treatment. METHODS: A 53-year-old man with myelodysplastic syndrome (MDS) received DFO therapy due to elevated ferritin levels. He was then referred to ophthalmology clinic due to blurred vision. He was diagnosed as bilateral neurosensory retinal detachment of the macula. During follow up, best corrected visual acuity (BCVA), optical coherence tomography (OCT), fundus fluorescein angiography (FFA), and fundus autofluorescence (FAF) were evaluated. RESULTS: At first visit, OCT showed bilateral foveal neurosensory detachment. Hyperfluorescence of the macula and the peripapillary region were found on FFA. After discontinuation of DFO, BCVA improved from 20/120 to 20/60 with resolution of the foveal detachments on OCT scan. Four weeks later, FAF showed bilateral mottled hyperautofluorescence and hypoautofluorescence at the macula and the peripapillary region. CONCLUSION: Deferoxamine can cause acute retinal toxicity. Haematologists should be alert to visual complaints associated with DFO therapy, as early diagnosis and discontinuation of the medication allows recovery of visual function with residual fundus findings.


Subject(s)
Macular Degeneration , Photochemotherapy , Retinal Diseases , Male , Humans , Middle Aged , Deferoxamine/adverse effects , Tomography, Optical Coherence/methods , Photochemotherapy/methods , Photosensitizing Agents
3.
Transfus Apher Sci ; 62(3): 103702, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37055329

ABSTRACT

Drug-induced nephrolithiasis can arise from insoluble components within medications or crystallization of metabolites due to changes in metabolism and urinary pH. The connection between drugs utilized for iron chelation therapy (ICT) and nephrolithiasis is not well understood. In this report, we describe two pediatric patients diagnosed with nephrolithiasis while undergoing treatment with the chelating agents deferasirox, deferiprone, and deferoxamine for iron overload secondary to repeat blood transfusion.


Subject(s)
Iron Overload , Nephrolithiasis , beta-Thalassemia , Humans , Child , Chelation Therapy/adverse effects , Iron Chelating Agents/adverse effects , Deferasirox/adverse effects , Deferiprone/therapeutic use , Deferoxamine/adverse effects , Benzoates/adverse effects , Triazoles , Iron Overload/drug therapy , Iron Overload/etiology , Nephrolithiasis/chemically induced , Nephrolithiasis/complications , Nephrolithiasis/drug therapy , Iron/therapeutic use , beta-Thalassemia/therapy
4.
Cochrane Database Syst Rev ; 3: CD012349, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36877640

ABSTRACT

BACKGROUND: Regularly transfused people with sickle cell disease (SCD) and people with thalassaemia are at risk of iron overload. Iron overload can lead to iron toxicity in vulnerable organs such as the heart, liver and endocrine glands, which can be prevented and treated with iron-chelating agents. The intensive demands and uncomfortable side effects of therapy can have a negative impact on daily activities and wellbeing, which may affect adherence. OBJECTIVES: To identify and assess the effectiveness of different types of interventions (psychological and psychosocial, educational, medication interventions, or multi-component interventions) and interventions specific to different age groups, to improve adherence to iron chelation therapy compared to another listed intervention, or standard care in people with SCD or thalassaemia. SEARCH METHODS: We searched CENTRAL (Cochrane Library), MEDLINE, PubMed, Embase, CINAHL, PsycINFO, ProQuest Dissertations & Global Theses, Web of Science & Social Sciences Conference Proceedings Indexes and ongoing trial databases (13 December 2021). We searched the Cochrane Cystic Fibrosis and Genetic Disorders Group's Haemoglobinopathies Trials Register (1 August 2022). SELECTION CRITERIA: For trials comparing medications or medication changes, only randomised controlled trials (RCTs) were eligible for inclusion. For studies including psychological and psychosocial interventions, educational interventions, or multi-component interventions, non-randomised studies of interventions (NRSIs), controlled before-after studies, and interrupted time series studies with adherence as a primary outcome were also eligible for inclusion. DATA COLLECTION AND ANALYSIS: For this update, two authors independently assessed trial eligibility and risk of bias, and extracted data. We assessed the certainty of the evidence using GRADE. MAIN RESULTS: We included 19 RCTs and one NRSI published between 1997 and 2021. One trial assessed medication management, one assessed an education intervention (NRSI) and 18 RCTs were of medication interventions. Medications assessed were subcutaneous deferoxamine, and two oral chelating agents, deferiprone and deferasirox. We rated the certainty of evidence as very low to low across all outcomes identified in this review. Four trials measured quality of life (QoL) with validated instruments, but provided no analysable data and reported no difference in QoL. We identified nine comparisons of interest. 1. Deferiprone versus deferoxamine We are uncertain whether or not deferiprone affects adherence to iron chelation therapy (four RCTs, unpooled, very low-certainty evidence), all-cause mortality (risk ratio (RR) 0.47, 95% confidence interval (CI) 0.18 to 1.21; 3 RCTs, 376 participants; very low-certainty evidence), or serious adverse events (SAEs) (RR 1.43, 95% CI 0.83 to 2.46; 1 RCT, 228 participants; very low-certainty evidence).  Adherence was reported as "good", "high" or "excellent" by all seven trials, though the data could not be analysed formally: adherence ranged from 69% to 95% (deferiprone, mean 86.6%), and 71% to 93% (deferoxamine, mean 78.8%), based on five trials (474 participants) only. 2. Deferasirox versus deferoxamine We are uncertain whether or not deferasirox affects adherence to iron chelation therapy (three RCTs, unpooled, very low-certainty evidence), although medication adherence was high in all trials. We are uncertain whether or not there is any difference between the drug therapies in serious adverse events (SAEs) (SCD or thalassaemia) or all-cause mortality (thalassaemia). 3. Deferiprone versus deferasirox We are uncertain if there is a difference between oral deferiprone and deferasirox based on a single trial in children (average age 9 to 10 years) with any hereditary haemoglobinopathy in adherence, SAEs and all-cause mortality. 4. Deferasirox film-coated tablet (FCT) versus deferasirox dispersible tablet (DT) One RCT compared deferasirox in different tablet forms. There may be a preference for FCTs, shown through a trend for greater adherence (RR 1.10, 95% CI 0.99 to 1.22; 1 RCT, 88 participants), although medication adherence was high in both groups (FCT 92.9%; DT 85.3%). We are uncertain if there is a benefit in chelation-related AEs with FCTs. We are uncertain if there is a difference in the incidence of SAEs, all-cause mortality or sustained adherence. 5. Deferiprone and deferoxamine combined versus deferiprone alone We are uncertain if there is a difference in adherence, though reporting was usually narrative as triallists report it was "excellent" in both groups (three RCTs, unpooled). We are uncertain if there is a difference in the incidence of SAEs and all-cause mortality.  6. Deferiprone and deferoxamine combined versus deferoxamine alone We are uncertain if there is a difference in adherence (four RCTs), SAEs (none reported in the trial period) and all-cause mortality (no deaths reported in the trial period). There was high adherence in all trials. 7. Deferiprone and deferoxamine combined versus deferiprone and deferasirox combined There may be a difference in favour of deferiprone and deferasirox (combined) in rates of adherence (RR 0.84, 95% CI 0.72 to 0.99) (one RCT), although it was high (> 80%) in both groups. We are uncertain if there is a difference in SAEs, and no deaths were reported in the trial, so we cannot draw conclusions based on these data (one RCT). 8. Medication management versus standard care We are uncertain if there is a difference in QoL (one RCT), and we could not assess adherence due to a lack of reporting in the control group. 9. Education versus standard care One quasi-experimental (NRSI) study could not be analysed due to the severe baseline confounding. AUTHORS' CONCLUSIONS: The medication comparisons included in this review had higher than average adherence rates not accounted for by differences in medication administration or side effects, though often follow-up was not good (high dropout over longer trials), with adherence based on a per protocol analysis. Participants may have been selected based on higher adherence to trial medications at baseline. Also, within the clinical trial context, there is increased attention and involvement of clinicians, thus high adherence rates may be an artefact of trial participation. Real-world, pragmatic trials in community and clinic settings are needed that examine both confirmed or unconfirmed adherence strategies that may increase adherence to iron chelation therapy. Due to lack of evidence this review cannot comment on intervention strategies for different age groups.


Subject(s)
Anemia, Sickle Cell , Drug-Related Side Effects and Adverse Reactions , Thalassemia , Child , Humans , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/drug therapy , Chelating Agents , Chelation Therapy , Deferoxamine/adverse effects , Iron
5.
Clin Neurol Neurosurg ; 227: 107634, 2023 04.
Article in English | MEDLINE | ID: mdl-36857886

ABSTRACT

BACKGROUND: Intracerebral hemorrhage (ICH) is a stroke with a high morbidity and mortality rate. Deferoxamine (DFX) is thought to be effective in treating Intracerebral Hemorrhage. In our study, we performed a meta-analysis to evaluate the treatment effects of DFX. METHODS: We systematically searched PubMed, Embase, Web of Science, the Cochrane Central Register of Controlled Trials, and Chinese Biomedical Literature Database in Jan 2022 for studies on DFX for ICH patients. Outcome measures included relative hematoma volume, relative edema volume, good neurological functional outcome and adverse events. Odds risk (OR) and weighted mean difference (WMD) were used to evaluate clinical outcomes. RESULTS: After searching 636 articles, 4 RCTs, 2 NRCTs, and 1cohort study were included. We found that DFX was effective in hematoma absorption on day 7 after onset, but the difference was not significant on day 14. DFX could suppress edema expansion on days 3, 7, and 14 after onset. DFX did not contribute to better outcomes after 3 and 6 months when used the modified Rankin Scale and the Glasgow Outcome Scale to evaluate neurological prognosis. The pooled results showed no statistically significant difference in Serious adverse events between the experimental and control groups. CONCLUSIONS: DFX could limit edema expansion on days 3, 7, and 14 after commencement and facilitate hematoma absorption at week 1 without significantly increasing the risk of adverse events, but it did not improve neurological prognosis.


Subject(s)
Deferoxamine , Stroke , Humans , Deferoxamine/adverse effects , Siderophores/pharmacology , Siderophores/therapeutic use , Cerebral Hemorrhage/drug therapy , Stroke/drug therapy , Hematoma/drug therapy
6.
Front Biosci (Elite Ed) ; 14(3): 18, 2022 07 12.
Article in English | MEDLINE | ID: mdl-36137990

ABSTRACT

Beta thalassaemia major (TM), a potentially fatal haemoglobinopathy, has transformed from a fatal to a chronic disease in the last 30 years following the introduction of effective, personalised iron chelation protocols, in particular the use of oral deferiprone, which is most effective in the removal of excess iron from the heart. This transition in TM has been achieved by the accessibility to combination therapy with the other chelating drugs deferoxamine and deferasirox but also therapeutic advances in the treatment of related co-morbidities. The transition and design of effective personalised chelation protocols was facilitated by the development of new non-invasive diagnostic techniques for monitoring iron removal such as MRI T2*. Despite this progress, the transition in TM is mainly observed in developed countries, but not globally. Similarly, potential cures of TM with haemopoietic stem cell transplantation and gene therapy are available to selected TM patients but potentially carry high risk of toxicity. A global strategy is required for the transition efforts to become available for all TM patients worldwide. The same strategy could also benefit many other categories of transfusional iron loaded patients including other thalassaemias, sickle cell anaemia, myelodysplasia and leukaemia patients.


Subject(s)
Iron Chelating Agents , Thalassemia , Benzoates/adverse effects , Deferasirox , Deferiprone/therapeutic use , Deferoxamine/adverse effects , Humans , Iron , Iron Chelating Agents/adverse effects , Iron Chelating Agents/therapeutic use , Pyridones/adverse effects , Risk Assessment , Thalassemia/chemically induced , Thalassemia/drug therapy , Triazoles/adverse effects , Triazoles/therapeutic use
7.
Drug Chem Toxicol ; 45(1): 93-102, 2022 Jan.
Article in English | MEDLINE | ID: mdl-31905029

ABSTRACT

We aimed to investigate the role of urinary kidney injury molecule-1 (KIM-1) in detection of subclinical nephrotoxicity in patients with Beta-thalassemia (ß-TM) in relation to chelation therapy and to correlate the urinary KIM-1 level with other clinical and laboratory findings. We conducted a cross-sectional study on 66 thalassemic patients. Their ages range from 7 to 22 years. Routine kidney indices and novel urinary KIM/creatinine ratio (UKIM-1/Cr) were measured. Estimated glomerular filtration rate (eGFR) was calculated. Results indicate that the level of serum creatinine was significantly higher in patients on deferasirox therapy than patients on deferoxamine and deferiprone therapy [median(IQR), 0.85(0.63-0.99), 0.50(0.34-0.58) and 0.44(0.36-0.45)] mg/dL, respectively, p < 0.001]. The median(IQR) level of eGFR was significantly lower in patients on deferasirox therapy than patients on deferoxamine and deferiprone therapy [63.3(56.5-92.1), 117.3(91.9-162) and 136.7(109.4-157.6)] ml/min/1.73 m2, respectively, p < 0.001]. The mean level of UKIM-1/Cr was significantly higher in patients on deferasirox therapy than patients on deferoxamine and deferiprone therapy (7.0 ± 1.9, 4.1 ± 1.7 and 4.2 ± 1.5) ng/mg creatinine, respectively, p < 0.001). We concluded that urinary KIM-1 is an early predictive biomarker for decline in eGFR in patients with ß-TM on deferasirox therapy. The appropriate chelation therapy and good monitoring of those patients are intensely needed for early detection of renal dysfunction and timely intervention.


Subject(s)
Iron Overload , beta-Thalassemia , Adolescent , Adult , Child , Cross-Sectional Studies , Deferasirox , Deferoxamine/adverse effects , Humans , Iron Chelating Agents/adverse effects , Kidney , Young Adult , beta-Thalassemia/complications , beta-Thalassemia/drug therapy
8.
Blood Adv ; 6(4): 1243-1254, 2022 02 22.
Article in English | MEDLINE | ID: mdl-34847228

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.


Subject(s)
Anemia, Sickle Cell , Iron Overload , Thalassemia , Adolescent , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/drug therapy , Blood Transfusion , Deferiprone/therapeutic use , Deferoxamine/adverse effects , Female , Humans , Iron Chelating Agents/adverse effects , Iron Overload/drug therapy , Iron Overload/etiology , Male , Pyridones/adverse effects , Thalassemia/complications , Thalassemia/drug therapy , Transferases
9.
Braz. J. Pharm. Sci. (Online) ; 58: e19409, 2022. graf
Article in English | LILACS | ID: biblio-1383963

ABSTRACT

Abstract Naringin has been shown to exhibit satisfying iron chelation capacity. Considering the side effects of routinely-used iron chelator (desferrioxamine, DFO), we decided to evaluate the iron chelation potency of naringin to discover whether or not it can be a promising natural substitute for treatment of excessive iron-related diseases. 35 mice were classified into five groups of 7 and subjected to iron dextran administration to induce the iron-overload condition. Iron-overloaded mice were then treated with normal saline (as control), naringin or DFO Morphology changes, and iron deposition in liver tissues were studied using H&E and Perl's staining. The results revealed that naringin is more potent than DFO in removing excessive iron ions deposited in liver tissues, indicating that naringin is a promising natural compound for therapy of iron overload disorders


Subject(s)
Animals , Male , Mice , Iron Overload/complications , Flavanones/analysis , Organization and Administration , Deferoxamine/adverse effects
10.
BMC Ophthalmol ; 21(1): 277, 2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34256738

ABSTRACT

BACKGROUND: Deferoxamine retinopathy is the informally designated term used to describe a characteristic pattern of outer retinal degeneration in iron-overloaded chronic anemia patients who are treated with deferoxamine. We hypothesize that insufficiently treated iron overloading and not only deferoxamine is the cause of the retinal degeneration. Our case report is based on exposure histories of two anemia patients and literature review. CASE PRESENTATION: Both anemia patients presented with bilateral visual loss secondary to photoreceptor and retinal pigment epithelium degeneration. Chart review showed that visual loss came after a year-long slow, and rather monotonous rise in plasma ferritin concentrations, with no obvious relation to iron chelator exposure. In one patient, the onset of symptomatic visual loss came after a bout of fever followed by two additional febrile episodes, all accompanied by plasma ferritin spikes. Adjustment of iron chelation therapy did not improve visual function. Experimental studies clearly show that both systemic and intraocular exposure to iron ions can induce retinal degeneration. CONCLUSION: The available evidence indicates that retinal degeneration in chronic anemia patients treated by deferoxamine is cause by insufficient iron chelation, not by deferoxamine. The actual role of iron chelating agents may be to promote a long enough survival to allow the slow development of retinal siderosis.


Subject(s)
Iron Overload , Retinal Degeneration , beta-Thalassemia , Deferoxamine/adverse effects , Humans , Iron Chelating Agents/adverse effects , Iron Overload/chemically induced , Iron Overload/complications , Iron Overload/drug therapy , Retinal Degeneration/chemically induced , Retinal Pigment Epithelium
11.
Rev. bras. oftalmol ; 80(4): e0026, 2021. graf
Article in English | LILACS | ID: biblio-1288636

ABSTRACT

ABSTRACT A 10-year-old Malay girl with underlying HbE/beta-thalassemia, on regular blood transfusion and deferoxamine iron chelation therapy, presented with two-month history of bilateral blurring of vision. On examination, her vision was 6/36 both eyes. Other optic nerve functions were normal. Anterior segment examination of both eyes was unremarkable. Fundus examination of both eyes revealed dull foveal reflex. Optical coherence tomography of both maculae showed increased central subfield thickness. Fundus fluorescence angiography showed patchy hypofluorescence over macular region for both eyes and late staining, indicating retinal pigment epithelium anomalies. A diagnosis of iron-chelation-therapy-related bilateral maculopathy was made. Patient was co-managed with pediatric hematology team to adjust the dose of deferoxamine, and was given three monthly appointments to monitor the progression of maculopathy at the ophthalmology clinic. However patient defaulted ophthalmology follow-up after the first visit.


RESUMO Uma menina malaia de 10 anos de idade com doença de base- B/beta-talassemia, em transfusão de sangue regular e terapia quelante de ferro deferoxamina, apresentou história de dois meses de visão turva bilateral. Ao exame, sua visão era de 6/36 em ambos os olhos. Outras funções do nervo óptico estavam normais. O exame do segmento anterior de ambos os olhos foi normal. Exame do fundo de ambos os olhos revelou reflexo foveal opaco. A tomografia de coerência óptica de ambas as máculas mostrou aumento da espessura do subcampo central. A angiografia de fluorescência do fundo mostrou hipofluorescência irregular sobre a região macular de ambos os olhos e coloração tardia, indicando anomalias de epitélio pigmentar da retina. Um diagnóstico de maculopatia bilateral relacionada à terapia quelante de ferro foi feito. A paciente foi avaliada em conjunto com a equipe de hematologia pediátrica para ajustar a dose de deferoxamina, e foram oferecidas três consultas mensais na clínica oftalmológica, para monitorar a progressão da maculopatia. No entanto, ela não compareceu para acompanhamento oftalmológico após a primeira visita.


Subject(s)
Humans , Female , Child , Siderophores/adverse effects , beta-Thalassemia/drug therapy , Deferoxamine/adverse effects , Transfusion Reaction , Macular Degeneration/complications , Blood Transfusion , Siderophores/therapeutic use , beta-Thalassemia/diagnosis , Deferoxamine/therapeutic use
12.
Cochrane Database Syst Rev ; 11: CD009280, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33236783

ABSTRACT

BACKGROUND: Stroke is the second leading cause of death and a major cause of morbidity worldwide. Retrospective clinical and animal studies have demonstrated neuroprotective effects of iron chelators in people with haemorrhagic or ischaemic stroke. This is the first update of the original Cochrane Review published in 2012. OBJECTIVES: To evaluate the effectiveness and safety of iron-chelating drugs in people with acute stroke. SEARCH METHODS: We searched the Cochrane Stroke Group Trials Register (2 September 2019), the Cochrane Central Register of Controlled Trials (CENTRAL) (The Cochrane Library 2019, Issue 9; 2 September 2019), MEDLINE Ovid (2 September 2019), Embase Ovid (2 September 2019), and Science Citation Index (2 September 2019). We also searched ongoing trials registers. SELECTION CRITERIA: We included randomised controlled trials (RCTs) of iron chelators versus no iron chelators or placebo for the treatment of acute stroke, including subarachnoid haemorrhage. DATA COLLECTION AND ANALYSIS: Two review authors independently screened the search results. We obtained the full texts of potentially relevant studies and evaluated them for eligibility. We assessed risk of bias using the Cochrane 'Risk of bias' tool, and the certainty of evidence using the GRADE approach. MAIN RESULTS: Two RCTs (333 participants) were eligible for inclusion; both compared the iron-chelating agent deferoxamine against placebo. Both studies evaluated participants with spontaneous intracerebral haemorrhage. We assessed one study to have a low risk of bias; the other study had potential sources of bias. The limited and heterogeneous data did not allow for meta-analysis of the outcome parameters. The evidence suggests that administration of deferoxamine may result in little to no difference in deaths (8% in placebo vs 8% in deferoxamine at 180 days; 1 RCT, 291 participants; low-certainty evidence). These RCTs suggest that there may be little to no difference in good functional outcome (modified Rankin Scale score 0 to 2) between groups at 30, 90 and 180 days (placebo vs deferoxamine: 67% vs 57% at 30 days and 36% vs 45% at 180 days; 2 RCTs, 333 participants; low-certainty evidence). One RCT suggests that administration of deferoxamine may not increase the number of serious adverse events or deaths (placebo vs deferoxamine: 33% vs 27% at 180 days; risk ratio 0.81, 95 % confidence interval 0.57 to 1.16; 1 RCT, 291 participants; low-certainty evidence). No data were available on any deaths within the treatment period. Deferoxamine may result in little to no difference in the evolution of National Institute of Health Stroke Scale scores from baseline to 90 days (placebo vs deferoxamine: 13 to 4 vs 13 to 3; P = 0.37; 2 RCTs, 333 participants; low-certainty evidence). Deferoxamine may slightly reduce relative oedema surrounding intracerebral haemorrhage at 15 days (placebo vs deferoxamine: 1.91 vs 10.26; P = 0.042; 2 RCTs, 333 participants; low-certainty evidence). Neither study reported quality of life. AUTHORS' CONCLUSIONS: We identified two eligible RCTs for assessment. We could not demonstrate any benefit for the use of iron chelators in spontaneous intracerebral haemorrhage. The added value of iron-chelating therapy in people with ischaemic stroke or subarachnoid haemorrhage remains unknown.


Subject(s)
Deferoxamine/therapeutic use , Hemorrhagic Stroke/drug therapy , Iron Chelating Agents/therapeutic use , Neuroprotective Agents/therapeutic use , Acute Disease , Bias , Deferoxamine/adverse effects , Hemorrhagic Stroke/mortality , Humans , Iron Chelating Agents/adverse effects , Neuroprotective Agents/adverse effects , Placebos/therapeutic use , Randomized Controlled Trials as Topic
13.
Int J Mol Sci ; 21(22)2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33233561

ABSTRACT

Thalassemia syndromes are characterized by the inability to produce normal hemoglobin. Ineffective erythropoiesis and red cell transfusions are sources of excess iron that the human organism is unable to remove. Iron that is not saturated by transferrin is a toxic agent that, in transfusion-dependent patients, leads to death from iron-induced cardiomyopathy in the second decade of life. The availability of effective iron chelators, advances in the understanding of the mechanism of iron toxicity and overloading, and the availability of noninvasive methods to monitor iron loading and unloading in the liver, heart, and pancreas have all significantly increased the survival of patients with thalassemia. Prolonged exposure to iron toxicity is involved in the development of endocrinopathy, osteoporosis, cirrhosis, renal failure, and malignant transformation. Now that survival has been dramatically improved, the challenge of iron chelation therapy is to prevent complications. The time has come to consider that the primary goal of chelation therapy is to avoid 24-h exposure to toxic iron and maintain body iron levels within the normal range, avoiding possible chelation-related damage. It is very important to minimize irreversible organ damage to prevent malignant transformation before complications set in and make patients ineligible for current and future curative therapies. In this clinical case-based review, we highlight particular aspects of the management of iron overload in patients with beta-thalassemia syndromes, focusing on our own experience in treating such patients. We review the pathophysiology of iron overload and the different ways to assess, quantify, and monitor it. We also discuss chelation strategies that can be used with currently available chelators, balancing the need to keep non-transferrin-bound iron levels to a minimum (zero) 24 h a day, 7 days a week and the risk of over-chelation.


Subject(s)
Deferoxamine/administration & dosage , Iron Chelating Agents/administration & dosage , Iron Overload/drug therapy , Iron/metabolism , Transfusion Reaction/complications , beta-Thalassemia/therapy , Adult , Blood Transfusion , Cardiomyopathies/blood , Cardiomyopathies/etiology , Cardiomyopathies/physiopathology , Cardiomyopathies/prevention & control , Chelation Therapy/adverse effects , Chelation Therapy/methods , Deferoxamine/adverse effects , Drug Monitoring/instrumentation , Drug Monitoring/methods , Female , Heart/drug effects , Heart/physiopathology , Humans , Iron/toxicity , Iron Chelating Agents/adverse effects , Iron Overload/blood , Iron Overload/complications , Iron Overload/physiopathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Middle Aged , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Transferrin/metabolism , Transfusion Reaction/blood , Transfusion Reaction/physiopathology , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
14.
Taiwan J Obstet Gynecol ; 59(1): 120-122, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32039778

ABSTRACT

OBJECTIVE: To report cases of use of chelation therapy during pregnancy which resulted in favorable outcomes for the babies. MATERIALS AND METHODS: In this retrospective cohort study, we described the evolution and outcome of 9 pregnancies in Italian thalassemic women who received deferoxamine (DFO) inadvertently during early pregnancy. RESULTS: The use of deferoxamine during first trimester did not lead to adverse effects on the fetus or cause major complications for the gestation, although an increase in iron burden was observed after suspending chelation therapy. CONCLUSION: In our experience, iron-chelation therapy might be administrated in pregnancy where the benefits to the mother outweigh the potential risks to the baby.


Subject(s)
Chelation Therapy/adverse effects , Deferoxamine/adverse effects , Maternal Exposure/adverse effects , Pregnancy Complications, Hematologic/drug therapy , Siderophores/adverse effects , beta-Thalassemia/drug therapy , Adult , Deferoxamine/administration & dosage , Female , Humans , Live Birth , Maternal-Fetal Exchange/drug effects , Pregnancy , Pregnancy Trimester, First , Retrospective Studies , Siderophores/administration & dosage
15.
Ear Nose Throat J ; 98(8): NP125-NP130, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31142160

ABSTRACT

Ocular and ophthalmological adverse effects may be seen in ß-thalassemia major (BTM) patients treated with regular blood transfusions and iron-chelating agents. We hypothesized that olfactory dysfunction may be present in this population. In this study, we aimed to investigate olfactory dysfunction in patients with BTM and determine the etiological factors. A total of 43 patients with BTM were included in the study. Forty-three patients without any nasal complaints, history of facial trauma, or nasal surgery were included as the controls. All participants had nasal endoscopy. The iron-chelating agents used, their duration of use, as well as hemoglobin and ferritin levels of the BTM patients were recorded. Sniffin' Sticks test (SST) was used to assess olfactory functions, and BTM and control groups were compared for the results. The correlations of SST scores with the other study parameters were analyzed. Eight (18.6%) of 43 patients in the BTM group had hyposmia while none of the patients in the control group had hyposmia (P < .001). Older age, low-hemoglobin level, and longer use of deferoxamine were found to be correlated with olfactory dysfunction. Olfactory dysfunction may be seen in BTM patients treated with iron-chelating agents. The results of this study suggest that screening for olfactory function may be needed in routine follow-up of BTM patients.


Subject(s)
Deferoxamine/therapeutic use , Iron Chelating Agents/therapeutic use , Olfaction Disorders/etiology , beta-Thalassemia/blood , beta-Thalassemia/drug therapy , Adolescent , Adult , Age Factors , Deferoxamine/adverse effects , Female , Ferritins/blood , Hemoglobins/metabolism , Humans , Iron Chelating Agents/adverse effects , Male , Young Adult , beta-Thalassemia/complications
16.
Lancet Neurol ; 18(5): 428-438, 2019 05.
Article in English | MEDLINE | ID: mdl-30898550

ABSTRACT

BACKGROUND: Iron from haemolysed blood is implicated in secondary injury after intracerebral haemorrhage. We aimed to assess the safety of the iron chelator deferoxamine mesylate in patients with intracerebral haemorrhage and to establish whether the drug merits investigation in a phase 3 trial. METHODS: We did a multicentre, futility-design, randomised, placebo-controlled, double-blind, phase 2 trial at 40 hospitals in Canada and the USA. Adults aged 18-80 years with primary, spontaneous, supratentorial intracerebral haemorrhage were randomly assigned (1:1) to receive deferoxamine mesylate (32 mg/kg per day) or placebo (saline) infusions for 3 consecutive days within 24 h of haemorrhage onset. Randomisation was done via a web-based trial-management system centrally in real time, and treatment allocation was concealed from both participants and investigators. The primary outcome was good clinical outcome, which was defined as a modified Rankin Scale score of 0-2 at day 90. We did a futility analysis: if the 90% upper confidence bound of the absolute risk difference between the two groups in the proportion of participants with a good clinical outcome was less than 12% in favour of deferoxamine mesylate, then to move to a phase 3 efficacy trial would be futile. Primary outcome and safety data were analysed in the modified intention-to-treat population, comprising only participants in whom the study infusions were initiated. This trial is registered with ClinicalTrials.gov, number NCT02175225, and is completed. FINDINGS: We recruited 294 participants between Nov 23, 2014, and Nov 10, 2017. The modified intention-to-treat population consisted of 144 patients assigned to the deferoxamine mesylate group and 147 assigned to the placebo group. At day 90, among patients with available data for the primary outcome, 48 (34%) of 140 participants in the deferoxamine mesylate group, and 47 (33%) of 143 patients in the placebo group, had modified Rankin Scale scores of 0-2 (adjusted absolute risk difference 0·6% [90% upper confidence bound 6·8%]). By day 90, 70 serious adverse events were reported in 39 (27%) of 144 patients in the deferoxamine mesylate group, and 78 serious adverse events were reported in 49 (33%) of 147 patients in the placebo group. Ten (7%) participants in the deferoxamine mesylate and 11 (7%) in the placebo group died. None of the deaths were judged to be treatment related. INTERPRETATION: Deferoxamine mesylate was safe. However, the primary result showed that further study of the efficacy of deferoxamine mesylate with anticipation that the drug would significantly improve the chance of good clinical outcome (ie, mRS score of 0-2) at day 90 would be futile. FUNDING: US National Institutes of Health and US National Institute of Neurological Disorders and Stroke.


Subject(s)
Cerebral Hemorrhage/drug therapy , Deferoxamine/therapeutic use , Iron Chelating Agents/therapeutic use , Aged , Deferoxamine/adverse effects , Double-Blind Method , Female , Humans , Infusions, Intravenous , Iron Chelating Agents/adverse effects , Male , Medical Futility , Middle Aged , Negative Results , Prospective Studies , Risk Assessment , Treatment Outcome
17.
Ann Nucl Med ; 33(2): 119-127, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30406361

ABSTRACT

OBJECTIVE: We conducted the present phase I/IIa positron emission tomography (PET) imaging study with 89Zr conjugated with desferroxamine-IAB2M (89Zr-Df-IAB2M), an anti-prostate-specific membrane-antigen minibody, to assess its safety and feasibility in patients with urological cancer. METHODS: 89Zr-Df-IAB2M was synthetized by IBA Molecular (Somerset, NJ, USA) and transported by air to Tsukuba Molecular Imaging Center (Tsukuba, Ibaraki, Japan).17 patients received 74 MBq (2 mCi) of 89Zr-Df-IAB2M at total mass doses of 10 mg. Whole-body and plasma clearance, normal-organ and lesion uptake, and radiation absorbed dose were estimated. We also preliminarily tested the performance of 89Zr-immuno-PET imaging for 13 patients with prostate cancer and 4 patients with other urological cancer. RESULTS: The administration of 89Zr-Df-IAB2M was well-tolerated, and no infusion-related reactions were observed in any patient. No adverse events were noted in the laboratory parameters, vital signs, or other parameters. The plasma clearance was biphasic, with an initial rapid phase (t 1/2 fast: 10.1 ± 3.4 h) followed by a slow phase (t 1/2 slow: 49.0 ± 22.7 h). The half-life of radioactivity in the whole body (WB t1/2) was 237 ± 9 h. The highest absorbed radiation dose was 1.67 mGy/MBq, observed in the liver and kidney. The effective dose was 0.68 ± 0.08 mSv/MBq. The radiation dose rate at 0.5 m distance from the patient was 8.67 µSv/h on day 1, and decreased to 2.26 µSv/h at 5 days after injection. Both bone and lymph node metastases were detected with 89Zr-Df-IAB2M by 24 or 48 h imaging. CONCLUSIONS: Administration of 89Zr-Df-IAB2M was well-tolerated and safe in terms of adverse events and radiation exposure and protection. 89Zr-Df-IAB2M is feasible for usage by long-distance transportation. Further studies are warranted for analysis of its use for tumor lesion detection (UMIN000015356).


Subject(s)
Deferoxamine , Positron-Emission Tomography , Radioisotopes , Urologic Neoplasms/diagnostic imaging , Zirconium , Aged , Deferoxamine/adverse effects , Feasibility Studies , Female , Humans , Isotope Labeling , Male , Middle Aged , Safety
18.
BMC Ophthalmol ; 18(1): 246, 2018 Sep 12.
Article in English | MEDLINE | ID: mdl-30208862

ABSTRACT

BACKGROUND: Deferoxamine (DFO) is one of the most commonly used chelation treatments for transfusional hemosiderosis. Pattern dystrophies constitute a distinct entity of retinal disorders that has been occasionally identified in association with deferoxamine. CASE PRESENTATION: We report two cases of bilateral macular pattern dystrophy in transfusion dependent patients undergoing chronic chelation therapy with deferoxamine due to thalassemias. Our patients were evaluated with multimodal imaging and the results are presented. Both patients had normal cone and rod responses in the full-field electroretinogram and continued the prescribed chelation therapy, after hematology consult. The patients were followed up every 3 months for 2 and 4 years respectively for possible deterioration. Their best corrected visual acuity remained stable with no anatomic change on Optical Coherence Tomography findings. CONCLUSION: Multimodal imaging of our patients allowed a better evaluation and possibly earlier detection of the DFO-related changes. Screening and close follow up of patients under chronic chelating therapy is important in order to promptly diagnose and manage possible toxicity either with discontinuation of the offending agent or dose modification.


Subject(s)
Deferoxamine/adverse effects , Retina/diagnostic imaging , Retinal Degeneration/chemically induced , Thalassemia/drug therapy , Deferoxamine/administration & dosage , Electroretinography , Female , Humans , Infusions, Subcutaneous/adverse effects , Middle Aged , Retinal Degeneration/diagnosis , Siderophores/administration & dosage , Siderophores/adverse effects , Tomography, Optical Coherence
19.
Expert Rev Clin Pharmacol ; 11(6): 641-650, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29727586

ABSTRACT

BACKGROUND: To compare the efficacy and safety of desferrioxamine (DFO), deferiprone (DFP), deferasirox (DFX) and silymarin in patients with either thalassemia or sickle cell disorder through network meta-analysis. METHODS: Electronic databases were searched for appropriate randomized clinical trials comparing iron chelators in patients with iron overload. Random effects model was used to generate direct, indirect and mixed treatment comparison pooled estimates for the following outcomes: serum ferritin, liver iron concentration (LIC), changes in serum ferritin, mortality, urine iron excretion, adverse events, neutropenia, agranulocytosis and number of patients withdrawing the chelating therapy. RESULTS: Thirty-two clinical trials were included in the meta-analysis. DFX/DFO was associated with better serum ferritin levels compared to DFO, DFX, DFO/Silymarin and DFP/DFO. DFX/DFO also lower LIC significantly compared to DFO. DFP/DFO was associated with higher LVEF, low risk of adverse events and reduced end of serum ferritin compared to DFO. Combination of silymarin with either DFP or DFX was observed with reduced end of treatment serum ferritin compared to using either of the drugs alone. DFP was observed with better effects in sickle cell disease. The strength of evidence was very low for most of the comparisons. CONCLUSION: Relative estimates between the individual iron chelators have been established. However, this evidence should be considered preliminary and may change with the results of future head-to-head clinical trials.


Subject(s)
Anemia, Sickle Cell/drug therapy , Iron Chelating Agents/therapeutic use , Thalassemia/drug therapy , Benzoates/administration & dosage , Benzoates/adverse effects , Benzoates/therapeutic use , Deferasirox , Deferiprone , Deferoxamine/administration & dosage , Deferoxamine/adverse effects , Deferoxamine/therapeutic use , Drug Therapy, Combination , Humans , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/adverse effects , Iron Overload , Network Meta-Analysis , Pyridones/administration & dosage , Pyridones/adverse effects , Pyridones/therapeutic use , Randomized Controlled Trials as Topic , Silymarin/administration & dosage , Silymarin/adverse effects , Silymarin/therapeutic use , Triazoles/administration & dosage , Triazoles/adverse effects , Triazoles/therapeutic use
20.
Am J Hematol ; 93(7): 943-952, 2018 07.
Article in English | MEDLINE | ID: mdl-29635754

ABSTRACT

Red blood cell transfusions have become standard of care for the prevention of life-threatening anemia in patients with ß-thalassemia and sickle cell disease (SCD). However, frequent transfusions can lead to accumulation of iron that can result in liver cirrhosis, diabetes mellitus, arthritis, arrhythmias, cardiomyopathy, heart failure, and hypogonadotropic hypogonadism. Iron chelation therapy has been shown to reduce serum ferritin levels and liver iron content, but limitations of trial design have prevented any demonstration of improved survival. The objective of this systematic review was to investigate the impact of iron chelation therapy on overall and event-free survival in patients with ß-thalassemia and SCD. Eighteen articles discussing survival in ß-thalassemia and 3 in SCD were identified. Overall iron chelation therapy resulted in better overall survival, especially if it is instituted early and compliance is maintained. Comparative studies did not show any significant differences between available iron chelation agents, although there is evidence that deferiprone is better tolerated than deferoxamine and that compliance is more readily maintained with the newer oral drugs, deferiprone and deferasirox. Iron chelation therapy, particularly the second-generation oral agents, appears to be associated with improved overall and event-free survival in transfusion-dependent patients with ß-thalassemia and patients with SCD.


Subject(s)
Anemia, Sickle Cell/drug therapy , Iron Chelating Agents/therapeutic use , beta-Thalassemia/drug therapy , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/mortality , Blood Transfusion , Deferiprone/adverse effects , Deferiprone/therapeutic use , Deferoxamine/adverse effects , Deferoxamine/therapeutic use , Humans , Iron Chelating Agents/adverse effects , Medication Adherence , Survival Analysis , beta-Thalassemia/mortality
SELECTION OF CITATIONS
SEARCH DETAIL
...