Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Pediatr Blood Cancer ; 71(8): e31061, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38840429

ABSTRACT

Chronic hemolytic anemia and vascular occlusion are hallmarks of sickle cell disease (SCD). Blood transfusions are critical for supportive and preventive management of SCD complications. Patients with SCD are at risk for hyperhemolysis syndrome (HHS), a subtype of delayed hemolytic transfusion reactions. HHS management includes intravenous immunoglobulin, corticosteroids, and avoidance of further transfusions. Not all patients respond to first-line agents. Eculizumab, which blocks terminal complement activation, has been proposed as second-line management of HHS. We describe two patients who received eculizumab for refractory HHS. In our experience, eculizumab is a safe and effective option for refractory pediatric HHS.


Subject(s)
Anemia, Sickle Cell , Antibodies, Monoclonal, Humanized , Humans , Antibodies, Monoclonal, Humanized/therapeutic use , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/drug therapy , Male , Female , Child , Hemolysis/drug effects , Adolescent , Child, Preschool , Transfusion Reaction/drug therapy
2.
Medicine (Baltimore) ; 100(44): e27724, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34871268

ABSTRACT

RATIONALE: An allergic transfusion reaction is a common side effect of transfusions of red blood cells. Using washed red blood cells is the most effective method for preventing such a reaction. However, the availability of other washed transfusion components, including platelets, is limited. PATIENT CONCERNS: A 69-year-old patient with acute myeloid leukemia progressed from myelodysplastic syndrome and was treated with azacitidine. She experienced a minor reaction to platelet transfusion that initially responded to the administration of corticosteroids and antihistamines. However, she worsened even after subsequent preventive treatments and was referred to the emergency department due to anaphylaxis. The patient developed hypotension, chest pain, and dyspnea 10 minutes after the initiation of platelet transfusion. DIAGNOSES: She was diagnosed with platelet-induced anaphylaxis. INTERVENTIONS: In an attempt to prevent anaphylaxis, 150 mg of omalizumab was prescribed 1 week prior to transfusion. However, she experienced anaphylaxis again and was administered intramuscular epinephrine. For the following transfusion, we treated her with a 300 mg dose of omalizumab 24 hours before the transfusion. OUTCOMES: She tolerated well and continued to receive further chemotherapy and platelet transfusion with premedication. LESSONS: This case suggests that omalizumab is a good candidate for the management of severe allergic transfusion reactions.


Subject(s)
Anaphylaxis , Omalizumab/therapeutic use , Platelet Transfusion/adverse effects , Transfusion Reaction/drug therapy , Aged , Blood Transfusion , Female , Humans
4.
Transfus Apher Sci ; 60(2): 103041, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33455877

ABSTRACT

BACKGROUND: Anti-allergic agents (e.g. dexamethasone, chlorpheniramine or promethazine) are commonly administered to patients prior to blood product transfusions. However, the use of these agents is largely experience-based instead of evidence-based. This meta-analysis aimed to explore the evidence behind using anti-allergic agents to attenuate transfusion reactions. MATERIALS AND METHODS: The Pubmed, EMBASE, Cochrane Library, Wanfang, Chinese National Knowledge Infrastructure (CNKI), and Chinese Biomedical literature (CMB) databases were all queried for related articles. Data from groups treated with and without anti-allergic agents were collected for meta-analysis using RevMan 5.3. Baseline characteristics and univariate statistics between groups were compared using SPSS 19.0. RESULTS: Eight eligible articles (six case control studies and two randomized controlled trials, all with high risks of bias) were identified (22060 total cases). Administered anti-allergic agents in these studies only included dexamethasone, chlorpheniramine or promethazine. Baseline characteristics showed no significant age or gender differences between treatment or control groups. There were no significant differences between the pooled experimental or control groups (for each of the three medications) in terms of fever, pruritis, rash, airway spasm or overall transfusion reaction rates. CONCLUSION: There is no evidence that dexamethasone, chlorpheniramine or promethazine can prevent transfusion reactions. Avoiding the arbitrary use of such anti-allergic agents before blood transfusions may potentially avoid needless adverse drug reactions.


Subject(s)
Anti-Allergic Agents/therapeutic use , Blood Transfusion/methods , Transfusion Reaction/drug therapy , Adult , Anti-Allergic Agents/pharmacology , Female , Humans , Male , Middle Aged
5.
Hematology Am Soc Hematol Educ Program ; 2020(1): 160-166, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33275743

ABSTRACT

The introduction of regular red cell transfusions 60 years ago transformed ß-thalassemia major from a fatal childhood illness into a chronic disorder. Further advances in the prevention of transfusion-transmitted infections and management of iron overload have allowed survival and quality of life to approach normal. However, transfusion therapy for some other thalassemia syndromes continues to challenge clinical decision-making. Nearly one-half of the patients with E ß thalassemia are transfusion-dependent, yet the criteria for initiating transfusions or hemoglobin targets are not well defined. Patients with thalassemia intermedia who begin transfusions as adults are at very high risk for developing red cell alloimmunization and serious hemolytic transfusion reactions. In the growing number of survivors of Bart hydrops fetalis, the approach to transfusion therapy and iron chelation is rapidly evolving. A collaboration between hematology and transfusion medicine specialists will be essential to improving patient care and developing evidence-based guidelines.


Subject(s)
Blood Transfusion , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Quality of Life , Transfusion Reaction/drug therapy , beta-Thalassemia/therapy , Child , Child, Preschool , Female , Humans , Iron Overload/etiology , Male
7.
Transfus Med Rev ; 34(3): 172-177, 2020 07.
Article in English | MEDLINE | ID: mdl-32703665

ABSTRACT

Hyperhemolytic transfusion reaction (HHTR) is a severe, life-threatening hemolytic transfusion reaction where hemoglobin value after red blood cell (RBC) transfusion is lower than the pre-transfusion value. When HHTR occurs, mainly in patients with hemoglobinopathy, complement activation up to membrane attack complex (MAC) is strongly suspected. However, our knowledge of HHTR in patients without hemoglobinopathy is limited. In the present study, we retrospectively reviewed patients with the diagnosis of HHTR who were attended at our hospital between 2013 and 2016. We also performed a literature search to identify other reported cases of HHTR. Finally, the role of terminal complement pathway activation in the pathogenesis of HHTR was assessed by exposing endothelial cells in vitro to activated-patient plasma to analyze C5b-9 deposits by immunofluorescence. HHTR was diagnosed in 3 patients according to current criteria. Patients were treated with intravenous immunoglobulins (alone or in conjunction with rituximab and plasma exchange), and all of them recovered successfully. We retrieved from literature search 10 patients without hemoglobinopathy who developed HHTR. A marked increase of C5b-9 (MAC) deposition on endothelial cells (almost 2.5-fold increase versus control, P < .05) was observed with the plasma sample obtained from one of our patients. In conclusion, HHTR was a rare transfusion reaction that occurred in patients without hemoglobinopathy. We add more evidence that complement cascade activation up to MAC might play a role in the pathogenesis of HHTR.


Subject(s)
Complement Activation , Hemolysis/immunology , Transfusion Reaction/immunology , Aged , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Factors/therapeutic use , Male , Middle Aged , Retrospective Studies , Transfusion Reaction/diagnosis , Transfusion Reaction/drug therapy , Transfusion Reaction/etiology
8.
Trends Mol Med ; 26(7): 683-697, 2020 07.
Article in English | MEDLINE | ID: mdl-32589936

ABSTRACT

Hemolysis and accumulation of cell-free hemoglobin (Hb) in the circulation or in confined tissue compartments such as the subarachnoid space is an important driver of disease. Haptoglobin is the Hb binding and clearance protein in human plasma and an efficient antagonist of Hb toxicity resulting from physiological red blood cell turnover. However, endogenous concentrations of haptoglobin are insufficient to provide protection against Hb-driven disease processes in conditions such as sickle cell anemia, sepsis, transfusion reactions, medical-device associated hemolysis, or after a subarachnoid hemorrhage. As a result, there is increasing interest in developing haptoglobin therapeutics to target 'toxic' cell-free Hb exposures. Here, we discuss key concepts of Hb toxicity and provide a perspective on the use of haptoglobin as a therapeutic protein.


Subject(s)
Haptoglobins/pharmacology , Haptoglobins/therapeutic use , Hemoglobins/toxicity , Anemia, Sickle Cell/drug therapy , Animals , Erythrocytes/drug effects , Hemolysis/drug effects , Humans , Sepsis/drug therapy , Transfusion Reaction/drug therapy
9.
Transfusion ; 60(1): 30-35, 2020 01.
Article in English | MEDLINE | ID: mdl-31642065

ABSTRACT

BACKGROUND: Hyperhemolysis syndrome (HHS) is a posttransfusion complication most frequently seen in sickle cell disease (SCD), characterized by rapid destruction of transfused and autologous red blood cells (RBCs), resulting in reticulocytopenia and a decrease in hemoglobin to below pretransfusion levels. Additional RBC transfusion can be life threatening. Most patients improve with intravenous immune globulin and steroids, but in refractory cases, hyperhemolysis may result in multiorgan failure and death in the absence of salvage therapy. The exact pathophysiology of HHS remains uncertain, yet new insights suggest that RBC destruction is driven by activated macrophages. Therefore, we propose that antimacrophage therapy may represent an effective treatment. CASE REPORT: A case of life-threatening HHS, refractory to intravenous immune globulin and steroids, in a patient with SCD is presented. Marked elevation in ferritin, an indirect marker of macrophage activation, a negative direct antiglobulin test, and the absence of RBC alloantibodies was noted. A hemoglobin nadir of 2.1 g/dL and resultant hypoxemia-induced organ failure prompted the use of tocilizumab, an interleukin-6 receptor monoclonal antibody. Hemoglobin-based oxygen carrier-201, a cell-free polymerized bovine hemoglobin, was used to support the patient during critical anemia. RESULTS: Hemolysis resolved and ferritin dramatically decreased after administration of tocilizumab, which was well tolerated. A full recovery was achieved. CONCLUSION: This case highlights both a novel and successful approach to managing refractory transfusion-induced hyperhemolysis with tocilizumab and provides further evidence supporting the role for macrophage activation in the destruction of RBCs in antibody-negative HHS. We propose that tocilizumab is an effective and rapid salvage therapy for refractory HHS.


Subject(s)
Anemia, Sickle Cell , Antibodies, Monoclonal, Humanized/administration & dosage , Erythrocyte Transfusion/adverse effects , Hemolysis/drug effects , Macrophage Activation/drug effects , Macrophages , Transfusion Reaction , Adult , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/therapy , Blood Substitutes , Female , Humans , Macrophages/pathology , Transfusion Reaction/blood , Transfusion Reaction/drug therapy , Transfusion Reaction/etiology
10.
Transfusion ; 60(2): 430-435, 2020 02.
Article in English | MEDLINE | ID: mdl-31859413

ABSTRACT

BACKGROUND: Vancomycin-resistant enterococci (VRE) are antibiotic-resistant organisms associated with both colonization and serious life-threatening infection in health care settings. Contamination of platelet concentrates (PCs) with Enterococcus can result in transfusion-transmitted infection. CASE PRESENTATION: This report describes the investigation of a septic transfusion case involving a 27-year-old male patient with relapsed acute leukemia who was transfused with a 5-day-old buffy coat PC pool and developed fever and rigors. DISCUSSION: Microbiology testing and pulse-field gel electrophoresis (PFGE) was done on patient blood cultures obtained from peripheral and central lines. Microbiology and molecular testing were also performed on the remaining posttransfusion PC pool, which was refrigerated for 24 hours before microbiology testing. Red blood cell (RBC) and plasma units associated with the implicated PCs were screened for microbial contamination. Patient blood cultures obtained from peripheral and central lines yielded vancomycin-resistant Enterococcus faecium. Gram stain of a sample from the platelet pool was negative but coagulase-negative Staphylococcus (CNST) and VRE were isolated on culture. Antibiotic sensitivity and PFGE profiles of several VRE isolates from the patient before and after transfusion, and the PC pool, revealed that all were closely related. Associated RBC and plasma components tested negative for microbial contamination. CONCLUSIONS: Microbiological and molecular investigations showed a relationship between VRE isolated from the patient before and after transfusion, and therefore it is postulated that a patient-to-PC retrograde contamination (from either blood or skin) occurred. As the CNST isolated from the PC pool was not isolated from patient samples, its implication in the transfusion event is unknown.


Subject(s)
Enterococcus faecium/pathogenicity , Transfusion Reaction/diagnosis , Transfusion Reaction/microbiology , Vancomycin-Resistant Enterococci/pathogenicity , Adult , Anti-Bacterial Agents/therapeutic use , Enterococcus faecium/drug effects , Humans , Male , Microbial Sensitivity Tests , Transfusion Reaction/drug therapy , Vancomycin-Resistant Enterococci/drug effects
11.
Transfus Med Rev ; 33(4): 225-230, 2019 10.
Article in English | MEDLINE | ID: mdl-31672341

ABSTRACT

Transfusion-related hemolysis is classically the result of an interaction between antibodies produced by the recipient and blood group antigens carried by the donor red blood cells. This reaction may be life threatening, especially in sickle cell patients when they develop hyperhemolysis with concomitant accelerated clearance of their own red blood cells. The complement system is a key participant in the pathophysiology of post-transfusion hemolysis. Complement can trigger the hemolytic reaction, amplify the inflammatory response and increase tissue damage. Complement is activated by the classical pathway but may also be activated by the alternative pathway in sickle cell disease. The hemolysis-derived products permanently released by sickle cell patients with chronic hemolytic anemia may affect the potency of complement activation. All the observations in sickle cell patients as well as in vitro experiments and in vivo data in animal models support the conclusion that complement is key disease driver and a promising therapeutic target in the context of transfusion-related hemolysis and hyperhemolysis.


Subject(s)
Complement System Proteins/physiology , Hemolysis/immunology , Transfusion Reaction/immunology , Anemia, Hemolytic/immunology , Anemia, Sickle Cell/immunology , Anemia, Sickle Cell/therapy , Animals , Blood Group Incompatibility/immunology , Complement Activation/physiology , Complement Inactivating Agents/therapeutic use , Complement System Proteins/immunology , Erythrocytes/immunology , Humans , Isoantibodies/blood , Transfusion Reaction/drug therapy , Transfusion Reaction/physiopathology
12.
Tunis Med ; 97(1): 149-152, 2019 Jan.
Article in English | MEDLINE | ID: mdl-31535708

ABSTRACT

Since the elimination of indigenous transmission of malaria in Tunisia in 1979, almost all the cases observed are imported cases related to travel. We report a recent case of highly probable post-transfusion malaria (PTM) in a 27-year-old Tunisian who has never left Tunisia. He has been allografted and has received of the globular pellets and the platelet units along with his hospitalization. The evolution was marked by the appearance of a fever resistant to antibiotics 15 days later. On day 11 of fever, a thick drop (TD) and a blood smear (BS) showed trophozoites of Plasmodium falciparum with 20% parasitaemia. The evolution was favorable under quinine. The epidemiological survey concluded that among blood donors an African donor from Ivory Coast, in Tunisia for 2 months, had a TD, a BS, a rapid test and a nested PCR for P. falciparum species were negative, only the serology was positive by indirect immunofluorescence (1/20). Real-time PCR was positive for P. falciparum, and the diagnosis of highly probable PTM was retained. Blood transfusion is a transmission pathway for Plasmodium and contamination can occur with a very few parasites. As a result, the PTM must be considered for any unexplained fever arising in the aftermath of a blood transfusion that and establish strict prevention recommendations for PTM in our country.


Subject(s)
Malaria, Falciparum/diagnosis , Malaria, Falciparum/etiology , Transfusion Reaction/diagnosis , Adult , Antimalarials/therapeutic use , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/transmission , Male , Plasmodium falciparum/isolation & purification , Transfusion Reaction/drug therapy , Transfusion Reaction/epidemiology , Tunisia/epidemiology
14.
Transfus Clin Biol ; 26(2): 130-134, 2019 May.
Article in English | MEDLINE | ID: mdl-30979566

ABSTRACT

Red blood cell (RBC) transfusion support represents a critical component of sickle cell disease (SCD) management. However, as with any therapeutic intervention, RBC transfusion is not without risk. Repeat exposure to allogeneic RBCs can result in the development of RBC alloantibodies that can make it difficult to find compatible RBCs for future transfusions and can directly increase the risk of developing acute or delayed hemolytic transfusion reactions, which can be further complicated by hyperhemolysis. Several prophylactic and treatment strategies have been employed in an effort to reduce or prevent hemolytic transfusion reactions. However, conflicting data exist regarding the efficacy of many of these approaches. We will explore the challenges associated with predicting, preventing and treating different types of hemolytic transfusion reactions in patients with SCD in addition to describing future strategies that may aid in the management of the complex transfusion requirements of SCD patients.


Subject(s)
Anemia, Sickle Cell/therapy , Erythrocyte Transfusion/adverse effects , Transfusion Reaction/prevention & control , Adrenal Cortex Hormones/therapeutic use , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Blood Group Antigens/immunology , Blood Group Incompatibility/complications , Blood Grouping and Crossmatching , Bortezomib/therapeutic use , Erythrocytes/immunology , Hemolysis , Humans , Immunoglobulins, Intravenous/therapeutic use , Isoantibodies/immunology , Transfusion Reaction/drug therapy , Transfusion Reaction/etiology
15.
Curr Opin Hematol ; 26(3): 139-144, 2019 05.
Article in English | MEDLINE | ID: mdl-30855336

ABSTRACT

PURPOSE OF REVIEW: For individuals who have transfusion-dependent anemia, iron overload is the long-term complication, which results in significant morbidity. Ameliorating this is now the biggest unmet need. This review specifically addresses this issue. RECENT FINDINGS: Over the last decade or so, major advances in the treatment of these individuals, has resulted from novel strategies aimed at reducing transfusion requirement as well as optimizing chelation therapy. This review will summarize these advances and provide insights into some of the therapies in the pipeline. Strategies aimed at reducing transfusion requirement include modulation of erythropoietic regulation by reducing ineffective red cell production through activin trapping, as well as stem cell gene modification approaches, which aim for a cure, and transfusion independence. Refined means of assessing tissue iron and the introduction of oral chelators have facilitated tailoring chelation regimens with closer monitoring and improved compliance. Newer approaches to ameliorate iron toxicity have focused on the hepcidin pathway, all of which would result in increased hepcidin levels and reduction of iron absorption from the intestine, sequestration of iron in normal storage sites and reduced exposure of more susceptible organs, such as the heart and endocrine organs, to the toxic effects of increased iron. SUMMARY: These advances offer the promise of improved management of transfusion-dependent individuals.


Subject(s)
Blood Transfusion , Iron Chelating Agents/therapeutic use , Iron Overload/drug therapy , Iron/metabolism , Transfusion Reaction/drug therapy , Administration, Oral , Gene Expression Regulation/drug effects , Hepcidins/metabolism , Humans , Iron Overload/etiology , Iron Overload/metabolism , Iron Overload/pathology , Transfusion Reaction/metabolism , Transfusion Reaction/pathology
17.
PLoS One ; 14(2): e0211942, 2019.
Article in English | MEDLINE | ID: mdl-30811439

ABSTRACT

BACKGROUND: Iron overload, resulting from blood transfusions in patients with chronic anemias, has historically been controlled with regular deferoxamine, but its parenteral requirement encouraged studies of orally-active agents, including deferasirox and deferiprone. Deferasirox, licensed by the US Food and Drug Administration in 2005 based upon the results of randomized controlled trials, is now first-line therapy worldwide. In contrast, early investigator-initiated trials of deferiprone were prematurely terminated after investigators raised safety concerns. The FDA declined market approval of deferiprone; years later, it licensed the drug as "last resort" therapy, to be prescribed only if first-line drugs had failed. We undertook to evaluate the long-term effectiveness and toxicities of deferiprone and deferasirox in one transfusion clinic. METHODS AND FINDINGS: Under an IRB-approved study, we retrospectively inspected the electronic medical records of consented iron-loaded patients managed between 2009 and 2015 at The University Health Network (UHN), Toronto. We compared changes in liver and heart iron, adverse effects and other outcomes, in patients treated with deferiprone or deferasirox. RESULTS: Although deferiprone was unlicensed in Canada, one-third (n = 41) of locally-transfused patients had been switched from first-line, licensed therapies (deferoxamine or deferasirox) to regimens of unlicensed deferiprone. The primary endpoint of monitoring in iron overload, hepatic iron concentration (HIC), increased (worsened) during deferiprone monotherapy (mean 10±2-18±2 mg/g; p < 0.0003), exceeding the threshold for life-threatening complications (15 mg iron/g liver) in 50% patients. During deferasirox monotherapy, mean HIC decreased (improved) (11±1-6±1 mg/g; p < 0.0001). Follow-up HICs were significantly different following deferiprone and deferasirox monotherapies (p < 0.0000002). Addition of low-dose deferoxamine (<40 mg/kg/day) to deferiprone did not result in reductions of HIC to <15 mg/g (baseline 20±4 mg/g; follow-up, 18±4 mg/g; p < 0.2) or in reduction in the proportion of patients with HIC exceeding 15 mg/g (p < 0.2). During deferiprone exposure, new diabetes mellitus, a recognized consequence of inadequate iron control, was diagnosed in 17% patients, most of whom had sustained HICs exceeding 15 mg/g for years; one woman died after 13 months of a regimen of deferiprone and low-dose deferasirox. During deferiprone exposure, serum ALT increased over baseline in 65% patients. Mean serum ALT increased 6.6-fold (p < 0.001) often persisting for years. During deferasirox exposure, mean ALT was unchanged (p < 0.84). No significant differences between treatment groups were observed in the proportions of patients estimated to have elevated cardiac iron. CONCLUSIONS: Deferiprone showed ineffectiveness and significant toxicity in most patients. Combination with low doses of first-line therapies did not improve the effectiveness of deferiprone. Exposure to deferiprone, over six years while the drug was unlicensed, in the face of ineffectiveness and serious toxicities, demands review of the standards of local medical practice. The limited scope of regulatory approval of deferiprone, worldwide, should restrict its exposure to the few patients genuinely unable to tolerate the two effective, first-line therapies.


Subject(s)
Administration, Oral , Deferasirox , Electronic Health Records , Iron Chelating Agents , Iron Overload/drug therapy , Transfusion Reaction/drug therapy , Anemia, Diamond-Blackfan/therapy , Blood Transfusion , Deferasirox/administration & dosage , Deferasirox/adverse effects , Deferiprone/administration & dosage , Deferiprone/adverse effects , Female , Humans , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/adverse effects , Iron Overload/etiology , Male , Retrospective Studies , beta-Thalassemia/therapy
18.
Hematology ; 24(1): 238-246, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30558524

ABSTRACT

OBJECTIVE: The study evaluates the long-term deferasirox treatment of adult and pediatric patients with chronic transfusional iron overload in clinical practice. METHODS: In this non-interventional study, patients were observed for up to 3 years from initiation of deferasirox treatment both prospectively and retrospectively for up to 1 year prior to enrollment. The primary end points were the proportion of patients with ≥1 notable increase in serum creatinine (SCr), and ≥1 notable increase in alanine aminotransferase (ALT). RESULTS: Overall, 120 patients were enrolled and 51 completed the study, with a limited 3-year dropout rate of 12.5% due to adverse events (AEs). Increase in SCr > 33% above baseline and the age-adjusted ULN (upper limit of normal) was observed in 14 patients (95%CI, 7.1-19.2). The ALT levels >5 × ULN was observed in 1 patient. Most frequent AEs reported during treatment with deferasirox include gastrointestinal disturbances. CONCLUSIONS: The long-term treatment with deferasirox was manageable in most transfusion-dependent patients with no unexpected safety findings. Regular monitoring and an adjusted deferasirox dosing strategy per local labels allowed continued iron chelation treatment and control of transfusional iron in the majority of patients on study.


Subject(s)
Blood Transfusion , Deferasirox/administration & dosage , Hemosiderosis/drug therapy , Iron Overload/drug therapy , Transfusion Reaction/drug therapy , Adolescent , Adult , Aged , Female , Hemosiderosis/blood , Hemosiderosis/etiology , Humans , Iron Overload/blood , Iron Overload/etiology , Male , Middle Aged , Retrospective Studies , Transfusion Reaction/blood
19.
PLoS One ; 13(12): e0208102, 2018.
Article in English | MEDLINE | ID: mdl-30507954

ABSTRACT

Chronic blood transfusions are responsible to cause iron overload, which leads to several complications to end organs and osteoporosis. Iron chelation is needed to remove iron excess and to contain bone-mass loss. Deferasirox is the most recent oral iron chelator that prevents transfusion related iron overload complications. Recently Eltrombopag (ELT) iron chelating properties are emerging. ELT is an agonist at Thrombopoietin receptor, used in treatment of thrombocytopenia. We tested ELT and Deferasirox in iron overloaded osteoclasts from thalassemic patients and donors measuring intracellular iron, TRAP expression and osteoclast activity. We confirmed ELT iron chelation capacity also in bone tissue and a synergic effect when used with Deferasirox. Moreover, having demonstrated its effects on osteoclast activity, we suggest for the first time that ELT could ameliorate bone tissue's health reducing bone mass loss.


Subject(s)
Benzoates/pharmacology , Hydrazines/pharmacology , Iron Chelating Agents/pharmacology , Iron Overload/drug therapy , Osteoporosis/prevention & control , Pyrazoles/pharmacology , Thalassemia/therapy , Transfusion Reaction/drug therapy , Adult , Benzoates/therapeutic use , Blood Transfusion , Bone and Bones/drug effects , Bone and Bones/metabolism , Cell Differentiation , Deferasirox/pharmacology , Deferasirox/therapeutic use , Drug Evaluation , Female , Ferritins/blood , Healthy Volunteers , Humans , Hydrazines/therapeutic use , Iron/analysis , Iron/metabolism , Iron Chelating Agents/therapeutic use , Iron Overload/blood , Iron Overload/complications , Leukocytes, Mononuclear , Osteoclasts/drug effects , Osteoclasts/physiology , Osteoporosis/etiology , Primary Cell Culture , Pyrazoles/therapeutic use , Receptors, Thrombopoietin/antagonists & inhibitors , Thalassemia/complications , Transfusion Reaction/blood , Transfusion Reaction/complications
20.
J Ayub Med Coll Abbottabad ; 30(3): 351-355, 2018.
Article in English | MEDLINE | ID: mdl-30465364

ABSTRACT

BACKGROUND: Donor notification of reactive status is important to prevent the spread of disease. Response of reactive donors to seek confirmation and treatment is a direct reflection of their knowledge and attitudes towards transfusion transmittable infections. METHODS: A cross sectional observational study was conducted from August 2014 to July 2015 at the blood bank of a tertiary care hospital, Karachi, Pakistan. Reactive donors' notification and responses were noted with reasons of failure. A cross-sectional analytical survey with non-probability purposive sampling was done on 350 potential consenting blood donors using a pre-tested questionnaire to assess their knowledge and attitude about disease awareness, transmission routes, financial implications and disease sensitization. RESULTS: Out of 16660 donations, 5.57% were rejected on positive screening tests. Repeat donors (69.5%) with primary to secondary qualifications constituted the bulk of reactive donors. Donor notification rate were 54.25% whereas 28.68% donors responded to blood bank in person. The survey showed limited awareness about transfusion transmitted infections. Respondents who were ignorant of disease spread through blood transfusion comprised of 48%. 96.6% donors did not know the financial impact of treatment and 69.7% were unable to afford it. Moreover, 94.9% donors were not protected against hepatitis B. Participants with secondary education had significantly less odds of being adequately knowledgeable (OR=0.372, 95% CI: 0.203-0.681, p-value <0.01) but more likely to have a positive attitude. CONCLUSIONS: There is need for structured pre-donation counselling to sensitize donors about transfusion related diseases in resource limited countries where treatment costs are high and out of reach for most donors..


Subject(s)
Blood Donors/psychology , Health Knowledge, Attitudes, Practice , Transfusion Reaction/prevention & control , Adult , Blood Transfusion , Cross-Sectional Studies , Donor Selection , Educational Status , Female , Humans , Male , Pakistan , Perception , Surveys and Questionnaires , Transfusion Reaction/diagnosis , Transfusion Reaction/drug therapy , Transfusion Reaction/economics , Truth Disclosure , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...