1
|
Jones JM, Wool J, Crowe EP, Bloch EM, Pecker LH, Lanzkron S. Longitudinal outcomes of chronically transfused adults with sickle cell disease and a history of childhood stroke. Transfusion 2024; 64:2260-2269. [PMID: 39501512 PMCID: PMC11637247 DOI: 10.1111/trf.18041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/18/2024] [Accepted: 09/24/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Many children with sickle cell disease (SCD) who suffer a stroke receive chronic transfusion therapy (CTT) indefinitely; however, their adulthood neurologic outcomes have not been reported. Understanding these outcomes is critical to inform decisions regarding curative therapy in childhood. STUDY DESIGN AND METHODS In this retrospective study, we described a cohort of adults with SCD and a history of childhood stroke who received care at a single center and compared their outcomes with matched subjects without childhood stroke using chi2 and Mann-Whitney U tests. RESULTS Of 42 subjects with childhood stroke, all received CTT for secondary stroke prophylaxis. Five (11%) developed recurrent stroke. The rate of stroke was similar in subjects with and without childhood stroke (0.7 vs. 1.1 per 100 person·years, p = .63). Both cohorts exhibited evidence of iron overload (median ferritin 2227 vs. 1409 ng/dL, p = .10) and alloimmunization (45% vs. 45%, p = 1.0), despite receiving care in a comprehensive SCD program. DISCUSSION For adults with SCD who had a childhood stroke, our results suggest CTT returns the risk of stroke to that of age-matched stroke naïve patients with SCD.
Collapse
Affiliation(s)
- Jennifer M. Jones
- Division of Transfusion Medicine, Department of Pathology, Michigan MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Julia Wool
- Division of Hematology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Elizabeth P. Crowe
- Division of Transfusion Medicine, Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Evan M. Bloch
- Division of Transfusion Medicine, Department of PathologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Lydia H. Pecker
- Division of Hematology, Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Sophie Lanzkron
- Division of Hematology, Department of MedicineThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Goldenberg M, Lanzkron S, Pecker LH. Late effects of hemopoietic stem cell transplant for sickle cell disease: monitoring and management. Expert Rev Hematol 2024; 17:891-905. [PMID: 39499235 DOI: 10.1080/17474086.2024.2423368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 10/27/2024] [Indexed: 11/07/2024]
Abstract
INTRODUCTION Allogeneic hemopoietic stem cell transplantation (HSCT) is a curative therapy for sickle cell disease (SCD). Exposure to both SCD and HSCT conditioning regimens is associated with late health effects. AREAS COVERED This review addresses post-HSCT outcomes and late health effects among individuals with SCD exposed to allogeneic HSCT regimens, summarizes recommendations for long-term care, and identifies future survivorship research needs. EXPERT OPINION Individuals with SCD exposed to HSCT and gene therapy require multidisciplinary care to monitor late health effects. To optimize care, multi-disciplinary clinics that include experts in late effects of HSCT exposure, SCD, complex chronic pain, mental health, and social work are needed. Research defining the late effects of exposure is needed to inform patient management and build clinical care infrastructure.
Collapse
Affiliation(s)
- Marti Goldenberg
- Pediatric Hematology Program, Division of Pediatric Hematology, Bloomberg Children's Center at John's Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sophie Lanzkron
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lydia H Pecker
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Baskin-Miller J, Carson S, Jaffray J, Fletcher C, Singer J, Freyer DR, Wood J, Coates TD, Denton CC. Transfusional hemosiderosis in childhood cancer patients and survivors. Pediatr Blood Cancer 2024; 71:e31220. [PMID: 39096194 DOI: 10.1002/pbc.31220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/21/2024] [Accepted: 07/10/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Children treated for cancer are at risk for adverse effects of iron due to transfusions administered during prolonged marrow suppression, which may increase exposure to toxic forms of iron, extrahepatic iron accumulation, and long-term organ damage. OBJECTIVE This study aimed to characterize the severity and organ distribution of clinically significant, multisystem iron overload (IO) in an at-risk cohort of pediatric cancer patients. METHODS This was a retrospective, cross-sectional study of childhood cancer patients who underwent a magnetic resonance imaging (MRI) due to clinical concern for IO. Data regarding cancer type and treatment, transfusion history, MRI and laboratory results, and treatment for IO were collected. Severity of IO was analyzed by non-parametric tests with respect to clinical characteristics. RESULTS Of the 103 patients, 98% of whom had a Cancer Intensity Treatment Rating (ITR-3) of 3 or higher, 53% (54/102) had moderate or greater hepatic siderosis, 80% (77/96) had pancreatic siderosis, 4% (3/80) had cardiac siderosis, and 45% (13/29) had pituitary siderosis and/or volume loss. Pancreatic iron was associated with both cardiac (p = .0043) and pituitary iron (p = .0101). In the 73 off-therapy patients, ferritin levels were lower (p = .0008) with higher correlation with liver iron concentration (LIC) (p = .0016) than on-therapy patients. Fifty-eight subjects were treated for IO. CONCLUSION In this heavily treated cohort of pediatric cancer patients, more than 80% had extrahepatic iron loading, which occurs with significant exposure to toxic forms of iron related to decreased marrow activity in setting of transfusions. Further studies should examine the effects of exposure to reactive iron on long-term outcomes and potential strategies for management.
Collapse
Affiliation(s)
- Jacquelyn Baskin-Miller
- Division of Pediatric Hematology Oncology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Susan Carson
- Children's Center for Cancer, Blood Diseases and Bone Marrow Transplantation, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - Julie Jaffray
- Division of Pediatric Hematology Oncology, Rady Children's Hospital, San Diego, California, USA
| | - Craig Fletcher
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Jessie Singer
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - David R Freyer
- Children's Center for Cancer, Blood Diseases and Bone Marrow Transplantation, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - John Wood
- Division of Cardiology, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Thomas D Coates
- Children's Center for Cancer, Blood Diseases and Bone Marrow Transplantation, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - Christopher C Denton
- Children's Center for Cancer, Blood Diseases and Bone Marrow Transplantation, Children's Hospital of Los Angeles, Los Angeles, California, USA
| |
Collapse
|
4
|
M Ross J, Forté S, Mercure-Corriveau N, Lemay AS, Rioux-Massé B, Potter BJ, Soulières D. Automated red blood cell exchange with a post-procedure haematocrit targeted at 34% in the chronic management of sickle cell disease. Br J Haematol 2024; 205:1556-1564. [PMID: 39081092 DOI: 10.1111/bjh.19674] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/17/2024] [Indexed: 10/18/2024]
Abstract
Optimal targets for red blood cell exchange (RCE) are not well defined in the chronic management of sickle cell disease. We analysed transfusion requirements and iron-related outcomes in 101 patients on chronic RCE with a post-procedure haematocrit (Ht) targeted at 34%, which is higher than typically used. A majority were of HbSS/HbSβ0 genotype (n = 72) and enrolled for neurological complications (n = 53). Fifty patients had a positive Ht balance with RCE (>2% mean increase from pre-procedure level), while 43 patients maintained a neutral balance. The first group required fewer red blood cell units/year (65 vs. 80, p < 0.001), but a significant proportion were iron overloaded based on R2* with liver MRI (32% vs. none performed) and prescription of iron chelation (52% vs. 0%, p < 0.001, after a median of 19 months). The second group was more likely to receive iron supplementation (6% vs. 56%, p < 0.001). Chronic automated RCE with a post-procedure Ht targeted at 34% is not iron-neutral, and personalized Ht goals may be more appropriate in certain settings. This higher target should be compared with a lower Ht strategy in individuals with similar baseline red cell volumes to assess iron homeostasis and blood product requirements.
Collapse
Affiliation(s)
- Jules M Ross
- Hematology-Oncology Division, Departement of Medicine, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Stéphanie Forté
- Hematology-Oncology Division, Departement of Medicine, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Nicolas Mercure-Corriveau
- Hematology-Oncology Division, Departement of Medicine, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Anne-Sophie Lemay
- Hematology-Oncology Division, Departement of Medicine, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Benjamin Rioux-Massé
- Hematology-Oncology Division, Departement of Medicine, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Brian J Potter
- Cardiology Division, Department of Medicine, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Denis Soulières
- Hematology-Oncology Division, Departement of Medicine, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| |
Collapse
|
5
|
Winzent-Oonk S, Staley A, Alami V, Bradley J, Harvey S, Pounds A, Kuldanek S, Pacenta H, Winters AC, McKinney C. Risk of transfusion-related iron overload varies based on oncologic diagnosis and associated treatment: Retrospective analysis from a single pediatric cancer center. Pediatr Blood Cancer 2024:e31254. [PMID: 39118250 DOI: 10.1002/pbc.31254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Transfusion-related iron overload (TRIO) is a widely acknowledged late effect of antineoplastic therapy in pediatric cancer survivors, but firm guidelines as to screening protocols or at-risk populations are lacking in the literature. PROCEDURE We performed retrospective analysis of all oncology patients diagnosed at our center from 2014 to 2019, who underwent TRIO screening as part of an internal quality improvement project. Correlations of MRI-confirmed TRIO with patient-, disease-, and treatment-specific features were evaluated. RESULTS We show that a tiered screening algorithm for TRIO, when followed as intended, led to the identification of the highest proportion of patients with TRIO. We confirm that cardiac TRIO is quite rare in the oncology patient population. However, accepted surrogate markers including red blood cell transfused volume and ferritin only modestly correlated with TRIO in our patient cohort. Instead, we found that older age, leukemia diagnosis, anthracycline exposure, and receipt of stem cell transplant were most strongly associated with risk for TRIO. CONCLUSIONS We describe associations between TRIO and patient, disease, and treatment characteristics in a multivariate risk model that could lead to an improved risk stratification of off-therapy patients, and which should be validated in a prospective manner.
Collapse
Affiliation(s)
- Shelby Winzent-Oonk
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Alyse Staley
- Biostatistics and Bioinformatics Shared Resource, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Vida Alami
- Biostatistics and Bioinformatics Shared Resource, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Julie Bradley
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Susan Harvey
- Hematology, Children's National Medical Center, Washington, District of Columbia, USA
| | - Aneisia Pounds
- Pediatric Hematology, Oncology, BMT, UF Health Shands Children's Hospital, Gainesville, Florida, USA
| | - Susan Kuldanek
- Pediatric Hematology, Children's Minnesota, Minneapolis, Minnesota, USA
| | - Holly Pacenta
- Pediatric Hematology/Oncology, Cook Children's, Fort Worth, Texas, USA
| | - Amanda C Winters
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Chris McKinney
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
6
|
Castro OL, De Franceschi L, Ganz T, Kanter J, Kato GJ, Pasricha SR, Rivella S, Wood JC. Iron restriction in sickle cell disease: When less is more. Am J Hematol 2024; 99:1349-1359. [PMID: 38400590 DOI: 10.1002/ajh.27267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024]
Abstract
Primum non nocere! Can iron deficiency, an abnormality that causes anemia, benefit people with sickle cell disease (SCD) who already have an anemia? The published literature we review appears to answer this question in the affirmative: basic science considerations, animal model experiments, and noncontrolled clinical observations all suggest a therapeutic potential of iron restriction in SCD. This is because SCD's clinical manifestations are ultimately attributable to the polymerization of hemoglobin S (HbS), a process strongly influenced by intracellular HbS concentration. Even small decrements in HbS concentration greatly reduce polymerization, and iron deficiency lowers erythrocyte hemoglobin concentration. Thus, iron deficiency could improve SCD by changing its clinical features to those of a more benign anemia (i.e., a condition with fewer or no vaso-occlusive events). We propose that well-designed clinical studies be implemented to definitively determine whether iron restriction is a safe and effective option in SCD. These investigations are particularly timely now that pharmacologic agents are being developed, which may directly reduce red cell hemoglobin concentrations without the need for phlebotomies to deplete total body iron.
Collapse
Affiliation(s)
- Oswaldo L Castro
- Center for Sickle Cell Disease, Howard University, Washington, District of Columbia, USA
| | | | - Tomas Ganz
- Center for Iron Disorders, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, USA
| | - Julie Kanter
- Division of Hematology and Oncology, University of Alabama, Birmingham, Birmingham, Alabama, USA
| | - Gregory J Kato
- Hematology Therapeutic Area, CSL Behring, King of Prussia, Pennsylvania, USA
| | - Sant-Rayn Pasricha
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Stefano Rivella
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA; RNA Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- RNA Gene Therapeutics Group, RNA Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - John C Wood
- Division of Cardiology, Department of Pediatrics and Radiology, Children's Hospital of Los Angeles, Los Angeles, California, USA
| |
Collapse
|
7
|
Alan S, Kanter J. Advances in pharmacotherapy for sickle cell disease: what is the current state of play? Expert Opin Pharmacother 2024; 25:1325-1334. [PMID: 38973339 DOI: 10.1080/14656566.2024.2377711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
INTRODUCTION Despite over 100 years of neglect and insufficient funding, sickle cell disease has risen to the top of the discussions due to the recent approval of two new genetic therapies. Prior to these approvals, there were only four prior approved medications for sickle cell disease in spite of being the most common inherited blood disorder. The advent and expense of these new genetic therapies have finally brought the trials and tribulations associated with SCD including the suffering and early mortality of affected individuals to the much-needed limelight. Presently, questions about how these therapies will be used and what that means for ongoing pharmaceutical development remain. AREAS COVERED Here, we wish to highlight the current medications and treatments for SCD using already published literature as well as scrutinize the tedious process of implementation for these newly approved commercial genetic therapies. EXPERT OPINION In our expert opinion, despite the progress we have made, significant challenges remain and the most important requirement for any of these treatments is ensuring all affected individuals have access to a sickle cell specialist who can provide comprehensive care.
Collapse
Affiliation(s)
- Sheinei Alan
- Inova Adult Sickle Cell Center, University of Virginia School of Medicine, Inova Fairfax Medical Campus, Fairfax, VA, USA
| | - Julie Kanter
- Lifespan Comprehensive Sickle Cell Center, University of Alabama Birmingham, Birmingham, AL, USA
| |
Collapse
|
8
|
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: 0] [Impact Index Per Article: 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
|
9
|
Xi C, Palani C, Takezaki M, Shi H, Horuzsko A, Pace BS, Zhu X. Simvastatin-Mediated Nrf2 Activation Induces Fetal Hemoglobin and Antioxidant Enzyme Expression to Ameliorate the Phenotype of Sickle Cell Disease. Antioxidants (Basel) 2024; 13:337. [PMID: 38539870 PMCID: PMC10968127 DOI: 10.3390/antiox13030337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 06/04/2024] Open
Abstract
Sickle cell disease (SCD) is a pathophysiological condition of chronic hemolysis, oxidative stress, and elevated inflammation. The transcription factor Nrf2 is a master regulator of oxidative stress. Here, we report that the FDA-approved oral agent simvastatin, an inhibitor of hydroxymethyl-glutaryl coenzyme A reductase, significantly activates the expression of Nrf2 and antioxidant enzymes. Simvastatin also induces fetal hemoglobin expression in SCD patient primary erythroid progenitors and a transgenic mouse model. Simvastatin alleviates SCD symptoms by decreasing hemoglobin S sickling, oxidative stress, and inflammatory stress in erythroblasts. Particularly, simvastatin increases cellular levels of cystine, the precursor for the biosynthesis of the antioxidant reduced glutathione, and decreases the iron content in SCD mouse spleen and liver tissues. Mechanistic studies suggest that simvastatin suppresses the expression of the critical histone methyltransferase enhancer of zeste homolog 2 to reduce both global and gene-specific histone H3 lysine 27 trimethylation. These chromatin structural changes promote the assembly of transcription complexes to fetal γ-globin and antioxidant gene regulatory regions in an antioxidant response element-dependent manner. In summary, our findings suggest that simvastatin activates fetal hemoglobin and antioxidant protein expression, modulates iron and cystine/reduced glutathione levels to improve the phenotype of SCD, and represents a therapeutic strategy for further development.
Collapse
Affiliation(s)
- Caixia Xi
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA 30912, USA; (C.X.); (C.P.)
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA (A.H.)
| | - Chithra Palani
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA 30912, USA; (C.X.); (C.P.)
| | - Mayuko Takezaki
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA 30912, USA; (C.X.); (C.P.)
| | - Huidong Shi
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA (A.H.)
| | - Anatolij Horuzsko
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA (A.H.)
| | - Betty S. Pace
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA 30912, USA; (C.X.); (C.P.)
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA (A.H.)
| | - Xingguo Zhu
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA 30912, USA; (C.X.); (C.P.)
- Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA (A.H.)
| |
Collapse
|
10
|
Oyedeji CI, Artz AS, Cohen HJ. How I treat anemia in older adults. Blood 2024; 143:205-213. [PMID: 36827619 PMCID: PMC10808247 DOI: 10.1182/blood.2022017626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/31/2023] [Accepted: 02/18/2023] [Indexed: 02/26/2023] Open
Abstract
ABSTRACT With the global growing older adult population, clinicians face the common, yet complex challenge of how to evaluate and manage anemia in this population. Older age predisposes to common causes of anemia such as nutritional deficiencies, inflammatory disorders, chronic kidney disease, and hematologic malignancies. Failure to diagnose and appropriately manage anemia may result in decreased quality of life, impaired cognition, impaired mobility, and increased mortality. Anemia diagnosis in older adults presents a diagnostic conundrum because anemia may have a single cause, may be multifactorial, or may have no apparent cause even after an extensive evaluation. We believe a systematic approach to diagnosis ensures appropriate testing and avoids the pitfall of undertreatment and overtreatment. In this article we present our recommended approach through common scenarios for the management of anemia in the older adult.
Collapse
Affiliation(s)
- Charity I. Oyedeji
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC
- Duke Claude D. Pepper Older Americans Independence Center, Durham, NC
| | | | - Harvey Jay Cohen
- Duke Claude D. Pepper Older Americans Independence Center, Durham, NC
- Center for the Study of Aging and Human Development, Duke University Medical Center, Durham, NC
| |
Collapse
|
11
|
Kelly S. Logistics, risks, and benefits of automated red blood cell exchange for patients with sickle cell disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:646-652. [PMID: 38066846 PMCID: PMC10905321 DOI: 10.1182/hematology.2023000498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Red blood cell (RBC) transfusions treat and prevent severe complications of sickle cell disease (SCD) and can be delivered as a simple or exchange transfusion. During an exchange, some of the patient's abnormal hemoglobin (Hb) S (HbS) RBCs are removed. An apheresis device can accomplish an automated RBC exchange, simultaneously removing patient's RBCs while returning other blood components along with normal RBCs. Automated RBC exchange is therefore an isovolemic transfusion that can efficiently decrease HbS RBCs while limiting iron loading and hyperviscosity. However, specialized equipment, trained personnel, appropriate vascular access, and increased RBC exposure are required compared to simple or manual RBC exchange. Therefore, risks and benefits must be balanced to make individualized decisions for patients with SCD who require transfusion.
Collapse
Affiliation(s)
- Shannon Kelly
- UCSF Benioff Children's Hospital Oakland, Oakland, CA
| |
Collapse
|
12
|
Inusa BP, Atoyebi W, Andemariam B, Hourani JN, Omert L. Global burden of transfusion in sickle cell disease. Transfus Apher Sci 2023; 62:103764. [PMID: 37541800 DOI: 10.1016/j.transci.2023.103764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 08/06/2023]
Abstract
Sickle cell disease (SCD) is the most common hereditary hemoglobinopathy. The underlying pathophysiology of the red blood cell (RBC) leads to pan-systemic complications which manifest at an early age. While curative and disease-modifying treatments exist for SCD, a key intervention in the management and treatment of SCD is RBC transfusion, which can alleviate or prevent many complications. SCD patients often require chronic RBC transfusion therapy which can result in complications, such as iron overload, alloimmunization and infection. In low- and middle-income countries (LMICs), SCD patients lack appropriate access to healthcare such as newborn screening, health education, prophylaxis for infection, and treatments to reduce both mortality and SCD-related adverse effects. Poor access to RBCs for transfusion, coupled with donated blood not meeting safety standards set by the World Health Organization, presents a significant barrier for patients requiring chronic transfusions in LMICs. Unmet needs associated with blood collection, blood component processing and recipient matching all pose a serious problem in many LMICs, although this varies depending on geographic location, political organizations and economy. This review aims to provide an overview of the global burden of SCD, focusing on the availability of current treatments and the burden of chronic RBC transfusions in patients with SCD.
Collapse
Affiliation(s)
- Baba Pd Inusa
- Guy's and Saint Thomas' NHS Foundation Trust, London, UK.
| | | | - Biree Andemariam
- New England Sickle Cell Institute, University of Connecticut Health, Farmington, CT, USA
| | | | | |
Collapse
|
13
|
Meloni A, Pistoia L, Lupi A, Righi R, Vallone A, Missere M, Renne S, Fina P, Riva A, Gamberini MR, Cecinati V, Sorrentino F, Rosso R, Messina G, Ricchi P, Positano V, Mavrogeni S, Quaia E, Cademartiri F, Pepe A. Impact of the COVID-19 Pandemic on Iron Overload Assessment by MRI in Patients with Hemoglobinopathies: The E-MIOT Network Experience. Tomography 2023; 9:1711-1722. [PMID: 37736989 PMCID: PMC10514849 DOI: 10.3390/tomography9050136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND The E-MIOT (Extension-Myocardial Iron Overload in Thalassemia) project is an Italian Network assuring high-quality quantification of tissue iron overload by magnetic resonance imaging (MRI). We evaluated the impact of the COVID-19 pandemic on E-MIOT services. METHODS The activity of the E-MIOT Network MRI centers in the year 2020 was compared with that of 2019. A survey evaluated whether the availability of MRI slots for patients with hemoglobinopathies was reduced and why. RESULTS The total number of MRI scans was 656 in 2019 and 350 in 2020, with an overall decline of 46.4% (first MRI: 71.7%, follow-up MRI: 36.9%), a marked decline (86.9%) in the period March-June 2020, and a reduction in the gap between the two years in the period July-September. A new drop (41.4%) was recorded in the period October-December for two centers, due to the general reduction in the total amount of MRIs/day for sanitization procedures. In some centers, patients refused MRI scans for fear of getting COVID. Drops in the MRI services >80% were found for patients coming from a region without an active MRI site. CONCLUSIONS The COVID-19 pandemic had a strong negative impact on MRI multi-organ iron quantification, with a worsening in the management of patients with hemoglobinopathies.
Collapse
Affiliation(s)
- Antonella Meloni
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.); (F.C.)
- Unità Operativa Complessa Bioingegneria, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| | - Laura Pistoia
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.); (F.C.)
| | - Amalia Lupi
- Istituto di Radiologia, Dipartimento di Medicina, Università di Padova, 35128 Padova, Italy; (A.L.); (E.Q.)
| | - Riccardo Righi
- Diagnostica per Immagini e Radiologia Interventistica, Ospedale del Delta, 44023 Lagosanto, Italy;
| | - Antonino Vallone
- Reparto di Radiologia, Azienda Ospedaliera “Garibaldi” Presidio Ospedaliero Nesima, 95126 Catania, Italy;
| | - Massimiliano Missere
- Unità Operativa Complessa Radiodiagnostica, Gemelli Molise SpA, Fondazione di Ricerca e Cura “Giovanni Paolo II”, 86100 Campobasso, Italy;
| | - Stefania Renne
- Struttura Complessa di Cardioradiologia, Presidio Ospedaliero “Giovanni Paolo II”, 88046 Lamezia Terme, Italy;
| | - Priscilla Fina
- Unità Operativa Complessa Diagnostica per Immagini, Ospedale “Sandro Pertini”, 00157 Roma, Italy;
| | - Ada Riva
- Struttura Complessa di Radiologia, Ospedale “SS. Annunziata” ASL Taranto, 74121 Taranto, Italy;
| | - Maria Rita Gamberini
- Unità Operativa di Day Hospital della Talassemia e delle Emoglobinopatie, Dipartimento della Riproduzione e dell’Accrescimento, Azienda Ospedaliero-Universitaria “S. Anna”, 44124 Cona, Italy;
| | - Valerio Cecinati
- Struttura Semplice di Microcitemia, Ospedale “SS. Annunziata” ASL Taranto, 74100 Taranto, Italy;
| | - Francesco Sorrentino
- Unità Operativa Semplice Dipartimentale Day Hospital Talassemici, Ospedale “Sant’Eugenio”, 00143 Roma, Italy;
| | - Rosamaria Rosso
- Unità Operativa Talassemie ed Emoglobinopatie, Azienda Ospedaliero-Universitaria Policlinico “Vittorio Emanuele”, 95100 Catania, Italy;
| | - Giuseppe Messina
- Centro Microcitemie, Grande Ospedale Metropolitano “Bianchi-Melacrino-Morelli”, 89100 Reggio Calabria, Italy;
| | - Paolo Ricchi
- Unità Operativa Semplice Dipartimentale Malattie Rare del Globulo Rosso, Azienda Ospedaliera di Rilievo Nazionale “A. Cardarelli”, 80131 Napoli, Italy;
| | - Vincenzo Positano
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.); (F.C.)
- Unità Operativa Complessa Bioingegneria, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy
| | | | - Emilio Quaia
- Istituto di Radiologia, Dipartimento di Medicina, Università di Padova, 35128 Padova, Italy; (A.L.); (E.Q.)
| | - Filippo Cademartiri
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (L.P.); (V.P.); (F.C.)
| | - Alessia Pepe
- Istituto di Radiologia, Dipartimento di Medicina, Università di Padova, 35128 Padova, Italy; (A.L.); (E.Q.)
| |
Collapse
|
14
|
Pathophysiological characterization of the Townes mouse model for sickle cell disease. Transl Res 2023; 254:77-91. [PMID: 36323381 DOI: 10.1016/j.trsl.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/17/2022] [Accepted: 10/23/2022] [Indexed: 11/07/2022]
Abstract
A deeper pathophysiologic understanding of available mouse models of sickle cell disease (SCD), such as the Townes model, will help improve preclinical studies. We evaluated groups of Townes mice expressing either normal adult human hemoglobin (HbA), sickle cell trait (HbAS), or SCD (HbS), comparing younger versus older adults, and females versus males. We obtained hematologic parameters in steady-state and hypoxic conditions and evaluated metabolic markers and cytokines from serum. Kidney function was evaluated by measuring the urine protein/creatinine ratio and urine osmolality. In vivo studies included von Frey assay, non-invasive plethysmography, and echocardiography. Histopathological evaluations were performed in lung, liver, spleen, and kidney tissues. HbS mice displayed elevated hemolysis markers and white blood cell counts, with some increases more pronounced in older adults. After extended in vivo hypoxia, hemoglobin, platelet counts, and white blood cell counts decreased significantly in HbS mice, whereas they remained stable in HbA mice. Cytokine analyses showed increased TNF-alpha in HbS mice. Kidney function assays revealed worsened kidney function in HbS mice. The von Frey assay showed a lower threshold to response in the HbS mice than controls, with more noticeable differences in males. Echocardiography in HbS mice suggested left ventricular hypertrophy and dilatation. Plethysmography suggested obstructive lung disease and inflammatory changes in HbS mice. Histopathological studies showed vascular congestion, increased iron deposition, and disruption of normal tissue architecture in HbS mice. These data correlate with clinical manifestations in SCD patients and highlight analyses and groups to be included in preclinical therapeutic studies.
Collapse
|
15
|
Oyedeji CI, Hodulik KL, Telen MJ, Strouse JJ. Management of Older Adults with Sickle Cell Disease: Considerations for Current and Emerging Therapies. Drugs Aging 2023; 40:317-334. [PMID: 36853587 PMCID: PMC10979738 DOI: 10.1007/s40266-023-01014-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 03/01/2023]
Abstract
People with sickle cell disease (SCD) are living longer than ever before, with the median survival increasing from age 14 years in 1973, beyond age 40 years in the 1990s, and as high as 61 years in recent cohorts from academic centers. Improvements in survival have been attributed to initiatives, such as newborn screening, penicillin prophylaxis, vaccination against encapsulated organisms, better detection and treatment of splenic sequestration, and improved transfusion support. There are an estimated 100,000 people living with SCD in the United States and millions of people with SCD globally. Given that the number of older adults with SCD will likely continue to increase as survival improves, better evidence on how to manage this population is needed. When managing older adults with SCD (defined herein as age ≥ 40 years), healthcare providers should consider the potential pitfalls of extrapolating evidence from existing studies on current and emerging therapies that have typically been conducted with participants at mean ages far below 40 years. Older adults with SCD have historically had little to no representation in clinical trials; therefore, more guidance is needed on how to use current and emerging therapies in this population. This article summarizes the available evidence for managing older adults with SCD and discusses potential challenges to using approved and emerging drugs in this population.
Collapse
Affiliation(s)
- Charity I Oyedeji
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
- Duke Claude D. Pepper Older Americans Independence Center, Durham, NC, USA.
- Department of Medicine, and Duke Comprehensive Sickle Cell Center, Duke University School of Medicine, 315 Trent Dr., Suite 266, DUMC Box 3939, Durham, NC, 27710, USA.
| | - Kimberly L Hodulik
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Department of Pharmacy, Duke University Hospital, Durham, NC, USA
| | - Marilyn J Telen
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - John J Strouse
- Division of Hematology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
- Duke Claude D. Pepper Older Americans Independence Center, Durham, NC, USA
- Department of Medicine, and Duke Comprehensive Sickle Cell Center, Duke University School of Medicine, 315 Trent Dr., Suite 266, DUMC Box 3939, Durham, NC, 27710, USA
- Division of Pediatric Hematology-Oncology, Duke University, Durham, NC, USA
| |
Collapse
|
16
|
Tang AY, Zhou M, Maillis AN, Lai KW, Lane PA, Snyder AB. Trends in blood transfusion, hydroxyurea use, and iron overload among children with sickle cell disease enrolled in Medicaid, 2004-2019. Pediatr Blood Cancer 2023; 70:e30152. [PMID: 36579749 DOI: 10.1002/pbc.30152] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/07/2022] [Accepted: 11/22/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND There have been significant changes in clinical guidelines for sickle cell disease (SCD) over the past two decades, including updated indications for hydroxyurea, transfusions, and iron overload management. In practice however, there are few studies that examine SCD care utilization over time. METHODS We conducted a serial cross-sectional cohort study of pediatric SCD patients from 2004 to 2019 using Georgia Medicaid claims data. For each year, we reported receipt of any transfusion, chronic transfusion, or three or more filled hydroxyurea prescriptions. For children receiving chronic transfusion (six or more annual transfusions), we evaluated iron overload diagnosis, monitoring, and chelation use. Among children with sickle cell anemia (SCA), we examined rates of transfusions and hydroxyurea use. The Cochran-Armitage test was used to assess trend. RESULTS There were 5316 unique children 2-18 years old with SCD enrolled in Georgia Medicaid from 2004 to 2019. Children receiving any transfusion increased from 2004 to 2010, then stabilized. In SCA patients, chronic transfusions initially increased from 2004 to 2010, then stabilized from 2010 to 2019. For chronically transfused children, monitoring of iron burden and filled chelator prescriptions both increased significantly. Hydroxyurea use in SCA patients increased from 12% to 37%, with increases noted within each age group, most notably from 21% to 60% in the 13-18-year-old cohort. CONCLUSION We demonstrated changes in SCD care utilization over time, including increased hydroxyurea use, changes in transfusion rates, and increased attention to iron overload management. While trends in clinical management do follow updates in treatment guidelines, there is still delayed and suboptimal uptake of guideline recommendations in pediatric SCD patients.
Collapse
Affiliation(s)
- Amy Y Tang
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.,Department of Pediatrics, Division of Hematology/Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mei Zhou
- Georgia Health Policy Center, Andrew Young School of Policy Studies, Georgia State University, Atlanta, Georgia, USA
| | - Alexander N Maillis
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Kristina W Lai
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Peter A Lane
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA.,Department of Pediatrics, Division of Hematology/Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Angela B Snyder
- Georgia Health Policy Center, Andrew Young School of Policy Studies, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
17
|
Bruzzese A, Martino EA, Mendicino F, Lucia E, Olivito V, Bova C, Filippelli G, Capodanno I, Neri A, Morabito F, Gentile M, Vigna E. Iron chelation therapy. Eur J Haematol Suppl 2023; 110:490-497. [PMID: 36708354 DOI: 10.1111/ejh.13935] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/29/2023]
Abstract
Iron overload is a pathological condition resulting from a congenital impairment of its regulation, increased intestinal iron absorption secondary to bone marrow erythroid hyperplasia, or a chronic transfusional regimen. In normal conditions, intracellular and systemic mechanisms contribute to maintaining iron balance. When this complex homeostatic mechanism fails, an iron overload could be present. Detecting an iron overload is not easy. The gold standard remains the liver biopsy, even if it is invasive and dangerous. Identifying iron using noninvasive techniques allowed a better understanding of the rate of iron overload in different organs, with a low risk for the patient. Estimating serum ferritin (mg/L) is the easiest and, consequently, the most employed diagnostic tool for assessing body iron stores, even if it could be a not specific method. The most common hematological causes of iron overload are myelodysplastic syndromes, sickle cell disease, and thalassemia. In all of these conditions, three drugs have been approved for the treatment of iron overload: deferiprone, deferoxamine, and deferasirox. These chelators have been demonstrated to help lower tissue iron levels and prevent iron overload complications, improving event-free survival (EFS). Nowadays, the decision to start chelation and which chelator to choose remains the joint decision of the clinician and patient.
Collapse
Affiliation(s)
- Antonella Bruzzese
- Hematology Unit, Department of Onco-hematology, A.O. of Cosenza, Cosenza, Italy
| | | | - Francesco Mendicino
- Hematology Unit, Department of Onco-hematology, A.O. of Cosenza, Cosenza, Italy
| | - Eugenio Lucia
- Hematology Unit, Department of Onco-hematology, A.O. of Cosenza, Cosenza, Italy
| | - Virginia Olivito
- Hematology Unit, Department of Onco-hematology, A.O. of Cosenza, Cosenza, Italy
| | - Carlo Bova
- Internal Medicine Department, AO of Cosenza, Cosenza, Italy
| | | | - Isabella Capodanno
- SOC Ematologia Azienda USL-IRCSS di Reggio Emilia, Reggio Emilia, Emilia Romagna, Italy
| | - Antonino Neri
- Scientific Direction Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Emilia Romagna, Italy
| | - Fortunato Morabito
- Biotechnology Research Unit, Aprigliano, A.O./ASP of Cosenza, Cosenza, Italy
| | - Massimo Gentile
- Hematology Unit, Department of Onco-hematology, A.O. of Cosenza, Cosenza, Italy.,Department of Pharmacy, Health and Nutritional Science, University of Calabria, Rende, Italy
| | - Ernesto Vigna
- Hematology Unit, Department of Onco-hematology, A.O. of Cosenza, Cosenza, Italy
| |
Collapse
|
18
|
Light J, Boucher M, Baskin-Miller J, Winstead M. Managing the Cerebrovascular Complications of Sickle Cell Disease: Current Perspectives. J Blood Med 2023; 14:279-293. [PMID: 37082003 PMCID: PMC10112470 DOI: 10.2147/jbm.s383472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
The importance of protecting brain function for people with sickle cell disease (SCD) cannot be overstated. SCD is associated with multiple cerebrovascular complications that threaten neurocognitive function and life. Without screening and preventive management, 11% of children at 24% of adults with SCD have ischemic or hemorrhagic strokes. Stroke screening in children with SCD is well-established using transcranial Doppler ultrasound (TCD). TCD velocities above 200 cm/s significantly increase the risk of stroke, which can be prevented using chronic red blood cell (RBC) transfusion. RBC transfusion is also the cornerstone of acute stroke management and secondary stroke prevention. Chronic transfusion requires long-term management of complications like iron overload. Hydroxyurea can replace chronic transfusions for primary stroke prevention in a select group of patients or in populations where chronic transfusions are not feasible. Silent cerebral infarction (SCI) is even more common than stroke, affecting 39% of children and more than 50% of adults with SCD; management of SCI is individualized and includes careful neurocognitive evaluation. Hematopoietic stem cell transplant prevents cerebrovascular complications, despite the short- and long-term risks. Newer disease-modifying agents like voxelotor and crizanlizumab, as well as gene therapy, may treat cerebrovascular complications, but these approaches are investigational.
Collapse
Affiliation(s)
- Jennifer Light
- Pediatric Hematology-Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maria Boucher
- Pediatric Hematology-Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jacquelyn Baskin-Miller
- Pediatric Hematology-Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mike Winstead
- Pediatric Hematology-Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Correspondence: Mike Winstead, Division of Pediatric Hematology-Oncology, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC, USA, Tel +1 919-966-1178, Fax +1 919-966-7629, Email
| |
Collapse
|
19
|
Azar SS, Gopal S. Serious Blood Disorders: A Focus on Sickle Cell Disease and Hemophilia. PALLIATIVE CARE IN HEMATOLOGIC MALIGNANCIES AND SERIOUS BLOOD DISORDERS 2023:37-54. [DOI: 10.1007/978-3-031-38058-7_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
Yang J, Tang Q, Zeng Y. Melatonin: Potential avenue for treating iron overload disorders. Ageing Res Rev 2022; 81:101717. [PMID: 35961513 DOI: 10.1016/j.arr.2022.101717] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/10/2022] [Accepted: 08/08/2022] [Indexed: 02/08/2023]
Abstract
Iron overload as a highly risk factor, can be found in almost all human chronic and common diseases. Iron chelators are often used to treat iron overload; however, patient adherence to these chelators is poor due to obvious side effects and other disadvantages. Numerous studies have shown that melatonin has a high iron chelation ability and direct free radical scavenging activity, and can inhibit the lipid peroxidation process caused by iron overload. Therefore, melatonin may become potential complementary therapy for iron overload-related disorders due to its iron chelating and antioxidant activities. Here, the research progress of iron overload is reviewed and the therapeutic potential of melatonin in the treatment of iron overload is analyzed. In addition, studies related to the protective effects of melatonin on oxidative damage induced by iron overload are discussed. This review provides a foundation for preventing and treating iron homeostasis disorders with melatonin.
Collapse
Affiliation(s)
- Jiancheng Yang
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qinghua Tang
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yuhong Zeng
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
21
|
Amanor E, Kwarteng A, Larbi A, Fordjour FA, Koranteng KK, Sackey DS, Bannor E, Osei FA, Mohammed A, Ackah EB, Odoom SF, Nguah SB, Paintsil V, Osei‐Akoto A. Iron stores in steady-state sickle cell disease children accessing care at a sickle cell disease clinic in Kumasi, Ghana: A cross-sectional study. Health Sci Rep 2022; 5:e934. [PMID: 36439047 PMCID: PMC9686355 DOI: 10.1002/hsr2.934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/29/2022] [Accepted: 11/02/2022] [Indexed: 11/26/2022] Open
Abstract
Background and Aims Children with sickle cell disease (SCD) have an increased risk of multiple hemotransfusions and this can predispose them to elevated iron stores. The objectives of the study were to determine the extent of elevated iron stores and the associated risk factors in a population of steady-state SCD children in Ghana. Methods This cross-sectional study was conducted at the pediatric sickle cell clinic at the Komfo Anokye Teaching Hospital. Complete blood count and serum ferritin assay were performed for (n = 178) steady-state SCD children. Descriptive and multivariate logistic regression analysis were performed. Elevated iron stores were defined as serum ferritin levels >300 ng/ml. Statistical significance was considered at p < 0.05. Results The mean (standard deviation) age of the participants was 9.61 (±4.34) years, and 51% of them were males. About 17% of SCD children had elevated iron stores and receiving at least three hemotransfusions during the last 12 months was strongly associated with elevated iron stores (p < 0.001). History of chronic hemotransfusion increased the odds of having elevated iron store (adjusted odds ratio [aOR] = 11.41; 95% confidence interval [CI] = 3.11-30.85; p < 0.001) but SCD patients on hydroxyurea treatment had reduced-odds of having elevated iron stores (aOR = 0.18; 95% CI = 0.06-0.602; p = 0.006). Moreover, red blood cell (Coef. = -0.84; 95% CI = -0.37, -1.32; p = 0.001), hemoglobin (Coef. = -0.83; 95% CI = -0.05, -1.61; p = 0.04), hematocrit (Coef. = -0.85; 95% CI = -0.08, -1.63; p = 0.03), mean cell volume (Coef. = 0.02; 95% CI = 0.01, 0.03; p = 0.001) and mean cell hemoglobin (Coef. = 0.04; 95% CI = 0.01, 0.07; p = 0.002) could significantly predict serum ferritin levels. Conclusion The magnitude of elevated iron stores was high among children with SCD in steady-state. Red cell indices could provide invaluable information regarding the risk of elevated iron stores. SCD children who have a history of chronic hemotransfusion or had received at least three hemotransfusions in a year should be monitored for elevated iron stores.
Collapse
Affiliation(s)
- Ernest Amanor
- Department of Biochemistry and Biotechnology, College of ScienceKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Alexander Kwarteng
- Department of Biochemistry and Biotechnology, College of ScienceKwame Nkrumah University of Science and TechnologyKumasiGhana
- Tropical Infections and Non‐communicable Disease Research GroupKumasi Center for Collaborative Research in Tropical MedicineKumasiGhana
| | - Amma Larbi
- Department of Biochemistry and Biotechnology, College of ScienceKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Fatima Amponsah Fordjour
- Department of Biochemistry and Biotechnology, College of ScienceKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Kelvin Kwaku Koranteng
- Department of Biochemistry and Biotechnology, College of ScienceKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - David Sebbie Sackey
- Department of Hematology, Laboratory Services DirectorateKomfo Anokye Teaching HospitalKumasiGhana
| | - Emmanuel Bannor
- Department of Hematology, Laboratory Services DirectorateKomfo Anokye Teaching HospitalKumasiGhana
| | - Francis Adjei Osei
- Department of International Health, School of Public HealthKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Aliyu Mohammed
- Department of Biostatistics and Epidemiology, School of Public HealthKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Ezekiel Bonwin Ackah
- Department of Medical Diagnostics, School of Public HealthKwame Nkrumah University of Science and TechnologyKumasiGhana
| | | | - Samuel Blay Nguah
- Child Health DirectorateKomfo Anokye Teaching HospitalKumasiGhana
- Department of Child Health, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Vivian Paintsil
- Child Health DirectorateKomfo Anokye Teaching HospitalKumasiGhana
- Department of Child Health, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Alex Osei‐Akoto
- Child Health DirectorateKomfo Anokye Teaching HospitalKumasiGhana
- Department of Child Health, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| |
Collapse
|
22
|
Buban KR, Lawrence CE, Zhu XJ, Tobian AAR, Gehrie EA, Vozniak S, Shrestha R, Lokhandwala PM, Bloch EM. Algorithm‐based selection of automated red blood cell exchange procedure goals reduces blood utilization in chronically transfused adults with sickle cell disease. J Clin Apher 2022; 37:468-475. [DOI: 10.1002/jca.22004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/11/2022] [Accepted: 06/22/2022] [Indexed: 11/08/2022]
Affiliation(s)
- Kristen R. Buban
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | - Courtney E. Lawrence
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | - Xianming Joshua Zhu
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | - Aaron A. R. Tobian
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | | | - Sonja Vozniak
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | - Ruchee Shrestha
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| | | | - Evan M. Bloch
- Division of Transfusion Medicine, Department of Pathology Johns Hopkins University Baltimore Maryland USA
| |
Collapse
|
23
|
Ross JM, Forté S, Soulières D. Emerging drugs for the treatment of sickle cell disease: a review of phase II/III trials. Expert Opin Emerg Drugs 2022; 27:211-224. [PMID: 35912835 DOI: 10.1080/14728214.2022.2105835] [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] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The substitution of glutamic acid by valine on the ß-globin gene produces the hemoglobin S variant responsible for sickle cell disease (SCD), a disorder that affects millions of people worldwide and leads to acute and cumulative organ damage. Even though life expectancy has significantly improved where the best medical care is available, there are still few therapeutic options for SCD and those are limited by their availability, cost, and individual toxicities. AREAS COVERED This review summarizes the clinical data on current treatments for SCD and emerging therapies studied in the acute setting as well as potential disease-modifying agents, with an emphasis on the FDA-approved agents. EXPERT OPINION Hydroxyurea has been a gold standard for two decades, showing benefits in acute complications and overall survival in sickle cell anemia, although data is lacking for certain genotypes such as hemoglobin SC. As progress is made in our understanding of the pathophysiological networks characterizing SCD, numerous pathways appear to be targetable, with L-glutamine, Crizanlizumab and Voxelotor now approved by the FDA. Pursuing a multi-agent approach could alter the disease course in a more effective fashion and provide an alternative option to curative therapies, but longer clinical studies are needed.
Collapse
Affiliation(s)
- Jules M Ross
- Centre Universitaire de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Stéphanie Forté
- Centre Universitaire de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| | - Denis Soulières
- Centre Universitaire de l'Université de Montréal (CHUM), Montreal, Quebec, Canada
| |
Collapse
|
24
|
Jones JM, Swett AD, Crowe EP, Lawrence C, Bloch EM, Lanzkron SM. Implementation of national blood conservation recommendations at an adult sickle cell center. Transfusion 2022; 62:1763-1771. [PMID: 35837727 DOI: 10.1111/trf.17007] [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: 03/03/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Due to the national blood supply crisis caused by the COVID-19 pandemic, the American Society of Hematology proposed guidance to decrease blood utilization for sickle cell patients on chronic transfusion therapy (CTT). Little evidence exists to support the efficacy and safety of these blood conservation strategies. STUDY DESIGN AND METHODS Through retrospective analysis, we sought to describe outcomes following implementation of these recommendations in 58 adult sickle cell patients on chronic exchange transfusions. The strategies employed included: relaxing the goal fraction of cells remaining (FCR) to 30%-50%, utilizing depletion exchanges in select patients, and transitioning select patients to monthly simple transfusions. We compared hemoglobin S%, hemoglobin values, and other laboratory parameters, acute care visits, and red blood cell usage during the first year of the COVID-19 pandemic to the year prior using Wilcoxon signed rank test. RESULTS Of 53 patients who remained on chronic exchanges during the pandemic, use of depletion exchange increased (15%-23%) and FCR increased (34.9 [SD 4.7] vs. 37.6 [SD 4.5], p < .05). These changes resulted in 854 units conserved without clinically significant changes to pre-exchange laboratory parameters, including hemoglobin S%, or number of acute care presentations. In contrast, five patients who transitioned to predominantly simple transfusions, experienced difficulty maintaining hemoglobin S% less than 30 and worsening anemia. DISCUSSION Our data suggest that in a blood shortage crisis, optimizing the exchange procedure itself may be the safest means of conserving blood in a population of adult patients with sickle cell disease.
Collapse
Affiliation(s)
- Jennifer M Jones
- Department of Hematology, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Ariel D Swett
- Department of Hematology, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Elizabeth P Crowe
- Division of Transfusion Medicine, Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Courtney Lawrence
- Division of Transfusion Medicine, Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland, USA.,Division of Pediatric Hematology, Department of Pediatrics, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Evan M Bloch
- Division of Transfusion Medicine, Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Sophie M Lanzkron
- Department of Hematology, Johns Hopkins Hospital, Baltimore, Maryland, USA
| |
Collapse
|
25
|
Meloni A, Barbuto L, Pistoia L, Positano V, Renne S, Peritore G, Fina P, Spasiano A, Allò M, Messina G, Casini T, Massa A, Romano L, Pepe A, Cademartiri F. Frequency, pattern, and associations of renal iron accumulation in sickle/β-thalassemia patients. Ann Hematol 2022; 101:1941-1950. [PMID: 35821343 DOI: 10.1007/s00277-022-04915-7] [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: 04/01/2022] [Accepted: 07/04/2022] [Indexed: 11/29/2022]
Abstract
We evaluated frequency, pattern, and associations of renal iron accumulation in sickle/β-thalassemia. Thirty-three sickle/β-thalassemia patients (36.5 ± 14.7 years; 13 females), 14 homozygous sickle cell disease (SCD) patients, and 71 thalassemia major (TM) patients, enrolled in the E-MIOT Network, underwent magnetic resonance imaging. Iron overload (IO) was quantified by the T2* technique. Sickle/β-thalassemia patients had a significantly lower frequency of renal IO (T2* < 31 ms) than homozygous SCD patients (9.1% vs. 57.1%; P = 0.001), besides having similar hepatic, cardiac and pancreatic IO. Kidney T2* values were comparable between regularly transfused sickle/β-thalassemia and TM patients but were significantly lower in regularly transfused homozygous SCD patients than in the other two groups. In sickle/β-thalassemia patients, global renal T2* values were not associated with age, gender, splenectomy, and presence of regular transfusions or chelation. No correlation was detected between renal T2* values and serum ferritin levels or iron load in the other organs. Global renal T2* values were not associated with serum creatinine levels but showed a significant inverse correlation with serum lactate dehydrogenase (R = - 0.709; P < 0.0001) and indirect bilirubin (R = - 0.462; P = 0.012). Renal IO is not common in sickle/β-thalassemia patients, with a prevalence significantly lower compared to that of homozygous SCD patients, but with a similar underlying mechanism due to the chronic hemolysis.
Collapse
Affiliation(s)
- Antonella Meloni
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1 - 56124, Pisa, Italy.,U.O.C. Bioingegneria, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | - Luigi Barbuto
- U.O.C. Radiologia Generale E Di Pronto Soccorso, Azienda Ospedaliera Di Rilievo Nazionale "A. Cardarelli", Naples, Italy
| | - Laura Pistoia
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1 - 56124, Pisa, Italy
| | - Vincenzo Positano
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1 - 56124, Pisa, Italy.,U.O.C. Bioingegneria, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | - Stefania Renne
- Struttura Complessa Di Cardioradiologia-UTIC, Presidio Ospedaliero "Giovanni Paolo II", Lamezia Terme, Italy
| | - Giuseppe Peritore
- Unità Operativa Complessa Di Radiologia, "ARNAS" Civico, Di Cristina Benfratelli, Palermo, Italy
| | - Priscilla Fina
- Unità Operativa Complessa Diagnostica Per Immagini, Ospedale "Sandro Pertini", Rome, Italy
| | - Anna Spasiano
- U.O.S.D. Malattie Rare del Globulo Rosso, Azienda Ospedaliera Di Rilievo Nazionale "A. Cardarelli", Naples, Italy
| | - Massimo Allò
- Ematologia Microcitemia, Ospedale San Giovanni di Dio - ASP Crotone, Crotone, Italy
| | - Giuseppe Messina
- Centro Microcitemie, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Tommaso Casini
- Centro Talassemie Ed Emoglobinopatie, Ospedale "Meyer", Florence, Italy
| | - Antonella Massa
- Servizio Trasfusionale, Ospedale "Giovanni Paolo II", Olbia, Italy
| | - Luigia Romano
- U.O.C. Radiologia Generale E Di Pronto Soccorso, Azienda Ospedaliera Di Rilievo Nazionale "A. Cardarelli", Naples, Italy
| | - Alessia Pepe
- Institute of Radiology, Department of Medicine, University of Padua, Padua, Italy
| | - Filippo Cademartiri
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1 - 56124, Pisa, Italy.
| |
Collapse
|
26
|
Pradhan-Sundd T, Kato GJ, Novelli EM. Molecular Mechanisms of Hepatic Dysfunction in Sickle Cell Disease: Lessons From The Townes Mouse Model. Am J Physiol Cell Physiol 2022; 323:C494-C504. [PMID: 35759437 PMCID: PMC9359658 DOI: 10.1152/ajpcell.00175.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Sickle cell disease (SCD) is an autosomal-recessive-genetic disorder that affects ~100,000 Americans and millions of people worldwide. Erythrocyte sickling, vaso-occlusion, sterile inflammation and hemolysis are the major pathophysiological pathways leading to liver injury in SCD. Although hepatic dysfunction affects up to 10-40% of SCD patients, therapeutic approaches to prevent liver injury in SCD are not known, and the molecular mechanisms promoting progressive liver injury in SCD remain poorly understood. Animal models have been beneficial in bridging the gap between preclinical and translational research in SCD. Recent advances in methodology have allowed the development of several humanized mouse models to address various aspects of SCD related liver diseases. This review provides an overview of current knowledge of the molecular mechanisms and potential therapeutic options of SCD associated liver dysfunction using the Townes mouse model.
Collapse
Affiliation(s)
- Tirthadipa Pradhan-Sundd
- Pittsburgh Heart, Liver and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Enrico M Novelli
- Pittsburgh Heart, Liver and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
27
|
Raj A, McGowan K, Knapp E, Zhao J, Shah S. Iron Chelation Therapy With Deferasirox in Sickle Cell Disease With End-Stage Renal Disease. Cureus 2022; 14:e24146. [PMID: 35582552 PMCID: PMC9107315 DOI: 10.7759/cureus.24146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2022] [Indexed: 11/15/2022] Open
Abstract
Patients with transfusion‐dependent sickle cell disease (SCD) are at risk of iron overload and its complications. Iron overload is a significant risk factor for chronic liver disease in patients who are dependent on hemodialysis secondary to end-stage renal disease (ESRD). Deferasirox is being increasingly used as an iron-chelating agent for the treatment of iron overload in both adults and children. There are limited reports on its use in pediatric patients with ESRD. Here, we discuss the use of deferasirox to treat iron overload in a 15-year-old male with SCD, ESRD from granulomatosis with polyangiitis, and dependent on hemodialysis. We also review the literature on similar uses of deferasirox in adult patients with ESRD.
Collapse
|
28
|
The Toxic Influence of Excess Free Iron on Red Blood Cells in the Biophysical Experiment: An In Vitro Study. J Toxicol 2022; 2022:7113958. [PMID: 35256882 PMCID: PMC8898121 DOI: 10.1155/2022/7113958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/10/2022] [Indexed: 11/18/2022] Open
Abstract
Iron is needed for life-essential processes, but free iron overload causes dangerous clinical consequences. The study of the role of red blood cells (RBCs) in the influence of excess free iron in the blood on the pathological consequences in an organism is relevant. Here, in a direct biophysical experiment in vitro, we studied the action of free iron overload on the packed red blood cell (pRBC) characteristics. In experiments, we incubated pRBCs with the ferrous sulfate solution (Fe2+). Wе used free iron in a wide range of concentrations. High Fe2+ concentrations made us possible to establish the pattern of the toxic effect of excess iron on pRBCs during a reduced incubation time in a biophysical experiment in vitro. It was found that excess free iron causes changes in pRBC morphology, the appearance of bridges between cells, and the formation of clots, increasing the membrane stiffness and methemoglobin concentration. We created a kinetic model of changes in the hemoglobin derivatives. The complex of simultaneous distortions of pRBCs established in our experiments can be taken into account when studying the mechanism of the toxic influence of excess free iron in the blood on pathological changes in an organism.
Collapse
|
29
|
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
|
30
|
Soulières D, Mercier-Ross J, Fradette C, Rozova A, Tsang YC, Tricta F. The pharmacokinetic and safety profile of single-dose deferiprone in subjects with sickle cell disease. Ann Hematol 2022; 101:533-539. [PMID: 34981144 PMCID: PMC8810455 DOI: 10.1007/s00277-021-04728-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/18/2021] [Indexed: 12/16/2022]
Abstract
Abstract
Patients with sickle cell disease (SCD) who undergo repeated blood transfusions often develop iron overload. Deferiprone (Ferriprox®) is an oral iron chelator indicated for the treatment of transfusional iron overload due to thalassemia syndromes and has been recently approved as a treatment for iron overload in adult and pediatric patients with SCD and other anemias. The present study aims to characterize the pharmacokinetic (PK) profile of deferiprone (DFP) in adult subjects with SCD. In this phase I, open-label study, subjects with SCD were administered a single 1500 mg dose of DFP. Blood and urine samples were collected for PK assessments of DFP and its main metabolite, deferiprone 3-O-glucuronide (DFP-G). Eight subjects were enrolled and completed the study. Following drug administration, serum levels of DFP and DFP-G rose to maximum concentrations at 1.0 and 2.8 h post-dose, respectively. The half-lives of DFP and DFP-G were 1.5 and 1.6 h, respectively. The majority of administered drug was metabolized and excreted as DFP-G, with less than 4% excreted unchanged in urine up to 10 h post-dose. Subjects received a safety assessment 7 (± 3) days post-dose. Two subjects reported mild adverse events unrelated to the study drug, and no other safety concerns were reported. The PK profile of DFP in SCD subjects is consistent with previous reports in healthy adult volunteers, suggesting no special dosing adjustments are indicated for this population. These findings provide valuable insight for treating iron overload in patients with SCD, who have limited chelation therapy treatment options (trial registration number: NCT01835496, date of registration: April 19, 2013).
Collapse
|
31
|
Soliman AT, De Sanctis V, Yassin M, Alshurafa A, Ata F, Nashwan A. Blood transfusion and iron overload in patients with Sickle Cell Disease (SCD): Personal experience and a short update of diabetes mellitus occurrence. ACTA BIO-MEDICA : ATENEI PARMENSIS 2022; 93:e2022291. [PMID: 36043959 PMCID: PMC9534241 DOI: 10.23750/abm.v93i4.13330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 06/16/2022] [Indexed: 01/26/2023]
Abstract
The conventional treatment of β-thalassemia (β-TM) patients is based on the correction of anemia through regular blood transfusions and iron chelation therapy. However, allogeneic hematopoietic stem cell transplantation (HSCT) remains the only currently available technique that has curative potential. Variable frequency and severity of long-term growth and endocrine changes after conventional treatment as well as after HSCT have been reported by different centers. The goal of this mini-review is to summarize and update knowledge about long-term growth and endocrine changes after HSCT in patients with β-TM in comparison to those occurring in β-TM patients on conventional treatment. Regular surveillance, early diagnosis, treatment, and follow-up in a multi-disciplinary specialized setting are suggested to optimize the patient's quality of life (www.actabiomedica.it).
Collapse
Affiliation(s)
| | - Vincenzo De Sanctis
- Pediatric and Adolescent Outpatient Clinic, Quisisana Hospital, Ferrara, Italy
| | - Mohamed Yassin
- Department of Hematology, Hamad Medical Center, Doha, Qatar
| | - Awni Alshurafa
- Department of Hematology, Hamad Medical Center, Doha, Qatar
| | - Fateen Ata
- Department of Hematology, Hamad Medical Center, Doha, Qatar
| | | |
Collapse
|
32
|
Elbostany EA, Elghoroury EA, Thabet EH, Rashad AA, Rasheed EA, El-Saeed GSM, Abdelhalim DA, Abdelfattah SN, Salama II, Salama N. Oxidative stress and hepcidin expression in pediatric sickle cell anemia with iron overload. Hematol Oncol Stem Cell Ther 2021; 16:238-244. [PMID: 34883086 DOI: 10.1016/j.hemonc.2021.11.003] [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: 06/07/2021] [Revised: 10/10/2021] [Accepted: 11/14/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Blood transfusion (BT) is essential in treating sickle cell disease (SCD); however, it leads to iron overload (IO) and oxidative stress. We studied the relationship between oxidative stress, iron status parameters, hepcidin mRNA gene expression, and IO in SCD patients. METHODS We classified all SCD patients (n = 90) into two groups: Group I, 45 children (s.ferritin ≥ 938 ng/mL) and Group II, 45 children (s.ferritin < 938 ng/mL). A total of 55 children, age and sex matched, participated as a control group. Malondialdehyde (MDA), nitrite, s.iron, s.total iron-binding capacity (sTIBC), transferrin saturation %, s.ferritin, s.hepcidin, and hepcidin mRNA gene expression were assessed. RESULTS Among SCD BT-dependent patients (>3 times/year), 63% were from Group I and 37% from Group II, p < .01. The two patient groups had significantly lower s.hepcidin and hepcidin gene expression than controls (p < .001). TIBC, s.iron, s.ferritin, transferrin saturation %, ferritin/hepcidin ratio, and MDA levels were higher among SCD patients than controls (p < .001). Group I had higher mean level of ferritin/hepcidin ratio and MDA than Group II (p < .01). The higher level of MDA and increased frequency of BT were the significant predicting risk factors for IO (p < .05). A receiver-operating characteristic curve indicates that MDA is the outstanding significant biomarker for high level of s.ferritin with subsequent IO progression. CONCLUSION MDA may serve as a biomarker of oxidative stress and IO in SCD patients. This result paid attention for urgent initiation of antioxidant and chelation therapy on detecting increased MDA level.
Collapse
Affiliation(s)
- Eman A Elbostany
- Department of Pediatrics, National Research Centre, Cairo, Egypt
| | - Eman A Elghoroury
- Department of Clinical and Chemical Pathology, National Research Centre, Cairo, Egypt.
| | - Eman H Thabet
- Department of Clinical and Chemical Pathology, National Research Centre, Cairo, Egypt
| | - Alaa A Rashad
- Department of Pediatrics, National Research Centre, Cairo, Egypt
| | - Enas A Rasheed
- Department of Clinical and Chemical Pathology, National Research Centre, Cairo, Egypt
| | | | - Dalia A Abdelhalim
- Department of Clinical and Chemical Pathology, National Research Centre, Cairo, Egypt
| | - Safa N Abdelfattah
- Department of Clinical and Chemical Pathology, National Research Centre, Cairo, Egypt
| | - Iman I Salama
- Department of Community Medicine Research, National Research Centre, Cairo, Egypt
| | - Niveen Salama
- Department of pediatrics, Faculty of Medicine, Cairo University, Egypt
| |
Collapse
|
33
|
Allam MFAB, Samra MFA, Rahman ASMA. The added value of chemical shift imaging in evaluation of bone marrow changes in sickle cell disease. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2021. [DOI: 10.1186/s43055-020-00384-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The aim of this study was to assess the added value of chemical shift imaging when used with routine MRI study in evaluation of bone marrow changes in SCD. Forty-two patients with SCD and bone pain were included in the study; they underwent CSI and routine MRI study on the symptomatic anatomic part of the skeleton.
Results
Four patterns of diffuse bone marrow changes were recognized; they varied from persistent red marrow to diffuse hypointense patterns with abnormal signal loss percentage on CSI that suggest presence of iron overload (n = 28, 66.6%). Serum ferritin level was increasing in accordance to the degree of signal changes found on CSI with significant high negative correlation between the percentage of signal loss on CSI obtained from IP-OP/IP formula and serum ferritin level. In focal marrow lesions, all T1 hyperintense lesions demonstrated corresponding hyperintensity on IP and OP; the detection frequency on CSI was relatively higher on OP compared with IP images.
Conclusion
CSI has high diagnostic performance in detecting diffuse marrow changes and development of iron overload in SCD. In SCD-related focal marrow lesions, CSI could have a complementary role in detection of T1 hyperintensity and lesion conspicuity.
Collapse
|
34
|
Alkindi S, Panjwani V, Al-Rahbi S, Al-Saidi K, Pathare AV. Iron Overload in Patients With Heavily Transfused Sickle Cell Disease-Correlation of Serum Ferritin With Cardiac T2 * MRI (CMRTools), Liver T2 * MRI, and R2-MRI (Ferriscan®). Front Med (Lausanne) 2021; 8:731102. [PMID: 34760898 PMCID: PMC8573209 DOI: 10.3389/fmed.2021.731102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/17/2021] [Indexed: 11/13/2022] Open
Abstract
The treatment of sickle cell disease (SCD) is mainly supportive, except for a minority, who receive bone marrow transplantation (BMT). Serum ferritin (SF) is routinely available but is notoriously unreliable as a tool for iron-overload assessment since it is an acute-phase reactant. Although blood transfusion is one of the most effective ways to deal with specific acute and chronic complications of SCD, this strategy is often associated with alloimmunization, iron overload, and hemolytic reactions. This study, thus, aims to evaluate iron overload in patients with SCD on chronic blood transfusions and specifically, correlate SF with the current standard of care of iron-overload assessment using MRI-based imaging techniques. Amongst a historic cohort of 58 chronically transfused patients with SCD, we were able to evaluate 44 patients who are currently alive and had multiple follow-up testing. Their mean age (±SD) was 35 (9) years and comprised of 68.2% of women. The studied iron-overload parameters included cardiac T2* MRI, liver iron concentration (LIC) by Liver T2* MRI, and serial SF levels. Additionally, in a smaller cohort, we also studied LIC by FerriScan© R2-MRI. Chronic blood transfusions were necessary for severe vaso-occlusive crisis (VOC) (38.6%), severe symptomatic anemia (38.6%), past history of stroke (15.9%), and recurrent acute chest syndrome (6.9%). About 14 (24%) patients among the original cohort died following SCD-related complications. Among the patients currently receiving chelation, 26 (96%) are on Deferasirox (DFX) [Jadenu® (24) or Exjade® (2)], with good compliance and tolerance. However, one patient is still receiving IV deferoxamine (DFO), in view of the significantly high systemic iron burden. In this evaluable cohort of 44 patients, the mean SF (±SD) reduced marginally from 4,311 to 4,230 ng/ml, mean Liver T2* MRI dropped from 12 to 10.3 mg/gm dry weight, while the mean cardiac T2*MRI improved from 36.8 to 39.5 ms. There was a mild to moderate correlation between the baseline and final values of SF ng/ml, r = 0.33, p = 0.01; Cardiac T2* MRI ms, r = 0.3, p = 0.02 and Liver T2* MRI mg/kg dry weight, r = 0.6, p < 0.001. Overall, there was a positive correlation between SF and Liver T2* MRI (Pearson's r = 0.78, p < 0.001). Cardiac T2*MRI increased with the decreasing SF concentration, showing a negative correlation which was statistically significant (Pearson's r = -0.6, p < 0.001). Furthermore, there was an excellent correlation between SF ng/ml and LIC by FerriScan© R2-MRI mg/g or mmol/kg (Spearmen's rho = -0.723, p < 0.008) in a small subset of patients (n = 14) who underwent the procedure. In conclusion, our study demonstrated a good correlation between serial SF and LIC by either Liver MRI T2* or by FerriScan© R2-MRI, even though SF is an acute-phase reactant. It also confirms the cardiac sparing effect in patients with SCD, even with the significant transfusion-related iron burden. About 14 (24%) patients of the original cohort died over the past 15 years, indicative of a negative impact of iron overload on disease morbidity and mortality.
Collapse
Affiliation(s)
- Salam Alkindi
- Department of Hematology, Sultan Qaboos University Hospital, Muscat, Oman.,College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Vinodh Panjwani
- Department of Hematology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Sarah Al-Rahbi
- Department of Hematology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Khalid Al-Saidi
- Department of Hematology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Anil V Pathare
- Department of Hematology, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
35
|
Al-Khateeb RS, Althagafy HS, ElAssouli MZ, Nori DA, AlFattani M, Al-Najjar SA, Al Amri T, Hashem AM, Harakeh S, Helmi N. Iron Chelation Reduces DNA Damage in Sickle Cell Anemia. Clin Appl Thromb Hemost 2021; 27:10760296211047230. [PMID: 34633875 PMCID: PMC8521763 DOI: 10.1177/10760296211047230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Sickle cell anemia (SCA) is a blood condition that causes severe pain. One of the therapeutic agents used for the treatment of SCA is hydroxyurea, which reduces the episodes of pain but causes DNA damage to white blood cells. The aim of this study was to evaluate the efficacy of the combination of hydroxyurea and iron chelation therapy in relation to the extent of DNA-associated damage. Blood samples were collected from 120 subjects from five groups. Various hematological parameters of the obtained serum were analyzed. The amount of damage caused to their DNA was detected using the comet assay and fluorescent microscopy techniques. The percentage of DNA damage in the group that was subjected to the combination therapy (target group) was 1.32% ± 1.51%, which was significantly lower (P < .05) than that observed in the group treated with hydroxyurea alone (6.36% ± 2.36%). While the target group showed comparable levels of hemoglobin F and lactate dehydrogenase compared to the group that was treated with hydroxyurea alone, highly significant levels of transferrin receptors and ferritin were observed in the target group. The results of this study revealed that the administration of iron chelation drugs with hydroxyurea may help improve patients' health and prevent the DNA damage caused to white blood cells due to hydroxyurea. Further studies are needed to better understand the underlying mechanisms that are involved in this process.
Collapse
Affiliation(s)
- Rawan S Al-Khateeb
- 37848Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia, KSA
| | | | - Mohammad Zaki ElAssouli
- 37848Vaccines and Immunotherapy Unit, King Fahd Medical Research Center (KFMRC); Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, KSA
| | - Dunya A Nori
- 37848Department of Biochemistry, King Abdulaziz University, Jeddah, Saudi Arabia, KSA
| | | | | | - Turki Al Amri
- 37848Family and Community Medicine Department, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anwar M Hashem
- 37848Vaccines and Immunotherapy Unit, King Fahd Medical Research Center (KFMRC); Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, KSA
| | - Steve Harakeh
- 37848Special Infectious Agents Unit, and Yousef Abdullatif Jameel Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, KSA
| | - Nawal Helmi
- 441424Department of Biochemistry, University of Jeddah, Jeddah, KSA
| |
Collapse
|
36
|
Leal LB, Nogueira MS, Mageski JGA, Martini TP, Barauna VG, Dos Santos L, de Carvalho LFDCES. Diagnosis of Systemic Diseases Using Infrared Spectroscopy: Detection of Iron Overload in Plasma-Preliminary Study. Biol Trace Elem Res 2021; 199:3737-3751. [PMID: 33415581 DOI: 10.1007/s12011-020-02510-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Despite the important role of iron in cellular homeostasis, iron overload (IO) is associated with systemic and tissue deposits which damage several organs. In order to reduce the impact caused by IO, invasive diagnosis exams (e.g., biopsies) and minimally invasive methods were developed including computed tomography and magnetic resonance imaging. However, current diagnostic methods are still time-consuming and expensive. A cost-effective solution is using Fourier-transform infrared spectroscopy (FTIR) for real-time and molecular-sensitive biofluid analysis during conventional laboratory exams. In this study, we performed the first evaluation of the accuracy of FTIR for IO diagnosis. The study was performed by collecting FTIR spectra of plasma samples of five rats intravenously injected with iron-dextran and five control rats. We developed a classification model based on principal component analysis and supervised methods including J48, random forest, multilayer perceptron, and radial basis function network. We achieved 100% accuracy for the classification of the IO status and provided a list of possible biomolecules related to the vibrational modes detected. In this preliminary study, we give a first step towards real-time diagnosis for acute IO or intoxication. Furthermore, we have expanded the literature knowledge regarding the pathophysiological changes induced by iron overload.
Collapse
Affiliation(s)
- Leonardo Barbosa Leal
- Department of Physiological Sciences, Federal University of Espírito Santo (UFES), Marechal Campos Ave, 1468, Maruípe, Vitória, Espírito Santo, 29040-090, Brazil.
| | - Marcelo Saito Nogueira
- Tyndall National Institute/University College Cork - Lee Maltings Complex, Dyke Parade, Cork, T12R5CP, Ireland
| | - Jandinay Gonzaga Alexandre Mageski
- Department of Physiological Sciences, Federal University of Espírito Santo (UFES), Marechal Campos Ave, 1468, Maruípe, Vitória, Espírito Santo, 29040-090, Brazil
| | - Thiago Pereira Martini
- Institute of Science and Technology, Federal University of Sao Paulo, São José dos Campos, Brazil
| | - Valério Garrone Barauna
- Department of Physiological Sciences, Federal University of Espírito Santo (UFES), Marechal Campos Ave, 1468, Maruípe, Vitória, Espírito Santo, 29040-090, Brazil
| | - Leonardo Dos Santos
- Department of Physiological Sciences, Federal University of Espírito Santo (UFES), Marechal Campos Ave, 1468, Maruípe, Vitória, Espírito Santo, 29040-090, Brazil
| | - Luis Felipe das Chagas E Silva de Carvalho
- Universidade de Taubaté. R. dos Operários, 09 - Centro, Taubaté, São Paulo, 12020-340, Brazil
- Centro Universitário Braz Cubas, Av. Francisco Rodrigues Filho, 1233 - Vila Mogilar, Mogi das Cruzes, São Paulo, Brazil
| |
Collapse
|
37
|
Wilson SR, Sears M, Williams E, Drapekin J, Sivakumar I, Padrino S, Desai PC, Hager W, Manwani D, Clay LJ, Field JJ, Kanter J, Cohen AJ, Saif Ur Rehman S, Lanzkron S, Little JA. Gaps in the diagnosis and management of iron overload in sickle cell disease: a 'real-world' report from the GRNDaD registry. Br J Haematol 2021; 195:e157-e160. [PMID: 34423416 DOI: 10.1111/bjh.17762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Samuel R Wilson
- Department of Medicine, Division of Hematology, University of North Carolina, Chapel Hill, NC, USA.,Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of North Carolina, Chapel Hill, NC, USA.,UNC Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Matthew Sears
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Medicine, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth Williams
- Department of Medicine, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jesse Drapekin
- School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Department of Medicine, The University of Chicago Medical Center, Chicago, IL, USA
| | - Ishwarya Sivakumar
- Department of Medicine, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susan Padrino
- School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Payal C Desai
- Department of Hematology, The Ohio State University, Columbus, OH, USA
| | - Ward Hager
- Department of Pediatrics, UCSF Benioff Children's Hospital, Oakland, CA, USA
| | - Deepa Manwani
- Division of Pediatric Hematology, Oncology, Marrow and Blood Cell Transplantation, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Leila J Clay
- Division of Hematology, Johns Hopkins All Children's Hospital, Johns Hopkins University School of Medicine, St. Petersburg, FL, USA
| | - Joshua J Field
- Blood Center of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Julie Kanter
- Division of Hematology and Oncology, University of Alabama Birmingham, Birmingham, AL, USA
| | | | - Sana Saif Ur Rehman
- Department of Medicine, Division of Hematology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sophie Lanzkron
- Department of Medicine, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jane A Little
- Department of Medicine, Division of Hematology, University of North Carolina, Chapel Hill, NC, USA.,UNC Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | | |
Collapse
|
38
|
Linder GE, Chou ST. Red cell transfusion and alloimmunization in sickle cell disease. Haematologica 2021; 106:1805-1815. [PMID: 33792218 PMCID: PMC8252926 DOI: 10.3324/haematol.2020.270546] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Indexed: 01/23/2023] Open
Abstract
Red cell transfusion remains a critical component of care for acute and chronic complications of sickle cell disease. Randomized clinical trials demonstrated the benefits of transfusion therapy for prevention of primary and secondary strokes and postoperative acute chest syndrome. Transfusion for splenic sequestration, acute chest syndrome, and acute stroke are guided by expert consensus recommendations. Despite overall improvements in blood inventory safety, adverse effects of transfusion are prevalent among patients with sickle cell disease and include alloimmunization, acute and delayed hemolytic transfusion reactions, and iron overload. Judicious use of red cell transfusions, optimization of red cell antigen matching, and the use of erythrocytapheresis and iron chelation can minimize adverse effects. Early recognition and management of hemolytic transfusion reactions can avert poor clinical outcomes. In this review, we discuss transfusion methods, indications, and complications in sickle cell disease with an emphasis on alloimmunization.
Collapse
Affiliation(s)
- Grace E Linder
- Department of Pathology and Lab Medicine, Children's Hospital of Philadelphia
| | - Stella T Chou
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA.
| |
Collapse
|
39
|
Samuel Olufemi A, Abiodun Folashade A, Tinuade Adetutu O. Burden of iron overload among non-chronically blood transfused preschool children with sickle cell anaemia. Afr Health Sci 2021; 21:753-758. [PMID: 34795732 PMCID: PMC8568236 DOI: 10.4314/ahs.v21i2.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Sickle cell disease is the commonest genetic disorder of haemoglobin due to inheritance of mutant haemoglobin genes from both parents. The disorder is characterized by chronic haemolysis which results in increased availability of iron from red blood cell destructions. OBJECTIVE To determine the prevalence of iron overload among non-chronically blood transfused preschool children with sickle cell anaemia. METHODS Serum ferritin was assayed and transferrin saturation derived in 97 steady state sickle cell anaemia children. Elevated iron stores were defined as serum ferritin level >300ng/ml, and transferrin saturation >45%. RESULTS Serum ferritin level was greater than 300 mg/ml in 14 (14.4%) subjects and transferrin saturation >45% in six (6.2%) subjects with sickle cell anaemia. The prevalence of iron overload was 20.6%. The prevalence of iron overload was higher among subjects in older age group, female, with history of blood transfusion, and with single blood transfusion session. CONCLUSION Iron overload is prevalent in older children; the number of blood transfusion sessions notwithstanding. Regular assessment of serum ferritin is recommended.
Collapse
|
40
|
MRI-based R2* mapping in patients with suspected or known iron overload. Abdom Radiol (NY) 2021; 46:2505-2515. [PMID: 33388804 DOI: 10.1007/s00261-020-02912-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 01/19/2023]
Abstract
PURPOSE R2* relaxometry is a quantitative method for assessment of iron overload. The purpose is to analyze the cross-sectional relationships between R2* in organs across patients with primary and secondary iron overload. Secondary analyses were conducted to analyze R2* according to treatment regimen. METHODS This is a retrospective, cross-sectional, institutional review board-approved study of eighty-one adult patients with known or suspected iron overload. R2* was measured by segmenting the liver, spleen, bone marrow, pancreas, renal cortex, renal medulla, and myocardium using breath-hold multi-echo gradient-recalled echo imaging at 1.5 T. Phlebotomy, transfusion, and chelation therapy were documented. Analyses included correlation, Kruskal-Wallis, and post hoc Dunn tests. p < 0.01 was considered significant. RESULTS Correlations between liver R2* and that of the spleen, bone marrow, pancreas, and heart were respectively 0.49, 0.33, 0.27, and 0.34. R2* differed between patients with primary and secondary overload in the liver (p < 0.001), spleen (p < 0.001), bone marrow (p < 0.01), renal cortex (p < 0.001), and renal medulla (p < 0.001). Liver, spleen, and bone marrow R2* were higher in thalassemia than in hereditary hemochromatosis (all p < 0.01). Renal cortex R2* was higher in sickle cell disease than in hereditary hemochromatosis (p < 0.001) and in thalassemia (p < 0.001). Overall, there was a trend toward lower liver R2* in patients assigned to phlebotomy and higher liver R2* in patients assigned to transfusion and chelation therapy. CONCLUSION R2* relaxometry revealed differences in degree or distribution of iron overload between organs, underlying etiologies, and treatment.
Collapse
|
41
|
Blood donation and heavy metal poisoning in developing nations: Any link? Transfus Apher Sci 2021; 60:103067. [PMID: 33541762 DOI: 10.1016/j.transci.2021.103067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/04/2021] [Accepted: 01/09/2021] [Indexed: 11/24/2022]
Abstract
Long term health effects of heavy metal exposure from both occupational and environmental settings involve multi-organ toxicities including but not limited to disturbances of neurological, cognitive, and metabolic processes, immune system dysregulation, carcinogenesis and sometimes permanent disabilities. Humans are exposed to toxic metals through various sources and routes of entry. The risk of heavy metal poisoning from donor blood has been the subject of many scientific investigations. In this review we highlight how the access to a safe and adequate blood transfusion with minimal risk of toxic metals to recipients is a public health challenge, especially in developing nations. For quality assurance purposes, blood donors are screened for various blood-borne pathogens, but screening for toxic metal levels is not routine. Evidence from scientific studies used in this review lends credence to the risk of heavy metal poisoning from donors with high blood concentrations of these heavy metals. The risk of toxicity is exceptionally high in vulnerable populations such as neonates and preterm infants, as well as in pregnant women and other individuals with conditions requiring multiple blood transfusions. This is worse in developing countries where some members of the population engage in illegal refining and artisanal mining activities. In order to reduce toxic metal exposure in vulnerable populations, blood meant for transfusion in vulnerable subjects, e.g. children, should be routinely screened for heavy metal concentrations. Patients receiving multiple blood transfusions should also be monitored for iron overload and its attendant toxicities.
Collapse
|
42
|
Garcia AJ, Okeagu CN, Kaye AD, Abd-Elsayed A. Metabolism, Pathophysiology, and Clinical Considerations of Iron Overload, a Comprehensive Review. ESSENTIALS OF BLOOD PRODUCT MANAGEMENT IN ANESTHESIA PRACTICE 2021:289-299. [DOI: 10.1007/978-3-030-59295-0_28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
43
|
The VP1u of Human Parvovirus B19: A Multifunctional Capsid Protein with Biotechnological Applications. Viruses 2020; 12:v12121463. [PMID: 33352888 PMCID: PMC7765992 DOI: 10.3390/v12121463] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
The viral protein 1 unique region (VP1u) of human parvovirus B19 (B19V) is a multifunctional capsid protein with essential roles in virus tropism, uptake, and subcellular trafficking. These functions reside on hidden protein domains, which become accessible upon interaction with cell membrane receptors. A receptor-binding domain (RBD) in VP1u is responsible for the specific targeting and uptake of the virus exclusively into cells of the erythroid lineage in the bone marrow. A phospholipase A2 domain promotes the endosomal escape of the incoming virus. The VP1u is also the immunodominant region of the capsid as it is the target of neutralizing antibodies. For all these reasons, the VP1u has raised great interest in antiviral research and vaccinology. Besides the essential functions in B19V infection, the remarkable erythroid specificity of the VP1u makes it a unique erythroid cell surface biomarker. Moreover, the demonstrated capacity of the VP1u to deliver diverse cargo specifically to cells around the proerythroblast differentiation stage, including erythroleukemic cells, offers novel therapeutic opportunities for erythroid-specific drug delivery. In this review, we focus on the multifunctional role of the VP1u in B19V infection and explore its potential in diagnostics and erythroid-specific therapeutics.
Collapse
|
44
|
Pinto VM, Forni GL. Management of Iron Overload in Beta-Thalassemia Patients: Clinical Practice Update Based on Case Series. Int J Mol Sci 2020; 21:E8771. [PMID: 33233561 PMCID: PMC7699680 DOI: 10.3390/ijms21228771] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 12/29/2022] Open
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.
Collapse
Affiliation(s)
- Valeria Maria Pinto
- Centro della Microcitemia e delle Anemie Congenite Ente Ospedaliero Ospedali Galliera, Via Volta 6, 16128 Genoa, Italy;
| | | |
Collapse
|
45
|
Özbolat G, Alizadeh Yegani A. Synthesis, characterization, biological activity and electrochemistry studies of iron(III) complex with curcumin-oxime ligand. Clin Exp Pharmacol Physiol 2020; 47:1834-1842. [PMID: 32497256 DOI: 10.1111/1440-1681.13359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 12/17/2022]
Abstract
Iron overload is a key target in drug development. This study aimed to investigate the coordination of Fe(III) ions with a curcumin-oxime ligand that may be used in the treatment of iron overload. The synthesis of the curcumin-oxime ligand and curcumin-oxime-Fe(III) complex was successfully made and characterized in its solid-state and solution-state using FT-IR, UV-Vis, elemental analysis, and 1 H-NMR. However, in this study, we investigated the apoptotic effects of the curcumin-oxime Fe (III) complex on SW480. SW480 cells were exposed to 99.2% medium for 48 hours. After 48 hours, the incubation period, cells were harvested by centrifugation and washed in phosphate-buffered saline (PBS) and lysed in radio-immunoprecipitation assay (RIPA) buffer for 20 minutes and supernatants were taken and pellets were discarded. ELISA test was used to examine the expression, and activity of cleaved caspase-3, Bax, and Bcl-2 proteins in SW480 cells. ELISA test results indicated that the activities of apoptotic proteins Bax, caspase 3 and Bcl-2 in human SW480 cell lines significantly increased in 48 hours treatment. Also, the activity of Bcl-2 was observed to decrease significantly. Catalase activities of the complex were investigated. The findings showed that the complex has a catalase activity. The findings suggest that this type of complex may constitute a new and interesting basis for the future search of new and more potent drugs. The SOD activity of the result showed that the complexes possessed a considerable SOD activity with an IC50 value of 7.685 µM. Also, when compared with the control, a complex increased the SOD levels (P < .05). Electrochemistry studies in the literature have shown that the Fe3+ /Fe2+ couple redox process occurs in low potential. This value is within the range of compounds that are expected to show superoxide dismutase activity. The Ipc /Ipa shows that one electron transport takes place in the complex. Our results suggest that curcumin-oxime may represent a new approach in the treatment of iron overload.
Collapse
Affiliation(s)
| | - Arash Alizadeh Yegani
- Department of Pharmacology, Faculty of Veterinary, Mustafa Kemal University, Hatay, Turkey
| |
Collapse
|
46
|
Sharma D, Ogbenna AA, Kassim A, Andrews J. Transfusion support in patients with sickle cell disease. Semin Hematol 2020; 57:39-50. [PMID: 32892842 DOI: 10.1053/j.seminhematol.2020.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Indexed: 12/23/2022]
Abstract
Blood transfusions are an integral component of the management of acute and chronic complications of sickle cell disease. Red cells can be administered as a simple transfusion, part of a modified exchange procedure involving manual removal of autologous red cells and infusion of donor red cells, and part of an automated red cell exchange procedure using apheresis techniques. Individuals with sickle cell disease are at risk of multiple complications of blood transfusions, including transfusional hemosiderosis, auto- and alloimmunization to minor red cell and human leukocyte antigens, delayed hemolytic transfusion reactions, and hyper-hemolysis. In low- and middle-income countries in sub-Saharan Africa, where a directed donor system is prevalent and limited laboratory methods are in place to perform extended red cell phenotyping, leukodepletion of cellular products, and infectious disease screening, there are additional challenges to providing safe and adequate transfusion support for this patient population. We review current indications for acute and chronic transfusions in sickle cell disease that are derived primarily from randomized controlled trials and observational studies in children living in high-income countries. We will highlight populations with unique transfusion needs, such as pregnant women and children, as well as the role of the transfusion medicine consultative service for individuals with sickle cell disease planning to have curative hematopoietic stem cell transplantation or gene therapy. Finally, we will discuss risk factors for alloimmunization in individuals with sickle cell disease, emerging new strategies to prevent alloimmunization in this population, and critical gaps in the implementation of transfusion guidelines for sickle cell disease in high- and low-income countries.
Collapse
Affiliation(s)
- Deva Sharma
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Transfusion Medicine, Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ann Abiola Ogbenna
- Department of Hematology and Blood Transfusion, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Adetola Kassim
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt-Meharry Sickle Cell Center of Excellence, Vanderilt University Medical Center, Nashville, TN, USA.
| | - Jennifer Andrews
- Division of Transfusion Medicine, Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Hematology and Oncology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
47
|
Mamsen LS, Kristensen SG, Pors SE, Bøtkjær JA, Ernst E, Macklon KT, Gook D, Kumar A, Kalra B, Andersen CY. Consequences of β-Thalassemia or Sickle Cell Disease for Ovarian Follicle Number and Morphology in Girls Who Had Ovarian Tissue Cryopreserved. Front Endocrinol (Lausanne) 2020; 11:593718. [PMID: 33519708 PMCID: PMC7844814 DOI: 10.3389/fendo.2020.593718] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/26/2020] [Indexed: 01/18/2023] Open
Abstract
Women with β-thalassemia (BT) and sickle cell disease (SCD) have a high risk of infertility and premature ovarian insufficiency. Different fertility preserving strategies, including ovarian tissue cryopreservation (OTC) and oocyte cryopreservation has been considered, and healthy babies have been born after successful OTC and transplantation. We evaluated follicle number and follicle health in ovarian tissue from a cohort of BT and SCD patients who underwent OTC before the age of 18 years. Patients undergoing OTC from 2002 to 2019 were included. A total of 14 girls and adolescents with BT and four with SCD, aged 2.8-17.4 years at OTC were included together with a reference group of 43 girls and adolescents with non-anemia diseases considered to have normal ovaries aged 0.6-17.9 years at OTC. Ovarian follicle density was measured in cortex biopsies and compared to the reference group. Expression of proteins associated with follicular health was evaluated using immunohistochemistry. Follicles were detected in the ovarian cortex biopsies from all patients with BT and SCD. The follicle densities were within the 95% prediction interval of the reference group in all cases. A similar expression of six proteins essential for follicular health was detected using immunohistochemistry in BT, SCD, and references. OTC should be considered an option for young girls and adolescents with BT and SCD.
Collapse
Affiliation(s)
- Linn Salto Mamsen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- *Correspondence: Linn Salto Mamsen,
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Erik Ernst
- Department of Obstetrics and Gynaecology, Regional Hospital Horsens, Horsens, Denmark
| | - Kirsten Tryde Macklon
- The Fertility Clinic, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Debra Gook
- Reproductive Services and Melbourne IVF, Royal Women’s Hospital, Parkville, VIC, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
| | - Ajay Kumar
- Ansh Labs LLC, Webster, TX, United States
| | | | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children, and Reproduction, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
48
|
Coates TD. Iron overload in transfusion-dependent patients. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:337-344. [PMID: 31808901 PMCID: PMC6913424 DOI: 10.1182/hematology.2019000036] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Before the advent of effective iron chelation, death from iron-induced cardiomyopathy occurred in the second decade in patients with transfusion-dependent chronic anemias. The advances in our understanding of iron metabolism; the ability to monitor iron loading in the liver, heart, pancreas and pituitary; and the availability of several effective iron chelators have dramatically improved survival and reduced morbidity from transfusion-related iron overload. Nevertheless, significantly increased survival brings about new complications such as malignant transformation resulting from prolonged exposure to iron, which need to be considered when developing long-term therapeutic strategies. This review discusses the current biology of iron homeostasis and its close relation to marrow activity in patients with transfusion-dependent anemias, and how biology informs clinical approach to treatment.
Collapse
Affiliation(s)
- Thomas D Coates
- Hematology Section, Children's Center for Cancer, Blood Diseases and Bone Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA
| |
Collapse
|
49
|
Wurschi GW, Mentzel HJ, Herrmann KH, Krumbein I, Beck JF, Reichenbach JR, Kentouche K. MRI as an alternative to serum ferritin for diagnosis of iron overload in children in the context of immune response after stem cell transplantation. Pediatr Transplant 2019; 23:e13583. [PMID: 31535426 DOI: 10.1111/petr.13583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/18/2019] [Accepted: 08/07/2019] [Indexed: 12/30/2022]
Abstract
Multiple blood cell transfusions may cause iron overload or even liver fibrosis, requiring early diagnosis and intervention. SF is the standard for estimating iron levels in the body, but it also increases with inflammation. We hypothesized that T2 * magnetic resonance (MR) relaxometry is a more accurate alternative for follow-up in pediatric patients before and after allogenic SCT. Twenty-three children (mean age 10.2 years, 10 female, 13 male) were evaluated prospectively before SCT as well as at least 1 year after SCT with T2 * relaxometry on a 1.5 T MR-scanner to estimate liver iron concentrations from the T2 * values ("MR-Fe"). The results were compared with SF, while also considering CRP, and correlated with the number of transfusions. Overall, 24.3 transfusions were administered in average, mainly within 100 days of SCT (mean 10.5 units). Both MR-Fe and SF increased after SCT and decreased in the absence of new transfusions 1 year later without chelate therapy. This suggests regeneration of LP and iron loss, although the original states were not reached. Additionally, simultaneous peaks of CRP and SF were observed directly after SCT. MR-Fe did neither reveal these peaks nor was it associated with CRP (P = .39). We postulate that these early CRP and SF peaks after SCT are probably related to inflammatory reactions and not to iron overload. Thus, SF is not reliable for iron overload diagnosis after SCT in every condition. Beside this interaction, SF and MR-Fe revealed similar accuracy. MRI, however, has practical and economical disadvantages in routine estimation of iron.
Collapse
Affiliation(s)
- Georg W Wurschi
- Section of Pediatric Radiology, Institute of Diagnostic and Interventional Radiology, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Hans-Joachim Mentzel
- Section of Pediatric Radiology, Institute of Diagnostic and Interventional Radiology, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Karl-Heinz Herrmann
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Ines Krumbein
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - James F Beck
- Section of Pediatric Hematology and Oncology, Department of Pediatrics, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Juergen R Reichenbach
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Karim Kentouche
- Section of Pediatric Hematology and Oncology, Department of Pediatrics, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
50
|
GSTM1 and Liver Iron Content in Children with Sickle Cell Anemia and Iron Overload. J Clin Med 2019; 8:jcm8111878. [PMID: 31694285 PMCID: PMC6912836 DOI: 10.3390/jcm8111878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/30/2019] [Accepted: 10/31/2019] [Indexed: 01/15/2023] Open
Abstract
Chronic blood transfusions in patients with sickle cell anemia (SCA) cause iron overload, which occurs with a degree of interpatient variability in serum ferritin and liver iron content (LIC). Reasons for this variability are unclear and may be influenced by genes that regulate iron metabolism. We evaluated the association of the copy number of the glutathione S-transferase M1 (GSTM1) gene and degree of iron overload among patients with SCA. We compared LIC in 38 children with SCA and ≥12 lifetime erythrocyte transfusions stratified by GSTM1 genotype. Baseline LIC was measured using magnetic resonance imaging (MRI), R2*MRI within 3 months prior to, and again after, starting iron unloading therapy. After controlling for weight-corrected transfusion burden (mL/kg) and splenectomy, mean pre-chelation LIC (mg/g dry liver dry weight) was similar in all groups: GSTM1 wild-type (WT) (11.45, SD±6.8), heterozygous (8.2, SD±4.52), and homozygous GSTM1 deletion (GSTM1-null; 7.8, SD±6.9, p = 0.09). However, after >12 months of chelation, GSTM1-null genotype subjects had the least decrease in LIC compared to non-null genotype subjects (mean LIC change for GSTM1-null = 0.1 (SD±3.3); versus -0.3 (SD±3.0) and -1.9 (SD±4.9) mg/g liver dry weight for heterozygous and WT, respectively, p = 0.047). GSTM1 homozygous deletion may prevent effective chelation in children with SCA and iron overload.
Collapse
|