1
|
Udeze C, Dovizio M, Veronesi C, Esposti LD, Li N, Dang TXMP, Forni GL. Mortality, Clinical Complications, and Healthcare Resource Utilization Associated with Managing Transfusion-Dependent β-Thalassemia and Sickle Cell Disease with Recurrent Vaso-occlusive Crises in Italy. PHARMACOECONOMICS - OPEN 2024:10.1007/s41669-024-00532-4. [PMID: 39468000 DOI: 10.1007/s41669-024-00532-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/30/2024]
Abstract
OBJECTIVE To examine the clinical burden and healthcare resource utilization (HCRU) among patients with transfusion-dependent β-thalassemia (TDT) and patients with sickle cell disease (SCD) with recurrent vaso-occlusive crises (VOCs) in Italy. METHODS Eligible patients were identified from an administrative claims database from 1 January 2010 and 1 February 2019. Patients with TDT had ≥ 1 iron chelation treatment, ≥ 8 red blood cell transfusions (RBCTs) during any 12-month period, and ≥ 12 months of available data pre- and post-index (i.e., first RBCT claim). Patients with SCD with recurrent VOCs had ≥ 2 VOCs/year in ≥ 2 consecutive years and ≥ 12 months of available data pre- and post-index (second VOC claim in the second of 2 consecutive years). Patients were propensity score matched to five controls by age, sex, geographic area, and index year. Clinical and HCRU outcomes were evaluated post-index. RESULTS In total, 214 patients with TDT and 111 patients with SCD with recurrent VOCs were matched to 1070 and 555 controls, respectively. Both patient groups had substantially higher mortality rates than controls (TDT: 4.8 versus 0.8 deaths per 100 person-years; SCD: 1.6 versus 0.4 deaths per 100 person-years). Clinical complications were prevalent in both patient groups. Compared with controls, both patient groups had significantly higher mean rates of all-cause hospitalizations (TDT: 1.4 versus 0.1; SCD: 2.0 versus 0.1) and outpatient services (TDT: 21.9 versus 1.6; SCD: 6.2 versus 1.0) per patient per year (all: p < 0.05). CONCLUSIONS Management of TDT and SCD in Italy is associated with significant clinical and health system burden, highlighting the need for new treatments that eliminate RBCTs and VOCs.
Collapse
Affiliation(s)
- Chuka Udeze
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, MA, 02210, USA.
| | | | | | | | - Nanxin Li
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, MA, 02210, USA
| | | | | |
Collapse
|
2
|
Bou-Fakhredin R, Cappellini MD, Taher AT, De Franceschi L. Hypercoagulability in hemoglobinopathies: Decoding the thrombotic threat. Am J Hematol 2024. [PMID: 39400943 DOI: 10.1002/ajh.27500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/15/2024]
Abstract
Beta (β)-thalassemia and sickle cell disease (SCD) are characterized by a hypercoagulable state, which can significantly influence organ complication and disease severity. While red blood cells (RBCs) and erythroblasts continue to play a central role in the pathogenesis of thrombosis in β-thalassemia and SCD, additional factors such as free heme, inflammatory vasculopathy, splenectomy, among other factors further contribute to the complexity of thrombotic risk. Thus, understanding the role of the numerous factors driving this hypercoagulable state will enable healthcare practitioners to enhance preventive and treatment strategies and develop novel therapies for the future. We herein describe the pathogenesis of thrombosis in patients with β-thalassemia and SCD. We also identify common mechanisms underlying the procoagulant profile of hemoglobinopathies translating into thrombotic events. Finally, we review the currently available prevention and clinical management of thrombosis in these patient populations.
Collapse
Affiliation(s)
- Rayan Bou-Fakhredin
- Department of Clinical Sciences and Community, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, SC Medicina ad Indirizzo Metabolico, Milan, Italy
| | - Maria Domenica Cappellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, SC Medicina ad Indirizzo Metabolico, Milan, Italy
| | - Ali T Taher
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Lucia De Franceschi
- Department of Medicine, University of Verona, and Azienda Ospedaliera Universitaria Verona, Verona, Italy
| |
Collapse
|
3
|
Aydinok Y, Purushotham S, Yucel A, Glassberg M, Deshpande S, Potrata B, Trapali M, Shah F. Systematic literature review of the indirect costs and humanistic burden of β-thalassemia. Ther Adv Hematol 2024; 15:20406207241270872. [PMID: 39297078 PMCID: PMC11409307 DOI: 10.1177/20406207241270872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 06/17/2024] [Indexed: 09/21/2024] Open
Abstract
Background β-Thalassemia is an inherited blood disorder requiring lifetime management of anemia and its complications. Objective This study aimed to determine the indirect costs and humanistic burden of β-thalassemia. Design A systematic literature review was conducted. Data sources and methods Searches were conducted in Embase, MEDLINE, MEDLINE In-Process, and EconLit (November 1, 2010, to November 25, 2020). Studies reporting indirect costs and health-related quality of life (HRQoL) for patients with β-thalassemia were eligible. Results Seventy-five publications were included. Mean annual days lost due to transfusion-related absenteeism ranged from 15.6 to 35 days. Patients spent a mean of 592 min (standard deviation (SD): 349) daily on disease management on transfusion days and 91 min (SD: 221) daily on non-transfusion days. Patients with non-transfusion-dependent β-thalassemia (NTDT) showed worse HRQoL versus those with transfusion-dependent β-thalassemia (TDT) on the 36-item Short Form Health Survey (75.8 vs 66.5; p = 0.021). Caregivers of patients with TDT had more severe stress compared with patients (20.17 vs 18.95; p = 0.006), as measured by the standardized Cohen Perceived Stress Questionnaire. Conclusion TDT is associated with substantial indirect costs and caregiver burden, and NTDT is associated with worse HRQoL. There is an unmet need for novel treatments in both TDT and NTDT that minimize patient and caregiver burden.
Collapse
Affiliation(s)
- Yesim Aydinok
- Department of Pediatric Hematology, Faculty of Medicine, Ege University Hospital, Kazımdirik, Ankara Asfaltı, 35100 Bornova, ĺzmir, Turkey
| | | | | | | | | | | | | | - Farrukh Shah
- Department of Haematology, Whittington Health NHS Trust, London, UK
| |
Collapse
|
4
|
Musallam KM, Sheth S, Cappellini MD, Forni GL, Maggio A, Taher AT. Anemia and iron overload as prognostic markers of outcomes in β-thalassemia. Expert Rev Hematol 2024; 17:631-642. [PMID: 39037857 DOI: 10.1080/17474086.2024.2383420] [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: 06/04/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Ineffective erythropoiesis and subsequent anemia as well as primary and secondary (transfusional) iron overload are key drivers for morbidity and mortality outcomes in patients with β-thalassemia. AREAS COVERED In this review, we highlight evidence from observational studies evaluating the association between measures of anemia and iron overload versus outcomes in both non-transfusion-dependent and transfusion-dependent forms of β-thalassemia. EXPERT OPINION Several prognostic thresholds have been identified with implications for patient management. These have also formed the basis for the design of novel therapy clinical trials by informing eligibility and target endpoints. Still, several data gaps persist in view of the challenge of assessing prospective long-term outcomes in a chronic disease. Pooling insights on the prognostic value of different measures of disease mechanism will be key to design future scoring systems that can help optimize patient management.
Collapse
Affiliation(s)
- Khaled M Musallam
- Center for Research on Rare Blood Disorders (CR-RBD), Burjeel Medical City, Abu Dhabi, United Arab Emirates
- Division of Hematology/Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Sujit Sheth
- Division of Hematology/Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Maria Domenica Cappellini
- Department of Clinical Sciences and Community, University of Milan, Ca' Granda Foundation IRCCS Maggiore Policlinico Hospital, Milan, Italy
| | | | - Aurelio Maggio
- Campus of Haematology Franco and Piera Cutino, AOOR Villa Sofia-V. Cervello, Palermo, Italy
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
5
|
Baldwin J, Udeze C, Li N, Boulmerka L, Dahal L, Pesce G, Quignot N, Jiang H, Galactéros F. Clinical burden and healthcare resource utilization associated with managing transfusion-dependent β-thalassemia in France. Curr Med Res Opin 2024; 40:1289-1295. [PMID: 38873781 DOI: 10.1080/03007995.2024.2368197] [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: 02/08/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/15/2024]
Abstract
OBJECTIVE To describe the clinical burden and healthcare resource utilization associated with managing transfusion-dependent β-thalassemia (TDT) in France. METHODS We used the French National Health Data System (système national des données de santé) to identify eligible patients from January 1, 2012, to March 1, 2019. Inclusion criteria were a diagnosis of β-thalassemia, ≥8 red blood cell (RBC) transfusion episodes per year in ≥2 consecutive years following the diagnosis, and ≥1 year of follow-up data. Patients were excluded if medical records showed evidence of sickle cell disease, α-thalassemia, hereditary persistence of fetal hemoglobin, or hematopoietic stem cell transplant. Clinical complications, mortality, treatment use, and healthcare resource utilization were evaluated. RESULTS Overall, 331 eligible patients with TDT were identified. Mean age was 26.1 (standard deviation [SD]: 18.0) years, and 50.5% were male. Common clinical complications were endocrine (26.0%), hepatobiliary (22.7%), and cardiopulmonary (18.7%). Fifteen (4.5%) patients died during follow-up, with a mortality rate of 1.16 deaths per 100 person-years (mean age of death: 52.5 years [SD: 22]). Patients had a mean of 13.5 (SD: 5.2) RBC transfusion episodes and 11.2 (SD: 5.3) iron chelation therapy treatments per year. Healthcare resource utilization was substantial, with a mean of 14.8 inpatient hospitalizations (including 13.8 mean inpatient day cases) and 16.9 outpatient prescriptions per patient per year. CONCLUSIONS Patients with TDT in France experience significant clinical complications, elevated mortality, and substantial healthcare resource utilization driven by frequent RBC transfusion episodes and inpatient hospitalizations. These results reinforce the need for disease-modifying therapies for this patient population.
Collapse
Affiliation(s)
| | - Chuka Udeze
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - Nanxin Li
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - Lyes Boulmerka
- Formerly of Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | - Lila Dahal
- Vertex Pharmaceuticals Incorporated, Boston, MA, USA
| | | | | | | | - Frédéric Galactéros
- Sickle Cell Referral Center, Internal Medicine Unit, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris, U-PEC, Créteil, France
| |
Collapse
|
6
|
Musallam KM, Barella S, Origa R, Ferrero GB, Lisi R, Pasanisi A, Longo F, Gianesin B, Forni GL. Revisiting iron overload status and change thresholds as predictors of mortality in transfusion-dependent β-thalassemia: a 10-year cohort study. Ann Hematol 2024; 103:2283-2297. [PMID: 38503936 DOI: 10.1007/s00277-024-05715-x] [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: 02/08/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024]
Abstract
Data on iron overload status and change thresholds that can predict mortality in patients with transfusion-dependent β-thalassemia (TDT) are limited. This was a retrospective cohort study of 912 TDT patients followed for up to 10 years at treatment centers in Italy (median age 32 years, 51.6% female). The crude mortality rate was 2.9%. Following best-predictive threshold identification through receiver operating characteristic curve analyses, data from multivariate Cox-regression models showed that patients with Period Average Serum Ferritin (SF) > 2145 vs ≤ 2145 ng/mL were 7.1-fold (P < 0.001) or with Absolute Change SF > 1330 vs ≤ 1330 ng/mL increase were 21.5-fold (P < 0.001) more likely to die from any cause. Patients with Period Average Liver Iron Concentration (LIC) > 8 vs ≤ 8 mg/g were 20.2-fold (P < 0.001) or with Absolute Change LIC > 1.4 vs ≤ 1.4 mg/g increase were 27.6-fold (P < 0.001) more likely to die from any cause. Patients with Index (first) cardiac T2* (cT2*) < 27 vs ≥ 27 ms were 8.6-fold (P < 0.001) more likely to die from any cause. Similarly, results at varying thresholds were identified for death from cardiovascular disease. These findings should support decisions on iron chelation therapy by establishing treatment targets, including safe iron levels and clinically meaningful changes over time.
Collapse
Affiliation(s)
- Khaled M Musallam
- Center for Research On Rare Blood Disorders (CR-RBD), Burjeel Medical City, Abu Dhabi, United Arab Emirates
| | - Susanna Barella
- S.C. Centro Delle Microcitemie E Anemie Rare, ASL Cagliari, Cagliari, Italy
| | - Raffaella Origa
- Università Di Cagliari, S.C. Centro Delle Microcitemie E Anemie Rare, ASL Cagliari, Cagliari, Italy
| | - Giovanni Battista Ferrero
- Hemoglobinopathies and Rare Anemia Reference Center, Department of Biological and Clinical Sciences, San Luigi Gonzaga University Hospital, University of Turin, Turin, Italy
| | - Roberto Lisi
- Thalassemia Unit, ARNAS Garibaldi, Catania, Italy
| | - Annamaria Pasanisi
- Centro Della Microcitemia A.Quarta, Hematology Unit, A. Perrino Hospital, Brindisi, Italy
| | - Filomena Longo
- Day Hospital Della Talassemia E Delle Emoglobinopatie, Azienda Ospedaliero Universitaria S. Anna, Ferrara, Italy
| | | | | |
Collapse
|
7
|
Chun GY, Ng SSM, Islahudin F, Selvaratnam V, Mohd Tahir NA. Polypharmacy and medication regimen complexity in transfusion-dependent thalassaemia patients: a cross- sectional study. Int J Clin Pharm 2024; 46:736-744. [PMID: 38551751 DOI: 10.1007/s11096-024-01716-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/14/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Medication burden and complexity have been longstanding problems in chronically ill patients. However, more data are needed on the extent and impact of medication burden and complexity in the transfusion-dependent thalassaemia population. AIM The aim of this study was to determine the characteristics of medication complexity and polypharmacy and determine their relationship with drug-related problems (DRP) and control of iron overload in transfusion-dependent thalassaemia patients. METHOD Data were derived from a cross-sectional observational study on characteristics of DRPs conducted at a Malaysian tertiary hospital. The medication regimen complexity index (MRCI) was determined using a validated tool, and polypharmacy was defined as the chronic use of five or more medications. The receiver operating characteristic curve analysis was used to determine the optimal cut-off value for MRCI, and logistic regression analysis was conducted. RESULTS The study enrolled 200 adult patients. The MRCI cut-off point was proposed to be 17.5 (Area Under Curve = 0.722; sensitivity of 73.3% and specificity of 62.0%). Approximately 73% and 64.5% of the patients had polypharmacy and high MRCI, respectively. Findings indicated that DRP was a full mediator in the association between MRCI and iron overload. CONCLUSION Transfusion-dependent thalassaemia patients have high MRCI and suboptimal control of iron overload conditions in the presence of DRPs. Thus, future interventions should consider MRCI and DRP as factors in serum iron control.
Collapse
Affiliation(s)
- Geok Ying Chun
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Sharon Shi Min Ng
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Farida Islahudin
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Veena Selvaratnam
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Nurul Ain Mohd Tahir
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia.
| |
Collapse
|
8
|
Ling L, Wang F, Li Y, He S, Wu F, Yang L, Xu L, Wang T, Zhou S, Yang F, Wei Z, Yang L, Yang Z, Fang X, Zhou Y, Xue J, Yin X, Wei H, Yu D. Depletion of miR-144/451 alleviates anemia in β-thalassemic mice. Blood Adv 2024; 8:2565-2570. [PMID: 37285799 PMCID: PMC11145754 DOI: 10.1182/bloodadvances.2022008757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 02/23/2023] [Accepted: 03/13/2023] [Indexed: 06/09/2023] Open
Affiliation(s)
- Ling Ling
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Fangfang Wang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
- Department of Hematology, Yangzhou University Clinical Medical College, Yangzhou, China
| | - Yaoyao Li
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
- Central Laboratory, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Sheng He
- Guangxi Key Laboratory of Birth Defects Research and Prevention, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Zhuang Autonomous Region Women and Children Care Hospital, Nanning, Guangxi, China
| | - Fan Wu
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Lei Yang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Lei Xu
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
- Central Laboratory, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Ting Wang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Shuting Zhou
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Fan Yang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Zichen Wei
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Lan Yang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Zhe Yang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Xiao Fang
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
| | - Yali Zhou
- Department of Hematology, The 923 Hospital of the Joint Logistics Support Force of the People's Liberation Army, Nanning, Guangxi, China
| | - Jun Xue
- Department of Hematology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiaolin Yin
- Department of Hematology, The 923 Hospital of the Joint Logistics Support Force of the People's Liberation Army, Nanning, Guangxi, China
| | - Hongwei Wei
- Guangxi Key Laboratory of Birth Defects Research and Prevention, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Zhuang Autonomous Region Women and Children Care Hospital, Nanning, Guangxi, China
| | - Duonan Yu
- Institute of Translational Medicine, Yangzhou University Medical College, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou, China
- Guangxi Key Laboratory of Birth Defects Research and Prevention, Guangxi Key Laboratory of Reproductive Health and Birth Defects Prevention, Guangxi Zhuang Autonomous Region Women and Children Care Hospital, Nanning, Guangxi, China
| |
Collapse
|
9
|
Bizri M, Koleilat R, Akiki N, Dergham R, Mihailescu AM, Bou-Fakhredin R, Musallam KM, Taher AT. Quality of life, mood disorders, and cognitive impairment in adults with β-thalassemia. Blood Rev 2024; 65:101181. [PMID: 38341336 DOI: 10.1016/j.blre.2024.101181] [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: 01/08/2024] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Advances in understanding the disease process in β-thalassemia supported development of various treatment strategies that resulted in improved survival. Improved survival, however, allowed multiple morbidities to manifest and cemented the need for frequent, lifelong treatment. This has directly impacted patients' health-related quality of life and opened the door for various psychiatric and cognitive disorders to potentially develop. In this review, we summarize available evidence on quality of life, depression and anxiety, suicidality, and cognitive impairment in adult patients with β-thalassemia while sharing our personal insights from experience in treating patients with both transfusion-dependent and non-transfusion-dependent forms.
Collapse
Affiliation(s)
- Maya Bizri
- Department of Psychiatry, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rawan Koleilat
- Department of Psychiatry, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nathalie Akiki
- Department of Haematology, King's College Hospital, London, United Kingdom
| | - Reem Dergham
- Department of Psychiatry, American University of Beirut Medical Center, Beirut, Lebanon
| | | | - Rayan Bou-Fakhredin
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Khaled M Musallam
- Center for Research on Rare Blood Disorders (CR-RBD), Burjeel Medical City, Abu Dhabi, United Arab Emirates
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
10
|
Wang P, Zheng L, Yan S, Xuan X, Yang Y, Qi X, Dong H. Understanding the role of red blood cells in venous thromboembolism: A comprehensive review. Am J Med Sci 2024; 367:296-303. [PMID: 38278361 DOI: 10.1016/j.amjms.2024.01.011] [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: 04/02/2023] [Revised: 12/07/2023] [Accepted: 01/19/2024] [Indexed: 01/28/2024]
Abstract
Traditionally, red blood cells (RBCs) have been perceived as passive entities within the fibrin network, without any significant role in the pathophysiology of venous thromboembolism (VTE). This review explores the involvement of RBCs in the VTE process, summarizing previous study findings and providing a comprehensive review of the latest theories. At first, it explores the influence of abnormal RBC counts (as seen in polycythemia vera and with erythropoietin use) and the exposure of RBCs to phosphatidylserine (Ptd-L-Ser) in the pathophysiology of VTE. The mechanisms of endothelial injury induced by RBCs and their adhesion to the endothelium under different disease models are then demonstrated. We explore the role of physical and chemical interactions between RBCs and platelets, as well as the interactions between RBCs and neutrophils - particularly the neutrophil extracellular traps (NETs) released by neutrophils - in the process of VTE. Additionally, we investigate the effect of RBCs on thrombin activation through two pathways, namely, the FXIIa-FXI-FIX pathway and the prekallikrein-dependent pathway. Lastly, we discuss the impact of RBCs on clot volume. In conclusion, we propose several potential methods aimed at unraveling the role of RBCs and their interaction with other components in the vascular system in the pathogenesis of VTE.
Collapse
Affiliation(s)
- Ping Wang
- Department of Vascular Surgery, The Second Hospital, Shanxi Medical University, Taiyuan 030001, China; Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Lin Zheng
- Department of Vascular Surgery, The Second Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - Sheng Yan
- Department of Vascular Surgery, The Second Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - Xuezhen Xuan
- Department of Vascular Surgery, The Second Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - Yusi Yang
- Department of Cardiology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xiaotong Qi
- Department of Vascular Surgery, The Second Hospital, Shanxi Medical University, Taiyuan 030001, China
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
11
|
Soubrier C, Jean E, De Sainte Marie B, Agouti I, Seguier J, Lavoipierre V, Clapasson C, Iline N, Gonin J, Giorgi R, Schleinitz N, Thuret I, Badens C, Bernit E. [Health status and quality of life in β-thalassemia adults in Marseille, France]. Rev Med Interne 2024; 45:187-193. [PMID: 38519305 DOI: 10.1016/j.revmed.2024.01.005] [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: 07/08/2023] [Revised: 11/17/2023] [Accepted: 01/01/2024] [Indexed: 03/24/2024]
Abstract
INTRODUCTION The life expectancy of β-thalassemia patients has increased over the last 20 years. In this study, we evaluated the current health status and quality of life of these patients managed in a reference center in Marseille. METHODS This is a single-center, descriptive study conducted between June and August 2019 in patients over 18 years of age with β-thalassemia major or intermedia. Clinical and paraclinical data were collected retrospectively and the SF-36 health survey questionnaire was proposed to each patient. RESULTS 43 of 64 selected patients were included and divided into 2 groups: 35 patients with transfusion-dependent β-thalassemia and 8 patients with non-transfusion-dependent β-thalassemia. Liver iron overload is the most frequent complication, present in 80% of transfusion-dependent and 62.5% of non-transfusion-dependent patients. Cardiac iron overload is present only in the transfusion dependent β-thalassemia group (20%). Hypogonadotropic hypogonadism remains the most common endocrine disorder (41.9%) followed by osteoporosis (37.2%). Among the 31 patients who completed the SF-36 questionnaire, physical and mental quality of life scores were lowered in transfusion dependent (respectively 42.7 and 46.8) as in non-transfusion-dependent patients (respectively 43.8 and 28.9). CONCLUSION Despite an improvement in medical care, our patients with β-thalassemia show an alteration in their quality of life that will need to be characterized in the entire French cohort.
Collapse
Affiliation(s)
- C Soubrier
- Service de médecine interne et médecine polyvalente, centre hospitalier d'Ajaccio Notre-Dame de la Miséricorde, site du Stilettu, 1180, route A.-Madunuccia, 20090 Ajaccio, France.
| | - E Jean
- Département de médecine interne, AP-HM, hôpital Timone, 264, rue Saint-Pierre, 13385 Marseille cedex, France
| | - B De Sainte Marie
- Département de médecine interne, AP-HM, hôpital Timone, 264, rue Saint-Pierre, 13385 Marseille cedex, France
| | - I Agouti
- Centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rares du globule rouge et de l'érythropoïèse, hôpital de la Timone, AP-PH, 264, rue Saint-Pierre, 13385 Marseille cedex, France
| | - J Seguier
- Département de médecine interne, AP-HM, hôpital Timone, 264, rue Saint-Pierre, 13385 Marseille cedex, France
| | - V Lavoipierre
- Service de médecine interne, centre hospitalier de Martigues, 3, boulevard des Rayettes, BP 50248, 13698 Martigues cedex, France
| | - C Clapasson
- Établissement français du sang, région PACA, 149, boulevard Baille, 13005 Marseille, France
| | - N Iline
- AP-HM, Hop Timone, BioSTIC, biostatistiques et technologies de l'information et de la communication, Marseille, France
| | - J Gonin
- AP-HM, Hop Timone, BioSTIC, biostatistiques et technologies de l'information et de la communication, Marseille, France
| | - R Giorgi
- AP-HM, Hop Timone, BioSTIC, biostatistiques et technologies de l'information et de la communication, Marseille, France; Aix Marseille université, AP-HM, Inserm, IRD, SESSTIM, sciences économiques & sociales de la santé & traitement de l'information médicale, ISSPAM, Marseille, France
| | - N Schleinitz
- Département de médecine interne, AP-HM, hôpital Timone, 264, rue Saint-Pierre, 13385 Marseille cedex, France; Aix-Marseille université, Marseille, France
| | - I Thuret
- Centre de référence des syndromes drépanocytaires majeurs, thalassémies et autres pathologies rares du globule rouge et de l'érythropoïèse, hôpital de la Timone, AP-PH, 264, rue Saint-Pierre, 13385 Marseille cedex, France
| | - C Badens
- Laboratoire de biochimie, AP-HM, hôpital Timone, 264, rue Saint-Pierre, 13385 Marseille cedex, France
| | - E Bernit
- Unité transversale de la drépanocytose, centre de référence Antilles-Guyane pour la drépanocytose, les thalassémies et les maladies constitutives du globule rouge et de l'érythropoïèse, CHU Guadeloupe, pôle parents-enfants, hôpital Ricou, BP465, 97159 Pointe-à-Pitre cedex, France
| |
Collapse
|
12
|
Corbacioglu S, Frangoul H, Locatelli F, Hobbs W, Walters M. Defining curative endpoints for transfusion-dependent β-thalassemia in the era of gene therapy and gene editing. Am J Hematol 2024; 99:422-429. [PMID: 38100154 DOI: 10.1002/ajh.27166] [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: 06/23/2023] [Revised: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 02/15/2024]
Abstract
β-thalassemia is a monogenic disease that results in varying degrees of anemia. In the most severe form, known as transfusion-dependent β-thalassemia (TDT), the clinical hallmarks are ineffective erythropoiesis and a requirement of regular, life-long red blood cell transfusions, with the development of secondary clinical complications such as iron overload, end-organ damage, and a risk of early mortality. With the exception of allogeneic hematopoietic cell transplantation, current treatments for TDT address disease symptoms and not the underlying cause of disease. Recently, a growing number of gene addition and gene editing-based treatments for patients with TDT with the potential to provide a one-time functional cure have entered clinical trials. A key challenge in the design and evaluation of these trials is selecting endpoints to evaluate if these novel genetic therapies have a curative versus an ameliorative effect. Here, we present an overview of the pathophysiology of TDT, review emerging gene addition or gene editing therapeutic approaches for TDT currently in clinical trials, and identify a series of endpoints that can quantify therapeutic effects, including a curative outcome.
Collapse
Affiliation(s)
| | - Haydar Frangoul
- Sarah Cannon Research Institute and the Children's Hospital at TriStar Centennial, Nashville, Tennessee, USA
| | - Franco Locatelli
- IRCCS, Ospedale Pediatrico Bambino, Gesù Rome, Catholic University of the Sacred Heart, Rome, Italy
| | - William Hobbs
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts, USA
| | - Mark Walters
- Department of Pediatrics, UCSF Benioff Children's Hospital Oakland, Oakland, California, USA
| |
Collapse
|
13
|
Musallam KM, Cappellini MD, Coates TD, Kuo KHM, Al-Samkari H, Sheth S, Viprakasit V, Taher AT. Αlpha-thalassemia: A practical overview. Blood Rev 2024; 64:101165. [PMID: 38182489 DOI: 10.1016/j.blre.2023.101165] [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: 10/30/2023] [Revised: 12/19/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
α-Thalassemia is an inherited blood disorder characterized by decreased synthesis of α-globin chains that results in an imbalance of α and β globin and thus varying degrees of ineffective erythropoiesis, decreased red blood cell (RBC) survival, chronic hemolytic anemia, and subsequent comorbidities. Clinical presentation varies depending on the genotype, ranging from a silent or mild carrier state to severe, transfusion-dependent or lethal disease. Management of patients with α-thalassemia is primarily supportive, addressing either symptoms (eg, RBC transfusions for anemia), complications of the disease, or its transfusion-dependence (eg, chelation therapy for iron overload). Several novel therapies are also in development, including curative gene manipulation techniques and disease modifying agents that target ineffective erythropoiesis and chronic hemolytic anemia. This review of α-thalassemia and its various manifestations provides practical information for clinicians who practice beyond those regions where it is found with high frequency.
Collapse
Affiliation(s)
- Khaled M Musallam
- Center for Research on Rare Blood Disorders (CR-RBD), Burjeel Medical City, Abu Dhabi, United Arab Emirates
| | - M Domenica Cappellini
- Department of Clinical Sciences and Community, University of Milan, Ca' Granda Foundation IRCCS Maggiore Policlinico Hospital, Milan, Italy
| | - Thomas D Coates
- Hematology Section, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Kevin H M Kuo
- Division of Hematology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Hanny Al-Samkari
- Center for Hematology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sujit Sheth
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Vip Viprakasit
- Department of Pediatrics & Thalassemia Center, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
14
|
Rahimi-Dehkordi N, Heidari-Soureshjani S, Sherwin CMT. The Effects and Safety of Silymarin on β-thalassemia in Children and Adolescents: A Systematic Review based on Clinical Trial Studies. Rev Recent Clin Trials 2024; 19:242-255. [PMID: 38818907 DOI: 10.2174/0115748871305325240511122602] [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: 01/18/2024] [Revised: 04/06/2024] [Accepted: 04/27/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND β-thalassemia imposes significant complications on affected patients. Silymarin, a natural flavonoid complex, has potential therapeutic properties. OBJECTIVE This systematic review aims to comprehensively evaluate the literature on the mechanistic effects of Silymarin on β-thalassemia outcomes in children and adolescents. METHODS A systematic search of electronic databases, including MEDLINE/PubMed, Embase, Scopus, Cochrane Library, and Web of Science (WOS), was done to identify relevant clinical trials before January 2024. Various data were extracted, including study characteristics, outcomes measured (hematological parameters, oxidative stress markers, iron metabolism, and other outcomes), proposed mechanisms, and safety. RESULTS By iron chelation effects, Silymarin can reduce reactive oxygen species (ROS) production, increase intracellular antioxidant enzyme glutathione (GSH), and insert antioxidant effects. It also attenuated inflammation through reduced tumor necrosis factor-alpha (TNF-α), transforming growth factor-β1 (TGF-β1), interferon-gamma (IFNγ), C-reactive protein (CRP), interleukin 6 (IL-6), IL-17, and IL-23 levels and increase in IL-4 and IL-10 levels. By reducing iron overload conditions, Silymarin indicates modulatory effects on immune abnormalities, inhibits red blood cell (RBC) hemolysis, increases RBC count, and minimizes the need for a transfusion. Moreover, it reduces myocardial and hepatic siderosis, improves liver function tests, and modifies abnormal enzymes, particularly for aspartate transaminase (AST), alanine transaminase (ALT), alkaline phosphatase (ALP), total bilirubin, and total protein levels. Silymarin also reduces iron overload, increases antioxidant and anti-inflammatory capacity in cardiomyocytes, and reveals antioxidant effects. CONCLUSION Silymarin indicates promising effects on various aspects of children and adolescents with β-thalassemia and has no serious side effects on the investigated dosage.
Collapse
Affiliation(s)
- Nasim Rahimi-Dehkordi
- Department of Pediatrics, School of Medicine, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Catherine M T Sherwin
- Pediatric Clinical Pharmacology and Toxicology, Department of Pediatrics, Wright State University Boonshoft School of Medicine, Dayton Children's Hospital, One Children's Plaza, Dayton, Ohio, USA
| |
Collapse
|
15
|
El-Beshlawy A, Dewedar H, Hindawi S, Alkindi S, Tantawy AA, Yassin MA, Taher AT. Management of transfusion-dependent β-thalassemia (TDT): Expert insights and practical overview from the Middle East. Blood Rev 2024; 63:101138. [PMID: 37867006 DOI: 10.1016/j.blre.2023.101138] [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: 08/29/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023]
Abstract
β-Thalassemia is one of the most common monogenetic diseases worldwide, with a particularly high prevalence in the Middle East region. As such, we have developed long-standing experience with disease management and devising solutions to address challenges attributed to resource limitations. The region has also participated in the majority of clinical trials and development programs of iron chelators and more novel ineffective erythropoiesis-targeted therapy. In this review, we provide a practical overview of management for patients with transfusion-dependent β-thalassemia, primarily driven by such experiences, with the aim of transferring knowledge to colleagues in other regions facing similar challenges.
Collapse
Affiliation(s)
- Amal El-Beshlawy
- Department of Pediatric Hematology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hany Dewedar
- Thalassemia Center, Latifa Hospital, Dubai, United Arab Emirates
| | - Salwa Hindawi
- Department of Hematology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salam Alkindi
- Department of Hematology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Azza A Tantawy
- Department of Pediatrics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed A Yassin
- Department of Hematology and Oncology, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
16
|
Hodroj MH, Akiki N, Bou-Fakhredin R, Taher AT. Beta-thalassemia: is cure still a dream? Minerva Med 2023; 114:850-860. [PMID: 37534831 DOI: 10.23736/s0026-4806.23.08501-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
β-thalassemia is a monogenic disorder characterized by decreased hemoglobin production, resulting in chronic anemia. There are several factors affecting the clinical presentation of patients with β-thalassemia, and several complications such as iron overload or ineffective erythropoiesis have been linked to this disease. Until nowadays, several conservative therapies namely blood transfusions, iron chelation, and the FDA-approved drug Luspatercept have been adopted alongside other debatable permanent cures. Other clinical trials are being conducted to develop better and safer management techniques for these patients. This review will discuss the different treatment strategies of β-thalassemia including novel therapies, besides all possible curative therapies that are being developed for this disease.
Collapse
Affiliation(s)
- Mohammad H Hodroj
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Nathalie Akiki
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rayan Bou-Fakhredin
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Ali T Taher
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon -
| |
Collapse
|
17
|
Hodroj MH, Taher A. Thalassemia and malignancies: Updates from the literature. Ann N Y Acad Sci 2023; 1529:14-20. [PMID: 37676814 DOI: 10.1111/nyas.15061] [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] [Indexed: 09/09/2023]
Abstract
Thalassemia management has undergone significant development with the advancement in iron chelation therapy, which has led to a prolonged life expectancy. This has been accompanied by the emergence of several new morbidities and chronic diseases, including cancer. Over the years, multiple cases of solid and hematologic malignancies in thalassemia patients have been reported in the literature, with no clear mechanism for the development of cancer in these patients despite a number of potential mechanisms. However, the results of many studies have been contradictory regarding the risk of development of malignancies in thalassemia. The present review aims to discuss the available data on cancer and thalassemia in the literature, with the latest updates regarding possible malignancy development mechanisms, risks, and the most commonly reported types.
Collapse
Affiliation(s)
- Mohammad Hassan Hodroj
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Taher
- Division of Hematology and Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
18
|
Saliba AN, Musallam KM, Taher AT. How I treat non-transfusion-dependent β-thalassemia. Blood 2023; 142:949-960. [PMID: 37478396 PMCID: PMC10644094 DOI: 10.1182/blood.2023020683] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/23/2023] Open
Abstract
The intricate interplay of anemia and iron overload under the pathophysiological umbrella of ineffective erythropoiesis in non-transfusion-dependent β-thalassemia (NTDT) results in a complex variety of clinical phenotypes that are challenging to diagnose and manage. In this article, we use a clinical framework rooted in pathophysiology to present 4 common scenarios of patients with NTDT. Starting from practical considerations in the diagnosis of NTDT, we delineate our strategy for the longitudinal care of patients who exhibit different constellations of symptoms and complications. We highlight the use of transfusion therapy and novel agents, such as luspatercept, in the patient with anemia-related complications. We also describe our approach to chelation therapy in the patient with iron overload. Although tackling every specific complication of NTDT is beyond the scope of this article, we touch on the management of the various morbidities and multisystem manifestations of the disease.
Collapse
Affiliation(s)
| | - Khaled M. Musallam
- Thalassemia Center, Burjeel Medical City, Abu Dhabi, United Arab Emirates
| | - Ali T. Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
19
|
Grewal A, Kakkar S, Dewan P, Bansal N, Sobti PC, Eleftheriou P. Prevalence, Severity, and Determinants of Pain in Thalassemia. Hemoglobin 2023; 47:191-197. [PMID: 37982258 DOI: 10.1080/03630269.2023.2269846] [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: 07/29/2022] [Accepted: 10/07/2023] [Indexed: 11/21/2023]
Abstract
As the life expectancy in thalassemia is improving, pain is being recognized as an emerging problem. To document the pain prevalence and severity in patients with transfusion-dependent thalassemia all transfusion-dependent thalassemia patients >10 years of age (n = 165) attending the Thalassemia Day Care Center were assessed for pain prevalence, severity, and its effect on various life activities using the Brief Pain Inventory. Their medical records were reviewed for the presence of various co-morbidities. Pain was reported by 62.4% of participants with 35.2% and 59.4% of participants, reporting pain in the past 1 and 4 weeks respectively. A significantly higher pain prevalence was reported in females (p = .037), patients residing in urban areas (p = .038), and employed participants (p = .038). The commonest sites of pain were the lower back and calves. General activity (p = .02) and enjoyment of life (p = .02) were significantly affected due to pain in patients between 21 and 30 years of age. Female participants reported interference of pain with mood (p = .03). A significant correlation of pain prevalence was found with higher average serum ferritin (p = .015), moderate to severe liver iron concentration (p = .04), and lower levels of 25 hydroxyvitamin D levels (p = .03). Pain is an emerging cause of morbidity in thalassemia. The study found a significant association of pain with modifiable factors such as serum ferritin, LIC, and 25 (OH) vitamin D levels.
Collapse
Affiliation(s)
- Amanat Grewal
- Dayanand Medical College and Hospital, Ludhiana, India
| | - Shruti Kakkar
- Department of Pediatrics, Dayanand Medical College and Hospital, Ludhiana, India
| | | | - Namita Bansal
- Dayanand Medical College and Hospital, Ludhiana, India
| | | | | |
Collapse
|
20
|
Sawh RN. The patient perspective: The struggles of living with thalassemia as an adult. Ann N Y Acad Sci 2023; 1527:20-24. [PMID: 37468233 DOI: 10.1111/nyas.15040] [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] [Indexed: 07/21/2023]
Abstract
Today it has become the norm for individuals diagnosed with severe forms of thalassemia who have access to hypertransfusion regimens, chelation therapy, and annual surveillance to survive well beyond childhood. However, with this improvement in prognosis and subsequent transition to adult care, it has become apparent that most adult healthcare providers, including many adult hematologists and primary care providers, are ill-prepared to care for these patients and the complications that accompany their survival into adulthood. Collaborative efforts are needed to develop comprehensive approaches to contend with the challenges faced by adult patients to ensure they are properly managed and supported.
Collapse
Affiliation(s)
- Radhika N Sawh
- The Joan H. Marks Graduate Program in Human Genetics, Sarah Lawrence College, Bronxville, New York, USA
| |
Collapse
|
21
|
Yu X, Peng Y, Nie T, Sun W, Zhou Y. Diabetes and two kinds of primary tumors in a patient with thalassemia: a case report and literature review. Front Oncol 2023; 13:1207336. [PMID: 37637036 PMCID: PMC10455928 DOI: 10.3389/fonc.2023.1207336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/21/2023] [Indexed: 08/29/2023] Open
Abstract
Background Thalassemia is a group of common genetic hematologic disorders characterized by deficient synthesis of the hemoglobin chain. Due to effective blood transfusion and optimization of chelate therapy, the survival of thalassemia patients and their overall quality of life have improved noticeably in the past few decades. As a consequence, the longer life expectancy has led to the manifestation of several concomitant morbidities, including heart disease, infections, cirrhosis, endocrine abnormalities, various malignancies, and so on. In this context, the probability and updated literature about some malignancy cases in patients with thalassemia build new scenarios for the next few years. We describe the first report of a thalassemic patient developing diabetes and head and neck cancer and try to summarize the possible predisposing factors and mechanisms behind their phenomenon. Case presentation The current case report describes a 50-year-old Asian man who has been diagnosed with thalassemia since childhood. In early 2017, he was also diagnosed with diabetes and started on insulin-hypoglycemic treatment. The patient was then diagnosed with primary non-keratinizing undifferentiated carcinoma of the nasopharynx in late February 2013. A biopsy of the left tongue revealed squamous cell carcinoma (SCC) in late March 2019. Conclusions We report the first case of a thalassemic patient developing diabetes and squamous cell carcinoma of the head and neck and discuss the possibility of a link between the three diseases. This specific case should alert physicians to the possibility of endocrinopathy and malignancy in thalassemic patients.
Collapse
Affiliation(s)
- Xiaoyan Yu
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Peng
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tingting Nie
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenjia Sun
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yajuan Zhou
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
22
|
Kalamara TVN, Zarkada EG, Kasimatis ED, Kofinas AG, Klonizakis PI, Vlachaki EC. Kidney transplantation in an adult with transfusion-dependent beta thalassemia: A challenging case report and literature review. Arch Clin Cases 2023; 10:97-101. [PMID: 37359087 PMCID: PMC10289047 DOI: 10.22551/2023.39.1002.10250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023] Open
Abstract
The markedly increased survival of transfusion-dependent beta thalassemia patients has led to the recognition of new complications, such as renal disorders. Kidney transplantation is nowadays the preferred treatment option for end-stage kidney disease (ESKD). We describe a case of a 49-year-old woman with β-Transfusion Dependent Thalassemia, who developed ESKD as a result of focal segmental glomerulosclerosis and received a deceased-donor kidney transplant following hemodialysis for over a decade. The particular challenges of this case are discussed, including the long-term survival in hemodialysis. Our patient had to overcome multiple obstacles, including hypercoagulability issues presented in the form of thromboembolism, infections, such as hepatitis C and gastroenteritis, and the acute T-cell-mediated rejection, which had to be managed postoperatively. A review of the current literature revealed only one previous report of a thalassemia patient who successfully underwent renal transplantation. More than a year after the transplantation our patient presents with a normal glomerular filtration rate (GFR=62ml/min/1.73m2) and creatinine level (Cr=0.96mg/dL) and is transfused every 3 weeks. In conclusion, renal transplantation is possible in patients with TDT and should not be discouraged. Regular transfusions and optimal follow-up for the elimination of post-transplant complications are required.
Collapse
Affiliation(s)
- Tsampika-Vasileia N. Kalamara
- Adults Thalassemia Unit, Second Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Evangelia G. Zarkada
- Adults Thalassemia Unit, Second Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Efstratios D. Kasimatis
- Department of Nephrology, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Athanasios G. Kofinas
- Department of Transplantation and Surgery, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Philippos I. Klonizakis
- Adults Thalassemia Unit, Second Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Efthymia C. Vlachaki
- Adults Thalassemia Unit, Second Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| |
Collapse
|
23
|
Musallam KM, Taher AT, Kattamis A, Kuo KHM, Sheth S, Cappellini MD. Profile of Luspatercept in the Treatment of Anemia in Adults with Non-Transfusion-Dependent β-Thalassemia (NTDT): Design, Development and Potential Place in Therapy. Drug Des Devel Ther 2023; 17:1583-1591. [PMID: 37255740 PMCID: PMC10226484 DOI: 10.2147/dddt.s368584] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/20/2023] [Indexed: 06/01/2023] Open
Abstract
Over the past decade, evidence has been mounting on the detrimental clinical sequelae of untreated anemia in patients with non-transfusion-dependent β-thalassemia (NTDT). There are no pharmacologic agents that are specifically approved for the management of anemia in NTDT, and available options such as splenectomy, transfusion therapy, and hydroxyurea each come with their own shortcomings, especially for long-term use. Luspatercept is an erythroid maturation agent that has been evaluated in a Phase 2, randomized trial and showed a significant benefit in raising hemoglobin level by at least 1 g/dL in adults with NTDT and a baseline hemoglobin level ≤10 g/dL. These data led to luspatercept's approval by the European Commission for the treatment of anemia in adults with NTDT and presents the first evidence-based approach for a novel agent that is able to ameliorate anemia in this patient population.
Collapse
Affiliation(s)
- Khaled M Musallam
- Thalassemia Center, Burjeel Medical City, Abu Dhabi, United Arab Emirates
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Antonis Kattamis
- First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| | - Kevin H M Kuo
- Division of Hematology, University of Toronto, Toronto, ON, Canada
| | - Sujit Sheth
- Division of Hematology and Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Maria Domenica Cappellini
- Department of Clinical Sciences and Community, University of Milan, Ca’ Granda Foundation IRCCS Maggiore Policlinico Hospital, Milan, Italy
| |
Collapse
|
24
|
Hossain MJ, Islam MW, Munni UR, Gulshan R, Mukta SA, Miah MS, Sultana S, Karmakar M, Ferdous J, Islam MA. Health-related quality of life among thalassemia patients in Bangladesh using the SF-36 questionnaire. Sci Rep 2023; 13:7734. [PMID: 37173392 PMCID: PMC10182078 DOI: 10.1038/s41598-023-34205-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Thalassemia is one of the most common autosomal recessive hereditary blood disorders worldwide, especially in developing countries, including Bangladesh. Thus, this study aimed to determine HRQoL and its determinants of thalassemia patients (TP) in Bangladesh. A cross-sectional survey was performed on 356 randomly selected thalassemia patients. Participants were invited to face-to-face interviews. Descriptive statistics (frequencies and percentages), independent t-test, ANOVA, and multivariate (linear and logistic regression) analysis was performed to analyze the data. Our demographic data showed that among 356 patients, 54% and 46% were male and female, respectively, with an average age of 19.75 (SD = 8.02) years. Most were transfusion-dependent (91%), 26% had comorbidities, and 52% were from low-income families. In the case of HRQoL, male patients showed significantly higher scores of bodily pains and physical health summaries than female patients. Lower income, high blood transfusion status, disease severity, comorbidities, and medical expenses (p < 0.05; CI 95%) are significantly associated with lower SF-36 scores. This study found an association between lower income, blood transfusion, disease severity, comorbidities, as well as medical expenses, and the deterioration of HRQoL among TP. Male patients experienced poorer HRQoL than females. National action plans are required to guarantee the holistic welfare of thalassemia patients.
Collapse
Affiliation(s)
- Md Jubayer Hossain
- Population Health Studies Division, Center for Health Innovation, Research, Action, and Learning-Bangladesh (CHIRAL Bangladesh), 9-10 Chittaranjan Avenue, Dhaka, 1100, Bangladesh
| | - Md Wahidul Islam
- Population Health Studies Division, Center for Health Innovation, Research, Action, and Learning-Bangladesh (CHIRAL Bangladesh), 9-10 Chittaranjan Avenue, Dhaka, 1100, Bangladesh
| | - Ummi Rukaiya Munni
- Population Health Studies Division, Center for Health Innovation, Research, Action, and Learning-Bangladesh (CHIRAL Bangladesh), 9-10 Chittaranjan Avenue, Dhaka, 1100, Bangladesh
- Health Systems and Population Studies Division, International Centre for Diarrhoeal Disease Research, Bangladesh, 68, Shaheed Tajuddin Ahmed Sarani Mohakhali, Dhaka, 1212, Bangladesh
- BRAC James P Grant School of Public Health, BRAC University, 66, Mohakhali, Dhaka, 1212, Bangladesh
| | - Rubaiya Gulshan
- Population Health Studies Division, Center for Health Innovation, Research, Action, and Learning-Bangladesh (CHIRAL Bangladesh), 9-10 Chittaranjan Avenue, Dhaka, 1100, Bangladesh
| | - Sumaiya Akter Mukta
- Population Health Studies Division, Center for Health Innovation, Research, Action, and Learning-Bangladesh (CHIRAL Bangladesh), 9-10 Chittaranjan Avenue, Dhaka, 1100, Bangladesh
| | - Md Sharif Miah
- Population Health Studies Division, Center for Health Innovation, Research, Action, and Learning-Bangladesh (CHIRAL Bangladesh), 9-10 Chittaranjan Avenue, Dhaka, 1100, Bangladesh
| | - Sabia Sultana
- Population Health Studies Division, Center for Health Innovation, Research, Action, and Learning-Bangladesh (CHIRAL Bangladesh), 9-10 Chittaranjan Avenue, Dhaka, 1100, Bangladesh
- Department of Microbiology, Jagannath University, 9-10 Chittaranjan Avenue, Dhaka, 1100, Bangladesh
| | - Mousumi Karmakar
- Department of Microbiology, Bangladesh University of Health Sciences, 125, Technical Mor, 1 Darus Salam Rd, Dhaka, 1216, Bangladesh
| | - Jannatul Ferdous
- Department of Transfusion Medicine, Mugda Medical College and Hospital, Hazi Kadam Ali Rd, Dhaka, Bangladesh
- Bangladesh Thalassemia Foundation, Chamelibagh, Shantinagar, Dhaka, 1217, Bangladesh
| | - Mohammad Ariful Islam
- Department of Microbiology, Jagannath University, 9-10 Chittaranjan Avenue, Dhaka, 1100, Bangladesh.
| |
Collapse
|
25
|
Algeri M, Lodi M, Locatelli F. Hematopoietic Stem Cell Transplantation in Thalassemia. Hematol Oncol Clin North Am 2023; 37:413-432. [PMID: 36907612 DOI: 10.1016/j.hoc.2022.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only consolidated, potentially curative treatment for patients with transfusion-dependent thalassemia major. In the past few decades, several new approaches have reduced the toxicity of conditioning regimens and decreased the incidence of graft-versus-host disease, improving patients' outcomes and quality of life. In addition, the progressive availability of alternative stem cell sources from unrelated or haploidentical donors or umbilical cord blood has made HSCT a feasible option for an increasing number of subjects lacking an human leukocyte antigen (HLA)-identical sibling. This review provides an overview of allogeneic hematopoietic stem cell transplantation in thalassemia, reassesses current clinical results, and discusses future perspectives.
Collapse
Affiliation(s)
- Mattia Algeri
- Department of Hematology/Oncology, Cell and Gene Therapy - IRCCS, Bambino Gesù Children's Hospital, Rome, Italy.
| | - Mariachiara Lodi
- Department of Hematology/Oncology, Cell and Gene Therapy - IRCCS, Bambino Gesù Children's Hospital, Rome, Italy
| | - Franco Locatelli
- Department of Hematology/Oncology, Cell and Gene Therapy - IRCCS, Bambino Gesù Children's Hospital, Rome, Italy; Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
26
|
Forni GL, Gianesin B, Musallam KM, Longo F, Rosso R, Lisi R, Gamberini MR, Pinto VM, Graziadei G, Vitucci A, Bonetti F, Musto P, Piga A, Cappellini MD, Borgna-Pignatti C. Overall and complication-free survival in a large cohort of patients with β-thalassemia major followed over 50 years. Am J Hematol 2023; 98:381-387. [PMID: 36588408 DOI: 10.1002/ajh.26798] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/26/2022] [Accepted: 11/26/2022] [Indexed: 01/03/2023]
Abstract
We report data on survival and complications for a longitudinal cohort of 709 transfusion-dependent β-thalassemia major patients (51.1% males) born between 1970 and 1997 and followed through 2020 at seven major centers in Italy. Overall survival probability at 30 years was 83.6% (95%CI: 78.5-89.1) in the oldest birth cohort (1970-1974) compared with 93.3% (95%CI: 88.6-98.3) in the youngest birth cohort (1985-1997) (p = 0.073). Females showed better survival than males (p = 0.022). There were a total of 93 deaths at a median age of 23.2 years with the most frequent disease-related causes being heart disease (n = 53), bone marrow transplant (BMT) complication (n = 10), infection (n = 8), liver disease (n = 4), cancer (n = 3), thromboembolism (n = 2) and severe anemia (n = 1). There was a steady decline in the number of deaths due to heart disease from the year 2000 onwards and no death from BMT was observed after the year 2010. A progressive decrease in the median age of BMT was observed in younger birth cohorts (p < 0.001). A total of 480 (67.7%) patients developed ≥1 complication. Patients in younger birth cohorts demonstrated better complication-free survival (p < 0.001) which was comparable between sexes (p = 0.230). Independent risk factors for death in multivariate analysis included heart disease (HR: 4.63, 95%CI: 1.78-12.1, p = 0.002), serum ferritin >1000 ng/mL (HR: 15.5, 95%CI: 3.52-68.2, p < 0.001), male sex (HR: 2.75, 95%CI: 0.89-8.45, p = 0.078), and splenectomy (HR: 6.97, 95%CI: 0.90-54.0, p < 0.063). Survival in patients with β-thalassemia major continues to improve with adequate access to care, best practice sharing, continued research, and collaboration between centers.
Collapse
Affiliation(s)
- Gian Luca Forni
- Center for Microcythemia, Congenital Anemia and Iron Dysmetabolism, Galliera Hospital, Genoa, Italy
| | - Barbara Gianesin
- Center for Microcythemia, Congenital Anemia and Iron Dysmetabolism, Galliera Hospital, Genoa, Italy.,ForAnemia Foundation, Genoa, Italy
| | | | - Filomena Longo
- Reference Centre for Hemoglobinopathies, AOU San Luigi Gonzaga Hospital, Orbassano, Italy
| | - Rosamaria Rosso
- Thalassemia and Hemoglobinopathies Unit, AOU Policlinico "Vittorio Emanuele", Catania, Italy
| | - Roberto Lisi
- Thalassemia Unit, ARNAS "Garibaldi", Catania, Italy
| | - Maria Rita Gamberini
- Thalassemia and Hemoglobinopathies Day Unit, AOU S'Anna Hospital, Ferrara, Italy
| | - Valeria Maria Pinto
- Center for Microcythemia, Congenital Anemia and Iron Dysmetabolism, Galliera Hospital, Genoa, Italy
| | - Giovanna Graziadei
- Department of Medicine and Medical Specialties, IRCCS Ca' Granda Foundation, Maggiore Policlinico Hospital, Milan, Italy
| | - Angelantonio Vitucci
- Hematology and Transplant Unit, AOU Consorziale Policlinico, Bari, Italy.,Department of Emergency and Organ Transplantation, "Aldo Moro" University School of Medicine, Bari, Italy
| | - Federico Bonetti
- Department of Pediatric Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Pellegrino Musto
- Hematology and Transplant Unit, AOU Consorziale Policlinico, Bari, Italy
| | - Antonio Piga
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Maria Domenica Cappellini
- Department of Medicine and Medical Specialties, IRCCS Ca' Granda Foundation, Maggiore Policlinico Hospital, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | | |
Collapse
|
27
|
Hokland P, Daar S, Khair W, Sheth S, Taher AT, Torti L, Hantaweepant C, Rund D. Thalassaemia-A global view. Br J Haematol 2023; 201:199-214. [PMID: 36799486 DOI: 10.1111/bjh.18671] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/10/2023] [Accepted: 01/15/2023] [Indexed: 02/18/2023]
Abstract
The thalassaemias are a group of genetic disorders of haemoglobin which are endemic in the tropics but are now found worldwide due to migration. Basic standard of care therapy includes regular transfusions to maintain a haemoglobin level of around 10 g/dL, together with iron chelation therapy to prevent iron overload. Novel therapies, bone marrow transplantation, and gene therapy are treatment options that are unavailable in many countries with stressed economies. This Wider Perspectives article presents the strategies for management of an adolescent refugee patient with beta thalassaemia, as it would be performed by expert haematologists in six countries: Italy, Lebanon, Oman, the Sudan, Thailand and the United States. The experienced clinicians in each country have adapted their practice according to the resources available, which vary greatly. Even in the current modern era, providing adequate transfusions and chelation is problematic in many countries. On the other hand, ensuring adherence to therapy, particularly during adolescence, is a similar challenge seen in all countries. The concluding section highlights the disparities in available therapies and puts the role of novel therapies into a societal context.
Collapse
Affiliation(s)
- Peter Hokland
- Faculty of Health, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Shahina Daar
- College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Wael Khair
- Khartoum Oncology Hospital, Khartoum, Sudan
| | - Sujit Sheth
- Division of Hematology Oncology, Department of Pediatrics, Weill Cornell Medicine, New York City, New York, USA
| | - Ali T Taher
- Division of Hematology & Oncology, Department of Internal Medicine, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Lorenza Torti
- Hemoglobinopathies Unit, Hematology Department, S. Eugenio Hospital, (ASL Roma 2), Rome, Italy
| | - Chattree Hantaweepant
- Faculty of Medicine Siriraj Hospital, Division of Hematology, Department of Medicine, Mahidol University, Bangkok, Thailand
| | - Deborah Rund
- Department of Haematology, Hebrew University-Hadassah Medical Centre, Jerusalem, Israel
| |
Collapse
|
28
|
Cardiovascular Complications in β-Thalassemia: Getting to the Heart of It. THALASSEMIA REPORTS 2023. [DOI: 10.3390/thalassrep13010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Beta thalassemia is an inherited disorder resulting in abnormal or decreased production of hemoglobin, leading to hemolysis and chronic anemia. The long-term complications can affect multiple organ systems, namely the liver, heart, and endocrine. Myocardial iron overload is a common finding in β-thalassemia. As a result, different cardiovascular complications in the form of cardiomyopathy, pulmonary hypertension, arrhythmias, and vasculopathies can occur, and in extreme cases, sudden cardiac death. Each of these complications pertains to underlying etiologies and risk factors, which highlights the importance of early diagnosis and prevention. In this review, we will discuss different types of cardiovascular complications that can manifest in patients with β-thalassemia, in addition to the current diagnostic modalities, preventive and treatment modalities for these complications.
Collapse
|
29
|
Identification of novel mutations in β-thalassemia patients in Maysan Governorate, Iraq. Mol Biol Rep 2023; 50:3053-3062. [PMID: 36683082 DOI: 10.1007/s11033-023-08271-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/10/2023] [Indexed: 01/23/2023]
Abstract
BACKGROUND In homozygous β-thalassemia, the primary genetic modifiers affecting the clinical severity of β-thalassemia are genetic variants and the ability to reduce globin chain imbalance, thus resulting in a milder form of thalassemia. However, there are few reports on the molecular genetics of β-thalassemia in Iraq. METHODS We performed PCR and DNA sequencing on 40 Iraqi patients who were clinically suspected of having β-thalassemia. RESULTS The first genetic sequencing study was conducted in Maysan Governate, Iraq, using patients from various locations to identify novel mutations. There were five novel mutations: 294.T>C 12% (city center and Almajar district), 205. C>T 25% (city center, Alsalam, and Almashrah districts), 289.G>A 38% (Almaymuna and Gleat Salih districts), 49.T>C 32% (city center), and 624.C>A 32% (city center). These mutations were identified among β-thalassemia patients by two regions of HBB gene 696 bp and 861 bp. CONCLUSIONS The discovery of new genetic variants helps predict the severity of β-thalassemia disease. There are relatively few studies in molecular genetics of β-thalassemia in Iraq, and the new mutations reported here will provide valuable data for the prevention and control of β-thalassemia in Maysan Governate, Iraq. The results can lead to new genetic sequencing investigations for other Iraqi regions.
Collapse
|
30
|
Echocardiographic evaluation of left atrial strain for predicting iron overload in pediatric patients with β-thalassemia with preserved ejection fraction. Int J Cardiovasc Imaging 2023; 39:895-906. [PMID: 36607471 DOI: 10.1007/s10554-022-02788-5] [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: 11/06/2022] [Accepted: 12/24/2022] [Indexed: 01/07/2023]
Abstract
Pediatric patients with β-thalassemia (β-TM) with preserved ejection fraction may experience early myocardial damage. This prospective study aimed to investigate left atrial (LA) function restructure in pediatric patients with β-TM by two-dimensional speckle tracking echocardiography (2D-STE) and evaluate the value of LA strain for predicting myocardial iron overload (MIO). We recruited 50 β-TM pediatric patients and 30 healthy children aged 3-14 years. The patients were assigned to a normal left ventricular (LV) lesion group (n = 20) and an enlarged LV lesion group (n = 30). Subjects all underwent echocardiography to measure conventional cardiac function parameters and LA strain parameters. The results displayed that LA reservoir strain (LASr), conduit strain (LAScd), contractile strain (LASct) and strain rate were significantly reduced in pediatric patients with β-TM with preserved ejection fraction. LASr, LAScd, and LASct were negatively correlated with the E/e' ratio, of which LASr had the most significant correlation (r = - 0.69, P < 0.001). LASr and LASct correlated positively with T2* (r = 0.70 and 0.62, respectively, all P < 0.001). In the multiple regression, LASr and LASct were independent predictors for T2*. The areas under the curve for LASr and LASct were 0.87 (P < 0.001) and 0.78 (P = 0.004), respectively. Our results demonstrated that LA strains were dramatically impaired in pediatric patients with β-TM, and LASr is an efficient indicator for detecting LV early diastolic dysfunction in β-TM pediatric patients and reflects early myocardial damage. LASr and LASct were independently predictive of MIO, but LASr was a more sensitive predictor.
Collapse
|
31
|
Uysal A, Oktay G, Ural C, Kalkan NB. The effect of ferritin levels on distal femoral cartilage thickness in patients with beta thalassaemia major. J Bone Miner Metab 2023; 41:95-104. [PMID: 36422676 DOI: 10.1007/s00774-022-01384-w] [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: 06/07/2022] [Accepted: 10/21/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION To the best of our knowledge, the present study is the first in the literature to assess distal femoral cartilage thickness and its relationship with ferritin levels in adult patients with beta thalassaemia major (BTM). MATERIALS AND METHODS 45 patients with BTM and 45 healthy controls were included in the study. Ferritin and haemoglobin levels of the patient and healthy groups were determined by blood analysis and distal femoral cartilage thicknesses were measured via ultrasound. Then, the patient group was divided into subgroups according to whether their ferritin levels were below or above 2500 µg/L. They were then compared among themselves and with the healthy control group using the available data. RESULTS Distal femoral cartilage thickness values were statistically significantly lower in the BTM group compared to the healthy control group (p values < 0.001). Patients with a ferritin level below 2500 µg/L had statistically significantly higher right and left average distal femoral cartilage thickness values than the patients with a ferritin level above 2500 µg/L (p = 0.029 and p = 0.019, respectively). The right and left average distal femoral cartilage thickness values of the patient subgroup with low ferritin levels were statistically similar to the control group (p = 0.146 and p = 0.164, respectively). CONCLUSION Our study showed that thalassaemia patients are more likely to develop osteoarthritis (OA) than the normal population and possible OA development can be prevented by keeping the ferritin levels of these patients in the optimum range.
Collapse
Affiliation(s)
- Alper Uysal
- Physical Medicine and Rehabilitation Clinic, Hatay Training and Research Hospital, Güzelburç, 31001, Antakya, Hatay, Turkey.
| | - Gönül Oktay
- Thalassemia Center, Hatay Training and Research Hospital, Hatay, Turkey
| | - Cihan Ural
- Hematology Clinic, Hatay Training and Research Hospital, Hatay, Turkey
| | | |
Collapse
|
32
|
Chun GY, Mohd Tahir NA, Islahudin F, Selvaratnam V, Li SC. Drug-related problems among transfusion-dependent thalassemia patients: A real-world evidence study. Front Pharmacol 2023; 14:1128887. [PMID: 37153805 PMCID: PMC10157080 DOI: 10.3389/fphar.2023.1128887] [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: 12/21/2022] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
Introduction: Thalassemia is among the most common genetic disorders globally and many patients suffer from iron overload (IOL) complications that mainly affect the heart, liver and endocrine system. These events may be further complicated by drug-related problems (DRP), an inherent issue among patients with chronic diseases. Objective: The study aimed to evaluate the burden, associated factors and impacts of DRP in transfusion-dependent thalassemia (TDT) patients. Method: Eligible TDT patients under follow-up in a tertiary hospital between 01 March 2020 to 30 April 2021 were interviewed and their medical records were reviewed retrospectively to identify any DRP. DRPs were classified using the Pharmaceutical Care Network Europe (PCNE) classification version 9.1. The incidence and preventability of DRP were assessed and the associated risk factors were estimated by univariate and multivariate logistic regression. Results: A total of 200 patients were enrolled with a median (interquartile range: IQR) age of 28 years at enrolment. Approximately 1 in 2 patients were observed to suffer from thalassemia-related complications. Throughout the study period, 308 DRPs were identified among 150 (75%) participants, with a median DRP per participant of 2.0 (IQR 1.0-3.0). Of the three DRP dimensions, treatment effectiveness was the most common DRP (55.8%) followed by treatment safety (39.6%) and other DRP (4.6%). The median serum ferritin level was statistically higher in patients with DRP compared with patients without DRP (3833.02 vs. 1104.98 μg/L, p < 0.001). Three risk factors were found to be significantly associated with the presence of DRP. Patients with frequent blood transfusion, moderate to high Medication Complexity Index (MRCI) and of Malay ethnicity were associated with higher odds of having a DRP (AOR 4.09, 95% CI: 1.83, 9.15; AOR 4.50, 95% CI: 1.89, 10.75; and AOR 3.26, 95% CI: 1.43, 7.43, respectively). Conclusion: The prevalence of DRP was relatively high amongst TDT patients. Increased medication complexity, more severe form of the disease and Malay patients were more likely to experience DRP. Hence, more viable interventions targeted to these groups of patients should be undertaken to mitigate the risk of DRP and achieve better treatment outcomes.
Collapse
Affiliation(s)
- Geok Ying Chun
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Centre for Clinical Trial, Ampang Hospital, Ampang, Selangor, Malaysia
| | - Nurul Ain Mohd Tahir
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- *Correspondence: Nurul Ain Mohd Tahir,
| | - Farida Islahudin
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Shu Chuen Li
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
33
|
Yousuf R, Akter S, Wasek SM, Sinha S, Ahmad R, Haque M. Thalassemia: A Review of the Challenges to the Families and Caregivers. Cureus 2022; 14:e32491. [PMCID: PMC9747324 DOI: 10.7759/cureus.32491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2022] [Indexed: 12/15/2022] Open
|
34
|
Zhuang J, Zhao Z, Zhang C, Song X, Lu C, Tian X, Jiang H. Case report: Successful outcome of treatment using rituximab in an adult patient with refractory minimal change disease and β-thalassemia complicating autoimmune hemolytic anemia. Front Med (Lausanne) 2022; 9:1059740. [PMID: 36405580 PMCID: PMC9669371 DOI: 10.3389/fmed.2022.1059740] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 10/18/2022] [Indexed: 10/31/2023] Open
Abstract
Minimal change disease (MCD) is one of the common causes of idiopathic nephrotic syndrome (INS), accounting for 10-20% of INS in adults. Glucocorticoids are the most commonly used and effective drugs in the treatment of MCD, but there is still a proportion of adult patients with MCD who are characterized by glucocorticoid resistance, glucocorticoid dependence, and frequent relapse, which are defined as refractory nephrotic syndrome. Glucocorticoid combination with immunosuppressants is frequently used in patients with refractory nephrotic syndrome, and patients concerned about adverse effects caused by long-term high-dose glucocorticoid therapy. Recent studies have suggested that Rituximab (RTX), a chimeric monoclonal antibody targeted against the pan-B-cell marker CD20, combined with a small or medium dose of glucocorticoid has a beneficial effect with less adverse effects on adult patients with refractory MCD. β-thalassemia is an inherited hemoglobulin disorder caused by the mutation of genes that encode β-globin and results in ineffective erythropoiesis. We here report a case of an adult patient with refractory MCD complicated with β-thalassemia minor accompanied by autoimmune hemolytic anemia (AIHA). MCD relapsed several times despite treatment using glucocorticoid combined with or without different immunosuppressive agent regimens. The β-thalassemia minor was caused by heterozygosity for a 4-base deletion mutation [codons 41/42 (-TTCT) BETA0] of the β-globin gene. After the administration of RTX, MCD achieved clinical complete remission, and the anemia due to mild β-thalassemia recovered to normal as well. The disease situation remained stable during 36 months of follow-up. These findings suggest that RTX may contribute to the improvement of refractory MCD and anemia in β-thalassemia minor accompanied by AIHA.
Collapse
Affiliation(s)
- Jing Zhuang
- Division of Nephrology, Department of Internal, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Zhigang Zhao
- Division of Nephrology, Department of Internal, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Changrong Zhang
- Division of Nephrology, Department of Internal, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xue Song
- Division of Nephrology, Department of Internal, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Chen Lu
- Division of Nephrology, Department of Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xuefei Tian
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Hong Jiang
- Division of Nephrology, Department of Internal, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
35
|
Shash H. Non-Transfusion-Dependent Thalassemia: A Panoramic Review. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58101496. [PMID: 36295656 PMCID: PMC9608723 DOI: 10.3390/medicina58101496] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/08/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022]
Abstract
Non-transfusion-dependent thalassemia (NTDT) has been considered less severe than its transfusion-dependent variants. The most common forms of NTDT include β-thalassemia intermedia, hemoglobin E/beta thalassemia, and hemoglobin H disease. Patients with NTDT develop several clinical complications, despite their regular transfusion independence. Ineffective erythropoiesis, iron overload, and hypercoagulability are pathophysiological factors that lead to morbidities in these patients. Therefore, an early and accurate diagnosis of NTDT is essential to ascertaining early interventions. Currently, several conventional management options are available, with guidelines suggested by the Thalassemia International Federation, and novel therapies are being developed in light of the advancement of the understanding of this disease. This review aimed to increase clinicians’ awareness of NTDT, from its basic medical definition and genetics to its pathophysiology. Specific complications to NTDT were reviewed, along with the risk factors for its development. The indications of different therapeutic options were outlined, and recent advancements were reviewed.
Collapse
Affiliation(s)
- Hwazen Shash
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia;
- Department of Pediatrics, King Fahad Hospital of the University, Al-Khobar 31952, Saudi Arabia
| |
Collapse
|
36
|
Pellegrino C, Dragonetti G, Chiusolo P, Rossi M, Orlando N, Teofili L. Acute Promyelocytic Leukemia in a Woman with Thalassemia Intermedia: Case Report and Review of Literature on Hematological Malignancies in β-Thalassemia Patients. Hematol Rep 2022; 14:310-321. [PMID: 36278522 PMCID: PMC9590045 DOI: 10.3390/hematolrep14040045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 10/01/2022] [Accepted: 10/17/2022] [Indexed: 11/04/2022] Open
Abstract
Patients affected by transfusion-dependent β-thalassemia are prone to developing several clinical complications, mostly related to the iron overload. We report the case of a patient affected by transfusion-dependent β-thalassemia (TDT) developing acute promyelocytic leukemia (APL). In our case, the therapeutic management was significantly complicated not only by myocardial dysfunction, but also by the occurrence of the differentiation syndrome following all-trans retinoic acid (ATRA) administration. We carried out a careful revision of the current literature on the occurrence of hematological malignancies in β-thalassemia patients to investigate the major complications so far described. APL occurrence in β-thalassemia patients has been very rarely reported, and our experience suggests that TDT patients suffering pre-existing comorbidities may develop a potentially fatal complication during ATRA therapy.
Collapse
Affiliation(s)
- Claudio Pellegrino
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, I-00168 Roma, Italy
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, I-00168 Rome, Italy
- Correspondence: ; Tel.: +06-30156016
| | - Giulia Dragonetti
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, I-00168 Roma, Italy
| | - Patrizia Chiusolo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, I-00168 Roma, Italy
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, I-00168 Rome, Italy
| | - Monica Rossi
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, I-00168 Roma, Italy
| | - Nicoletta Orlando
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, I-00168 Roma, Italy
| | - Luciana Teofili
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, I-00168 Roma, Italy
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, I-00168 Rome, Italy
| |
Collapse
|
37
|
Hsu CC, Senussi NH, Fertrin KY, Kowdley KV. Iron overload disorders. Hepatol Commun 2022; 6:1842-1854. [PMID: 35699322 PMCID: PMC9315134 DOI: 10.1002/hep4.2012] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 04/06/2022] [Accepted: 04/16/2022] [Indexed: 01/19/2023] Open
Abstract
Iron overload disorders represent a variety of conditions that lead to increased total body iron stores and resultant end-organ damage. An elevated ferritin and transferrin-iron saturation can be commonly encountered in the evaluation of elevated liver enzymes. Confirmatory homeostatic iron regulator (HFE) genetic testing for C282Y and H63D, mutations most encountered in hereditary hemochromatosis, should be pursued in evaluation of hyperferritinemia. Magnetic resonance imaging with quantitative assessment of iron content or liver biopsy (especially if liver disease is a cause of iron overload) should be used as appropriate. A secondary cause for iron overload should be considered if HFE genetic testing is negative for the C282Y homozygous or C282Y/H63D compound heterozygous mutations. Differential diagnosis of secondary iron overload includes hematologic disorders, iatrogenic causes, or chronic liver diseases. More common hematologic disorders include thalassemia syndromes, myelodysplastic syndrome, myelofibrosis, sideroblastic anemias, sickle cell disease, or pyruvate kinase deficiency. If iron overload has been excluded, evaluation for causes of hyperferritinemia should be pursued. Causes of hyperferritinemia include chronic liver disease, malignancy, infections, kidney failure, and rheumatic conditions, such as adult-onset Still's disease or hemophagocytic lymphohistiocytosis. In this review, we describe the diagnostic testing of patients with suspected hereditary hemochromatosis, the evaluation of patients with elevated serum ferritin levels, and signs of secondary overload and treatment options for those with secondary iron overload.
Collapse
Affiliation(s)
- Christine C. Hsu
- Medstar Georgetown University HospitalMedstar Georgetown Transplant InstituteWashingtonDistrict of ColumbiaUSA
| | - Nizar H. Senussi
- Gastroenterology and HepatologyUniversity of New MexicoAlbuquerqueNew MexicoUSA
| | - Kleber Y. Fertrin
- Division of HematologyDepartment of MedicineUniversity of WashingtonWashingtonUSA
| | - Kris V. Kowdley
- Liver Institute Northwest and Elson S. Floyd College of MedicineWashington State UniversityWashingtonUSA
| |
Collapse
|
38
|
Locke M, Reddy PS, Badawy SM. Adherence to Iron Chelation Therapy among Adults with Thalassemia: A Systematic Review. Hemoglobin 2022; 46:201-213. [PMID: 35930250 PMCID: PMC9948767 DOI: 10.1080/03630269.2022.2072320] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Iron chelation therapy (ICT) is essential to prevent complications of iron overload in patients with transfusion-dependent thalassemia. However, the role that adherence to ICT plays in health-related outcomes is less well known. Our objectives were to identify adherence rates of ICT, and to assess methods of measurement, predictors of adherence, and adherence-related health outcomes in the literature published between 1980 and 2020. Of 543 articles, 43 met the inclusion criteria. Studies measured ICT adherence, predictors, and/or outcomes associated with adherence. Most studies were across multiple countries in Europe and North America (n = 8/43, 18.6%), recruited in clinics (n = 39/43, 90.7%), and focused on β-thalassemia (β-thal) (n = 25/43, 58.1%). Common methods of assessing ICT adherence included patient self-report (n = 24/43, 55.8%), pill count (n = 9/43, 20.9%), prescription refill history (n = 3/43, 7.0%), provider scoring (n = 3/43, 7.0%), and combinations of methods (n = 4/43, 9.3%). Studies reported adherence either in 'categories' with different levels of adherence (n = 24) or 'quantitatively' as a percentage of doses of medication taken out of those prescribed (n = 17). Adherence levels varied (median 91.7%, range 42.0-99.97%). Studies varied in sample size and methods of adherence assessment and reporting, which prohibited meta-analysis. Due to a lack of consensus on how adherence is defined, it is difficult to compare ICT adherence reporting. Further research is needed to establish guidelines for assessing adherence and identifying suboptimal adherence. Behavioral digital interventions have the potential to optimize ICT adherence and health outcomes.
Collapse
Affiliation(s)
- Margaret Locke
- Department of Internal Medicine, Zucker School of Medicine at Hofstra/Northwell Institute, Hempstead, NY, USA
| | - Paavani S. Reddy
- Department of Medical Education, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Sherif M. Badawy
- Division of Hematology, Oncology, and Stem Cell Transplant, Ann & Robert H. Lurie Children’s Hospital of Chicago, IL, USA,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
39
|
Aramayo-Singelmann C, Halimeh S, Proske P, Vignalingarajah A, Cario H, Christensen MO, Yamamoto R, Röth A, Reinhardt D, Reinhardt HC, Alashkar F. Screening and diagnosis of hemoglobinopathies in Germany: Current state and future perspectives. Sci Rep 2022; 12:9762. [PMID: 35697769 PMCID: PMC9192588 DOI: 10.1038/s41598-022-13751-8] [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: 02/21/2022] [Accepted: 05/12/2022] [Indexed: 11/21/2022] Open
Abstract
This monocentric study conducted at the Pediatric and Adult Hemoglobinopathy Outpatient Units of the University Hospital of Essen summarizes the results of hemoglobinopathies diagnosed between August 2018 and September 2021, prior to the introduction of a general newborn screening (NBS) for SCD in Germany (October 2021). In total, 339 patients (pts.), 182 pediatric [50.5% males (92/182)] and 157 adult pts. [75.8% females (119/157)] were diagnosed by molecular analysis. The most common (parental) descent among affected pts. were the Middle Eastern and North African/Turkey (Turkey: 19.8%, Syria: 11.8%, and Iraq: 5.9%), and the sub-Saharan African region (21.3%). Median age at diagnosis in pediatric carriers [N = 157; 54.1% males (85/157)] was 6.2 yrs. (range 1 (months) mos.–17.8 yrs.) and 31 yrs. (range 18–65 yrs.) in adults [N = 53; 75.2% females (115/153)]. Median age at diagnosis of homozygous or compound-heterozygous disease in pediatric pts. (72% (18/25) females) was 3.7 yrs., range 4 mos.–17 yrs. (HbSS (N = 13): 2.5 yrs., range 5 mos.–7.8 yrs.; HbS/C disease (N = 5): 8 yrs., range 1–8 yrs.; homozygous/compound heterozygous β-thalassemia (N = 5): 8 yrs., range 3–13 yrs.), in contrast to HbH disease (N = 5): 18 yrs. (median), range 12–40 yrs. Hemoglobinopathies represent a relevant health problem in Germany due to immigration and late diagnosis of second/third generation migrants. SCD-NBS will accelerate diagnosis and might result in reduction of disease-associated morbidity. However, diagnosis of carriers and/or disease-states (i.e. thalassemic syndromes) in newly immigrated and undiagnosed patients will further be delayed. A first major step has been taken, but further steps are required.
Collapse
Affiliation(s)
- Carmen Aramayo-Singelmann
- Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Susan Halimeh
- Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Coagulation Center Rhein-Ruhr, Duisburg, Germany
| | - Pia Proske
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Abinuja Vignalingarajah
- Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Holger Cario
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | | | | | - Alexander Röth
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dirk Reinhardt
- Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Hans Christian Reinhardt
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ferras Alashkar
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
40
|
Aprile A, Sighinolfi S, Raggi L, Ferrari G. Targeting the Hematopoietic Stem Cell Niche in β-Thalassemia and Sickle Cell Disease. Pharmaceuticals (Basel) 2022; 15:ph15050592. [PMID: 35631417 PMCID: PMC9146437 DOI: 10.3390/ph15050592] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 01/19/2023] Open
Abstract
In the last decade, research on pathophysiology and therapeutic solutions for β-thalassemia (BThal) and sickle cell disease (SCD) has been mostly focused on the primary erythroid defect, thus neglecting the study of hematopoietic stem cells (HSCs) and bone marrow (BM) microenvironment. The quality and engraftment of HSCs depend on the BM microenvironment, influencing the outcome of HSC transplantation (HSCT) both in allogeneic and in autologous gene therapy settings. In BThal and SCD, the consequences of severe anemia alter erythropoiesis and cause chronic stress in different organs, including the BM. Here, we discuss the recent findings that highlighted multiple alterations of the BM niche in BThal and SCD. We point out the importance of improving our understanding of HSC biology, the status of the BM niche, and their functional crosstalk in these disorders towards the novel concept of combined therapies by not only targeting the genetic defect, but also key players of the HSC–niche interaction in order to improve the clinical outcomes of transplantation.
Collapse
Affiliation(s)
- Annamaria Aprile
- San Raffaele-Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.S.); (L.R.)
- Correspondence: (A.A.); (G.F.)
| | - Silvia Sighinolfi
- San Raffaele-Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.S.); (L.R.)
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Laura Raggi
- San Raffaele-Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.S.); (L.R.)
- University of Milano Bicocca, 20126 Milan, Italy
| | - Giuliana Ferrari
- San Raffaele-Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (S.S.); (L.R.)
- Vita-Salute San Raffaele University, 20132 Milan, Italy
- Correspondence: (A.A.); (G.F.)
| |
Collapse
|
41
|
Mortality in β-thalassemia patients with confirmed pulmonary arterial hypertension on right heart catheterization. Blood 2022; 139:2080-2083. [PMID: 34986266 DOI: 10.1182/blood.2021014862] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/21/2021] [Indexed: 01/19/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening complication of β-thalassemia, especially in untransfused patients with thalassemia intermedia. Pinto and colleagues analyzed the outcome of 24 patients with PAH documented by right heart catheterization, and they report that with a median follow-up of 4 years, 54% died, most of which deaths were attributable to PAH. Patients who receive treatment that reduce their pulmonary pressures have improved survival, suggesting that improvement in monitoring and treatment are critical imperatives for these patients.
Collapse
|
42
|
Link between Genotype and Multi-Organ Iron and Complications in Children with Transfusion-Dependent Thalassemia. J Pers Med 2022; 12:jpm12030400. [PMID: 35330400 PMCID: PMC8950605 DOI: 10.3390/jpm12030400] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 01/27/2023] Open
Abstract
We evaluated the impact of the genotype on hepatic, pancreatic and myocardial iron content, and on hepatic, cardiac and endocrine complications in children with transfusion-dependent β-thalassemia (β-TDT). We considered 68 β-TDT patients (11.98 ± 3.67 years, 51.5% females) consecutively enrolled in the Extension-Myocardial Iron Overload in Thalassemia network. Iron overload was quantified by T2* technique and biventricular function by cine images. Replacement myocardial fibrosis was evaluated by late gadolinium enhancement technique. Three groups of patients were identified: homozygous β+ (N = 19), compound heterozygous β0β+ (N = 24), and homozygous β0 (N = 25). The homozygous β0 group showed significantly lower global heart and pancreas T2* values than the homozygous β+ group. Compared to patients with homozygous β+ genotype, β0β+ as well as β0β0 patients were more likely to have pancreatic iron overload (odds ratio = 6.53 and 10.08, respectively). No difference was detected in biventricular function parameters and frequency of replacement fibrosis. No patient had cirrhosis/fibrosis, diabetes or heart failure, and the frequency of endocrinopathies was comparable among the groups. In pediatric β-TDT patients, there is an association between genotype and cardiac and pancreatic iron overload. The knowledge of patients’ genotype can be valuable in predicting some patients’ phenotypic features and in helping the clinical management of β-TDT patients.
Collapse
|
43
|
Locatelli F, Thompson AA, Kwiatkowski JL, Porter JB, Thrasher AJ, Hongeng S, Sauer MG, Thuret I, Lal A, Algeri M, Schneiderman J, Olson TS, Carpenter B, Amrolia PJ, Anurathapan U, Schambach A, Chabannon C, Schmidt M, Labik I, Elliot H, Guo R, Asmal M, Colvin RA, Walters MC. Betibeglogene Autotemcel Gene Therapy for Non-β 0/β 0 Genotype β-Thalassemia. N Engl J Med 2022; 386:415-427. [PMID: 34891223 DOI: 10.1056/nejmoa2113206] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Betibeglogene autotemcel (beti-cel) gene therapy for transfusion-dependent β-thalassemia contains autologous CD34+ hematopoietic stem cells and progenitor cells transduced with the BB305 lentiviral vector encoding the β-globin (βA-T87Q) gene. METHODS In this open-label, phase 3 study, we evaluated the efficacy and safety of beti-cel in adult and pediatric patients with transfusion-dependent β-thalassemia and a non-β0/β0 genotype. Patients underwent myeloablation with busulfan (with doses adjusted on the basis of pharmacokinetic analysis) and received beti-cel intravenously. The primary end point was transfusion independence (i.e., a weighted average hemoglobin level of ≥9 g per deciliter without red-cell transfusions for ≥12 months). RESULTS A total of 23 patients were enrolled and received treatment, with a median follow-up of 29.5 months (range, 13.0 to 48.2). Transfusion independence occurred in 20 of 22 patients who could be evaluated (91%), including 6 of 7 patients (86%) who were younger than 12 years of age. The average hemoglobin level during transfusion independence was 11.7 g per deciliter (range, 9.5 to 12.8). Twelve months after beti-cel infusion, the median level of gene therapy-derived adult hemoglobin (HbA) with a T87Q amino acid substitution (HbAT87Q) was 8.7 g per deciliter (range, 5.2 to 10.6) in patients who had transfusion independence. The safety profile of beti-cel was consistent with that of busulfan-based myeloablation. Four patients had at least one adverse event that was considered by the investigators to be related or possibly related to beti-cel; all events were nonserious except for thrombocytopenia (in 1 patient). No cases of cancer were observed. CONCLUSIONS Treatment with beti-cel resulted in a sustained HbAT87Q level and a total hemoglobin level that was high enough to enable transfusion independence in most patients with a non-β0/β0 genotype, including those younger than 12 years of age. (Funded by Bluebird Bio; HGB-207 ClinicalTrials.gov number, NCT02906202.).
Collapse
Affiliation(s)
- Franco Locatelli
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Alexis A Thompson
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Janet L Kwiatkowski
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - John B Porter
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Adrian J Thrasher
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Suradej Hongeng
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Martin G Sauer
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Isabelle Thuret
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Ashutosh Lal
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Mattia Algeri
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Jennifer Schneiderman
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Timothy S Olson
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Ben Carpenter
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Persis J Amrolia
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Usanarat Anurathapan
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Axel Schambach
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Christian Chabannon
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Manfred Schmidt
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Ivan Labik
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Heidi Elliot
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Ruiting Guo
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Mohammed Asmal
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Richard A Colvin
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| | - Mark C Walters
- From IRCCS Ospedale Pediatrico Bambino Gesù, Sapienza, University of Rome, Rome (F.L., M. Algeri); Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago (A.A.T., J.S.); Children's Hospital of Philadelphia and Perelman School of Medicine at the University of Pennsylvania, Philadelphia (J.L.K., T.S.O.); University College London Hospital (J.B.P., B.C.) and University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust (A.J.T., P.J.A.) - all in London; Ramathibodi Hospital, Mahidol University, Bangkok, Thailand (S.H., U.A.); the Department of Pediatric Hematology, Oncology, and Stem Cell Transplantation in Children (M.G.S.) and the Institute of Experimental Hematology (A.S.), Hannover Medical School, Hannover, and GeneWerk, Heidelberg (M.S., I.L.) - both in Germany; Hôpital de la Timone (I.T.) and Institut Paoli-Calmettes Comprehensive Cancer Center (C.C.) - both in Marseille, France; the University of California, San Francisco, Benioff Children's Hospital, Oakland (A.L., M.C.W.); and the Division of Hematology-Oncology, Boston Children's Hospital, Harvard Medical School, Boston (A.S.), and Bluebird Bio, Cambridge (H.E., R.G., M. Asmal, R.A.C.) - all in Massachusetts
| |
Collapse
|
44
|
Bulgurcu SC, Canbolat Ayhan A, Emeksiz HC, Ovali F. Assessment of the Nutritional Status, Bone Mineralization, and Anthropometrics of Children with Thalassemia Major. Medeni Med J 2021; 36:325-332. [PMID: 34939399 PMCID: PMC8694160 DOI: 10.4274/mmj.galenos.2021.66915] [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: 01/19/2023] Open
Abstract
Objective: Children with thalassemia major (TM) are prone to growth failure and micronutrient deficiency. Thus, this study aimed to evaluate nutritional status, anthropometrics, and bone mineralization defects in patients with regular blood transfusion. Methods: Data obtained were analyzed by evaluating laboratory tests, anthropometric measures, and bone mineral density. Results: This study included 29 patients (62% male and 38% female) with a mean age of 12.26±4.74 years, mean pre-transfusion hemoglobin of 8.64±1.01 g/dL, and mean serum ferritin of 1158.6±556.8 ng/mL. Vitamin D (72.4%), selenium (72.4%), and folate (37.9%) deficiencies were most frequent. Hypocalcemia was observed in 17.2%, hypomagnesemia in 3.5%, and decreased ceruloplasmin in 10.3% of patients. Folate was higher between 2 and 6 years old (p=0.028). Ceruloplasmin was higher between 6 and 10 years old (p=0.018). Selenium was significantly higher in patients with a ferritin of ≥1,500 (p=0.008). No significant ferritin-related differences were found in other micronutrients (p>0.05). Body mass index (BMI) were <5 percentile (p) in 31% of patient, whereas none was >95 p. Height in 24.5% and weight in 20.7% of patients were <3 p, whereas none with >97 p. BMI of patients aged 10-18 years was significantly higher (p=0.001). Anthropometric percentiles did not significantly differ in the mean serum ferritin and micronutrient levels. Hypoparathyroidism was observed in 13.8% and hypothyroidism in 3.5% of patients. Low bone density was detected in 14.8% (2 osteopenic and 2 osteoporotic) of patients. Bone mineral density did not significantly differ in the ferritin and micronutrient levels. Conclusions: Nutritional support and deficiency prevention are important to minimize the burden of complications and increase the life expectancy and quality in patients with TM.
Collapse
Affiliation(s)
- Serap Cevher Bulgurcu
- Istanbul Medeniyet University Faculty of Medicine, Department of Pediatrics, Istanbul, Turkey
| | - Aylin Canbolat Ayhan
- Istanbul Medeniyet University Faculty of Medicine, Department of Pediatric Hematology-Oncology, Istanbul, Turkey
| | - Hamdi Cihan Emeksiz
- Istanbul Medeniyet University Faculty of Medicine, Department of Pediatric Endocrinology, Istanbul, Turkey
| | - Fahri Ovali
- Istanbul Medeniyet University Faculty of Medicine, Department of Pediatrics, Istanbul, Turkey
| |
Collapse
|
45
|
Tang A, Strat AN, Rahman M, Zhang H, Bao W, Liu Y, Shi D, An X, Manwani D, Shi P, Yazdanbakhsh K, Mendelson A. Murine bone marrow mesenchymal stromal cells have reduced hematopoietic maintenance ability in sickle cell disease. Blood 2021; 138:2570-2582. [PMID: 34329381 PMCID: PMC8678997 DOI: 10.1182/blood.2021012663] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/25/2021] [Indexed: 11/20/2022] Open
Abstract
Sickle cell disease (SCD) is characterized by hemolytic anemia, which can trigger oxidative stress, inflammation, and tissue injury that contribute to disease complications. Bone marrow mesenchymal stromal cells (MSCs) tightly regulate hematopoietic stem cell (HSC) homeostasis in health and disease, but their functionality in SCD remains unclear. We identified for the first time that murine SCD MSCs have altered gene signatures, reduced stem cell properties, and increased oxidative stress, due in part to hemolysis. Murine SCD MSCs had lower HSC maintenance ability in vitro and in vivo, as manifested by increased HSC mobilization and decreased HSC engraftment after transplant. Activation of Toll-like receptor-4 through p65 in MSCs further contributed to MSC dysfunction. Transfusions led to an improved MSC and HSC oxidative state in SCD mice. Improving the regulation between MSCs and HSCs has vital implications for enhancing clinical HSC transplantation and gene therapy outcomes and for identification of new molecular targets for alleviating SCD complications.
Collapse
Affiliation(s)
- Alice Tang
- Laboratory of Stem Cell Biology & Engineering
| | | | | | - Helen Zhang
- Laboratory of Stem Cell Biology & Engineering
| | - Weili Bao
- Laboratory of Complement Biology, and
| | | | - David Shi
- Laboratory of Stem Cell Biology & Engineering
| | - Xiuli An
- Laboratory of Membrane BiologyNew York Blood CenterNew YorkNY
| | - Deepa Manwani
- Department of Pediatrics, Montefiore Health Center, Albert Einstein College of Medicine, Children's Hospital at Montefiore, Bronx, NY; and
| | - Patricia Shi
- Sickle Cell Clinical Research Program, New York Blood Center, New York, NY
| | | | | |
Collapse
|
46
|
Musallam KM, Bou‐Fakhredin R, Cappellini MD, Taher AT. 2021 update on clinical trials in β-thalassemia. Am J Hematol 2021; 96:1518-1531. [PMID: 34347889 DOI: 10.1002/ajh.26316] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 01/19/2023]
Abstract
The treatment landscape for patients with β-thalassemia is witnessing a swift evolution, yet several unmet needs continue to persist. Patients with transfusion-dependent β-thalassemia (TDT) primarily rely on regular transfusion and iron chelation therapy, which can be associated with considerable treatment burden and cost. Patients with non-transfusion-dependent β-thalassemia (NTDT) are also at risk of significant morbidity due to the underlying anemia and iron overload, but treatment options in this patient subgroup are limited. In this review, we provide updates on clinical trials of novel therapies targeting the underlying pathology in β-thalassemia, including the α/non-α-globin chain imbalance, ineffective erythropoiesis, and iron dysregulation.
Collapse
Affiliation(s)
- Khaled M. Musallam
- Thalassemia Center, Burjeel Medical City Abu Dhabi United Arab Emirates
- International Network of Hematology London UK
| | - Rayan Bou‐Fakhredin
- Department of Internal Medicine American University of Beirut Medical Center Beirut Lebanon
| | - Maria Domenica Cappellini
- Department of Clinical Sciences and Community University of Milan, Ca’ Granda Foundation IRCCS Maggiore Policlinico Hospital Milan Italy
| | - Ali T. Taher
- Department of Internal Medicine American University of Beirut Medical Center Beirut Lebanon
| |
Collapse
|
47
|
Taher AT, Cappellini MD. Luspatercept for β-thalassemia: beyond red blood cell transfusions. Expert Opin Biol Ther 2021; 21:1363-1371. [PMID: 34404288 DOI: 10.1080/14712598.2021.1968825] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Red blood cell transfusions and iron chelation therapy are the cornerstone of treatment for β-thalassemia, with allogeneic hematopoietic stem cell transplantation and gene therapy offering further disease-management options for eligible patients. With up to 90% of severe cases of β-thalassemia occurring in resource-constrained countries, and estimates indicating that 22,500 deaths occur annually as a direct consequence of undertransfusion, provision of adequate treatment remains a major issue. AREAS COVERED In this review, we provide an overview of luspatercept, a first-in-class erythroid maturation agent, and present the available clinical data related to the treatment of β-thalassemia. EXPERT OPINION The recent approval of luspatercept offers a new, long-term therapeutic option for adult patients with transfusion-dependent β-thalassemia to reduce red blood cell transfusion burden, anemia, and iron overload.
Collapse
Affiliation(s)
- Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | | |
Collapse
|
48
|
Expression of the immune checkpoint receptors CTLA-4, LAG-3, and TIM-3 in β-thalassemia major patients: correlation with alloantibody production and regulatory T cells (Tregs) phenotype. Ann Hematol 2021; 100:2463-2469. [PMID: 34324022 DOI: 10.1007/s00277-021-04605-w] [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: 02/28/2021] [Accepted: 07/06/2021] [Indexed: 11/22/2022]
Abstract
Alloimmunization is a serious complication in β-thalassemia major patients as a result of repeated blood transfusion. The immune checkpoint receptors play an important role in regulating immune system homeostasis and the function of the immune cells. This study aimed to evaluate the expression of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), lymphocyte activation gene 3 (LAG-3), and T-cell immunoglobulin and mucin domain-containing protein-3 (TIM-3) immune checkpoint molecules in β-thalassemia major patients with and without alloantibody. For this purpose, 68 β-thalassemia major patients with (34 patients) and without (34 patients) alloantibody as well as 20 healthy controls were enrolled. The expression of these genes was evaluated in different groups of patients by SYBR Green real-time PCR method. Our results showed that the mean expression of LAG-3 was significantly increased in thalassemia patients compared to the control group (*P < 0.001). However, there was no significant difference in expression of the CTLA-4 and TIM-3 as well as LAG-3 genes between patients with and without alloantibody (P > 0.05). A positive correlation was observed between the level of LAG-3 expression with markers associated with Treg function including FOXP3 and GDF-15 genes in β-thalassemia major patients. Taken together, the LAG-3 molecule might have a more prominent role in the abnormality of the immune system in thalassemia patients especially the function of regulatory T cells (Tregs), prior to the CTLA-4 and TIM-3 genes.
Collapse
|
49
|
The European Medicines Agency Review of Luspatercept for the Treatment of Adult Patients With Transfusion-dependent Anemia Caused by Low-risk Myelodysplastic Syndromes With Ring Sideroblasts or Beta-thalassemia. Hemasphere 2021; 5:e616. [PMID: 34291195 PMCID: PMC8288896 DOI: 10.1097/hs9.0000000000000616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/08/2021] [Indexed: 01/19/2023] Open
Abstract
Luspatercept is a recombinant fusion protein that selectively binds to ligands belonging to the transforming growth factor-beta superfamily, resulting in erythroid maturation and differentiation. On June 25, 2020, a marketing authorization valid through the European Union (EU) was issued for luspatercept for the treatment of adult patients with transfusion-dependent anemia caused by very low-, low-, and intermediate-risk myelodysplastic syndromes (MDS) with ring sideroblasts, or those with transfusion-dependent beta thalassemia (BT). Luspatercept was evaluated in 2 separate phase 3, double-blind, placebo-controlled multicentre trials. The primary endpoints of these trials were the percentage of patients achieving transfusion independence over ≥8 weeks or longer for patients with MDS, and the percentage of patients achieving a ≥33% reduction in transfusion burden from baseline to week 13–24 for patients with BT. In the MDS trial, the percentage of responders was 37.91% versus 13.16%, P < 0.0001, for patients receiving luspatercept versus placebo, respectively. In the BT trial, the percentage of responders was 21.4% versus 4.5% (P < 0.0001) for luspatercept versus placebo, respectively. Treatment with luspatercept led to similar incidences of adverse events (AEs), but higher incidences of grade ≥3 AEs and serious AEs compared to placebo. The most frequently reported treatment-emergent AEs (≥15%) in the pooled luspatercept group were headache; back pain, bone pain, and arthralgia; diarrhea; fatigue; pyrexia; and cough. The aim of this article is to summarize the scientific review of the application, which led to the regulatory approval in the EU.
Collapse
|
50
|
Lin W, Zhang Q, Shen Z, Qu X, Wang Q, Wei L, Qiu Y, Yang J, Xu X, Lao J. Molecular and phenotype characterization of an elongated β-globin variant produced by HBB:C.313delA. Int J Lab Hematol 2021; 43:1620-1627. [PMID: 34271589 DOI: 10.1111/ijlh.13639] [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: 03/24/2021] [Revised: 05/25/2021] [Accepted: 06/02/2021] [Indexed: 11/27/2022]
Abstract
INTRODUCTION β-thalassemia is a severe hereditary hemolytic anemia. Due to the diversity of mutations spectrum, β-thalassemia manifests a highly heterogeneous clinical severity. We noted that a previous report characterized HBB:c.313delA, at the end of exon 2, as a β-thalassemia trait rather than dominant β-thalassemia, the classification given to similar mutations. We further explored the impact of this functional variant on globin structure through larger pedigree analysis and in vitro studies. METHODS Hematological analysis and molecular genotyping were conducted on the proband and his family members. We evaluated functional effects of the variant on β-globin gene in the proband's nucleated erythrocytes and transfected HEK-293T cells. Three-dimensional construction of protein structure was carried out in silico to demonstrate amino acid changes. RESULTS The thalassemia major proband was identified as a compound heterozygote of HBB:c.313delA and HBB:c.126_129delCTTT. Three family members with heterozygotes of HBB:c.313delA displayed microcytic hypochromic anemia. Molecular characterization demonstrated that the frameshift mutation could give rise to retro-positioning of the termination codon, resulting in an elongated β-globin chain with an extension of 10 amino acids. Clinical phenotype and functional experiments indicated that HBB:c.313delA led to β0 -thalassemia phenotype. CONCLUSION We concluded that the phenotype of HBB:c.313delA was mainly related to the stability of mutant mRNA, the degradation of mutant proteins, and production of inclusion bodies according to a systematic description of clinical phenotype and a series of molecular experiments.
Collapse
Affiliation(s)
- Wanying Lin
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Genetics Testing Engineering Research Center, Guangzhou, China.,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Qianqian Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Genetics Testing Engineering Research Center, Guangzhou, China.,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Zongrui Shen
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Genetics Testing Engineering Research Center, Guangzhou, China.,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Xiang Qu
- Department of Pediatrics, Liuzhou Worker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Qi Wang
- Department of Pediatrics, Liuzhou Worker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Liuyuan Wei
- Department of Pediatrics, Liuzhou Worker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Yuhao Qiu
- Department of Pediatrics, Liuzhou Worker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Jie Yang
- Department of Pediatrics, Liuzhou Worker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Xiangmin Xu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Guangdong Genetics Testing Engineering Research Center, Guangzhou, China.,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, China
| | - Jinquan Lao
- Department of Pediatrics, Liuzhou Worker's Hospital, Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| |
Collapse
|