1
|
Hayer S, Wong T, Feist C, Sun RC, Williams T, Shereck EB, Chon AH. Diagnosis and management of severe SPTA1-associated congenital anaemia in a family cohort affected by two founder variants. BMJ Case Rep 2025; 18:e265190. [PMID: 40355272 DOI: 10.1136/bcr-2025-265190] [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: 05/14/2025] Open
Abstract
Hereditary spherocytosis due to mutations in the SPTA gene is a rare cause of fetal anaemia which can present as hydrops fetalis. We present a unique family cohort demonstrating severe transfusion-dependent anaemia with prenatal onset due to two SPTA1 founder variants which were diagnosed over several years as genetic testing capabilities advanced. Each patient was managed antenatally with fetal blood sampling (FBS) and intrauterine transfusions (IUT) with varying levels of success. Out of six affected pregnancies, two resulted in fetal demise, two resulted in postnatal demise and two long-term survivors required bone marrow transplant. While FBS and IUT are key in the antenatal management, multidisciplinary care is essential given the long-term complex medical comorbidities among postnatal survivors.
Collapse
Affiliation(s)
- Sarena Hayer
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon, USA
| | - Trisha Wong
- Departments of Pediatrics and Pathology, Oregon Health & Science University, Portland, Oregon, USA
| | - Cori Feist
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon, USA
| | - Raphael C Sun
- Department of Surgery, Oregon Health & Science University, Portland, Oregon, USA
| | - Todd Williams
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, USA
| | - Evan B Shereck
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon, USA
| | - Andrew H Chon
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
2
|
Longitudinal proteomics study of serum changes after allogeneic HSCT reveals potential markers of metabolic complications related to aGvHD. Sci Rep 2022; 12:14002. [PMID: 35977993 PMCID: PMC9385631 DOI: 10.1038/s41598-022-18221-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/08/2022] [Indexed: 01/19/2023] Open
Abstract
Even though hematopoietic stem cell transplantation (HSCT) allows successful treatment for many malignant and non-malignant disorders, its curative potential remains limited by severe side effects, including infections and other transplant-related complications such as graft-versus-host disease (GvHD). This study examined changes in serum proteome via high-performance two-dimensional gel electrophoresis (2-DE) during HSCT to search for diagnostic biomarkers for post-HSCT complications. Longitudinal proteomic analysis revealed proteins related to metabolic complications and hemolytic anemia. Retinol-binding protein 4 (RBP4), a reliable marker of insulin resistance, was identified, and is possibly associated with the onset mechanism of acute graft-versus-host disease (aGvHD) and/or skin GvHD. Although the cause of insulin resistance is not fully understood, it is thought to be associated with adipocytes inflammation induced by RBP4, iron overload and hemolytic anemia after HSCT, as observed in this study. The present study has demonstrated that insulin resistance and metabolic complications could be immediate complications after transplantation and are associated with aGvHD. The biomarkers revealed in this study are promising tools to be used for improving the early diagnosis of HSCT-associated complications, especially aGvHD, possibly even before clinical manifestations.
Collapse
|
3
|
Duda K, Wieczorkiewicz-Kabut A, Spałek A, Koclęga A, Kopińska AJ, Woźniczka K, Helbig G. Room for Improvement: A 20-Year Single Center Experience with Allogeneic Stem Cell Transplantation for Myelodysplastic Syndromes. Indian J Hematol Blood Transfus 2021; 38:680-690. [PMID: 36258723 PMCID: PMC9569268 DOI: 10.1007/s12288-021-01508-8] [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: 06/18/2021] [Accepted: 11/29/2021] [Indexed: 11/24/2022] Open
Abstract
Allogeneic stem cell transplantation (allo-SCT) remains the only curative therapeutic approach for patients with myelodysplastic syndromes (MDS). The aim of the study was to assess the efficacy/safety of allo-SCT as well as to identify factors influencing post-transplant survival. One hundred and two MDS patients (median age: 48 years; 57 males) who underwent allo-SCT were retrospectively evaluated. Twenty seven patients were transplanted from HLA-matched sibling and 75 patients received grafts from unrelated donors. Peripheral blood was a source of stem cell for 79 patients. Reduced intensity conditioning was used in 64 subjects. Acute and chronic graft versus host disease (GvHD) developed in 61 and 19 of patients, respectively. In total, 61 patients have died. The causes of deaths included infectious complications (n = 30), steroid-resistant GvHD (n = 17), MDS relapse (n = 9) and transformation to AML (n = 5). Non-relapse mortality and cumulative incidence of relapse at 2 years were 49.8% and 9%, respectively. 41 patients are alive at last contact and present full donor chimerism. 38 patients remain in complete hematological remission (CHR), 3 patients had CHR with incomplete platelet recovery. Median follow-up from diagnosis of MDS and transplantation are 27.1 months and 7 months respectively. Overall survival and relapse-free survival were 41% at 2 years. Increased serum ferritin level > 1000 ng/ml, presence of acute GvHD, grades III–IV acute GvHD and high hematopoietic cell transplantation-comorbidity index were found to negatively influenced survival. Allo-SCT for MDS is feasible procedure with a proportion of patients to be cured.
Collapse
Affiliation(s)
- Katarzyna Duda
- Department of Hematology and Bone Marrow Transplantation, Medical School of Silesia, Silesian Medical University, Dąbrowski Street 25, 40-032 Katowice, Poland
| | - Agata Wieczorkiewicz-Kabut
- Department of Hematology and Bone Marrow Transplantation, Medical School of Silesia, Silesian Medical University, Dąbrowski Street 25, 40-032 Katowice, Poland
| | - Adrianna Spałek
- Department of Hematology and Bone Marrow Transplantation, Medical School of Silesia, Silesian Medical University, Dąbrowski Street 25, 40-032 Katowice, Poland
| | - Anna Koclęga
- Department of Hematology and Bone Marrow Transplantation, Medical School of Silesia, Silesian Medical University, Dąbrowski Street 25, 40-032 Katowice, Poland
| | - Anna J. Kopińska
- Department of Hematology and Bone Marrow Transplantation, Medical School of Silesia, Silesian Medical University, Dąbrowski Street 25, 40-032 Katowice, Poland
| | - Krzysztof Woźniczka
- Department of Hematology and Bone Marrow Transplantation, Medical School of Silesia, Silesian Medical University, Dąbrowski Street 25, 40-032 Katowice, Poland
| | - Grzegorz Helbig
- Department of Hematology and Bone Marrow Transplantation, Medical School of Silesia, Silesian Medical University, Dąbrowski Street 25, 40-032 Katowice, Poland
| |
Collapse
|
4
|
Unbalance in Iron Metabolism in Childhood Leukemia Converges with Treatment Intensity: Biochemical and Clinical Analysis. Cancers (Basel) 2021; 13:cancers13123029. [PMID: 34204310 PMCID: PMC8233795 DOI: 10.3390/cancers13123029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary In children undergoing therapy for acute leukemia or after hematopoietic cell transplantation, the following iron metabolism parameters were analyzed in the context of iron overload: (1) parameters measuring functional and storage iron pools: non-transferrin-bound iron (NTBI) and labile plasma iron (LPI) levels, iron, transferrin, total iron-binding capacity, ferritin, ferritin heavy and light chains; (2) proteins regulating iron absorption and its release from tissue stores: hepcidin, soluble hemojuvelin, soluble ferroportin-1; (3) proteins regulating the erythropoietic activity of bone marrow: erythroferrone, erythropoietin, soluble transferrin receptor. It has been shown that the occurrence of NTBI and LPI in the circulation and the intensification of disturbances in iron metabolism were associated with the intensity of anti-leukemic treatment and were the highest in the transplant group followed by the acute leukemia after treatment and de novo groups. In patients after transplantation, the most significant changes were found in NTBI, LPI, iron, ferritin, hepcidin, and ferroportin-1 levels. Abstract Objective: The aim of this study was to evaluate non-transferrin-bound iron (NTBI) and labile plasma iron (LPI) levels and other parameters of iron metabolism in children undergoing therapy for acute leukemia or after hematopoietic cell transplantation (HCT), in the context of iron overload. Patients: A total number of 85 children were prospectively included into four groups: controls, acute leukemia de novo, acute leukemia after intensive treatment, and after HCT. Methods: The following iron metabolism parameters were analyzed: (1) parameters measuring functional and storage iron pools: NTBI, LPI, iron, transferrin, total iron-binding capacity, ferritin, ferritin heavy and light chains; (2) proteins regulating iron absorption and its release from tissue stores: hepcidin, soluble hemojuvelin, soluble ferroportin-1; (3) proteins regulating the erythropoietic activity of bone marrow: erythroferrone, erythropoietin, soluble transferrin receptor. Results: Intensive treatment of leukemia in children was associated with the presence of serum NTBI and LPI, which was the highest in the HCT group followed by the acute leukemia after treatment and de novo groups. In patients after HCT, the most significant changes were found in NTBI, LPI, iron, ferritin, hepcidin, and ferroportin-1 levels. Conclusions: The occurrence of NTBI and LPI in the circulation and the intensification of disturbances in iron metabolism were associated with the intensity of the anti-leukemic treatment.
Collapse
|
5
|
Isidori A, Loscocco F, Visani G, Chiarucci M, Musto P, Kubasch AS, Platzbecker U, Vinchi F. Iron Toxicity and Chelation Therapy in Hematopoietic Stem Cell Transplant. Transplant Cell Ther 2021; 27:371-379. [PMID: 33969823 DOI: 10.1016/j.jtct.2020.11.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 01/19/2023]
Abstract
Many patients with hematologic malignancies receive RBC transfusion support, which often causes systemic and tissue iron toxicity. Because of their compromised bone marrow function, hematopoietic stem cell transplant (HSCT) recipients are especially vulnerable to excess iron levels. Iron toxicity may compromise transplant engraftment and eventually promote relapse by mediating oxidative and genotoxic stress in hematopoietic stem cells (HSCs) and further impairing the already dysfunctional bone marrow microenvironment in HSCT recipients. Iron toxicity is thought to be primarily mediated by its ability to induce reactive oxygen species and trigger inflammation. Elevated iron levels in the bone marrow can decrease the number of HSCs and progenitor cells, as well as their clonogenic potential, alter mesenchymal stem cell differentiation, and inhibit the expression of chemokines and adhesion molecules involved in hematopoiesis. In vivo, in vitro, and clinical studies support the concept that iron chelation therapy may limit iron toxicity in the bone marrow and promote hematologic improvement and engraftment in HSCT recipients. This review will provide an overview of the current knowledge of the detrimental impact of iron toxicity in the setting of HSCT in patients with hematologic malignancies and the use of iron restriction approaches to improve transplant outcome.
Collapse
Affiliation(s)
- Alessandro Isidori
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy.
| | - Federica Loscocco
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Martina Chiarucci
- Hematology and Stem Cell Transplant Center, AORMN Hospital, Pesaro, Italy
| | - Pellegrino Musto
- Unit of Hematology and Stem Cell Transplantation, Department of Emergency and Organ Transplantation, "Aldo Moro" University School of Medicine, AOU Consorziale Policlinico, Bari, Italy
| | - Anne-Sophie Kubasch
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Francesca Vinchi
- Iron Research Program, Lindsley Kimball Research Institute, New York Blood Center, New York, New York; Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
6
|
Huang Z, Xiao Y, Chen X, Li H, Gao J, Wei W, Zhang X, Feng X. Cotransplantation of Umbilical Cord Mesenchymal Stem Cells Promotes the Engraftment of Umbilical Cord Blood Stem Cells in Iron Overload NOD/SCID Mice. Transplant Cell Ther 2021; 27:230.e1-230.e7. [PMID: 35348116 DOI: 10.1016/j.jtct.2020.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/27/2020] [Indexed: 12/11/2022]
Abstract
Iron overload aggravates the difficulty of umbilical cord blood (UCB) stem cell engraftment and reduces the survival of patients undergoing hematopoietic stem cell (HSC) transplantation. Mesenchymal stem cells (MSCs) have been suggested to have a significant role in HSC engraftment. This study aimed to determine the effect of intra-bone marrow (IBM) and i.v. cotransplantation of UBC mononuclear cells (MNCs) and umbilical cord (UC) MSCs on engraftment and hematopoietic recovery in an iron overload hematopoietic microenvironment. The iron overload model was established by dose-escalation intraperitoneal injection of iron dextran in NOD/SCID mice. Iron deposition in the bone marrow, heart, and liver was examined using hematoxylin and eosin (H&E) staining. Serum levels of ferritin and iron status in the liver were measured. The iron overload NOD/SCID mice were sublethally irradiated and divided into 5 groups for transplantation: (1) control group; (2) IBM+ group: IBM injection of combined UCB-MNCs/UC-MSCs; (3) IV+ group: i.v. injection of combined UCB-MNCs/UC-MSCs; (4) IBM group: IBM injection of only UCB-MNCs; and (5) IV group: i.v. injection of UCB-MNCs. At 6 weeks after transplantation, the human CD45+ cells in the bone marrow were analyzed by flow cytometry. The semiquantitative analysis of vascular endothelial growth factor (VEGF-A), osteopontin (OPN), and stromal cell-derived factor-1a (SDF-1a) were examined by immunohistochemical staining (IHC). Compared with the IBM and IV groups, the survival rate and the percentages of human CD45+ cells and CD34+ cells and colony-forming units (CFU) in bone marrow were elevated in the IBM+ and IV+ groups. In addition, the levels of VEGF-A, OPN, and SDF-1a in bone marrow were all higher in the IBM+ and IV+ groups. Our data show that IBM and i.v. cotransplantation of UC-MSCs can improve the engraftment and proliferation of UCB-MNCs in iron overload NOD/SCID mice. The increased expression of VEGF-A, OPN, and SDF-1a in the bone marrow may be involved in improving the hematopoietic microenvironment and promoting the implantation of human UCB stem cells in the bone marrow with iron overload.
Collapse
Affiliation(s)
- Zhi Huang
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuhua Xiao
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaomin Chen
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huiping Li
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Gao
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Wei
- Experimental Center, Guangdong Cord Blood Bank, Guangzhou, China
| | - Xinyao Zhang
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoqin Feng
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Calabrese C, Panuzzo C, Stanga S, Andreani G, Ravera S, Maglione A, Pironi L, Petiti J, Shahzad Ali M, Scaravaglio P, Napoli F, Fava C, De Gobbi M, Frassoni F, Saglio G, Bracco E, Pergolizzi B, Cilloni D. Deferasirox-Dependent Iron Chelation Enhances Mitochondrial Dysfunction and Restores p53 Signaling by Stabilization of p53 Family Members in Leukemic Cells. Int J Mol Sci 2020; 21:ijms21207674. [PMID: 33081324 PMCID: PMC7589297 DOI: 10.3390/ijms21207674] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/02/2020] [Accepted: 10/10/2020] [Indexed: 12/11/2022] Open
Abstract
Iron is crucial to satisfy several mitochondrial functions including energy metabolism and oxidative phosphorylation. Patients affected by Myelodysplastic Syndromes (MDS) and acute myeloid leukemia (AML) are frequently characterized by iron overload (IOL), due to continuous red blood cell (RBC) transfusions. This event impacts the overall survival (OS) and it is associated with increased mortality in lower-risk MDS patients. Accordingly, the oral iron chelator Deferasirox (DFX) has been reported to improve the OS and delay leukemic transformation. However, the molecular players and the biological mechanisms laying behind remain currently mostly undefined. The aim of this study has been to investigate the potential anti-leukemic effect of DFX, by functionally and molecularly analyzing its effects in three different leukemia cell lines, harboring or not p53 mutations, and in human primary cells derived from 15 MDS/AML patients. Our findings indicated that DFX can lead to apoptosis, impairment of cell growth only in a context of IOL, and can induce a significant alteration of mitochondria network, with a sharp reduction in mitochondrial activity. Moreover, through a remarkable reduction of Murine Double Minute 2 (MDM2), known to regulate the stability of p53 and p73 proteins, we observed an enhancement of p53 transcriptional activity after DFX. Interestingly, this iron depletion-triggered signaling is enabled by p73, in the absence of p53, or in the presence of a p53 mutant form. In conclusion, we propose a mechanism by which the increased p53 family transcriptional activity and protein stability could explain the potential benefits of iron chelation therapy in terms of improving OS and delaying leukemic transformation.
Collapse
Affiliation(s)
- Chiara Calabrese
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
- Correspondence:
| | - Serena Stanga
- Department of Neuroscience Rita Levi Montalcini, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10126 Turin, Italy;
| | - Giacomo Andreani
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Silvia Ravera
- Human Anatomy Section, Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy;
| | - Alessandro Maglione
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Lucrezia Pironi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Jessica Petiti
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Muhammad Shahzad Ali
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Patrizia Scaravaglio
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Francesca Napoli
- Department of Oncology, University of Turin, 10043 Turin, Italy; (F.N.); (E.B.)
| | - Carmen Fava
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Marco De Gobbi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Francesco Frassoni
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Enrico Bracco
- Department of Oncology, University of Turin, 10043 Turin, Italy; (F.N.); (E.B.)
| | - Barbara Pergolizzi
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, 10043 Turin, Italy; (C.C.); (G.A.); (A.M.); (L.P.); (J.P.); (M.S.A.); (P.S.); (C.F.); (M.D.G.); (F.F.); (G.S.); (B.P.); (D.C.)
| |
Collapse
|
8
|
Sandler RD, Tattersall RS, Schoemans H, Greco R, Badoglio M, Labopin M, Alexander T, Kirgizov K, Rovira M, Saif M, Saccardi R, Delgado J, Peric Z, Koenecke C, Penack O, Basak G, Snowden JA. Diagnosis and Management of Secondary HLH/MAS Following HSCT and CAR-T Cell Therapy in Adults; A Review of the Literature and a Survey of Practice Within EBMT Centres on Behalf of the Autoimmune Diseases Working Party (ADWP) and Transplant Complications Working Party (TCWP). Front Immunol 2020; 11:524. [PMID: 32296434 PMCID: PMC7137396 DOI: 10.3389/fimmu.2020.00524] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022] Open
Abstract
Introduction: Secondary haemophagocytic lymphohistiocytosis (sHLH) or Macrophage Activation Syndrome (MAS) is a life-threatening hyperinflammatory syndrome that can occur in patients with severe infections, malignancy or autoimmune diseases. It is also a rare complication of haematopoetic stem cell transplantation (HSCT), with a high mortality. It may be associated with graft vs. host disease in the allogeneic HSCT setting. It is also reported following CAR-T cell therapy, but differentiation from cytokine release syndrome (CRS) is challenging. Here, we summarise the literature and present results of a survey of current awareness and practice in EBMT-affiliated centres of sHLH/MAS following HSCT and CAR-T cell therapy. Methods: An online questionnaire was sent to the principal investigators of all EBMT member transplant centres treating adult patients (18 years and over) inviting them to provide information regarding: number of cases of sHLH/MAS seen in their centre over 3 years (2016-2018 inclusive); screening strategies and use of existing diagnostic/classification criteria and treatment protocols. Results: 114/472 centres from 24 different countries responded (24%). We report estimated rates of sHLH/MAS of 1.09% (95% CI = 0.89-1.30) following allogeneic HSCT, 0.15% (95% CI = 0.09-5.89) following autologous HSCT and 3.48% (95% CI = 0.95-6.01) following CAR-T cell therapy. A majority of centres (70%) did not use a standard screening protocol. Serum ferritin was the most commonly used screening marker at 78% of centres, followed by soluble IL-2 receptor (24%), triglycerides (15%), and fibrinogen (11%). There was significant variation in definition of "clinically significant" serum ferritin levels ranging from 500 to 10,000 μg/mL. The most commonly used criteria to support diagnosis were HLH-2004 (43%) and the H score (15%). Eighty percent of responders reported using no standard management protocol, but reported using combinations of corticosteroids, chemotherapeutic agents, cytokine blockade, and monoclonal antibodies. Conclusions: There is a remarkable lack of consistency between EBMT centres in the approach to screening, diagnosis and management. Further research in this field is needed to raise awareness of and inform harmonised, evidence-based approaches to the recognition and treatment of sHLH/MAS following HSCT/CAR-T cell therapy.
Collapse
Affiliation(s)
- Robert David Sandler
- Department of Rheumatology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Rachel Scarlett Tattersall
- Department of Rheumatology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Helene Schoemans
- Department of Hematology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Raffaella Greco
- Haematology and BMT Unit, San Raffaele Hospital (IRCCS), Milan, Italy
| | - Manuela Badoglio
- EBMT Paris Study Office, Department of Haematology, Hôpital Saint-Antoine, Paris, France
| | - Myriam Labopin
- EBMT Paris Study Office, Department of Haematology, Hôpital Saint-Antoine, Paris, France
| | - Tobias Alexander
- Department of Rheumatology and Clinical Immunology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | | | - Montserrat Rovira
- BMT Unit, Department of Hematology, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Muhammad Saif
- Manchester Royal Infirmary, Manchester, United Kingdom
| | - Riccardo Saccardi
- Cell Therapy and Transfusion Medicine Unit, Careggi Hospital, Florence, Italy
| | - Julio Delgado
- BMT Unit, Department of Hematology, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Zinaida Peric
- School of Medicine, University of Zagreb, Zagreb, Croatia
- University Hospital Centre Zagreb, Zagreb, Croatia
| | - Christian Koenecke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hanover, Germany
| | - Olaf Penack
- Department of Hematology, Oncology and Tumorimmunology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Grzegorz Basak
- Department of Hematology, Oncology and Internal Medicine, University Clinical Center of the Medical University of Warsaw, Warsaw, Poland
| | - John Andrew Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| |
Collapse
|
9
|
Verna G, Liso M, De Santis S, Dicarlo M, Cavalcanti E, Crovace A, Sila A, Campiglia P, Santino A, Lippolis A, Serino G, Fasano A, Chieppa M. Iron Overload Mimicking Conditions Skews Bone Marrow Dendritic Cells Differentiation into MHCII lowCD11c +CD11b +F4/80 + Cells. Int J Mol Sci 2020; 21:ijms21041353. [PMID: 32079304 PMCID: PMC7072937 DOI: 10.3390/ijms21041353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 12/21/2022] Open
Abstract
Iron overload is an undesired effect of frequent blood transfusions or genetic diseases. Myelodysplastic syndrome (MDS) patients become transfusion dependent, but due to the combination of ineffective haematopoiesis and repeated blood transfusions they are often subject to iron overload. In this study, we demonstrate that iron-overload mimicking condition alters bone marrow progenitor differentiation towards dendritic cells (DCs). Cells cultured in iron-enriched culture medium for seven days fail to differentiate into conventional CD11c+MHCIIhi DCs and fail to efficiently respond to LPS (Lipopolysaccharides). Cells appear smaller than control DCs but vital and able to perform FITC-dextran (Fluorescein isothiocyanate-dextran) endocytosis. At molecular level, cells cultured in iron-enriched conditions show increased ARG1 and PU.1, and decreased IRF8 expression.
Collapse
Affiliation(s)
- Giulio Verna
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.V.); (M.L.); (M.D.); (E.C.); (A.C.); (A.S.); (A.L.); (G.S.)
- Department of Immunology and Cell Biology, European Biomedical Research Institute of Salerno (EBRIS), 84125 Salerno, Italy; (P.C.); (A.F.)
| | - Marina Liso
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.V.); (M.L.); (M.D.); (E.C.); (A.C.); (A.S.); (A.L.); (G.S.)
| | - Stefania De Santis
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy;
- Department of Pharmacy-Drug Science, University of Bari Aldo Moro, 70126 Bari, Italy
| | - Manuela Dicarlo
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.V.); (M.L.); (M.D.); (E.C.); (A.C.); (A.S.); (A.L.); (G.S.)
| | - Elisabetta Cavalcanti
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.V.); (M.L.); (M.D.); (E.C.); (A.C.); (A.S.); (A.L.); (G.S.)
| | - Alberto Crovace
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.V.); (M.L.); (M.D.); (E.C.); (A.C.); (A.S.); (A.L.); (G.S.)
| | - Annamaria Sila
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.V.); (M.L.); (M.D.); (E.C.); (A.C.); (A.S.); (A.L.); (G.S.)
| | - Pietro Campiglia
- Department of Immunology and Cell Biology, European Biomedical Research Institute of Salerno (EBRIS), 84125 Salerno, Italy; (P.C.); (A.F.)
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy;
| | - Angelo Santino
- Unit of Lecce, Institute of Sciences of Food Production C.N.R., via Monteroni, 73100 Lecce, Italy;
| | - Antonio Lippolis
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.V.); (M.L.); (M.D.); (E.C.); (A.C.); (A.S.); (A.L.); (G.S.)
| | - Grazia Serino
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.V.); (M.L.); (M.D.); (E.C.); (A.C.); (A.S.); (A.L.); (G.S.)
| | - Alessio Fasano
- Department of Immunology and Cell Biology, European Biomedical Research Institute of Salerno (EBRIS), 84125 Salerno, Italy; (P.C.); (A.F.)
- Harvard Medical School Division of Pediatric Gastroenterology and Nutrition and Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA 02114, USA
| | - Marcello Chieppa
- National Institute of Gastroenterology “S. de Bellis”, Research Hospital, Castellana Grotte, 70013 Bari, Italy; (G.V.); (M.L.); (M.D.); (E.C.); (A.C.); (A.S.); (A.L.); (G.S.)
- Department of Immunology and Cell Biology, European Biomedical Research Institute of Salerno (EBRIS), 84125 Salerno, Italy; (P.C.); (A.F.)
- Correspondence:
| |
Collapse
|
10
|
Kontoghiorghes GJ. How to manage iron toxicity in post-allogeneic hematopoietic stem cell transplantation? Expert Rev Hematol 2020; 13:299-302. [PMID: 32037922 DOI: 10.1080/17474086.2020.1719359] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science Technology, Environment and Medicine, Limassol, Cyprus
| |
Collapse
|
11
|
Sandler RD, Carter S, Kaur H, Francis S, Tattersall RS, Snowden JA. Haemophagocytic lymphohistiocytosis (HLH) following allogeneic haematopoietic stem cell transplantation (HSCT)-time to reappraise with modern diagnostic and treatment strategies? Bone Marrow Transplant 2020; 55:307-316. [PMID: 31455895 PMCID: PMC6995779 DOI: 10.1038/s41409-019-0637-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/28/2019] [Accepted: 06/07/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Robert David Sandler
- Department of Rheumatology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, S10 2JF, UK.
| | - Stuart Carter
- Department of Rheumatology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, S10 2JF, UK
| | - Harpreet Kaur
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, S10 2JF, UK
| | - Sebastian Francis
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, S10 2JF, UK
| | - Rachel Scarlett Tattersall
- Department of Rheumatology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, S10 2JF, UK
| | - John Andrew Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, S10 2JF, UK
| |
Collapse
|
12
|
Chawla LS, Beers-Mulroy B, Tidmarsh GF. Therapeutic Opportunities for Hepcidin in Acute Care Medicine. Crit Care Clin 2019; 35:357-374. [DOI: 10.1016/j.ccc.2018.11.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
13
|
Laribi K, Bolle D, Alani M, Ghnaya H, Le Bourdelles S, Besançon A, Farhi J, Denizon N, Baugier de Materre A. Prognostic impact of elevated pretreatment serum ferritin in patients with high-risk MDS treated with azacitidine. Exp Hematol 2018; 65:34-37. [DOI: 10.1016/j.exphem.2018.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/14/2018] [Accepted: 05/25/2018] [Indexed: 12/26/2022]
|
14
|
Abstract
Micronutrients are indispensable for adequate metabolism, such as biochemical function and cell production. The production of blood cells is named haematopoiesis and this process is highly consuming due to the rapid turnover of the haematopoietic system and consequent demand for nutrients. It is well established that micronutrients are relevant to blood cell production, although some of the mechanisms of how micronutrients modulate haematopoiesis remain unknown. The aim of the present review is to summarise the effect of Fe, Mn, Ca, Mg, Na, K, Co, iodine, P, Se, Cu, Li and Zn on haematopoiesis. This review deals specifically with the physiological requirements of selected micronutrients to haematopoiesis, showing various studies related to the physiological requirements, deficiency or excess of these minerals on haematopoiesis. The literature selected includes studies in animal models and human subjects. In circumstances where these minerals have not been studied for a given condition, no information was used. All the selected minerals have an important role in haematopoiesis by influencing the quality and quantity of blood cell production. In addition, it is highly recommended that the established nutrition recommendations for these minerals be followed, because cases of excess or deficient mineral intake can affect the haematopoiesis process.
Collapse
|
15
|
Magdaleno F, Ge X, Fey H, Lu Y, Gaskell H, Blajszczak CC, Aloman C, Fiel MI, Nieto N. Osteopontin deletion drives hematopoietic stem cell mobilization to the liver and increases hepatic iron contributing to alcoholic liver disease. Hepatol Commun 2018; 2:84-98. [PMID: 29404515 PMCID: PMC5776866 DOI: 10.1002/hep4.1116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 09/11/2017] [Accepted: 09/25/2017] [Indexed: 01/01/2023] Open
Abstract
The aim of this study was to investigate the role of osteopontin (OPN) in hematopoietic stem cell (HPSC) mobilization to the liver and its contribution to alcoholic liver disease (ALD). We analyzed young (14-16 weeks) and old (>1.5 years) wild-type (WT) littermates and global Opn knockout (Opn-/- ) mice for HPSC mobilization to the liver. In addition, WT and Opn-/- mice were chronically fed the Lieber-DeCarli diet for 7 weeks. Bone marrow (BM), blood, spleen, and liver were analyzed by flow cytometry for HPSC progenitors and polymorphonuclear neutrophils (PMNs). Chemokines, growth factors, and cytokines were measured in serum and liver. Prussian blue staining for iron deposits and naphthol AS-D chloroacetate esterase staining for PMNs were performed on liver sections. Hematopoietic progenitors were lower in liver and BM of young compared to old Opn-/- mice. Granulocyte colony-stimulating factor and macrophage colony-stimulating factor were increased in Opn-/- mice, suggesting potential migration of HPSCs from the BM to the liver. Furthermore, ethanol-fed Opn-/- mice showed significant hepatic PMN infiltration and hemosiderin compared to WT mice. As a result, ethanol feeding caused greater liver injury in Opn-/- compared to WT mice. Conclusion: Opn deletion promotes HPSC mobilization, PMN infiltration, and iron deposits in the liver and thereby enhances the severity of ALD. The age-associated contribution of OPN to HPSC mobilization to the liver, the prevalence of PMNs, and accumulation of hepatic iron, which potentiates oxidant stress, reveal novel signaling mechanisms that could be targeted for therapeutic benefit in patients with ALD. (Hepatology Communications 2018;2:84-98).
Collapse
Affiliation(s)
| | - Xiaodong Ge
- Department of PathologyUniversity of Illinois at ChicagoChicagoIL
- Division of Liver Diseases, Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNY
| | - Holger Fey
- Division of Digestive DiseasesRush University Medical CenterChicagoIL
| | - Yongke Lu
- Division of Liver Diseases, Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNY
| | - Harriet Gaskell
- Department of PathologyUniversity of Illinois at ChicagoChicagoIL
| | | | - Costica Aloman
- Division of Digestive DiseasesRush University Medical CenterChicagoIL
| | - M. Isabel Fiel
- Department of PathologyIcahn School of Medicine at Mount SinaiNew YorkNY
| | - Natalia Nieto
- Department of PathologyUniversity of Illinois at ChicagoChicagoIL
- Division of Liver Diseases, Department of MedicineIcahn School of Medicine at Mount SinaiNew YorkNY
- Department of Medicine, Division of Gastroenterology and HepatologyUniversity of Illinois at ChicagoChicagoIL
| |
Collapse
|
16
|
Shaw J, Chakraborty A, Nag A, Chattopadyay A, Dasgupta AK, Bhattacharyya M. Intracellular iron overload leading to DNA damage of lymphocytes and immune dysfunction in thalassemia major patients. Eur J Haematol 2017; 99:399-408. [PMID: 28815805 DOI: 10.1111/ejh.12936] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2017] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To investigate the cause and effects of intracellular iron overload in lymphocytes of thalassemia major patients. METHODS Sixty-six thalassemia major patients having iron overload and 10 age-matched controls were chosen for the study. Blood sample was collected, and serum ferritin, oxidative stress; lymphocyte DNA damage were examined, and infective episodes were also counted. RESULTS Case-control analysis revealed significant oxidative stress, iron overload, DNA damage, and rate of infections in thalassemia cases as compared to controls. For cases, oxidative stress (ROS) and iron overload (serum ferritin) showed good correlation with R2 = 0.934 and correlation between DNA damage and ROS gave R2 = 0.961. We also demonstrated that intracellular iron overload in thalassemia caused oxidative damage of lymphocyte DNA as exhibited by DNA damage assay. The inference is further confirmed by partial inhibition of such damage by chelation of iron and the concurrent lowering of the ROS level in the presence of chelator deferasirox. CONCLUSION Therefore, intracellular iron overload caused DNA fragmentation, which may ultimately hamper lymphocyte function, and this may contribute to immune dysfunction and increased susceptibility to infections in thalassemia patients as indicated by the good correlation (R2 = 0.91) between lymphocyte DNA damage and rate of infection found in this study.
Collapse
Affiliation(s)
- Jyoti Shaw
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Ayan Chakraborty
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Arijit Nag
- Institute of Haematology and Transfusion Medicine, Kolkata Medical College, Kolkata, India
| | - Arnab Chattopadyay
- Institute of Haematology and Transfusion Medicine, Kolkata Medical College, Kolkata, India
| | - Anjan K Dasgupta
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | | |
Collapse
|
17
|
Naoum FA, Espósito BP, Zotarelli Filho IJ. Impact of labile plasma iron and iron chelation on the viability of cultured mononuclear cells from patients undergoing autologous hematopoietic stem cell transplantation. Blood Res 2017; 52:135-136. [PMID: 28698851 PMCID: PMC5503892 DOI: 10.5045/br.2017.52.2.135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 09/27/2016] [Indexed: 11/20/2022] Open
|
18
|
Hilken A, Langebrake C, Wolschke C, Kersten JF, Rohde H, Nielsen P, Kröger N. Impact of non-transferrin-bound iron (NTBI) in comparison to serum ferritin on outcome after allogeneic stem cell transplantation (ASCT). Ann Hematol 2017; 96:1379-1388. [DOI: 10.1007/s00277-017-3034-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 05/25/2017] [Indexed: 01/19/2023]
|
19
|
Halawi R, Cappellini MD, Taher A. A higher prevalence of hematologic malignancies in patients with thalassemia: Background and culprits. Am J Hematol 2017; 92:414-416. [PMID: 28195443 DOI: 10.1002/ajh.24682] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 02/07/2017] [Accepted: 02/08/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Racha Halawi
- Emory University School of MedicineAtlanta Georgia USA
| | | | - Ali Taher
- American University of Beirut Medical CenterBeirut Lebanon
| |
Collapse
|
20
|
Yoo SH, Koh Y, Kim DY, Lee JH, Lee JH, Lee KH, Yoon SS, Park S, Park SK, Hong DS, Yi HG, Kim CS, Jang JE, Cheong JW, Moon J, Min YH, Sohn SK, Kim I. Salvage therapy for acute chemorefractory leukemia by allogeneic stem cell transplantation: the Korean experience. Ann Hematol 2017; 96:605-615. [PMID: 28091736 DOI: 10.1007/s00277-017-2919-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
Abstract
Little is known about the characteristics that make patients with acute leukemia suitable for undergoing salvage therapy by allogeneic hematopoietic stem cell transplantation (allo-HSCT). Here, we analyzed the clinical outcomes of 223 patients with acute leukemia who underwent allo-HSCT while not in complete remission (CR). The primary end points were overall survival (OS) and CR rate. CR was achieved in 79.8% of patients after allo-HSCT. Acute graft-versus-host disease (GVHD) was significantly associated with CR (P = 0.045). During a median follow-up of 30.1 months, the median OS was 6.1 months. OS was significantly longer in patients with good or standard risk cytogenetic characteristics than in those with poor risk cytogenetic characteristics (P = 0.029, P = 0.030, respectively). Patients who received allo-HSCT from a matched sibling donor had better survival than those with unrelated donors (P = 0.015). Primary chemorefractoriness was not associated with poor survival (P = 0.071). The number of chemotherapies before allo-HSCT was significantly correlated with outcome (P = 0.006). Chronic GVHD was a strong predictor of a longer OS (P = 0.025). In conclusion, survival of patients with primary chemorefractory acute leukemia is not lower when treated upfront with allo-HSCT. Hence, allo-HSCT should be actively considered in such patients. Acute and chronic GVHD is associated with better outcomes patients with acute leukemia who have undergone allo-HSCT and not achieved CR.
Collapse
Affiliation(s)
- Shin Hye Yoo
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Chongno-gu, Seoul, 110-744, South Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Chongno-gu, Seoul, 110-744, South Korea
| | - Dae-Young Kim
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jung-Hee Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Je-Hwan Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kyoo-Hyung Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sung-Soo Yoon
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Chongno-gu, Seoul, 110-744, South Korea
| | - Seonyang Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Chongno-gu, Seoul, 110-744, South Korea
| | - Sung-Kyu Park
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Bucheon, South Korea
| | - Dae-Sik Hong
- Department of Internal Medicine, Soonchunhyang University College of Medicine, Bucheon, South Korea
| | - Hyeon Gyu Yi
- Department of Internal Medicine, Inha University College of Medicine, Incheon, South Korea
| | - Chul-Soo Kim
- Department of Internal Medicine, Inha University College of Medicine, Incheon, South Korea
| | - Ji Eun Jang
- Department of Internal Medicine, Yonsei University Severance Hospital, 250 Sungsan-ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - June-Won Cheong
- Department of Internal Medicine, Yonsei University Severance Hospital, 250 Sungsan-ro, Seodaemun-gu, Seoul, 120-752, South Korea
| | - Joonho Moon
- Department of Internal Medicine, Kyung-Pook University Hospital, Daegu, South Korea
| | - Yoo Hong Min
- Department of Internal Medicine, Yonsei University Severance Hospital, 250 Sungsan-ro, Seodaemun-gu, Seoul, 120-752, South Korea.
| | - Sang Kyun Sohn
- Department of Internal Medicine, Kyung-Pook University Hospital, Daegu, South Korea. .,Department of Internal Medicine, Kyungpook National University Hospital, Kyungpook National University School of Medicine, 200 Dongduk-ro, Jung-gu, Daegu, 700-721, South Korea.
| | - Inho Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Chongno-gu, Seoul, 110-744, South Korea.
| | | |
Collapse
|
21
|
Cremers EMP, van Biezen A, de Wreede LC, Scholten M, Vitek A, Finke J, Platzbecker U, Beelen D, Schwerdtfeger R, Volin L, Harhalakis N, Blijlevens N, Nagler A, Kröger N, de Witte T. Prognostic pre-transplant factors in myelodysplastic syndromes primarily treated by high dose allogeneic hematopoietic stem cell transplantation: a retrospective study of the MDS subcommittee of the CMWP of the EBMT. Ann Hematol 2016; 95:1971-1978. [PMID: 27650829 PMCID: PMC5093200 DOI: 10.1007/s00277-016-2802-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/26/2016] [Indexed: 01/28/2023]
Abstract
Many pre-transplant factors are known to influence the outcome of allogeneic stem cell transplantation (SCT) treatment in myelodysplastic syndromes (MDS). However, patient cohorts are often heterogeneous by disease stage and treatment modalities, which complicates interpretation of the results. This study aimed to obtain a homogeneous patient cohort by including only de novo MDS patients who received upfront allogeneic SCT after standard high dose myelo-ablative conditioning. The effect of pre-transplant factors such as age, disease stage, transfusions, iron parameters and comorbidity on overall survival (OS), non-relapse mortality (NRM), and relapse incidence (RI) was evaluated in 201 patients. In this cohort, characterized by low comorbidity and a short interval between diagnosis and transplantation, NRM was the most determinant factor for survival after SCT (47 % after 2-year follow-up). WHO classification and transfusion burden were the only modalities with a significant impact on overall survival after SCT. Estimated hazard ratios (HR) showed a strongly increased risk of death, NRM and RI, in patients with a high transfusion-burden (HR 1.99; P = 0.006, HR of 1.89; P = 0.03 and HR 2.67; P = 0.03). The HR's for ferritin level and comorbidity were not significantly increased.
Collapse
Affiliation(s)
- E M P Cremers
- VU University Medical Center, Amsterdam, The Netherlands. .,Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - A van Biezen
- Leiden University Medical Center, Leiden, The Netherlands
| | - L C de Wreede
- Leiden University Medical Center, Leiden, The Netherlands
| | - M Scholten
- Leiden University Medical Center, Leiden, The Netherlands
| | - A Vitek
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - J Finke
- University of Freiburg, Freiburg, Germany
| | | | - D Beelen
- University Hospital, Essen, Germany
| | | | - L Volin
- Helsinki University Central Hospital, Helsinki, Finland
| | | | - N Blijlevens
- Radboud University Medical Centre, Nijmegen, The Netherlands
| | - A Nagler
- Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - N Kröger
- University Hospital Eppendorf, Hamburg, Germany
| | - T de Witte
- Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
22
|
Avdić A, Tucker S, Evans R, Smith A, Zimmerman MB. Comparing the ratio of mean red blood cell transfusion episode rate of 1 unit versus 2 units in hematopoietic stem cell transplant patients. Transfusion 2016; 56:2346-51. [DOI: 10.1111/trf.13708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 05/20/2016] [Accepted: 05/23/2016] [Indexed: 12/22/2022]
Affiliation(s)
| | | | | | - Anne Smith
- Department of Nursing Services and Patient Care
| | | |
Collapse
|
23
|
Murray C, De Gelder T, Pringle N, Johnson JC, Doherty M. Management of iron overload in the Canadian hematology/oncology population: Implications for nursing practice. Can Oncol Nurs J 2016; 26:19-28. [PMID: 31148725 PMCID: PMC6516318 DOI: 10.5737/236880762611928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Red blood cell (RBC) transfusions are vital for many patients with chronic anemias associated with oncologic/hematologic disorders. However, repeated transfusions over time can lead to iron overload, which, if left untreated, can increase the risk of further malignancy and end-organ damage. Nurses and other health care professionals may not be aware of the significant implications of RBC transfusions and iron overload in patients with hematological/oncological disorders. This article was developed by a group of Canadian nurse practitioners and specialized oncology nurses to help improve health care professionals' understanding of iron overload in oncology patients and its associated risks, as well as provide a practical guide for the management of patients receiving treatment for this potentially serious condition.
Collapse
Affiliation(s)
- Cindy Murray
- Nurse Practitioner, Blood and Marrow Disorder Program - Transfusion Outpatient Clinic, University Health Network (UHN) Princess Margaret Cancer Centre, Toronto, ON
| | - Tammy De Gelder
- Nurse Practitioner, Hamilton Health Sciences, Juravinski Hospital and Cancer Centre, Hamilton, ON
| | - Nancy Pringle
- Specialized Oncology Nurse, Leukemia Clinic, UHN Princess Margaret Cancer Centre, Toronto, ON
| | - J Colleen Johnson
- Nurse Practitioner, Red Blood Cell Disorders Clinic, UHN, Toronto General Division, Toronto, ON
| | - Mary Doherty
- Nurse Practitioner, Blood and Marrow Disorder Program - Transfusion Outpatient Clinic, University Health Network (UHN) Princess Margaret Cancer Centre, Toronto, ON
| |
Collapse
|
24
|
Murray C, De Gelder T, Pringle N, Johnson JC, Doherty M. Gestion de la surcharge en fer auprès des patients en hématologie et en oncologie: répercussions sur la pratique. Can Oncol Nurs J 2016; 26:29-39. [PMID: 31148744 PMCID: PMC6516315 DOI: 10.5737/236880762612939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Les transfusions de globules rouges sont vitales pour de nombreux patients aux prises avec de l’anémie chronique associée à des maladies oncologiques et hématologiques. Cependant, les transfusions répétées au fil du temps peuvent causer une surcharge en fer, laquelle, si elle n’est pas traitée, peut augmenter le risque de tumeur maligne et d’atteintes aux organes terminaux. Les infirmières et les autres professionnels de la santé peuvent ne pas être au fait de l’impact majeur des transfusions de globules rouges et de la surcharge en fer sur les patients en hématologie et en oncologie. Cet article a été élaboré par un groupe canadien d’infirmières praticiennes et d’infirmières spécialisées en oncologie pour aider les professionnels de soins de santé à mieux comprendre la surcharge en fer chez les patients atteints de cancer et les risques associés, et pour offrir un guide pratique de gestion des patients traités pour cet état potentiellement grave.
Collapse
Affiliation(s)
- Cindy Murray
- Infirmière praticienne (IP), programme sur les maladies du sang et de la moelle osseuse, consultation externe pour les transfusions, Réseau universitaire de santé (University Health Network, UHN), Centre de cancérologie Princess Margaret, Toronto, ON
| | - Tammy De Gelder
- Infirmière praticienne, Centre de cancérologie et Juravinski Hospital, Hamilton Health Sciences, Hamilton, ON
| | - Nancy Pringle
- Infirmière spécialisée en oncologie, clinique de leucémie, Réseau universitaire de santé, Centre de cancérologie Princess Margaret, Toronto, ON
| | - J Colleen Johnson
- Infirmière praticienne, clinique de maladies des globules rouges, Réseau universitaire de santé, Toronto General Division, Toronto, ON
| | - Mary Doherty
- Infirmière praticienne (IP), programme sur les maladies du sang et de la moelle osseuse, consultation externe pour les transfusions, Réseau universitaire de santé (University Health Network, UHN), Centre de cancérologie Princess Margaret, Toronto, ON
| |
Collapse
|
25
|
Hematopoietic Stem Cell Transplantation. PATHOLOGY OF TRANSPLANTATION 2016. [PMCID: PMC7124099 DOI: 10.1007/978-3-319-29683-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
26
|
Porter JB, de Witte T, Cappellini MD, Gattermann N. New insights into transfusion-related iron toxicity: Implications for the oncologist. Crit Rev Oncol Hematol 2015; 99:261-71. [PMID: 26806144 DOI: 10.1016/j.critrevonc.2015.11.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 09/28/2015] [Accepted: 11/24/2015] [Indexed: 01/19/2023] Open
Abstract
Iron overload is a potentially life-threatening consequence of multiple red-blood-cell transfusions. Here, we review factors affecting excess iron distribution and its damage to specific tissues, as well as mechanisms of oncogenesis by iron. Although consequences of transfusional iron overload are best described in thalassemia major and related inherited anemias, they are increasingly recognized in acquired conditions, such as myelodysplastic syndromes (MDS). Iron overload in MDS not only impacts on certain tissues, but may affect the clonal evolution of MDS through generation of reactive oxygen species. Iron overload may also influence hematopoietic-stem-cell-transplantation outcomes. Novel MRI methods for assessing body iron have impacted significantly on outcome in inherited anemias by allowing monitoring of iron burden and iron chelation therapy. This approach is increasingly being used in MDS and stem-cell-transplant procedures. Knowledge gained from managing transfusional iron overload in inherited anemias may be translated to general oncology, with potential for improved patient outcomes.
Collapse
Affiliation(s)
- John B Porter
- Department of Haematology, University College London, UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK.
| | - Theo de Witte
- Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | | | | |
Collapse
|
27
|
Akı ŞZ, Suyanı E, Cengiz M, Özenirler S, Elbeğ Ş, Paşaoğlu H, Sucak GT. Association between Plasma Endothelin-1, Transforming Growth Factor-β, Fibroblast Growth Factor, and Nitric Oxide Levels and Liver Injury in Hematopoietic Stem Cell Transplantation Recipients with Persistent Iron Overload after Transplantation. Biol Blood Marrow Transplant 2015; 21:948-953. [PMID: 25681034 DOI: 10.1016/j.bbmt.2015.02.002] [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: 11/16/2014] [Accepted: 02/03/2015] [Indexed: 01/19/2023]
Abstract
Graft-versus-host disease, iron overload, and infections are the major causes of liver dysfunction in allogeneic hematopoietic stem cell transplantation (AHSCT) recipients. We investigated the relationship between serum iron parameters and the levels of transforming growth factor-β (TGF-β), fibroblast growth factor (FGF), endothelin-1 (ET-1), and nitric oxide (NO) as predictors of chronic liver injury in 54 AHSCT recipients who survived at least a year after transplantation. Serum samples from patients were obtained for the evaluation of ET-1, TGF-β, FGF, NO, and nontransferrin bound iron at the first year follow-up visit using commercially available ELISA kits. Patients were categorized depending on serum ferritin and transferrin saturation levels. The parameters were compared between the groups, and survival analysis was also performed. Most of the AHSCT recipients (81.5%) were in complete remission during the study. After a median follow-up time of 73 months (range, 13 to 109 months), 72.2% of the patients were alive. Mean serum levels of ET-1, NO, TGF-β, and FGF were 81.54 ± 21.62 μmol/mL, 31.82 ± 26.42 μmol/mL, 2.56 ± 0.77 ng/mL, and 50.31 ± 32.69 pg/mL, respectively. Nineteen patients (35.2% of the cohort) had serum ferritin levels higher than 1000 ng/mL. Mean serum levels of ET-1, NO, TGF-β, and FGF were similar in patients with serum ferritin levels below or above 1000 ng/mL (P > .05). Serum ferritin levels were positively correlated with serum alanine aminotransferase (r = .284, P = .042) and γ-glutamyl transferase (r = .271, P = .05) levels and were negatively correlated with serum albumin levels (r = .295, P = .034). There was a significant positive correlation between serum transferrin saturation and alanine aminotransferase levels (r = .305, P = .03). Serum ET-1 level was positively correlated with alkaline phosphatase levels (r = .304, P = .026). In univariate Cox regression analysis serum levels of iron parameters, ET-1, NO, TGF-β, and FGF did not have an impact on overall survival (P > .05). The probability of progression-free survival was also similar in patients with ferritin levels above or below 1000 ng/mL (P = .275). The probability of survival was similar in patients with transferrin saturation ≥70% and <70% (P > .05). Serum iron parameters showed a positive correlation with liver injury. However, there was no correlation between fibrogenic cytokines and liver transaminases. Our results suggest that iron overload at least with the current levels of ferritin might have a relatively benign course. Prospective randomized trials will guide the actual role of iron chelation in the post-transplantation setting.
Collapse
Affiliation(s)
- Şahika Zeynep Akı
- Faculty of Medicine, Department of Haematology, Gazi University, Ankara, Turkey
| | - Elif Suyanı
- Faculty of Medicine, Department of Haematology, Gazi University, Ankara, Turkey
| | - Mustafa Cengiz
- Department of Gastroenterology, Dr. A.Y. Ankara Oncology Training and Research Hospital, Ankara, Turkey.
| | - Seren Özenirler
- Department of Gastroenterology, Gazi University, Ankara, Turkey
| | - Şehri Elbeğ
- Department of Biochemistry, Gazi University, Ankara, Turkey
| | | | - Gülsan Türköz Sucak
- Faculty of Medicine, Department of Haematology, Gazi University, Ankara, Turkey
| |
Collapse
|
28
|
Non transferrin bound iron (NTBI) in acute leukemias throughout conventional intensive chemotherapy: kinetics of its appearance and potential predictive role in infectious complications. Leuk Res 2014; 39:88-91. [PMID: 25435028 DOI: 10.1016/j.leukres.2014.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 10/18/2014] [Accepted: 11/08/2014] [Indexed: 11/24/2022]
Abstract
We analyzed appearance of non transferrin bound iron (NTBI) in 30 transplant eligible patients with acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) during conventional chemotherapy treatment program and evaluated possible relationship with transfusional body iron intake, iron parameters and clinical complications. For each course, serum samples for NTBI detection were taken prior to chemotherapy, during treatment and during subsequent bone marrow myelosuppression: NTBI was assessed by HPLC. Appearance of NTBI was observed from the start of induction treatment and was still detectable during bone marrow myelosuppression; the recovery of the bone marrow function coincided with the disappearance of NTBI. This kinetic was observed in all subsequent high doses chemotherapy courses, independently from confounding variables such as transfusional iron intake and transferrin saturation. NTBI seems to be a consequence of chemotherapy induced lysis of bone marrow cells and, partly, of hepatocytes after cytotoxic injury. The subsequent persistence of NTBI throughout bone marrow myelosuppression is related to the transient suspension of erythropoietic activity. Moreover, NTBI levels >2μM at the beginning of iatrogenic myelosuppression were associated with higher risk of sepsis caused by Gram negative Bacilli (RR 2.571), also compared with other infectious complications (RR 1.954).
Collapse
|
29
|
Merkel DG, Nagler A. Toward resolving the unsettled role of iron chelation therapy in myelodysplastic syndromes. Expert Rev Anticancer Ther 2014; 14:817-29. [PMID: 24641787 DOI: 10.1586/14737140.2014.896208] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Transfusion dependent low risk myelodysplastic syndromes (MDS) patients, eventually develop iron overload. Iron toxicity, via oxidative stress, can damage cellular components and impact organ function. In thalassemia major patients, iron chelation therapy lowered iron levels with recovery of cardiac and liver functions and significant improvement in survival. Several noncontrolled studies show inferior survival in MDS patients with iron overload, including an increase in transplant-related mortality and infection risk while iron chelation appears to improve survival in both lower risk MDS patients and in stem cell transplant settings. Collated data are presented on the pathophysiological impact of iron overload; measuring techniques and chelating agents' therapy positive impact on hematological status and overall survival are discussed. Although suggested by retrospective analyses, the lack of clear prospective data of the beneficial effects of iron chelation on morbidity and survival, the role of iron chelation therapy in MDS patients remains controversial.
Collapse
Affiliation(s)
- Drorit G Merkel
- Division of Hematology, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | | |
Collapse
|
30
|
Pullarkat V. Iron toxicity in hematopoietic stem cell transplantation: Strike while the iron is labile. Acta Haematol 2013; 131:220-1. [PMID: 24335205 DOI: 10.1159/000355827] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 09/19/2013] [Indexed: 11/19/2022]
Affiliation(s)
- Vinod Pullarkat
- Jane Ann Nohl Division of Hematology, University of Southern California Keck School of Medicine, Los Angeles, Calif., USA
| |
Collapse
|
31
|
Tachibana T, Tanaka M, Numata A, Matsumoto K, Tomita N, Fujimaki K, Taguchi J, Sakai R, Fujita H, Fujisawa S, Maruta A, Ishigatsubo Y, Kanamori H. Clinical significance of pre- and 1-year post-transplant serum ferritin among adult transplant recipients. Leuk Lymphoma 2013; 55:1350-6. [PMID: 24024470 DOI: 10.3109/10428194.2013.842981] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
To clarify the significance of post-transplant serum ferritin (SF), we retrospectively assessed pre- and post-transplant SF. Among 256 patients undergoing allogeneic stem cell transplant (SCT) for hematologic malignancies between 2000 and 2011, those who had relapsed within 1 year were excluded, and 110 patients surviving for more than 1 year were included in the analysis. The cut-off value of SF was 1000 ng/mL, and four pre- and post-SF groups were defined: low-low (n = 62), low-high (n = 12), high-low (n = 13) and high-high (n = 23). Outcomes at 5 years for each group were as follows: overall survival (OS) 88.2, 38.1, 92.3 and 76.7%, respectively, p = 0.004, and non-relapse mortality (NRM) 11.3, 53.6, 7.7 and 18.9%, respectively, p = 0.037. Patients receiving larger transfusion volumes or developing chronic graft-versus-host disease (GVHD) demonstrated higher 1-year SF values. In multivariate analysis for OS and NRM, low-high SF remained a significant predictor of OS (hazard ratio [HR] = 3.49, 95% confidence interval [CI]: 1.10-11.0, p = 0.032) and NRM (HR = 2.95, 95%CI: 1.04-8.36, p = 0.041). These results suggest that the elevation of SF at 1 year after SCT, which may reflect transfusion and the development of chronic GVHD, may have an aggravating influence on outcomes after SCT. This study provides a clue to clarifying the clinical significance of SF in a transplant setting.
Collapse
|
32
|
Özcan MA, Ilhan O, Ozcebe OI, Nalcaci M, Gülbas Z. Review of therapeutic options and the management of patients with myelodysplastic syndromes. Expert Rev Hematol 2013; 6:165-89. [PMID: 23547866 DOI: 10.1586/ehm.13.7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Myelodysplastic syndromes (MDS) are a poorly understood group of disorders caused by one or more genetic aberrations in the bone marrow-derived cell line responsible for hematopoiesis. Recent advances in genetic medicine have offered new insights into the epigenesis as well as the prognosis of MDS, but have not resulted in new or improved curative treatment options. Bone marrow transplantation, introduced before the advent of genetic medicine, is still the only potential cure. Advances in other medical and pharmaceutical areas have broadened the scope of supportive care and disease-modifying therapies, and treating physicians now have a broad range of disease management options depending on a patient's likely prognosis. There is now clear evidence that appropriate supportive care and therapeutic intervention can improve progression-free and overall survival of MDS patients.
Collapse
Affiliation(s)
- Mehmet A Özcan
- Department of Hematology, Dokuz Eylül University, Izmir, Turkey.
| | | | | | | | | |
Collapse
|
33
|
Adams RLC, Bird RJ. Safety and efficacy of deferasirox in the management of transfusion-dependent patients with myelodysplastic syndrome and aplastic anaemia: a perspective review. Ther Adv Hematol 2013; 4:93-102. [PMID: 23610617 DOI: 10.1177/2040620712472355] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Deferasirox is an orally administered, once-daily iron chelator with a generally good safety and efficacy profile. Reported adverse events in the older myelodysplastic population are somewhat different to the more intensively investigated and younger thalassaemic population. Renal impairment is the most concerning adverse event, but this is reversible if identified and the drug is withdrawn early. Gastrointestinal effects, particularly diarrhoea, can be troublesome for older patients, but can be minimized with tailored therapy. Negative iron balance can be achieved in most patients with a median dose of 20 mg/kg/day, and doses up to 40 mg/kg are possible in patients with severe iron overload, who are at risk of cardiac decompensation.
Collapse
|
34
|
Eisfeld AK, Krahl R, Jaekel N, Niederwieser D, Al-Ali HK. Kinetics of iron removal by phlebotomy in patients with iron overload after allogeneic hematopoietic cell transplantation. AMERICAN JOURNAL OF BLOOD RESEARCH 2012; 2:243-253. [PMID: 23226624 PMCID: PMC3512175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 11/13/2012] [Indexed: 06/01/2023]
Abstract
UNLABELLED Excess body iron could persist for years after allogeneic hematopoietic cell transplantation (HCT) with possible deleterious sequels. An iron depletive therapy with phlebotomy seems rational. Kinetics of iron removal by phlebotomy without erythropoietin support in non-thalassemic adult patients with iron overload after HCT and the impact of pre- and post-HCT hemochromatosis (HFE) genotype on iron mobilization were investigated. PATIENTS AND METHODS Phlebotomy was initiated in 61 recipients of allografts due to hematologic malignancies (median age 48 years) after a median of 18 months. The prephlebotomy median serum ferritin (SF) was 1697ng/ml and the median number of blood transfusions 28 units. Alanine aminotransferase (ALT)/aspartate aminotransferase (AST), alkaline phosphates (AP), and bilirubin were elevated in 55.7%, 64% and 11.5% patients respectively. HFE-genotype was elucidated by polymerase chain reaction using hybridization probes and melting curve analysis. RESULTS Phlebotomy was well-tolerated irrespective of age or conditioning. A negative iron balance in 80% of patients (median SF 1086 ng/ml) and a rise in hemoglobin were observed (p<0.0001). Higher transfusional burden and SF were associated with a greater iron mobilization per session (p=0.02). In 58% of patients, a plateau after an initial steady decline in SF was followed by a second decline under further phlebotomy. The improvement in ALT (p=0.002), AST (p=0.03), AP (p=0.01), and bilirubin (p<0.0001) did not correlate with the decline in SF. Mutant HFE-gene variants were detected in 14/55 (25%) pre-HCT and 22/55 (40%) patients post-HCT. Overall, dissimilar pre- and posttransplantational HFE-genotypes were detected in 20/55 (40%) patients. Posttransplantational mutant HFE variants correlated with a slower decline in SF (p=0.007). CONCLUSIONS Phlebotomy is a convenient therapy of iron overload in survivors of HCT. A negative iron balance and a rise in hemoglobin were observed in the majority of patients. Liver dysfunction improved irrespective of SF reduction suggesting a probable rapid decline of the deleterious labile plasma iron. In recipients of grafts with mutant HFE variants a "mixed chimerism" of HFE in body tissues might be created with a change in the set point for iron regulation. The transient plateau in SF after an initial decline might reflect iron mobilization from various tissues.
Collapse
|