Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Bone Marrow Transplant ; 54(7): 1121-1123, 2019 07.
Article En | MEDLINE | ID: mdl-30518979

To candidate children as bone marrow donors raises two main concerns: donor safety and adequate marrow cell dose. Data in the field are limited and guidelines for child donor care management are lacking. In this context, we herein report the experience collected in our center by comparing very-young donors (defined as age ≤ 3 years) with young donors (defined as age > 3 years) who donated bone marrow (BM) for patients affected by beta-globin disorders.


Bone Marrow Transplantation/methods , Tissue and Organ Harvesting/methods , Child, Preschool , Female , Humans , Male , Tissue Donors
2.
Blood Adv ; 2(3): 263-270, 2018 02 13.
Article En | MEDLINE | ID: mdl-29431621

We examined outcomes of haploidentical hematopoietic cell transplantation (haplo-HCT) using T-cell receptor αß+ (TCRαß+)/CD19+-depleted grafts (TCR group, 14 patients) in children with hemoglobinopathies. Patients received a preparative regimen consisting of busulfan, thiotepa, cyclophosphamide, and antithymocyte globulin preceded by fludarabine, hydroxyurea, and azathioprine. The median follow-up among surviving patients was 3.9 years. The 5-year probabilities of overall survival (OS) and disease-free survival (DFS) were 84% and 69%, respectively. The incidence of graft failure was 14%. We compared outcomes to a historical group of 40 patients with hemoglobinopathies who received CD34+-selected grafts (CD34 group). The median follow-up of surviving patients for the CD34 group was 7.5 years. The 5-year probabilities of OS and DFS were 78% and 39%, respectively. The CD34 group had a significantly higher incidence of graft failure (45%) than the TCR group (14%) (P = .048). The incidences of grades 2 to 4 acute graft-versus-host disease (GVHD) in the TCR and CD34 groups were 28% and 29%, respectively, and 21% and 10% (P = .1), respectively, for extensive chronic GVHD. Viral reactivation was common in both groups. The overall incidence of posttransplant lymphoproliferative disorders for the entire group was 16%. Among all patients, 5 developed autoimmune hemolytic anemia or thrombocytopenia, with the overall cumulative incidence of 11%. The 2 groups showed suboptimal CD4+ recovery within the first 6 months of transplantation with no significant difference between groups. These data demonstrate that TCRαß+/CD19+-depleted grafts are associated with a reduced incidence of graft failure, but delayed immune reconstitution and associated morbidity and mortality remain a significant challenge.


Graft Survival/immunology , Hematopoietic Stem Cell Transplantation/methods , Hemoglobinopathies/therapy , Lymphocyte Depletion/methods , Transplantation, Haploidentical/methods , Adolescent , Antigens, CD19 , Antigens, CD34 , Child , Child, Preschool , Graft Rejection , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Hemoglobinopathies/complications , Hemoglobinopathies/mortality , Humans , Male , Receptors, Antigen, T-Cell, alpha-beta , Retrospective Studies , Survival Analysis , Transplantation, Haploidentical/adverse effects , Treatment Outcome
3.
Biol Blood Marrow Transplant ; 23(9): 1531-1540, 2017 Sep.
Article En | MEDLINE | ID: mdl-28602890

Posterior reversible encephalopathy syndrome (PRES) is a serious adverse event associated with calcineurin inhibitors used for graft-versus-host disease (GVHD) prophylaxis. We compared the incidence of PRES in children with thalassemia (n = 222, 1.4 to 17.8 years old) versus sickle cell disease (SCD; n = 59, 2 to 17 years old) who underwent hematopoietic cell transplantation from HLA-matched siblings or alternative donors and analyzed the risk factors for PRES. Overall, 31 children developed calcineurin inhibitor-related PRES (11%), including 30 patients with seizures and 1 patient without seizures. PRES incidence was significantly higher in SCD patients (22%; 95% confidence interval [CI], 10% to 32%) than in thalassemia patients (8%; 95% CI, 5% to 12%;P = .002). In multivariate analysis, factors associated with PRES were hypertension (hazard ratio [HR], 5.87; 95% CI, 2.57 to 13.43; P = .0001), SCD (HR, 2.49; 95% CI, 1.25 to 4.99; P = .009), and acute GVHD (HR 2.27; 95% CI, 1.06 to 4.85; P= .031). In the entire cohort overall survival (OS) was significantly higher in patients without versus with PRES (90% versus 77%; P = .02). In a subgroup analysis that including matched sibling transplants, OS and disease-free survival (DFS) were similar in thalassemia patients without PRES (92% and 88%, respectively) and with PRES (82% and 73%, respectively), whereas SCD patients with PRES had significantly lower OS (67%) and DFS (67%) than patients without PRES (94% and 94%, respectively; P = .008). Thus, SCD patients had a significantly higher incidence of PRES than thalassemia patients, and hypertension and GVHD were the 2 main risk factors for PRES in patients with hemoglobinopathies. Although PRES did not significantly influence survival in patients with thalassemia, patients with SCD had significantly lower survival after PRES.


Anemia, Sickle Cell/therapy , Calcineurin Inhibitors/adverse effects , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/adverse effects , Posterior Leukoencephalopathy Syndrome/therapy , Seizures/therapy , beta-Thalassemia/therapy , Acute Disease , Adolescent , Anemia, Sickle Cell/immunology , Anemia, Sickle Cell/mortality , Anemia, Sickle Cell/pathology , Calcineurin Inhibitors/administration & dosage , Child , Child, Preschool , Cohort Studies , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Graft vs Host Disease/pathology , Graft vs Host Disease/prevention & control , Humans , Hypertension/diagnosis , Hypertension/physiopathology , Immunosuppressive Agents/administration & dosage , Infant , Male , Posterior Leukoencephalopathy Syndrome/chemically induced , Posterior Leukoencephalopathy Syndrome/immunology , Posterior Leukoencephalopathy Syndrome/mortality , Risk Factors , Seizures/chemically induced , Seizures/immunology , Seizures/mortality , Siblings , Survival Analysis , Transplantation, Homologous , Unrelated Donors , beta-Thalassemia/immunology , beta-Thalassemia/mortality , beta-Thalassemia/pathology
5.
Mol Diagn Ther ; 20(2): 161-73, 2016 Apr.
Article En | MEDLINE | ID: mdl-26897028

INTRODUCTION: Several DNA polymorphisms have been associated with high production of fetal hemoglobin (HbF), although the molecular basis is not completely understood. In order to identify and characterize novel HbF-associated elements, we focused on five probands and their four families (from Egypt, Iraq and Iran) with thalassemia major (either ß(0)-IVSII-1 or ß(0)-IVSI-1) and unusual HbF elevation (>98 %), congenital or acquired after rejection of bone marrow transplantation, suggesting an anticipated favorable genetic background to high HbF expression. METHODS: Patient recruitment, genomic DNA sequencing, western blotting, electrophoretic mobility shift assays, surface plasmon resonance (SPR) biospecific interaction analysis, bioinformatics analyses based on docking experiments. RESULTS: A polymorphism of the Aγ-globin gene is here studied in four families with ß(0)-thalassemia (ß(0)-IVSII-1 and ß(0)-IVSI-1) and expressing unusual high HbF levels, congenital or acquired after rejection of bone marrow transplantation. This (G→A) polymorphism is present at position +25 of the Aγ-globin genes, corresponding to a 5'-UTR region of the Aγ-globin mRNA and, when present, is physically linked in chromosomes 11 of all the familiar members studied to the XmnI polymorphism and to the ß(0)-thalassemia mutations. The region corresponding to the +25(G→A) polymorphism of the Aγ-globin gene belongs to a sequence recognized by DNA-binding protein complexes, including LYAR (Ly-1 antibody reactive clone), a zinc-finger transcription factor previously proposed to be involved in down-regulation of the expression of γ-globin genes in erythroid cells. CONCLUSION: We found a novel polymorphism of the Aγ-globin gene in four families with ß(0)-thalassemia and high levels of HbF expression. Additionally, we report evidence suggesting that the Aγ-globin gene +25(G→A) polymorphism decreases the efficiency of the interaction between this sequence and specific DNA binding protein complexes.


Fetal Hemoglobin/metabolism , Polymorphism, Single Nucleotide , beta-Thalassemia/genetics , gamma-Globins/genetics , Adolescent , Child , Child, Preschool , Chromosomes, Human, Pair 11/genetics , DNA-Binding Proteins/metabolism , Female , Fetal Hemoglobin/genetics , Humans , K562 Cells , Male , Pedigree , beta-Thalassemia/metabolism , gamma-Globins/chemistry , gamma-Globins/metabolism
6.
Transplantation ; 100(4): 925-32, 2016 Apr.
Article En | MEDLINE | ID: mdl-26457600

BACKGROUND: Bone marrow transplantation (BMT) for class 3 patients with thalassemia is challenging due to high rates of graft rejection and transplant-related mortality. Since the first studies of BMT in the late 1980s, a number of conditioning regimens have been designed to improve outcomes, but with suboptimal results. Here we report the outcome of transplantation in class 3 patients using a modified protocol. METHODS: Sixty-three patients between 5 and 16.7 years of age with class 3 thalassemia received HLA-matched sibling BMT following either the original protocol (26 patients) or the modified protocol (37 patients). Both regimens comprised preconditioning cytoreduction with hydroxyurea and azathioprine starting at -45 days pretransplant, and fludarabine from days -16 to -12. Conditioning was performed with busulfan and cyclophosphamide (original protocol) or with busulfan, thiotepa, and cyclophosphamide (modified protocol). RESULTS: The 2 groups showed similar patient demographics. At day 0, the degree of cytoreduction (lymphopenia, neuthropenia, and thrombocytopenia) achieved by the modified protocol was greater than the original protocol. The incidence of graft failure/rejection was significantly higher in the original group (15%; 95% confidence interval [95% CI], 5-32%) compared with the modified group (0%) (P = 0.014). The respective 5-year thalassemia-free survival rates were 73% (95% CI, 51-86%) and 92% (95% CI, 77-97%) (P = 0.047). Both groups showed similar incidences of grades II to IV acute graft-versus host disease. Modified protocol did not increase nonhematological toxicity or infectious complications. CONCLUSIONS: The modified treatment protocol effectively and safely prevented graft failure/rejection and significantly increased thalassemia-free survival of class 3 patients with thalassemia.


Bone Marrow Transplantation/methods , HLA Antigens/immunology , Histocompatibility , Living Donors , Siblings , Thalassemia/surgery , Adolescent , Age Factors , Bone Marrow Transplantation/adverse effects , Child , Child, Preschool , Disease-Free Survival , Drug Therapy, Combination , Female , Graft Rejection/epidemiology , Graft Rejection/immunology , Graft Survival , Graft vs Host Disease/epidemiology , Graft vs Host Disease/immunology , Histocompatibility Testing , Humans , Immunosuppressive Agents/administration & dosage , Incidence , Kaplan-Meier Estimate , Male , Predictive Value of Tests , Prospective Studies , Risk Factors , Rome/epidemiology , Thalassemia/diagnosis , Thalassemia/genetics , Thalassemia/immunology , Time Factors , Transplantation Conditioning , Treatment Outcome
7.
Niger Med J ; 56(3): 175-9, 2015.
Article En | MEDLINE | ID: mdl-26229224

BACKGROUND: Sickle cell anaemia (SCA) remains associated with high risks of morbidity and early death. Children with SCA are at high risk for ischaemic stroke and transient ischaemic attacks, secondary to intracranial arteriopathy involving carotid and cerebral arteries. Allogeneic haematopoietic stem cell transplantation (HSCT) is the only curative treatment for SCA. We report our experience with transplantation in a group of patients with the Black African variant of SCA. PATIENTS AND METHODS: This study included 31 consecutive SCA patients who underwent bone marrow transplantation from human leukocyte antigen (HLA)-identical sibling donors between 2010 and 2014 following a myeloablative-conditioning regimen. RESULTS: The median patient age was 10 years (range 2-17 years). Before transplantation, 14 patients had recurrent, painful, vaso-occlusive crisis; ten patients had recurrent painful crisis in association with acute chest syndrome; three patients experienced ischaemic stroke and recurrent vaso-occlusive crisis; two patients experienced ischaemic stroke; one patient exhibited leukocytosis; and one patient exhibited priapism. Of the 31 patients, 28 survived without sickle cell disease, with Lansky/Karnofsky scores of 100. All surviving patients remained free of any SCA-related events after transplantation. CONCLUSION: The protocols used for the preparation to the transplant in thalassaemia are very effective also in the other severe haemoglobinopathy as in the sickle cell anaemia with 90% disease free survival. Today, if a SCA patient has a HLA identical family member, the cellular gene therapy through the transplantation of the allogeneic haemopoietic cell should be performed. Tomorrow, hopefully, the autologous genetically corrected stem cell will break down the wall of the immunological incompatibility.

8.
Mediterr J Hematol Infect Dis ; 7(1): e2015006, 2015.
Article En | MEDLINE | ID: mdl-25574365

Sickle Cell Anaemia (SCA) is the most common inherited blood disorder and is associated with severe morbidity and decreased survival. Allogeneic Haematopoietic Stem Cell Transplantation (HSCT) is the only curative approach. Nevertheless the decision to perform a bone marrow transplant includes the risk of major complications and transplant-related mortality. Infections represent the leading cause of death in SCA patients undergoing HSCT. Invasive Pulmonary Aspergillosis (IPA) is a devastating opportunistic infection and remains a significant cause of morbidity and mortality in HSCT recipients. Data regarding IPA in the setting of SCA are lacking. In the present report, we describe a patient with SCA, who developed IPA after allogeneic bone marrow transplant. The fungal infection was treated by systemic antifungal therapy in addition to surgery, despite mild chronic graft versus host disease (GVHD) and continuing immunosuppressive therapy. This case shows that IPA occurring in bone marrow recipients with SCA can be successfully treated.

9.
Pediatr Blood Cancer ; 62(4): 680-6, 2015 Apr.
Article En | MEDLINE | ID: mdl-25557687

BACKGROUND: Busulfan (Bu) is an integral part of conditioning regimens for patients with sickle cell anemia (SCA) undergoing transplantation. Patients with SCA might predispose to transplant-related neurological and pulmonary toxicities due to pre-existing disease-related cerebrovascular and lung injury. Bu therapy appears to be an important contributing factor in this context. PROCEDURE: We studied the pharmacokinetics of intravenous Bu and clinical outcomes of 36 children with SCA undergoing bone marrow transplantation. Most patients had pre-existing organ system damage. Busulfan was administered every 6 hr for 4 days with pharmacokinetic-guided dose adjustment to target a conservative area under the concentration versus time curve (AUC) range of 900-1,350 µMol*min. RESULTS: We found that the first-dose Bu clearance was significantly higher (P < 0.0005) than the subsequent daily clearance, which remained unchanged during the following days. After the first-dose, 69% of patients achieved the target range. We adapted a new dose-adjustment strategy targeting exposures to the lower end (900 µMol*min) of the AUC range after the first dose of Bu to avoid unnecessary dose increases on subsequent days due to differences in clearance. This strategy enabled most patients to maintain the AUC within therapeutic range following dose adjustments. CONCLUSIONS: Differences in Bu clearance after the first-dose and subsequent daily doses in patients with SCA should be considered for pharmacokinetic-guided dose adjustment. Conservative AUC range and targeting exposures to the lower end of the range after the first dose was associated with negligible toxicity, and high engraftment and sickle cell-free survival rates.


Anemia, Sickle Cell/therapy , Bone Marrow Transplantation , Busulfan/administration & dosage , Busulfan/pharmacokinetics , Myeloablative Agonists/administration & dosage , Myeloablative Agonists/pharmacokinetics , Transplantation Conditioning/methods , Adolescent , Allografts , Anemia, Sickle Cell/mortality , Busulfan/adverse effects , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Infant , Infusions, Intravenous , Male , Myeloablative Agonists/adverse effects , Prospective Studies , Survival Rate , Time Factors
10.
Mediterr J Hematol Infect Dis ; 6(1): e2014066, 2014.
Article En | MEDLINE | ID: mdl-25408852

Allogeneic cellular gene therapy through hematopoietic stem cell transplantation is the only radical cure for congenital hemoglobinopathies like thalassemia and sickle cell anemia. Persistent mixed hematopoietic chimerism (PMC) has been described in thalassemia and sickle cell anemia. Here, we describe the clinical course of a 6-year-old girl who had received bone marrow transplant for sickle cell anemia. After the transplant, the patient showed 36% donor hematopoietic stem cells in the bone marrow, whereas in the peripheral blood there was evidence of 80% circulating donor red blood cells (RBC). The analysis of apoptosis at the Bone Marrow level suggests that Fas might contribute to the cell death of host erythroid precursors. The increase in NK cells and the regulatory T cell population observed in this patient suggests that these cells might contribute to the condition of mixed chimerism.

11.
Mediterr J Hematol Infect Dis ; 6(1): e2014054, 2014.
Article En | MEDLINE | ID: mdl-25045462

BACKGROUND AND PURPOSE: Allogeneic hematopoietic stem cell transplantation (HSCT) is the only curative treatment for sickle cell anemia (SCA). We report our experience with transplantation in children with the Black African variant of SCA and the effects of transplant on erythroid compartment in bone marrow (BM). PATIENTS AND METHODS: Twenty-seven consecutive patients who underwent BM transplantation from HLA-identical donors following a myeloablative conditioning regimen were included. Using both CD71 and FSC parameters, we obtained three erythroid populations: EryA-C. Ery A (CD71(high) FSC(high)) are basophilic; Ery B (CD71(high) FSC(low)) are late basophilic and polychromatic; and Ery C (CD71(low) FSC(low)) are orthochromatic erythroblasts and reticulocytes. To analyze the effect of transplantation on intramedullary apoptosis, we studied Fas (CD95+) and caspase-3 expression in erythroblast subpopulations. RESULTS: All patients experienced sustained engraftment, and all surviving patients remained free of SCA-related events after transplantation. The erythroid population showed expansion in the BM at baseline. After transplant, levels decreased, especially of Ery C, in parallel to reduced Fas expression and an initial caspase 3 increase in erythroid population, similar to reported later steps of "normal" erythroid maturation. CONCLUSIONS: The results suggest a good chance of cure for children with SCA, with an excellent survival rate. We also observed "normalization" of erythroid populations in parallel with a decreased intramedullary apoptosis rate, suggesting normal erythroid maturation in ex-SCA patients after HSCT.

12.
Blood ; 122(15): 2751-6, 2013 Oct 10.
Article En | MEDLINE | ID: mdl-23963044

Bone marrow transplantation (BMT) performance can be limited by a lack of ideal donors, and the role of alternative donor hematopoietic cell transplantation in thalassemia is not well established. Here we used a new treatment protocol (Pc 26.1) in 16 thalassemia patients to perform BMT using phenotypically HLA-identical or 1-antigen-mismatched relatives (related donors [RDs]). We compared these results with HLA-matched sibling (matched sibling donors [MSDs]) BMT in 66 patients. The entire RD group and 88% of MSD group had sustained engraftment. Rejection incidence was 0% in the RD and 12% (95% confidence interval [95% CI], 6%-21%) in MSD groups (P = .15), with respective thalassemia-free survival probabilities of 94% (95% CI, 63%-99%) and 82% (95% CI, 70%-89%) (P = .24). Transplant-related mortality was 6% (95% CI, 1%-26%) in the RD group and 8% (95% CI, 3%-16%) in the MSD group (P = .83). The intensified new protocol was not associated with increased nonhematologic toxicity. The present data show that the Pc 26.1 preparative regimen allows thalassemia patients to safely undergo BMT from RDs who are not HLA-matched siblings, with transplant outcomes similar to patients with MSD grafts.


Bone Marrow Transplantation/methods , Histocompatibility Testing , Histocompatibility , Thalassemia/therapy , Adolescent , Amniotic Fluid , Bone Marrow Transplantation/mortality , Child , Child, Preschool , Family , Female , Graft Rejection/mortality , Graft vs Host Disease/mortality , Humans , Infant , Male , Prospective Studies , Survival Rate , Thalassemia/mortality , Treatment Outcome , Young Adult
13.
Biol Blood Marrow Transplant ; 16(11): 1557-66, 2010 Nov.
Article En | MEDLINE | ID: mdl-20546907

To analyze immunohematologic reconstitution, particularly of natural killer (NK) cells, we evaluated 13 ß-thalassemia patients after 20 and 60 days and 1 year posttransplantation with T cell-depleted HLA-haploidentical stem cells. We assessed lymphocyte and bone marrow (BM) progenitor cell phenotype and differentiation capacity, spontaneous BM cytokine production, stromal cells, and stromal cell interleukin (IL)-7 production. A reduced clonogenic capability manifested at day +20. Patients had significantly lower CD4(+) T cells versus controls, mainly in the CD45RA(+)CD62L(+) subset. NKs were among the first lymphocytes to repopulate the peripheral blood. At day +60, an increase in primitive BM progenitor cells paralleled small increases in CD4(+), naïve CD4(+), and thymic naïve Th cells. A significant increase in CD4(+) and CD8(+) markers paralleled an increase in CD3⁻CD16(+) NKs, especially with full engraftment. In patients with stable mixed chimerism we observed very low levels of CD3(+) donor chimerism early after transplant that increased over time, but a stable population of high donor NK cells, suggesting a role of these cells on donor engraftment. Stromal cells secreted less IL-7 and displayed "macrophage-like" morphology. Patients initially manifested impaired stem/progenitor cell growth and differentiation capacity in parallel with altered T cell homeostasis and a reduced T cell naïve compartment. We hypothesize that T cell compartment damage partly arises from altered new T cell production from the hematopoietic stem/progenitor cells under stromal cytokine influence. NNK subset analysis might be useful for determining transplant outcome.


Hematopoietic Stem Cell Transplantation/methods , Histocompatibility, Maternal-Fetal , Lymphocyte Depletion , Lymphocytes/cytology , T-Lymphocytes/cytology , beta-Thalassemia/therapy , B-Lymphocytes/cytology , Blood Cells/cytology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Count , Child , Child, Preschool , Chimera/blood , Colony-Forming Units Assay , Graft Rejection/immunology , Graft Survival/immunology , HLA Antigens/genetics , HLA Antigens/immunology , Humans , Interleukin-2/metabolism , Interleukin-7/metabolism , Killer Cells, Natural/cytology , Living Donors , Lymphocyte Count , Mothers , Stromal Cells/cytology , Stromal Cells/metabolism , T-Lymphocyte Subsets/cytology , Transplants , Treatment Outcome , Tumor Necrosis Factor-alpha/metabolism
14.
Blood ; 115(6): 1296-302, 2010 Feb 11.
Article En | MEDLINE | ID: mdl-19897573

Fetomaternal microchimerism suggests immunological tolerance between mother and fetus. Thus, we performed primary hematopoietic stem cell transplantation from a mismatched mother to thalassemic patient without an human leukocyte antigen-identical donor. Twenty-two patients with thalassemia major were conditioned with 60 mg/kg hydroxyurea and 3 mg/kg azathioprine from day -59 to -11; 30 mg/m(2) fludarabine from day -17 to -11; 14 mg/kg busulfan starting on day -10; and 200 mg/kg cyclophosphamide, 10 mg/kg thiotepa, and 12.5 mg/kg antithymocyte globulin daily from day -5 to -2. Fourteen patients received CD34(+)-mobilized peripheral blood and bone marrow progenitor cells; 8 patients received marrow graft-selected peripheral blood stem cells CD34(+) and bone marrow CD3/CD19-depleted cells. T-cell dose was adjusted to 2 x 10(5)/kg by fresh marrow cell addback at the time of transplantation. Both groups received cyclosporine for graft-versus-host disease prophylaxis for 2 months after transplantation. Two patients died (cerebral Epstein-Barr virus lymphoma or cytomegalovirus pneumonia), 6 patients reject their grafts, and 14 showed full chimerism with functioning grafts at a median follow-up of 40 months. None of the 14 patients who showed full chimerism developed acute or chronic graft-versus-host disease. These results suggest that maternal haploidentical hematopoietic stem cell transplantation is feasible in patients with thalassemia who lack a matched related donor.


Antigens, CD34/metabolism , Bone Marrow Transplantation , Graft vs Host Disease/prevention & control , Lymphocyte Depletion , Peripheral Blood Stem Cell Transplantation , T-Lymphocytes , Thalassemia/therapy , Adolescent , Adult , Child , Child, Preschool , Feasibility Studies , Flow Cytometry , Graft Survival/immunology , HLA Antigens/metabolism , Humans , In Situ Hybridization, Fluorescence , Middle Aged , Mothers , Pilot Projects , Polymerase Chain Reaction , Prospective Studies , Transplantation Conditioning , Transplantation, Homologous , Young Adult
15.
Biol Blood Marrow Transplant ; 16(5): 662-71, 2010 May.
Article En | MEDLINE | ID: mdl-20026413

Little is known about late-onset hemorrhagic cystitis (HC) in children, its relationship to BK virus, and treatment with cidofovir (CDV) following hematopoietic stem cell transplantation (HSCT). We prospectively investigated BK virus reactivation in children who underwent HSCT from a matched related donor for thalassemia or sickle cell anemia following busulfan-cyclophosphamide-based conditioning regimens and analyzed risk factors for development of HC and its treatment with CDV. Grade 2-4 HC occurred in 30 patients with a cumulative incidence of 26% (95% confidence interval [CI] = 18%-34%). The cumulative incidences of BK viruria and viremia were 81% (95% CI = 69%-89%) and 28% (95% CI = 18%-40%), respectively. Multivariate analysis revealed that use of antithymocyte globulin (ATG) (hazard ratio [HR] = 10.5; P = .001), peak BK viruria >100,000 copies/mL (HR = 6.2; P = .004), and grade II-IV acute graft-versus-host disease (HR = 5.3; P = .007) were predictive factors for HC. Nineteen patients with HC were given CDV at 1.5 mg/kg/day 3 times a week, or 5 mg/kg/week. The median duration of therapy was 27 days (range, 21-180 days), and a median of 9 doses were given (range, 6-22). All patients had a complete clinical response (CCR), and 69% had a microbiological response at 4 weeks. Eleven patients with BK virus-related HC receiving supportive care also had CCR. The median duration of HC in these patients was similar to that in patients treated with CDV. None of the patients with HC cleared BK viruria when CCR was achieved. We conclude that late-onset HC is more prevalent in children with sustained high BK viruria who are treated with ATG or who develop graft-versus-host disease. Randomized clinical trials are urgently needed to better define the role of CDV in treating BK virus-related HC.


Anemia, Sickle Cell/complications , Cystitis/etiology , Cytosine/analogs & derivatives , Hematopoietic Stem Cell Transplantation/adverse effects , Organophosphonates/therapeutic use , Polyomavirus Infections/drug therapy , Thalassemia/complications , Adolescent , Adult , Anemia, Sickle Cell/therapy , BK Virus , Child , Child, Preschool , Cidofovir , Cystitis/drug therapy , Cystitis/virology , Cytosine/therapeutic use , Hemorrhage , Humans , Incidence , Infant , Middle Aged , Polyomavirus Infections/etiology , Prospective Studies , Thalassemia/therapy , Tumor Virus Infections , Young Adult
16.
Am J Hematol ; 84(6): 372-3, 2009 Jun.
Article En | MEDLINE | ID: mdl-19373892

An adult patient affected by beta(0)-thalassemia major underwent allogeneic bone marrow transplant (BMT) from a matched related donor. Forty days after transplant, allogeneic engraftment failure and autologous beta(0)-thalassemic bone marrow recovery were documented. Red blood cell transfusions were required until 118 days post-transplant. Thereafter, the haemoglobin (Hb) levels stabilized over 11.8 gr/dl throughout the ongoing 34-month follow-up, abolishing the need for transfusion support. The Hb electrophoresis showed 100% Hb Fetal (HbF). This unexplained case suggests full HbF production may occur in an adult patient with beta(0)-thalassemia major.


Bone Marrow Transplantation , Fetal Hemoglobin/biosynthesis , Graft Rejection/blood , beta-Thalassemia/blood , Adolescent , Humans , beta-Thalassemia/genetics , beta-Thalassemia/surgery
17.
Haematologica ; 90(6): 785-92, 2005 Jun.
Article En | MEDLINE | ID: mdl-15951291

BACKGROUND AND OBJECTIVES: Natural killer (NK) cells constitute an important area of research for hematologic malignancies. The anti-leukemic activity of NK cells against acute myeloid leukemia (AML) blasts has been described, but very few data are available for acute lymphoid leukemia (ALL). The present study was designed to investigate whether: (i) NK effectors could be expanded from adult and pediatric ALL patients in complete remission; (ii) the signal transduction machinery of these cells was preserved; (iii) NK cells showed cytotoxic activity against autologous blasts; (iv) interleukin (IL)-2, IL-12 and IL-15 were able to increase lytic activity in our in vitro model; (v) any differences in cytotoxic activity could be found between expanded effectors from adult and pediatric patients. DESIGN AND METHODS: We co-cultured patients' peripheral blood mononuclear cells (PBMC) with the feeder cell line RPMI 8866 and analyzed the NK cells' expansion capacity by cell count and cytofluorimetric analyses. Signal transduction of expanded effector cells was evaluated by Western blot. 51Cr release assays, before and after stimulation with activating cytokines, were performed to analyze the cytotoxic potential of effector cells against tumor cell lines and autologous blast cells. Data were analyzed with t-tests for paired data. RESULTS: We obtained an average 40-fold increase in NK cells. Signal transduction through the CD16 receptor was preserved. Patients' expanded cells showed cytotoxic activity against target cell lines comparable to that of normal donors. More significantly, these cells also exerted a lytic effect against autologous blasts. In addition, incubating these effectors for 24 hours with IL-2 + IL-15 significantly increased this cytotoxic function. No differences in expansion and cytotoxic activity were found between pediatric and adult patients. INTERPRETATION AND CONCLUSIONS: These findings document for the first time the possibility of expanding ex vivo cytotoxic effectors with autologous killing capacity from ALL patients in remission, and suggest a new potential immunotherapeutic strategy for the management of early disease recurrence or of residual disease.


Immunotherapy/methods , Killer Cells, Natural/cytology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adult , Child , Coculture Techniques , Cytokines/metabolism , Flow Cytometry , Humans , Interleukin-15/metabolism , Interleukin-2/metabolism , Killer Cells, Natural/metabolism , Receptors, IgG/metabolism , Remission Induction , Signal Transduction
...