Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Haematologica ; 98(4): 555-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23242593

ABSTRACT

Several studies have established an association between iron chelation therapy with deferasirox and hematopoietic improvement in patients with myelodysplastic syndromes. There are no data from patients with ß-thalassemia major. In a cross-sectional study, we evaluated the absolute number of several hematopoietic peripheral progenitors (colony-forming unit-granulocyte/macrophage, erythroid burst-forming units, colony-forming unit-granulocyte/erythrocyte/macrophage/megakaryocyte, and long-term culture-initiating cells) in 30 patients with ß-thalassemia major (median age 29.5 years, 40% males) and 12 age-matched controls. For the ß-thalassemia major patients, data on splenectomy status, the type of iron chelator used, and serum ferritin levels reflecting changes in iron status on the chelator were also retrieved. All patients had to be using the same iron chelator for at least 6 months with >80% compliance. The absolute number of all hematopoietic peripheral progenitors was higher in ß-thalassemia major patients than in controls, and varied between splenectomized and non-splenectomized patients (lower number of erythroid burst-forming units and higher numbers of colony-forming unit-granulocyte/macrophage, colony-forming unit-granulocyte/erythrocyte/macrophage/megakaryocyte, and long-term culture-initiating cells). The number of erythroid burst-forming units was significantly higher in patients taking deferasirox (n=10) than in those taking either deferoxamine (n=10) or deferiprone (n=10) (P<0.05). After adjusting for age, sex, splenectomy status, and serum ferritin changes, the association between a higher absolute number of erythroid burst-forming units in deferasirox-treated patients than in patients taking deferoxamine or deferiprone remained statistically significant (P=0.011). In conclusion, in ß-thalassemia major patients, compared with other iron chelators, deferasirox therapy is associated with higher levels of circulating erythroid burst-forming units. This variation is independent of iron status changes and is more likely to be due to the type of chelator.


Subject(s)
Chelation Therapy/methods , Hematopoietic Stem Cells/drug effects , Iron Chelating Agents/therapeutic use , beta-Thalassemia/drug therapy , Adult , Benzoates/therapeutic use , Blood Cell Count , Colony-Forming Units Assay , Cross-Sectional Studies , Deferasirox , Deferiprone , Deferoxamine/therapeutic use , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Female , Ferritins/blood , Flow Cytometry , Hematopoietic Stem Cells/cytology , Humans , Linear Models , Male , Multivariate Analysis , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/drug effects , Pyridones/therapeutic use , Splenectomy , Triazoles/therapeutic use , Young Adult , beta-Thalassemia/blood
2.
Blood ; 114(14): 3127-30, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19628707

ABSTRACT

Two putative types of circulating endothelial progenitor cells have been recently identified in vitro: (1) endothelial colony-forming cell (ECFC) and (2) colony-forming unit-endothelial cell (CFU-EC). Only the former is now recognized to belong to endothelial lineage. We have used the ECFC and CFU-EC assays to readdress the issue of the clonal relation between endothelial progenitor cells and hematopoietic stem cells in patients with Philadelphia-positive and Philadelphia-negative chronic myeloproliferative disorders. Both ECFCs and CFU-ECs were cultured from peripheral blood mononuclear cells, and either BCR-ABL rearrangement or JAK2-V617F mutation were assessed in both types of endothelial colonies. We found that ECFCs lack the disease-specific markers, which are otherwise present in CFU-ECs, thus reinforcing the concept that the latter belongs to the hematopoietic lineage, and showing that in chronic myeloproliferative disorders the cell that gives rise to circulating ECFC has a distinct origin from the cell of the hematopoietic malignant clone.


Subject(s)
Biomarkers, Tumor/genetics , Endothelial Cells/pathology , Fusion Proteins, bcr-abl/deficiency , Hematopoietic Stem Cells/pathology , Janus Kinase 2/deficiency , Myeloproliferative Disorders/genetics , Stem Cells/pathology , Adult , Aged , Cells, Cultured , Chronic Disease , Colony-Forming Units Assay , Female , Fusion Proteins, bcr-abl/genetics , Gene Rearrangement , Humans , Janus Kinase 2/genetics , Male , Middle Aged , Mutation/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology
3.
Lancet Oncol ; 9(9): 831-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18693069

ABSTRACT

BACKGROUND: Cord-blood transplants are associated with delayed or failed engraftment in about 20% of adult patients. The aim of this phase I/II study was to establish the safety and efficacy of a new administration route (intrabone) for cord-blood cells, measured by the donor-derived neutrophil and platelet engraftment. METHODS: Adult patients with acute leukaemia, for whom an unrelated stem-cell transplantation was indicated and no suitable unrelated human leucocyte antigen (HLA)-matched donor had been identified, were included in the study and underwent a cord-blood transplant in San Martino Hospital, Genoa, Italy. Eight patients were in first complete remission, ten in second complete remission, and 14 had advanced-stage, refractory disease. HLA matching was 5/6, 4/6, and 3/6 for 9, 22, and one patient, respectively. Cord-blood cells were concentrated in four 5-mL syringes, and were infused in the superior-posterior iliac crest under rapid general anaesthesia. Median transplanted cell dose was 2.6 x 10(7)/kg (range 1.4-4.2). The primary endpoint was the probability of neutrophil and platelet recovery after intrabone cord-blood transplantantion. Secondary endpoints included the incidence of acute graft-versus-host disease, relapse, and overall survival. This trial is registered on the ClinicalTrials.gov website, number NCT 00696046. FINDINGS: Between March 31, 2006, and Jan 25, 2008, 32 consecutive patients with acute myeloid leukaemia (n=20) or acute lymphoblastic leukaemia (n=12) underwent a cord-blood transplant (median age 36 years [range 18-66]). No complications occurred during or after the intrabone infusion of cells. Four patients with advanced-stage disease died within 12 days of the procedure. Median time to recovery of neutrophils in 28 patients (>/=0.5 x 10(9)/L) was 23 days (range 14-44) and median time to recovery of platelets in 27 patients (>/=20 x 10(9)/L) was 36 days (range 16-64). All patients were fully chimeric from 30 days after transplantation to the last follow-up visit, suggesting an early complete donor engraftment. No patient developed grade III-IV acute graft-versus-host disease. Causes of death were transplant related (n=5), infection (n=7), and relapse (n=4). 16 patients were alive and in haematological remission at a median follow-up of 13 months (range 3-23). INTERPRETATION: Our preliminary data suggest that direct intrabone cord-blood transplantation overcomes the problem of graft failure even when low numbers of HLA-mismatched cord-blood cells are transplanted, thus leading to the possibility of use of this technique in a large number of adult patients.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Leukemia, Myeloid, Acute/therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Aged , Humans , Infusions, Intraosseous , Middle Aged , Transplantation, Homologous , Treatment Outcome
4.
Rev. bras. hematol. hemoter ; 30(supl.2): 42-44, jun. 2008.
Article in English | LILACS | ID: lil-496444

ABSTRACT

Minimal residual disease (MRD) was monitored in 80 patients with acute lymphoid (ALL, n=44) or myeloid (AML, n=36) leukemia, undergoing allogeneic haemopoietic stem cell transplantations. MRD markers were IgH-VDJ and TCR gene re-arrangement for ALL, and Wilm's Tumor (WT1) expression for AML. The overall cumulative incidence (CI) of MRD was positive in 45 percent and the CI of hematologic relapse was 24 percent (36 percent in MRD+ vs. 16 percent in MRD patients, p=0.03). The median interval from transplant to first MRD positivity was 120 days and to hematologic relapse 203 days. Patients were divided in 3 MRD groups: MRD (n=44), MRD+ given donor lymphocyte infusions (DLI) (n=17) and MRD+ not given DLI (n=19): leukemia relapse rates in these 3 groups were 16 percent, 6 percent and 63 percent, respectively (p<0.0001); the actuarial 3-year survival rates were 78 percent, 80 percent and 26 percent (p=0.001). In multivariate COX analysis, the MRD group was predictor of relapse (p<0.0001) and survival (p=0.01), together with disease phase and chronic graft versus host disease. In MRD+ patients, DLI protected against relapse (p=0.003) and improved survival (p=0.01). In conclusion, MRD positivity post-transplant predicts leukemia relapse: however, when MRD+ patients are given DLI, their outcomes are comparable to MRD- patients.


A doença residual mínima (DRM) foi monitorada em 80 pacientes com leucemia linfóide aguda (n=44) e mielóide aguda (n=36) que foram submetidos ao transplante alogênico de célula-tronco. Marcadores da DRM foram a IgH-VDJ e rearranjo do TCR para a LLA e expressão do Tumor de Wills (WT1) para LMA. A incidência acumulada global (IC) para a DRM foi positiva em 45 por cento e a IC para recaída hematológica foi 24 por cento (36 por cento na DRM+ versus 16 por cento na DRM-, p=0.03). O intervalo mediano entre o TMO e a primeira DRM positividade foi dia +120, e para a recaída hematológica, dia +203. Os pacientes puderam ser divididos em três grupos: DRM-(n=44), DRM+ onde foi dada a infusão de linfócitos do doador (ILD) (n=17) e DRM+ não dado ILD (n=19): a recidiva nos três grupos foi de 16 por cento, 6 por cento e 63 por cento, respectivamente (p<0.0001); a sobrevida em três anos foi 78 por cento, 80 por cento e 26 por cento (p=0.001). No modelo de Cox, o grupo de DRM foi preditor de recidiva (p<0.0001) e sobrevida global (p=0.01), juntamente com a fase da doença e a doença do enxerto contra o hospedeiro. Na DRM+, IDL protegeu contra a recidiva (p=0.003) e melhorou a sobrevida (p=0.01). Em conclusão, a positividade para a DRM pós-transplante prediz recidiva da leucemia. Entretanto, quando é dada a ILD ao paciente DRM+, a evolução destes pacientes é comparável aos pacientes DRM-.


Subject(s)
Humans , Hematopoietic Stem Cell Transplantation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Lymphocytes , Neoplasm, Residual , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Transplantation, Homologous
5.
Exp Hematol ; 36(3): 340-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18082308

ABSTRACT

OBJECTIVE: Endothelial progenitor cells (EPCs) are involved in neovessel formation. So far, therapeutic angiogenesis is hampered by the low frequency and limited proliferative potential of these cells isolated from peripheral blood. Recently, it has been shown that cord blood-derived EPCs (CB EPCs) can be ex vivo expanded on a clinical scale. In this study, we evaluated the expansion potential of CB EPCs together with their phenotypic, functional, and chromosomal stability over time. MATERIALS AND METHODS: Flow cytometry, in vitro tube formation, and proliferation assays were performed to characterize CB EPC-derived cells. Chromosomal stability was evaluated by karyotype analysis. In vitro and in vivo tumorigenicity was evaluated by soft agar assay and injection into nonobese diabetic/severe combined immunodeficient mice, respectively. RESULTS: We showed that CB EPC-derived cells displayed phenotypic and functional features of EPCs, although a process of maturation was observed over time. Although we confirmed that CB EPCs have a greater expansion potential compared to peripheral blood EPCS, we observed a high incidence of cytogenetic alterations (71%) in the expanded endothelial cell population, even at early times of culture. In two cases, spontaneous transformation in vitro was documented, but none of the samples tested showed tumorigenic potential in vivo. Conversely, no karyotype alterations have been observed on peripheral blood EPCs-derived cells. CONCLUSIONS: We confirm that CB represents a good source for clinical ex vivo expansion of EPCs. However, because of high frequency of karyotype alterations, these cells cannot be considered free of risk in clinical application.


Subject(s)
Chromosome Aberrations , Endothelial Cells/cytology , Fetal Blood/cytology , Stem Cells/cytology , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Colony-Forming Units Assay , Endothelial Cells/pathology , Flow Cytometry , Humans , Immunophenotyping , Karyotyping , Risk Factors , Stem Cells/pathology
6.
Cancer ; 110(9): 2007-11, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17823911

ABSTRACT

BACKGROUND: Leukemias are dependent on Akt/NF-kappaB activation and angiogenesis. METHODS: The antiangiogenic Akt/NF-kappaB inhibitor xanthohumol (XN) has in vitro activity against acute and chronic myelogenous leukemia cell lines (AML, CML) and fresh samples from patients were investigated. RESULTS: Inhibition of cell proliferation is associated with induction of apoptosis and reduced VEGF secretion. Decreased cell invasion, metalloprotease production, and adhesion to endothelial cells observed in the presence of XN could prevent in vivo life-threatening complications of leukostasis and tissue infiltration. CONCLUSIONS: As endothelial cells and hematopoietic cells are mutually correlated in their development and growth, targeting both tumor cells and endothelial cells with agents possessing cytotoxic and antiangiogenic activities may lead to synergistic antitumor effects interrupting a reciprocal stimulatory loop between leukemia and endothelial cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Proliferation/drug effects , Hematologic Neoplasms , Neovascularization, Pathologic/drug therapy , Propiophenones/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Dose-Response Relationship, Drug , Flavonoids , Humans , NF-kappa B/antagonists & inhibitors , NF-kappa B/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/drug effects
8.
Haematologica ; 91(7): 935-40, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16818281

ABSTRACT

BACKGROUND AND OBJECTIVES: A proportion of patients develop poor graft function (PGF) following an allogeneic hemopoietic stem cell transplant (HSCT). It is uncertain whether a boost of donor marrow or blood cells is beneficial in terms of trilineage recovery and non-relapse-related mortality (NRM). DESIGN AND METHODS: The aim of this study was to compare outcomes in patients with PGF and full donor chimerism following an allogeneic HSCT who did or did not receive a boost of donor stem cells. The study included patients with primary PGF--i.e. those failing to achieve sustained graft function- and secondary PGF--i.e. those developing PGF after complete hematologic recovery. We studied 54 patients with PGF: 20 patients received no further donor cell infusion (group A), 14 received a boost of unmanipulated marrow or blood cells from the original donor, without further conditioning (group B), and 20 received donor cells after CD34 selection without conditioning (group C). The three groups were comparable for disease phase, patients' age, donor type, primary or secondary PGF, full donor chimerism and duration of PGF. RESULTS: Trilineage recovery was seen in 40%, 36% and 75% of the patients in, respectively, groups A, B and C (p=0.02). In multivariate Cox analysis trilineage recovery was more frequent in patients with secondary PGF (RR of complete recovery 2.82, p=0.01) and in patients receiving CD34+-selected cells (RR of complete recovery 3.0; p=0.007). There was no effect of donor type on hematologic recovery. The rate of NRM was 55%, 64%, 20% in groups A, B and C, respectively (p=0.06) and was highly correlated with trilineage recovery (RR 0.36, p<0.0001). PGF was the primary cause of death in 30%, 21% and 10% of the patients in the three groups, graft-versus-host disease (GVHD) in 5%, 36%, and 10%. INTERPRETATIONS AND CONCLUSIONS: In patients with poor graft function (a) a boost of CD34+-selected peripheral blood donor cells is associated with a high chance of trilineage recovery and a low risk of acute GVHD; (b) a boost of unmanipulated donor cells does not seem to offer a survival advantage over no infusion of cells; and (c) NRM is lower when using peripheral blood cells for the boost. These data may be useful when discussing second stem cell donations for patients with poor graft function.


Subject(s)
Antigens, CD34 , Graft Survival , Peripheral Blood Stem Cell Transplantation/methods , Adolescent , Adult , Female , Graft vs Host Disease/mortality , Humans , Male , Middle Aged , Peripheral Blood Stem Cell Transplantation/adverse effects , Peripheral Blood Stem Cell Transplantation/mortality , Transplantation, Homologous , Treatment Failure
9.
Exp Hematol ; 34(7): 934-42, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16797421

ABSTRACT

OBJECTIVE: Multipotent mesenchymal stromal cells (MSCs) are endowed with multilineage differentiative potential and immunomodulatory properties. It is still a matter of debate whether donor MSCs have sustained engraftment potential in host bone marrow (BM) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). The aim of this study was to analyze the donor/recipient origin of MSCs in children receiving allogeneic either BM or cord blood (CB) transplantation. METHODS: Thirty-seven pediatric patients undergoing allo-HSCT for either a malignant or a nonmalignant disorder were enrolled in the study; 19 received CB and 18 BM transplantation. Results were compared with those obtained in 14 adults given BM transplantation for either malignant or nonmalignant disorders. MSCs were grown from BM aspirates obtained 1-17 and 2-192 months after allo-HSCT in pediatric and adult patients, respectively. MSC samples at the third-fourth passage were phenotypically characterized. Donor/recipient origin of MSCs was assessed by amelogenin assay and microsatellite analysis. RESULTS: MSCs could be grown from 30 of 37 children; at the third-fourth passage MSCs resulted positive (> or = 98%) for CD73, CD105, CD106, CD29, CD13, CD44 and negative (< or = 1%) for CD34, CD45, CD14. Mixed chimerism with donor cells was observed in 4 BM and 5 CB transplantation recipients, respectively; full recipient chimerism was detected in the remaining children. Full recipient MSC chimerism was observed also in all assessable (12/14) adult patients. CONCLUSIONS: BM of pediatric patients might be a more favorable milieu than that of adults for sustained engraftment of transplanted MSCs. MSCs able to engraft in the host can be transferred with cryopreserved CB units.


Subject(s)
Bone Marrow Transplantation , Fetal Blood/transplantation , Mesoderm/cytology , Stromal Cells/cytology , Adolescent , Adult , Child , Child, Preschool , Humans , Infant
10.
Blood ; 108(5): 1661-7, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16684957

ABSTRACT

Vascular endothelial growth factor165 (VEGF165) and semaphorin3A (SEMA3A) elicit pro- and antiangiogenic signals respectively in endothelial cells (ECs) by binding to their receptors VEGFR-2, neuropilin-1 (NRP1), and plexin-A1. Here we show that the VEGF165-driven angiogenic potential of multiple myeloma (MM) ECs is significantly higher than that of monoclonal gammopathy of undetermined significance (MGUS) ECs (MGECs) and human umbilical vein (HUV) ECs. This is probably due to a constitutive imbalance of endogenous VEGF165/SEMA3A ratio, which leans on VEGF165 in MMECs but on SEMA3A in MGECs and HUVECs. Exogenous VEGF165 induces SEMA3A expression in MGECs and HUVECs, but not in MMECs. Moreover, by counteracting VEGF165 activity as efficiently as an anti-VEGFR-2 antibody, exogenous SEMA3A restrains the over-angiogenic potential of MMECs. Our data indicate that loss of endothelial SEMA3A in favor of VEGF165 could be responsible for the angiogenic switch from MGUS to MM.


Subject(s)
Bone Marrow Cells/pathology , Multiple Myeloma/pathology , Receptors, Vascular Endothelial Growth Factor/genetics , Semaphorin-3A/deficiency , Semaphorin-3A/genetics , Vascular Endothelial Growth Factor A/genetics , Cell Division , Cells, Cultured , Chemotaxis , DNA Primers , Endothelial Cells/pathology , Humans , Multiple Myeloma/genetics , Neovascularization, Physiologic , Reverse Transcriptase Polymerase Chain Reaction
11.
Exp Hematol ; 33(7): 819-27, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15963858

ABSTRACT

OBJECTIVE: To compare the suppressive effect of mesenchymal stem cells (MSC), derived from normal individuals or severe aplastic anemia patients (SAA), on T-cell activation. PATIENTS AND METHODS: We studied bone marrow MSC from 19 healthy donors and 23 SAA patients in different phases of the disease: at diagnosis (n = 3), following immunosuppressive therapy (IS) (n = 16), or after a bone marrow transplant (BMT) (n = 4). MSC were tested for T-cell suppression in the following assays: mixed lymphocyte reaction (MLR), phytohemaglutinin (PHA)-primed cultures, activation surface markers, gamma-IFN production, hematopoietic colony formation (CFC), production of cyclic ADP-ribose (cADPR). RESULTS: The abnormalities of SAA MSC included: 1) significantly lower suppression of T-cell proliferation induced by alloantigens (p = 0.009) or PHA (p = 0.006); 2) impaired capacity to suppress CD38 expression on PHA-primed T cells (p = 0.001); 3) impaired ability to suppress gamma-IFN production in PHA cultures, resulting in an 11-fold higher gamma-IFN concentration; 4) no preventive effect on T cell-mediated inhibition of CFC; and 5) significantly reduced (p = 0.009) production of cADPR, a universal calcium mobilizer. MSC-mediated suppression of PHA-induced T-cell proliferation was restored to control levels in 3 of 4 patients post-BMT. CONCLUSION: The ability of MSC to downregulate T-cell priming, proliferation, and cytokine release is deficient in patients with SAA, persists indefinitely after immunosuppressive therapy, but seems to be restored after BMT. Whether these abnormalities are relevant to the pathogenesis of aplastic anemia remains to be determined.


Subject(s)
Anemia, Aplastic/immunology , Anemia, Aplastic/pathology , Mesoderm/physiology , Stem Cells/physiology , T-Lymphocytes/immunology , Adolescent , Adult , Aged , Bone Marrow Cells/cytology , Bone Marrow Cells/pathology , Bone Marrow Transplantation/immunology , Female , Humans , Immunosuppression Therapy , Lymphocyte Activation , Lymphocyte Culture Test, Mixed , Male , Middle Aged , Phytohemagglutinins , Reference Values
12.
Haematologica ; 90(4): 516-25, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15820948

ABSTRACT

BACKGROUND AND OBJECTIVES: Experimental evidence and preliminary clinical studies have demonstrated that human mesenchymal stem cells (MSC) have an important immune modulatory function in the setting of allogeneic hematopoietic stem cell (HSC) transplantation. We extended the evaluation of mechanisms responsible for the immune regulatory effect derived from the interaction of human MSC with cells involved in alloantigen-specific immune response in mixed lymphocyte culture (MLC). DESIGN AND METHODS: Dendritic cell (DC) differentiation, T- and natural killer (NK)-lymphocyte expansion, alloantigen-specific cytotoxic activity and differentiation of CD4+ T-cell subsets co-expressing CD25 and/or CTLA4 molecules were assessed, comparing the effect observed using third-party MSC with that obtained employing MSC autologous to the MLC responder. RESULTS: We found that human MSC strongly inhibit alloantigen-induced DC1 differentiation, down-regulate alloantigen-induced lymphocyte expansion, especially that of CD8+ T cells and of NK lymphocytes, decrease alloantigen-specific cytotoxic capacity mediated by either cytotoxic T lymphocytes or NK cells and favor the differentiation of CD4+ T-cell subsets co-expressing CD25 and/or CTLA4. More effective suppressive activity on MLC-induced T-cell activation was observed when MSC were third-party, rather than autologous, with respect to MLC-responder cells. INTERPRETATION AND CONCLUSIONS: Our results strongly suggest that MSC-mediated inhibition of alloantigen-induced DC1 differentiation and preferential activation of CD4+ CD25+ T-cell subsets with presumed regulatory activity represent important mechanisms contributing to the immunosuppressive activity of MSC. Collectively, these data provide immunological support for the use of MSC to prevent immune complications related to both HSC and solid organ transplantation and to the theory that MSC are universal suppressors of immune reactivity.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Hematopoietic Stem Cell Transplantation/adverse effects , Isoantigens/immunology , Mesenchymal Stem Cells/cytology , T-Lymphocyte Subsets/cytology , Antibody Formation , Cell Communication , Cell Differentiation , Epitopes , Flow Cytometry , Gene Expression Regulation/genetics , Humans , Immunosuppression Therapy , Lymphocyte Culture Test, Mixed , Mesenchymal Stem Cells/immunology , Phenotype
13.
Haematologica ; 89(10): 1238-47, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15477210

ABSTRACT

BACKGROUND AND OBJECTIVES: Transplant-related mortality (TRM) following allogeneic hematopoietic stem cell transplantation (HSCT) has been reported to be related to disease stage, duratiion of disease and type of donor. Furthermore, the outcome of transplants performed in the 1990s appears to be better than that of transplants done in the previous decade. The aims of this study were to determine whether these relationships still hold and whether the outcome of transplants is continuing to improve. DESIGN AND METHODS: We analyzed 1180 consecutive patients with leukemia (n=979) or other hematologic malignancies (n=201) undergoing HSCT in 4 time periods: before 1990, 1991-1995, 1996-2000, and 2001-2002. Changes during these eras include increasing patient age, more unrelated transplants, more patients with advanced disease, different graft-versus-host disease (GvHD) prophylaxis, and different management of infections. RESULTS: The actuarial 2-year transplant-related mortality (TRM) differed significantly between the transplant eras (p<0.001) with a significant interaction with disease phase (p=0.018). In patients in first remission (n=585) TRM was 34%, 25%, 21% and 6% in the four transplant eras. The reduction in TRM was less evident in patients in second remission (n=284) (37%, 35%, 30%, 25%) and absent in relapsed patients (n=311) (TRM=45%, 41%, 29%, 51%). This is a consequence of reductions in GvHD, infections and multiorgan failure among patients in remission but not among those who relapse. The actuarial 2-year survival has improved significantly in patients in first remission (54%, 66%, 72%, 78%) but not in those in second remission (38%, 46%, 52%,45%), or relapsed patients (31%, 25%, 36%, 21%). INTERPRETATION AND CONCLUSIONS: In conclusion, TRM has been significantly reduced in first remission patients, suggesting an allograft should be considered in this phase, when appropriate, without delay. There has been no improvement in survival for patients beyond first remission, due to persisting high risk of infections and organ toxicity, a possible consequence of prolonged pre-transplant chemotherapy and neutropenia.


Subject(s)
Bone Marrow Transplantation/mortality , Hematologic Neoplasms/surgery , Transplantation, Homologous/mortality , Actuarial Analysis , Adult , Anti-Infective Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Transplantation/statistics & numerical data , Cause of Death , Combined Modality Therapy , Female , Graft vs Host Disease/etiology , Graft vs Host Disease/mortality , Graft vs Host Disease/prevention & control , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/mortality , Hematopoietic Stem Cell Transplantation/mortality , Hematopoietic Stem Cell Transplantation/statistics & numerical data , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Infection Control , Infections/etiology , Infections/mortality , Leukemia/drug therapy , Leukemia/mortality , Leukemia/surgery , Lung Diseases, Interstitial/etiology , Lung Diseases, Interstitial/mortality , Male , Middle Aged , Multiple Organ Failure , Neutropenia/chemically induced , Neutropenia/complications , Peripheral Blood Stem Cell Transplantation/adverse effects , Peripheral Blood Stem Cell Transplantation/methods , Peripheral Blood Stem Cell Transplantation/mortality , Peripheral Blood Stem Cell Transplantation/statistics & numerical data , Premedication , Remission Induction , Retrospective Studies , Salvage Therapy , Survival Analysis , Transplantation Conditioning/methods , Transplantation Conditioning/mortality , Transplantation, Homologous/methods , Transplantation, Homologous/statistics & numerical data , Treatment Outcome
14.
Exp Hematol ; 32(8): 782-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15308330

ABSTRACT

OBJECTIVE: Intravenous (IV) injection is currently the normal method for transplanting hematopoietic cells. However, the problem of seeding efficiency and homing is relevant especially when a limited number of stem cells is available. Intra-bone marrow (IBM) injection of bone marrow cells (BMCs) may overcome this problem. MATERIALS AND METHODS: Irradiated (750 cGy) C57BL/6J mice were transplanted with 1 x 10(5) BMCs harvested from transgenic mice expressing an enhanced version of the green fluorescent protein (EGFP+) via IBM or with 1 x 10(6) EGFP+ BMCs via IV. Irradiated (320 cGy) NOD/SCID mice were transplanted with 1 x 10(6) human cord blood (CB) cells via IBM or with 1 x 10(7) human CB cells via IV. RESULTS: In C57BL/6J mice after 90 days, the fraction of EGFP+ cells harvested was 37% and 53% in IV-treated and IBM-treated (contralateral tibia and femur in the IBM) mice, respectively: the expansion folds were 114 and 1760, respectively. In NOD/SCID mice, the percentages of CD45+ cells and CD45+/CD34+ cells were, at 30 days, 3.3% and 0.3% in IV-treated mice, and 4.4% and 1.1% in IBM-treated mice. At 60 days, the percentages of CD45+ cells and CD45+/CD34+ cells were 2.1% and 0.3% in IV-treated mice and 1.4% and 0.4% in IBM-treated mice. At day 90 the percentages of CD45+ cells and CD45+/CD34+ cells were 3% and 0.3% in IV-treated mice and 2.3% and 0.4% in IBM-treated mice. CONCLUSION: Our data demonstrate that IBM transplantation is associated with a seeding efficiency 15 times greater than IV transplantation. IBM transplantation may improve the results of transplant and may be useful in several settings: 1) when a limited number of hematopoietic progenitors are available; and 2) in experiments aiming to place in the bone marrow stem cells of other lineages (CNS, muscle, etc.).


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/methods , Animals , Cell Division , Green Fluorescent Proteins , Humans , Luminescent Proteins/analysis , Mice , Mice, Inbred C57BL
15.
Blood ; 102(3): 1138-41, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12689932

ABSTRACT

Delayed hematopoietic recovery is the main factor precluding a wider use of cord blood (CB) transplants. We hypothesized that this delayed engraftment might not be related to an insufficient number of stem cells in the graft, but to an intrinsic difficulty of these cells to undergo differentiation. To test our hypothesis, 2 groups of children were compared; 12 received a CB transplant and 12 an adult bone marrow (BM) transplant. We studied neutrophil and platelet recovery and, at a median time of approximately 1 year after transplantation, the frequency of colony-forming cells (CFCs) and long-term culture initiating cells (LTC-ICs) in the BM of the 2 groups. Recipients of BM transplants received 1-log more cells and had significantly faster neutrophil and platelet recovery. Conversely, the frequency of committed and early progenitors was significantly higher in the BM of children given CB cells compared with BM transplant recipients (median count of CFC/2 x 10(4) BM mononuclear cells, 20 versus 11, P =.007; median count of LTC-IC/10(6) BM mononuclear cells, 8.2 versus 0.2 P =.001). CB, but not adult BM stem cells, can better restore the host hematopoietic progenitor cell reservoir; the delayed engraftment after CB transplantation may reflect the difficulty of CB progenitors to reprogram themselves toward differentiation.


Subject(s)
Bone Marrow Transplantation/standards , Cord Blood Stem Cell Transplantation/standards , Graft Survival , Hematopoiesis , Adolescent , Cell Differentiation/physiology , Child , Child, Preschool , Female , Hematologic Diseases/therapy , Hematopoietic Stem Cells/cytology , Humans , Infant , Male
SELECTION OF CITATIONS
SEARCH DETAIL