Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Leuk Res ; 36(8): 982-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22524974

ABSTRACT

BACKGROUND: We previously demonstrated upregulation of c-myc, survivin, and cyclin D1 in CD34+ bone marrow mononuclear cells (BMMNCs) of patients with trisomy 8 and monosomy 7 myelodysplastic syndromes (MDS). "Knockdown" of cyclin D1 by RNA interference decreased trisomy 8 cell growth, suggesting that this might be a therapeutic target in MDS. EXPERIMENTAL DESIGN: We performed preclinical studies using BMMNCs from patients with MDS and AML to examine the effects of the styryl sulfone ON 01910.Na on cyclin D1 accumulation, aneuploidy, and CD34+ blast percentage. We next treated twelve patients with higher risk MDS and two trisomy 8 AML patients with ON 01910.Na on a phase I clinical protocol (NCT00533416). RESULTS: ON 01910.Na inhibited cyclin D1 expression, and was selectively toxic to trisomy 8 cells in vitro. Flow cytometry studies demonstrated increased mature CD15+ myeloid cells and decreased CD34+ blasts. Three patients treated with ON 01910.Na on a clinical had decreased bone marrow blasts by ≥ 50%, and three patients had hematologic improvements, one of which was sustained for 33 months. Patients with hematologic responses to ON 01910.Na had decreased cyclin D1 expression in their CD34+ cells. CONCLUSIONS: The preclinical results and responses of patients on a clinical trial warrant further investigation of ON 01910.Na as a potential novel targeted therapy for higher risk MDS patients.


Subject(s)
Cyclin D1/antagonists & inhibitors , Glycine/analogs & derivatives , Molecular Targeted Therapy/methods , Myelodysplastic Syndromes/drug therapy , Sulfones/therapeutic use , Aged , Aged, 80 and over , Algorithms , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Chromosomes, Human, Pair 8 , Dose-Response Relationship, Drug , Female , Glycine/administration & dosage , Glycine/adverse effects , Glycine/pharmacology , Glycine/therapeutic use , Humans , Male , Middle Aged , Molecular Targeted Therapy/adverse effects , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Sulfones/administration & dosage , Sulfones/adverse effects , Sulfones/pharmacology , Trisomy/pathology , Tumor Cells, Cultured
2.
Transfusion ; 52(3): 537-41, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21883270

ABSTRACT

BACKGROUND: Reports of Monosomy 7 in patients receiving granulocyte-colony-stimulating factor (G-CSF) have raised concerns that this cytokine may promote genomic instability. However, there are no studies addressing whether repeated administration of G-CSF produces Monosomy 7 aneuploidy in healthy donors. STUDY DESIGN AND METHODS: We examined Chromosomes 7 and 8 by fluorescent in situ hybridization (FISH) in CD34+ cells from 35 healthy hematopoietic stem cell transplant (HSCT) donors after G-CSF administration for 5 days and by spectral karyotyping analysis (SKY) in four individuals to assess chromosomal integrity. We also studied 38 granulocyte donors who received up to 42 doses of G-CSF and dexamethasone (Dex) using FISH for Chromosomes 7 and 8. RESULTS: We found no abnormalities in Chromosomes 7 and 8 in G-CSF-mobilized CD34+ cells when assessed by FISH or SKY, nor did we detect aneuploidy in G-CSF- and Dex-treated donors. CONCLUSION: G-CSF does not promote clinically detectable Monosomy 7 or Trisomy 8 aneuploidy in HSCT or granulocyte donors. These findings should be reassuring to healthy HSCT and granulocyte donors.


Subject(s)
Genomic Instability/drug effects , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocytes/drug effects , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Aneuploidy , Chromosome Deletion , Chromosomes, Human, Pair 7/drug effects , Dexamethasone/administration & dosage , Glucocorticoids/administration & dosage , Granulocytes/physiology , Granulocytes/transplantation , Hematopoietic Stem Cell Mobilization/adverse effects , Hematopoietic Stem Cell Mobilization/standards , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Humans , Tissue Donors
3.
JAMA ; 305(8): 814-9, 2011 Feb 23.
Article in English | MEDLINE | ID: mdl-21343581

ABSTRACT

Myelodysplastic syndromes (MDS) are a heterogeneous group of bone marrow disorders characterized by ineffective hematopoiesis and a tendency to develop leukemia. In some patients, laboratory and clinical evidence supports a role for the immune system in the pathogenesis of early MDS. Many younger patients who respond to immunosuppressive therapy with drugs such as antithymocyte globulin and cyclosporine have clonal expansions of cytotoxic CD8(+) T cells that suppress normal hematopoiesis, as well as expansion of CD4(+) helper T-cell subsets that promote and sustain autoimmunity. Immunosuppressive therapy can produce hematologic responses in some patients and may improve survival and halt leukemic progression. In this report, we describe a 56-year-old woman who presented with fatigue and easy bruising, eventually became pancytopenic, and was diagnosed with MDS. After treatment with a clinical protocol using alemtuzumab, an anti-CD52 antibody, her blood cell counts returned to normal and she has remained in complete remission for more than 2 years of follow-up. In this article, we review the pathobiology of immune dysregulation in MDS and summarize the role of immunosuppressive therapy in MDS.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Antineoplastic Agents/therapeutic use , Immune System/physiopathology , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/immunology , Alemtuzumab , Antibodies, Monoclonal, Humanized , Female , Humans , Immunosuppressive Agents/therapeutic use , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/epidemiology , Prognosis , Treatment Outcome
4.
Blood ; 117(9): 2691-9, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21097671

ABSTRACT

Clinical observations and laboratory evidence link bone marrow failure in myelodysplastic syndrome (MDS) to a T cell-mediated immune process that is responsive to immunosuppressive treatment (IST) in some patients. Previously, we showed that trisomy 8 MDS patients had clonally expanded CD8(+) T-cell populations that recognized aneuploid hematopoietic progenitor cells (HPC). Furthermore, microarray analyses showed that Wilms tumor 1 (WT1) gene was overexpressed by trisomy 8 hematopoietic progenitor (CD34(+)) cells compared with CD34(+) cells from healthy donors. Here, we show that WT1 mRNA expression is up-regulated in the bone marrow mononuclear cells of MDS patients with trisomy 8 relative to healthy controls and non-trisomy 8 MDS; WT1 protein levels were also significantly elevated. In addition, using a combination of physical and functional assays to detect the presence and reactivity of specific T cells, respectively, we demonstrate that IST-responsive MDS patients exhibit significant CD4(+) and CD8(+) T-cell responses directed against WT1. Finally, WT1-specific CD8(+) T cells were present within expanded T-cell receptor Vß subfamilies and inhibited hematopoiesis when added to autologous patient bone marrow cells in culture. Thus, our results suggest that WT1 is one of the antigens that triggers T cell-mediated myelosuppression in MDS.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunosuppression Therapy , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/therapy , WT1 Proteins/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Case-Control Studies , Chromosomes, Human, Pair 8/genetics , Chromosomes, Human, Pair 8/immunology , Gene Expression Regulation , HLA-A Antigens/chemistry , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Immunodominant Epitopes/immunology , Protein Structure, Quaternary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Trisomy/genetics , Trisomy/immunology , WT1 Proteins/genetics , WT1 Proteins/metabolism
5.
J Clin Oncol ; 28(35): 5166-73, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21041705

ABSTRACT

PURPOSE: Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis and progression to leukemia. Clinical and experimental evidence suggests an immune-mediated pathophysiology in some patients, in whom immunosuppressive therapy (IST) with horse antithymocyte globulin (h-ATG) and cyclosporine (CsA) can be effective. Because of the toxicities associated with h-ATG/CsA, we investigated an alternative regimen with alemtuzumab in MDS. PATIENTS AND METHODS: We conducted a nonrandomized, off-label, pilot, phase I/II study of alemtuzumab monotherapy in patients with MDS who were judged likely to respond to IST based on the following criteria: HLA-DR15-negative patients whose age plus the number of months of RBC transfusion dependence (RCTD) was less than 58; and HLA-DR15-positive patients whose age plus RCTD was less than 72. In total, 121 patients with MDS were screened, of whom 32 met eligibility criteria to receive alemtuzumab 10 mg/d intravenously for 10 days. Primary end points were hematologic responses at 3, 6, and 12 months after alemtuzumab. RESULTS: Seventeen (77%) of 22 evaluable intermediate-1 patients and four (57%) of seven evaluable intermediate-2 patients responded to treatment with a median time to response of 3 months. Four of seven evaluable responders with cytogenetic abnormalities before treatment had normal cytogenetics by 1 year after treatment. Five (56%) of nine responding patients evaluable at 12 months had normal blood counts, and seven (78%) of nine patients were transfusion independent. CONCLUSION: Alemtuzumab is safe and active in MDS and may be an attractive alternative to ATG in selected patients likely to respond to IST.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Antineoplastic Agents/therapeutic use , Myelodysplastic Syndromes/drug therapy , Adult , Aged , Alemtuzumab , Antibodies, Monoclonal, Humanized , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Myelodysplastic Syndromes/mortality , Off-Label Use , Pilot Projects , Treatment Outcome , Young Adult
6.
JAMA ; 304(12): 1358-64, 2010 Sep 22.
Article in English | MEDLINE | ID: mdl-20858879

ABSTRACT

CONTEXT: Critically short telomeres produce apoptosis, cell senescence, and chromosomal instability in tissue culture and animal models. Variations in telomere length have been reported in severe aplastic anemia but their clinical significance is unknown. OBJECTIVE: To investigate the relationship between telomere length and clinical outcomes in severe aplastic anemia. DESIGN, SETTING, AND PATIENTS: Single institution analysis of 183 patients with severe aplastic anemia who were treated in sequential prospective protocols at the National Institutes of Health from 2000 to 2008. The pretreatment leukocyte age-adjusted telomere length of patients with severe aplastic anemia consecutively enrolled in immunosuppression protocols with antithymocyte globulin plus cyclosporine for correlation with clinical outcomes were analyzed. MAIN OUTCOME MEASURES: Hematologic response, relapse, clonal evolution, and survival. RESULTS: There was no relationship between hematologic response and telomere length with response rates of 56.5% of 46 patients in the first, 54.3% of 46 in the second, 60% of 45 in the third, and 56.5% of 46 in the fourth quartiles. Multivariate analysis demonstrated that telomere length was associated with relapse, clonal evolution, and mortality. Evaluated as a continuous variable, telomere length inversely correlated with the probability of hematologic relapse (hazard ratio [HR], 0.16; 95% confidence interval [CI], 0.03-0.69; P = .01). The probability of clonal evolution was higher in patients in the first quartile (24.5%; 95% CI, 8.7%-37.5%) than in quartiles 2 through 4 (8.4%; 95% CI, 3.2%-13.3%; P = .009), and evolution to monosomy 7 or complex cytogenetics was more common in the first quartile (18.8%; 95% CI, 3.5%-31.6%) [corrected] than in quartiles 2 through 4 (4.5%; 95% CI, 0.5%-8.2%; P = .002) [corrected]. Survival between these 2 groups differed, with 66% (95% CI, 52.9%-82.5%) surviving 6 years in the first quartile compared with 83.8% (95% CI, 77.3%-90.9%) in quartiles 2 through 4 (P = .008). CONCLUSION: In a cohort of patients with severe aplastic anemia receiving immunosuppressive therapy, telomere length was unrelated to response but was associated with risk of relapse, clonal evolution, and overall survival.


Subject(s)
Anemia, Aplastic/genetics , Cell Transformation, Neoplastic , Chromosomal Instability , Telomere/ultrastructure , Adult , Anemia, Aplastic/blood , Anemia, Aplastic/drug therapy , Anemia, Aplastic/pathology , Cohort Studies , Female , Humans , Immunosuppressive Agents/therapeutic use , Leukocytes/ultrastructure , Male , Prognosis , Recurrence , Survival Analysis , Treatment Outcome
7.
Genes Cancer ; 1(4): 331-45, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20717479

ABSTRACT

Here we report the discovery of ON044580, an α-benzoyl styryl benzyl sulfide that possesses potent inhibitory activity against two unrelated kinases, JAK2 and BCR-ABL, and exhibits cytotoxicity to human tumor cells derived from chronic myelogenous leukemia (CML) and myelodysplasia (MDS) patients or cells harboring a mutant JAK2 kinase. This novel spectrum of activity is explained by the non-ATP-competitive inhibition of JAK2 and BCR-ABL kinases. ON044580 inhibits mutant JAK2 kinase and the proliferation of JAK2(V617F)-positive leukemic cells and blocks the IL-3-mediated phosphorylation of JAK2 and STAT5. Interestingly, this compound also directly inhibits the kinase activity of both wild-type and imatinib-resistant (T315I) forms of the BCR-ABL kinase. Finally, ON044580 effectively induces apoptosis of imatinib-resistant CML patient cells. The apparently unrelated JAK2 and BCR-ABL kinases share a common substrate, STAT5, and such substrate competitive inhibitors represent an alternative therapeutic strategy for development of new inhibitors. The novel mechanism of kinase inhibition exhibited by ON044580 renders it effective against mutant forms of kinases such as the BCR-ABL(T315I) and JAK2(V617F). Importantly, ON044580 selectively reduces the number of aneuploid cells in primary bone marrow samples from monosomy 7 MDS patients, suggesting another regulatory cascade amenable to this agent in these aberrant cells. Data presented suggest that this compound could have multiple therapeutic applications including monosomy 7 MDS, imatinib-resistant CML, and myeloproliferative neoplasms that develop resistance to ATP-competitive agents.

8.
Biol Blood Marrow Transplant ; 16(12): 1665-73, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20659573

ABSTRACT

Graft-versus-host disease (GVHD) is a major risk factor for secondary malignancy after hematopoietic stem cell transplantation. Squamous cell carcinoma (SCC) of the skin and mucous membranes are especially frequent in this setting where aneuploidy and tetraploidy are associated with aggressive disease. The current study is directed at the mechanism of neoplasia in this setting. Unmanipulated keratinocytes from areas of oral GVHD in 9 patients showed tetraploidy in 10% to 46% of cells when examined by florescein in situ hybridization (FISH). Keratinocytes isolated from biopsy sites of GVHD but not from normal tissue showed even greater numbers of tetraploid cells (mean = 78%, range: 15%-85%; N = 9) after culture. To mimic the inflammatory process in GVHD, allogeneic HLA-mismatched lymphocytes were mixed with normal keratinocytes. After 2 weeks, substantial numbers of aneuploid and tetraploid cells were evident in cultures with lymphocytes and with purified CD8 but not CD4 cells. Telomere length was substantially decreased in the lymphocyte-treated sample. No mutations were present in the p53 gene, although haploinsufficiency for p53 due to the loss of chromosome 17 was common in cells exposed to lymphocytes. These findings suggest that in GVHD, inflammation and repeated cell division correlate with the development of karyotypic abnormalities.


Subject(s)
Chromosome Aberrations , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Keratinocytes/pathology , Adult , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Chromosome Deletion , Chromosomes, Human, Pair 17 , Coculture Techniques , Female , Genes, p53 , Graft vs Host Disease/pathology , HLA Antigens/immunology , Humans , Keratinocytes/immunology , Leukemia/genetics , Leukemia/pathology , Leukemia/surgery , Lymphocytes/cytology , Lymphocytes/immunology , Male , Middle Aged , Ploidies , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Smith-Magenis Syndrome , Telomere/genetics , Young Adult
9.
Hematol Oncol Clin North Am ; 24(2): 331-41, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20359629

ABSTRACT

Laboratory evidence and clinical evidence suggest that some patients with myelodysplastic syndrome (MDS) have immunologically mediated disease. This article describes the laboratory evidence supporting a role for the immune system in the marrow failure of MDS and clinical trials using IST in these patients.


Subject(s)
Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Myelodysplastic Syndromes/drug therapy , Adult , Aged , Aged, 80 and over , Antilymphocyte Serum/therapeutic use , Apoptosis , Biomedical Research , Bone Marrow/pathology , Clinical Medicine , Clinical Trials as Topic/statistics & numerical data , Cyclosporine/therapeutic use , Cytokines/metabolism , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/pathology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology
10.
Haematologica ; 95(3): 382-7, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20207845

ABSTRACT

BACKGROUND: Pure red cell aplasia and moderate aplastic anemia are marrow failure states with an immune pathogenesis. Previously, we described short-term improvements in blood counts in two pilot studies treating moderate aplastic anemia (mAA) and pure red cell aplasia (PRCA) patients with daclizumab, a humanized monoclonal antibody to the interleukin-2 receptor; we now report our long-term experience with a larger cohort of patients. DESIGN AND METHODS: After a median follow-up period of 4.8 years, 19 of 45 (42%) evaluable mAA patients and 10 of 26 (38%) patients with PRCA responded by three months and 2 additional mAA patients responded by six months following administration of the drug. RESULTS: Seven of 28 (25%) mAA patients achieved long-term packed red blood cell PRBC transfusion independence, and all PRCA responders achieved long-term transfusion PRBC transfusion independence. CONCLUSIONS: Red cell transfusion-independence prior to treatment in mAA patients predicted response. The only significant adverse treatment-related events were transient rashes and arthralgias. Daclizumab is safe and effective, and produces lengthy remissions in patients with PRCA and mAA.


Subject(s)
Anemia, Aplastic/drug therapy , Antibodies, Monoclonal/therapeutic use , Immunoglobulin G/therapeutic use , Immunosuppressive Agents/therapeutic use , Red-Cell Aplasia, Pure/drug therapy , Antibodies, Monoclonal, Humanized , Daclizumab , Follow-Up Studies , Humans , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Safety , Survival Rate , Time Factors , Treatment Outcome
12.
Hematol Oncol Clin North Am ; 23(2): 347-60, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19327588

ABSTRACT

Myelodysplasia must be considered in the differential diagnosis of patients who have bone marrow failure, but bone marrow cellularity per se may not substantially affect either response to therapy or prognosis. It is unclear whether the primary pathophysiologic defect differs between hyper- and hypoplastic patients who have myelodysplasia. Cellularity does not seem to affect response to immunosuppressive therapy significantly and does not seem to be the major factor affecting improvements in response to lenalidomide, stem cell transplantation, or hematopoietic growth factors.


Subject(s)
Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/drug therapy , Age Factors , Animals , Antilymphocyte Serum/therapeutic use , Bone Marrow Cells/pathology , Cyclosporine/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/physiopathology , Prognosis
13.
Haematologica ; 94(3): 348-54, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19181786

ABSTRACT

BACKGROUND: We hypothesized that the addition of sirolimus to standard horse antithymocyte globulin (h-ATG) and cyclosporine (CsA) would improve response rates in severe aplastic anemia, due to its complementary and synergistic properties to cyclosporine A. DESIGN AND METHODS: To test this hypothesis, we conducted a prospective randomized study comparing hATG/CsA/sirolimus to standard h-ATG/CsA. A total of 77 patients were treated from June 2003 to November 2005; 35 received h-ATG/CsA/sirolimus and 42 h-ATG/CsA. The two groups were well matched demographically and in blood counts prior to therapy. The primary end-point was hematologic response rate at 3 months, defined as no longer meeting the criteria for severe aplastic anemia. The study was powered to show a superior hematologic response rate of h-ATG/CsA/sirolimus compared to standard h-ATG/CsA. RESULTS: The overall response rate at 3 months was 37% for h-ATG/CsA/sirolimus and 50% for h-ATG/CsA and at 6 months 51% for h-ATG/CsA/sirolimus and 62% for h-ATG/CsA. After a planned interim analysis of 30 evaluable patients in each arm, accrual to the h-ATG/CsA/sirolimus arm was closed, as the conditional power for rejecting the null hypothesis was less than 1%. The rate of relapse, clonal evolution, and survival (secondary outcomes) did not differ significantly between patients treated with the two different regimens. CONCLUSIONS: Despite a theoretical rationale for its use, sirolimus did not improve the response rate in patients with severe aplastic anemia when compared to standard h-ATG/CsA.


Subject(s)
Anemia, Aplastic/drug therapy , Antilymphocyte Serum/therapeutic use , Cyclosporine/therapeutic use , Sirolimus/therapeutic use , Adolescent , Adult , Aged , Anemia, Aplastic/pathology , Animals , Child , Child, Preschool , Drug Therapy, Combination , Female , Horses , Humans , Immunosuppressive Agents/therapeutic use , Kaplan-Meier Estimate , Male , Middle Aged , Proportional Hazards Models , Prospective Studies , Recurrence , Treatment Outcome , Young Adult
14.
Blood ; 113(4): 875-82, 2009 Jan 22.
Article in English | MEDLINE | ID: mdl-18922853

ABSTRACT

Primitive quiescent CD34(+) chronic myeloid leukemia (CML) cells are more biologically resistant to tyrosine kinase inhibitors than their cycling counterparts; however, graft-versus-leukemia (GVL) effects after allogeneic stem cell transplantation (SCT) probably eliminate even these quiescent cells in long-term surviving CML transplant recipients. We studied the progeny of CD34(+) cells from CML patients before SCT, which were cultured 4 days in serum-free media with hematopoietic growth factors. BCR-ABL expression was similar in both cycling and quiescent noncycling CD34(+) populations. Quiescent CD34(+) cells from CML patients were less susceptible than their cycling CD34(+) and CD34(-) counterparts to lysis by natural killer (NK) cells from their HLA-identical sibling donors. Compared with cycling populations, quiescent CD34(+) CML cells had higher surface expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptors DR4 and DR5. Bortezomib up-regulated TRAIL receptor expression on quiescent CD34(+) CML cells, and further enhanced their susceptibility to cytotoxicity by in vitro expanded donor NK cells. These results suggest that donor-derived NK cell-mediated GVL effects may be improved by sensitizing residual quiescent CML cells to NK-cell cytotoxicity after SCT. Such treatment, as an adjunct to donor lymphocyte infusions and pharmacologic therapy, may reduce the risk of relapse in CML patients who require treatment by SCT.


Subject(s)
Antigens, CD34/metabolism , Boronic Acids/pharmacology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Pyrazines/pharmacology , Adolescent , Adult , Bortezomib , Cell Separation , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic/genetics , Genetic Predisposition to Disease/genetics , Genotype , Humans , Killer Cells, Natural/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Male , Middle Aged , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Receptors, Death Domain/metabolism , Up-Regulation/drug effects
15.
Exp Hematol ; 36(12): 1616-24, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18954937

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired genetic disorder of the bone marrow that produces intravascular hemolysis, proclivity to venous thrombosis, and hematopoietic failure. Mutation in the PIG-A gene of a hematopoietic stem cell abrogates synthesis of glycosylphosphoinositol (GPI) anchors and expression of all GPI-anchored proteins on the surface of progeny erythrocytes, leukocytes, and platelets. Urokinase plasminogen activator receptor (uPAR), a GPI-linked protein expressed on neutrophils, mediates endogenous thrombolysis through a urokinase-dependent mechanism. Here we show that membrane GPI-anchored uPAR is decreased or absent on granulocytes and platelets of patients with PNH, while soluble uPAR (suPAR) levels are increased in patients' plasma. Serum suPAR concentrations correlated with the number of GPI-negative neutrophils and were highest in patients who later develop thrombosis. In vitro, suPAR is released from PNH hematopoietic cells and from platelets upon activation, suggesting that these cells are the probable source of plasma suPAR in the absence of GPI anchor synthesis and trafficking of uPAR to the cell membrane. In vitro, the addition of recombinant suPAR results in a dose-dependent decrease in the activity of single-chain urokinase. We hypothesized that suPAR, prevents the interaction of urokinase with membrane-anchored uPAR on residual normal cells.


Subject(s)
Fibrinolysin/analysis , Hematopoietic Stem Cells/metabolism , Hemoglobinuria, Paroxysmal/blood , Receptors, Urokinase Plasminogen Activator/blood , Thrombosis/blood , Urokinase-Type Plasminogen Activator/blood , Blood Cells/metabolism , Female , Gene Expression Regulation/genetics , Hematopoiesis/genetics , Hemoglobinuria, Paroxysmal/complications , Hemoglobinuria, Paroxysmal/genetics , Hemolysis/genetics , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Protein Transport/genetics , Receptors, Urokinase Plasminogen Activator/genetics , Solubility , Thrombosis/etiology , Thrombosis/genetics , Urokinase-Type Plasminogen Activator/genetics
16.
J Clin Oncol ; 26(15): 2505-11, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18413642

ABSTRACT

PURPOSE: Marrow failure in some patients with myelodysplastic syndrome (MDS) responds to immunosuppressive treatment (IST), but long-term outcome after IST has not been described. We evaluated patients with MDS treated with IST at our institution to determine their clinical course compared with a comparable supportive care only group. PATIENTS AND METHODS: One hundred twenty-nine patients with MDS received IST with a median follow-up of 3.0 years (range, 0.03 to 11.3 years), using antithymocyte globulin (ATG) or cyclosporine (CsA) in combination or singly. Variables affecting response and survival were studied and outcomes were compared with those of 816 patients with MDS reported to the International Myelodysplasia Risk Analysis Workshop (IMRAW) who received only supportive care. RESULTS: Thirty-nine (30%) of 129 patients receiving IST responded either completely or partially: 18 (24%) of 74 patients responded to ATG, 20 (48%) of 42 patients responded to ATG plus CsA, and one (8%) of 13 patients responded to CsA. Thirty-one percent (12 of 39) of the responses were complete, resulting in transfusion independence and near-normal blood counts. In multivariate analysis, younger age was the most significant factor favoring response to therapy. Other favorable factors affecting response were HLA-DR15 positivity and combination ATG plus CsA treatment (P = .001 and P = .048, respectively). In multivariate analysis of the combined IMRAW and IST cohorts, younger age, treatment with IST, and intermediate or low International Prognostic Scoring System score significantly favored survival. CONCLUSION: IST produced significant improvement in the pancytopenia of a substantial proportion of patients with MDS and was associated with improved overall and progression-free survival, especially in younger individuals with lower-risk disease.


Subject(s)
Antilymphocyte Serum/therapeutic use , Cyclosporine/therapeutic use , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/mortality , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Male , Middle Aged , Myelodysplastic Syndromes/complications , Palliative Care , Prognosis , Survival , Treatment Outcome
17.
Semin Hematol ; 45(1): 39-48, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18179968

ABSTRACT

Patients with myelodysplastic syndrome (MDS) have intrinsic, usually acquired genetic defects in their hematopoietic stem cells, but some others exhibit T-cell-mediated inhibition of hematopoiesis and good responses to immunosuppression. In these cases, MDS shares a similar pathophysiology with aplastic anemia (AA). Here, we review the evidence supporting a role of the immune system in the pathophysiology of MDS and the results of clinical trials of immunosuppressive agents.


Subject(s)
Immune System/physiopathology , Immunosuppressive Agents/therapeutic use , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/therapy , Antilymphocyte Serum/therapeutic use , Cyclosporine/therapeutic use , Humans , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Vaccines/immunology
19.
Curr Hematol Malig Rep ; 3(1): 23-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-20425443

ABSTRACT

Myelodysplastic syndromes (MDS) are often associated with autoimmune processes that contribute to the cytopenias that characterize this disease group. Ten years ago, several investigators described improvement in the cytopenia of MDS following immunosuppressive treatments (IST) with antithymocyte globulin or cyclosporine. These findings have since been widely confirmed. The consensus is that about 30% of unselected transfusion-dependent patients with MDS given IST have sustained hematologic responses and can regain transfusion independence. Over the past decade, prognostic features have been defined that accurately identify the subset of patients most likely to benefit from IST. With increasing knowledge about the pathophysiology of myelosuppression in MDS, we are beginning to define the immunosuppressive agents and schedules with the greatest chance of causing hematologic improvement. Significantly, long-term follow-up indicates that IST responders have a better progression-free survival than comparable patients with MDS who do not receive IST.


Subject(s)
Antilymphocyte Serum/therapeutic use , Cyclosporine/therapeutic use , Immunosuppressive Agents/therapeutic use , Myelodysplastic Syndromes/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Clinical Trials as Topic , Humans , Immunization , Immunosuppression Therapy , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/therapy , Stem Cell Transplantation , Treatment Outcome
20.
Exp Hematol ; 35(12): 1777-81, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17697745

ABSTRACT

OBJECTIVE: To study if telomere length can be used as a surrogate marker for the mitotic history in normal and affected hematopoietic cells from patients with paroxysmal nocturnal hemoglobinuria (PNH). METHODS: The telomere length was measured by automated multicolor flow fluorescence in situ hybridization in glycosyl-phosphatidyl-inositol anchored protein (GPI)-negative and GPI-positive peripheral blood leukocytes. Eleven patients were studied, two with predominantly hemolytic PNH and nine with PNH associated with marrow failure. RESULTS: Telomere length in GPI-negative cells was significantly shorter than in GPI-positive cells of the same patient (p < 0.01, n = 11). The difference in telomere length (telomere length in GPI-positive minus telomere length in GPI-negative cells) correlated with the percentage of GPI-negative white blood cells. CONCLUSION: Our results support the hypothesis that telomere length is correlated to the replicative history of GPI-positive and GPI-negative cells and warrant further studies of telomere length in relation to disease progression in PNH.


Subject(s)
Hemoglobinuria, Paroxysmal/genetics , Telomere , Flow Cytometry , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...