Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
1.
Transplant Cell Ther ; 27(5): 430.e1-430.e7, 2021 05.
Article in English | MEDLINE | ID: mdl-33965187

ABSTRACT

Prolonged thrombocytopenia occurs in up to 37% of patients after hematopoietic stem cell transplantation (HSCT) and is associated with adverse prognosis and increased risk of bleeding. Eltrombopag, a thrombopoietin receptor agonist, can increase platelet counts in thrombocytopenic patients. We conducted a phase II study, adaptively randomizing patients at ≥35 days post-HSCT to receive placebo or eltrombopag at a platelet count ≤20,000/µL for 7 days or platelet transfusion-dependent and a neutrophil count ≥1500/µL. Sixty patients were randomized to eltrombopag (n = 42) or placebo (n = 18) and received at least 1 dose. Fifteen patients (36%) in the eltrombopag arm achieved a platelet count of ≥30,000/µL, compared with 5 patients (28%) in the placebo arm, with a posterior probability of 0.75. (The protocol required this probability to be >0.975 to declare a winner; thus, the results are inconclusive.) However, 9 patients (21%) in the eltrombopag arm achieved a platelet count of ≥50,000/µL, compared with no patients in the placebo arm (P = .046). The overall survival, progression-free survival, relapse rate, and nonrelapse mortality were similar in the 2 arms. In conclusion, compared with placebo, treatment with eltrombopag led to a higher percentage of patients achieving a platelet count of ≥50,000/µL in patients with persistent thrombocytopenia after HSCT.


Subject(s)
Neoplasm Recurrence, Local , Thrombocytopenia , Benzoates/therapeutic use , Humans , Hydrazines/therapeutic use , Pyrazoles , Thrombocytopenia/drug therapy
4.
Science ; 359(6371): 97-103, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29097493

ABSTRACT

Preclinical mouse models suggest that the gut microbiome modulates tumor response to checkpoint blockade immunotherapy; however, this has not been well-characterized in human cancer patients. Here we examined the oral and gut microbiome of melanoma patients undergoing anti-programmed cell death 1 protein (PD-1) immunotherapy (n = 112). Significant differences were observed in the diversity and composition of the patient gut microbiome of responders versus nonresponders. Analysis of patient fecal microbiome samples (n = 43, 30 responders, 13 nonresponders) showed significantly higher alpha diversity (P < 0.01) and relative abundance of bacteria of the Ruminococcaceae family (P < 0.01) in responding patients. Metagenomic studies revealed functional differences in gut bacteria in responders, including enrichment of anabolic pathways. Immune profiling suggested enhanced systemic and antitumor immunity in responding patients with a favorable gut microbiome as well as in germ-free mice receiving fecal transplants from responding patients. Together, these data have important implications for the treatment of melanoma patients with immune checkpoint inhibitors.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunotherapy , Melanoma/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/therapy , Animals , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/genetics , Humans , Melanoma/immunology , Metagenome , Mice , Skin Neoplasms/immunology
5.
Leukemia ; 32(4): 960-970, 2018 04.
Article in English | MEDLINE | ID: mdl-28972595

ABSTRACT

Ibrutinib, a covalent inhibitor of Bruton Tyrosine Kinase (BTK), is approved for treatment of patients with relapsed/refractory or treatment-naïve chronic lymphocytic leukemia (CLL). Besides directly inhibiting BTK, ibrutinib possesses immunomodulatory properties through targeting multiple signaling pathways. Understanding how this ancillary property of ibrutinib modifies the CLL microenvironment is crucial for further exploration of immune responses in this disease and devising future combination therapies. Here, we investigated the mechanisms underlying the immunomodulatory properties of ibrutinib. In peripheral blood samples collected prospectively from CLL patients treated with ibrutinib monotherapy, we observed selective and durable downregulation of PD-L1 on CLL cells by 3 months post-treatment. Further analysis showed that this effect was mediated through inhibition of the constitutively active signal transducer and activator of transcription 3 (STAT3) in CLL cells. Similar downregulation of PD-1 was observed in CD4+ and CD8+ T cells. We also demonstrated reduced interleukin (IL)-10 production by CLL cells in patients receiving ibrutinib, which was also linked to suppression of STAT3 phosphorylation. Taken together, these findings provide a mechanistic basis for immunomodulation by ibrutinib through inhibition of the STAT3 pathway, critical in inducing and sustaining tumor immune tolerance. The data also merit testing of combination treatments combining ibrutinib with agents capable of augmenting its immunomodulatory effects.


Subject(s)
B-Lymphocytes, Regulatory/drug effects , B7-H1 Antigen/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Programmed Cell Death 1 Receptor/metabolism , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , STAT3 Transcription Factor/metabolism , Tumor Microenvironment/drug effects , Adenine/analogs & derivatives , Aged , B-Lymphocytes, Regulatory/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Female , Humans , Immune Tolerance/drug effects , Immunosuppressive Agents/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Middle Aged , Piperidines , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects
6.
Bone Marrow Transplant ; 52(4): 580-587, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27991894

ABSTRACT

We hypothesized that IV busulfan (Bu) dosing could be safely intensified through pharmacokinetic (PK-) dose guidance to minimize the inter-patient variability in systemic exposure (SE) associated with body-sized dosing, and that this should improve outcome of AML/MDS patients undergoing allogeneic stem cell transplantation. To test this hypothesis, we treated 218 patients (median age 50.7 years, male/female 50/50%) with fludarabine 40 mg/m2 once daily x4, each dose followed by IV Bu, randomized to 130 mg/m2 (N=107) or PK-guided to average daily SE, AUC of 6000 µM min (N=111), stratified for remission status and allo-grafting from HLA-matched donors. Toxicity and GvHD rates in the groups were similar; the risk of relapse or treatment-related mortality remained higher in the fixed-dose group throughout the 80-month observation period. Further, PK-guidance yielded safer disease control, leading to improved overall and PFS, most prominently in MDS patients and in AML patients not in remission at allogeneic stem cell transplantation. We conclude that AML/MDS patients receiving pretransplant conditioning treatment with our 4-day regimen may benefit significantly from PK-guided Bu dosing. This could be considered an alternative to fixed-dose delivery since it provides the benefit of precise dose delivery to a predetermined SE without increasing risk(s) of serious toxicity and/or GvHD.


Subject(s)
Busulfan/administration & dosage , Drug Monitoring/methods , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/therapy , Transplantation Conditioning/methods , Transplantation, Homologous/methods , Vidarabine/analogs & derivatives , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/toxicity , Busulfan/pharmacokinetics , Busulfan/toxicity , Female , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/mortality , Humans , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Myelodysplastic Syndromes/mortality , Recurrence , Survival Analysis , Transplantation Conditioning/mortality , Transplantation, Homologous/mortality , Treatment Outcome , Vidarabine/administration & dosage
8.
Am J Transplant ; 16(5): 1492-502, 2016 05.
Article in English | MEDLINE | ID: mdl-26608093

ABSTRACT

Nephropathy due to BK virus (BKV) infection is an evolving challenge in patients undergoing hematopoietic stem cell transplantation (HSCT). We hypothesized that BKV infection was a marker of kidney function decline and a poor prognostic factor in HSCT recipients who experience this complication. In this retrospective study, we analyzed all patients who underwent their first allogeneic HSCT at our institution between 2004 and 2012. We evaluated the incidence of persistent kidney function decline, which was defined as a confirmed reduction in estimated glomerular filtration rate of at least 25% from baseline using the Chronic Kidney Disease Epidemiology equation. Cox proportional hazard regression was used to model the cause-specific hazard of kidney function decline, and the Fine-Gray method was used to account for the competing risks of death. Among 2477 recipients of a first allogeneic HSCT, BK viruria was detected in 25% (n = 629) and kidney function decline in 944 (38.1%). On multivariate analysis, after adjusting for age, sex, acute graft-versus-host disease (GVHD), chronic GVHD, preparative conditioning regimen, and graft source, BK viruria remained a significant risk factor for kidney function decline (p < 0.001). In addition, patients with BKV infection and kidney function decline experienced worse overall survival. After allogeneic HSCT, BKV infection was strongly and independently associated with subsequent kidney function decline and worse patient survival after HSCT.


Subject(s)
BK Virus/pathogenicity , Graft vs Host Disease/mortality , Hematologic Diseases/mortality , Hematopoietic Stem Cell Transplantation/adverse effects , Kidney Diseases/mortality , Polyomavirus Infections/mortality , Tumor Virus Infections/mortality , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Follow-Up Studies , Glomerular Filtration Rate , Graft vs Host Disease/etiology , Hematologic Diseases/complications , Hematologic Diseases/therapy , Humans , Infant , Infant, Newborn , Kidney Diseases/virology , Kidney Function Tests , Male , Middle Aged , Polyomavirus Infections/virology , Prognosis , Retrospective Studies , Risk Factors , Survival Rate , Transplantation, Homologous , Tumor Virus Infections/virology , Young Adult
9.
Leukemia ; 30(4): 800-11, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26621337

ABSTRACT

Natural killer (NK) cells are key components of the innate immune system, providing potent antitumor immunity. Here, we show that the tumor growth factor-ß (TGF-ß)/SMAD signaling pathway is an important mechanism for NK cell immune evasion in childhood B-acute lymphoblastic leukemia (ALL). We characterized NK cells in 50 consecutive children with B-ALL at diagnosis, end induction and during maintenance therapy compared with age-matched controls. ALL-NK cells at diagnosis had an inhibitory phenotype associated with impaired function, most notably interferon-γ production and cytotoxicity. By maintenance therapy, these phenotypic and functional abnormalities partially normalized; however, cytotoxicity against autologous blasts remained impaired. We identified ALL-derived TGF-ß1 to be an important mediator of leukemia-induced NK cell dysfunction. The TGF-ß/SMAD signaling pathway was constitutively activated in ALL-NK cells at diagnosis and end induction when compared with healthy controls and patients during maintenance therapy. Culture of ALL blasts with healthy NK cells induced NK dysfunction and an inhibitory phenotype, mediated by activation of the TGF-ß/SMAD signaling pathway, and abrogated by blocking TGF-ß. These data indicate that by regulating the TGF-ß/SMAD pathway, ALL blasts induce changes in NK cells to evade innate immune surveillance, thus highlighting the importance of developing novel therapies to target this inhibitory pathway and restore antileukemic cytotoxicity.


Subject(s)
Cytotoxicity, Immunologic/immunology , Immune Evasion/immunology , Killer Cells, Natural/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Female , Flow Cytometry , Follow-Up Studies , Humans , Infant , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Male , Phosphorylation , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction , Tumor Cells, Cultured , Tumor Microenvironment/immunology
10.
Ann Oncol ; 26(12): 2490-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26416896

ABSTRACT

BACKGROUND: Cutaneous T-cell lymphomas (CTCLs) and its common variants mycosis fungoides (MF) and leukemic Sézary syndrome (SS) are rare extranodal non-Hodgkin's lymphomas. Patients who present with advanced disease and large-cell transformation (LCT) are incurable with standard treatments. In this article, we report the largest single-center experience with allogeneic stem-cell transplantation (SCT) for advanced CTCL. PATIENTS AND METHODS: This is a prospective case series of 47 CTCL patients who underwent allogeneic SCT after failure of standard therapy between July 2001 and September 2013. The Kaplan-Meier method was used to estimate overall survival (OS) and progression-free survival (PFS) curves. The method of Fine and Gray was used to fit regression models to the same covariates for these cumulative incidence data. RESULTS: The Kaplan-Meier estimates of OS and PFS at 4 years were 51% and 26%, respectively. There was no statistical difference in the OS in patients who had MF alone, SS, MF with LCT, or SS with LCT. PFS at 4 years was superior in patients who had SS versus those who did not (52.4% versus 9.9%; P = 0.02). The cumulative incidences of grade 2-4 acute graft-versus-host disease (GVHD) and chronic GVHD were 40% and 28%, respectively. The cumulative nonrelapse mortality rate was 16.7% at 2 years. CONCLUSION: Allogeneic SCT may result in long-term remissions in a subset of patients with advanced CTCL. Although post-SCT relapse rates are high, many patients respond to immunomodulation and achieve durable remissions. CLINICALTRIALSGOV: NCT00506129.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Lymphoma, T-Cell, Cutaneous/diagnosis , Lymphoma, T-Cell, Cutaneous/therapy , Skin Neoplasms/diagnosis , Skin Neoplasms/therapy , Adult , Aged , Disease-Free Survival , Female , Follow-Up Studies , Hematopoietic Stem Cell Transplantation/trends , Humans , Male , Middle Aged , Prospective Studies , Transplantation, Homologous/methods , Transplantation, Homologous/trends , Young Adult
11.
Bone Marrow Transplant ; 50 Suppl 2: S55-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26039209

ABSTRACT

We are entering a very exciting era in umbilical cord blood transplantation (UCBT), where many of the associated formidable challenges may become treatable by ex vivo graft manipulation and/or adoptive immunotherapy utilizing specific cellular products. We envisage the use of double UCBT rather than single UCBT for most patients; this allows for greater ability to treat larger patients as well as to manipulate the graft. Ex vivo expansion and/or fucosylation of one cord will achieve more rapid engraftment, minimize the period of neutropenia and also give certainty that the other cord will provide long-term engraftment/immune reconstitution. The non-expanded (and future dominant) cord could be chosen for characteristics such as better HLA matching to minimize GvHD, or larger cell counts to enable part of the unit to be utilized for the development of specific cellular therapies such as the production of virus-specific T-cells or chimeric-antigen receptor T-cells which are reviewed in this study.


Subject(s)
Cord Blood Stem Cell Transplantation , Graft Enhancement, Immunologic/methods , Graft Survival , Graft vs Host Disease/prevention & control , Tissue Engineering/methods , Humans
13.
Cell Death Differ ; 22(1): 96-107, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25168239

ABSTRACT

Natural killer cells (NK) are important effectors of anti-tumor immunity, activated either by the downregulation of HLA-I molecules on tumor cells and/or the interaction of NK-activating receptors with ligands that are overexpressed on target cells upon tumor transformation (including NKG2D and NKP30). NK kill target cells by the vesicular delivery of cytolytic molecules such as Granzyme-B and Granulysin activating different cell death pathways, which can be Caspase-3 dependent or Caspase-3 independent. Multiple myeloma (MM) remains an incurable neoplastic plasma-cell disorder. However, we previously reported the encouraging observation that cord blood-derived NK (CB-NK), a new source of NK, showed anti-tumor activity in an in vivo murine model of MM and confirmed a correlation between high levels of NKG2D expression by MM cells and increased efficacy of CB-NK in reducing tumor burden. We aimed to characterize the mechanism of CB-NK-mediated cytotoxicity against MM cells. We show a Caspase-3- and Granzyme-B-independent cell death, and we reveal a mechanism of transmissible cell death between cells, which involves lipid-protein vesicle transfer from CB-NK to MM cells. These vesicles are secondarily transferred from recipient MM cells to neighboring MM cells amplifying the initial CB-NK cytotoxicity achieved. This indirect cytotoxicity involves the transfer of NKG2D and NKP30 and leads to lysosomal cell death and decreased levels of reactive oxygen species in MM cells. These findings suggest a novel and unique mechanism of CB-NK cytotoxicity against MM cells and highlight the importance of lipids and lipid transfer in this process. Further, these data provide a rationale for the development of CB-NK-based cellular therapies in the treatment of MM.


Subject(s)
Immunity, Cellular , Killer Cells, Natural/immunology , Multiple Myeloma/immunology , Secretory Vesicles/immunology , Caspase 3/immunology , Female , Fetal Blood , Granzymes/immunology , Humans , K562 Cells , Killer Cells, Natural/pathology , Male , Multiple Myeloma/pathology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Natural Cytotoxicity Triggering Receptor 3/immunology , Reactive Oxygen Species/immunology
14.
Bone Marrow Transplant ; 49(6): 793-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24710569

ABSTRACT

Exact mechanism of action of umbilical cord blood (CB)-derived regulatory T cells (Tregs) in the prevention of GVHD remains unclear. On the basis of selective overexpression of peptidase inhibitor 16 in CB Tregs, we explored the related p53 pathway, which has been shown to negatively regulate miR15a/16 expression. Significantly lower levels of miR15a/16 were observed in CB Tregs when compared with conventional CB T cells (Tcons). In a xenogeneic GVHD mouse model, lower levels of miR15a/16 were also found in Treg recipients, which correlated with a better GVHD score. Forced overexpression of miR15a/16 in CB Tregs led to inhibition of FOXP3 and CTLA4 expression and partial reversal of Treg-mediated suppression in an allogeneic mixed lymphocyte reaction that correlated with the reversal of FOXP3 demethylation in CB Tregs. On the other hand, miR15a/16 knockdown in CB Tcons led to expression of FOXP3 and CTLA4 and suppression of allogeneic lymphocyte proliferation. Using a luciferase-based mutagenesis assay, FOXP3 was determined to be a direct target of miR15a and miR16. We propose that miR15a/16 has an important role in mediating the suppressive function of CB Tregs and these microRNAs may have a 'toggle-switch' function in Treg/Tcon plasticity.


Subject(s)
Fetal Blood/immunology , Fetal Blood/metabolism , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , MicroRNAs/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , CTLA-4 Antigen/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Fetal Blood/cytology , Forkhead Transcription Factors/immunology , Gene Expression , Gene Knockdown Techniques , Genes, p53 , Glycoproteins/genetics , Glycoproteins/metabolism , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Heterografts , Humans , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred NOD , Mice, SCID , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Mutagenesis, Site-Directed , T-Lymphocytes, Regulatory/cytology
15.
Leukemia ; 27(7): 1538-47, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23528871

ABSTRACT

Adoptive immunotherapy with ex vivo expanded T cells is a promising approach to prevent or treat leukemia. Myeloid leukemias express tumor-associated antigens (TAA) that induce antigen-specific cytotoxic T lymphocyte (CTL) responses in healthy individuals. We explored the feasibility of generating TAA-specific CTLs from stem cell donors of patients with myeloid leukemia to enhance the graft-versus-leukemia effect after stem cell transplantation. CTL lines were manufactured from peripheral blood of 10 healthy donors by stimulation with 15mer peptide libraries of five TAA (proteinase 3 (Pr3), preferentially expressed antigen in melanoma, Wilms tumor gene 1 (WT1), human neutrophil elastase (NE) and melanoma-associated antigen A3) known to be expressed in myeloid leukemias. All CTL lines responded to the mix of five TAA and were multi-specific as assessed by interferon-γ enzyme-linked immunospot. Although donors showed individual patterns of antigen recognition, all responded comparably to the TAAmix. Immunogenic peptides of WT1, Pr3 or NE could be identified by epitope mapping in all donor CTL lines. In vitro experiments showed recognition of partially human leukocyte antigen (HLA)-matched myeloid leukemia blasts. These findings support the development of a single clinical grade multi-tumor antigen-specific T-cell product from the stem cell source, capable of broad reactivity against myeloid malignancies for use in donor-recipient pairs without limitation to a certain HLA-type.


Subject(s)
Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid/therapy , Membrane Glycoproteins/immunology , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/immunology , Cell Line , Dendritic Cells/cytology , Dendritic Cells/immunology , Epitope Mapping , Histocompatibility Testing , Humans , Immunophenotyping , Leukemia, Myeloid/immunology , Leukocyte Elastase/immunology , Myeloblastin/immunology , Peptide Fragments/immunology , Recurrence , Tissue Donors , Transplantation, Homologous , WT1 Proteins/immunology
16.
Bone Marrow Transplant ; 48(1): 26-31, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22732703

ABSTRACT

We investigated the administration of i.v. BU combined with melphalan (Mel) in patients with ALL undergoing allogeneic hematopoietic SCT. Forty-seven patients with a median age of 33 years (range 20-61) received a matched sibling (n=27) or matched unrelated donor transplant (n=20) for ALL in first CR (n=26), second CR (n=13), or with more advanced disease (n=8). BU was infused daily for 4 days, either at a fixed dose of 130 mg/m² (5 patients) or using pharmacokinetic (PK) dose adjustment (42 patients), to target an average daily area-under-the-curve (AUC) of 5000 µmol/min, determined by a test dose of i.v. BU at 32 mg/m². This was followed by a rest day, then two daily doses of Mel at 70 mg/m². Stem cells were infused on the following day. The 2-year OS, PFS and non-relapse mortality (NRM) rates were 35% (95% confidence interval (CI), 23-51%), 31% (95% CI, 21-48%) and 37% (95% CI, 23-50%), respectively. Acute NRM at 100 days was favorable at 12% (95% CI, 5-24%); however, the 2-year NRM was significantly higher for patients older than 40 years, 58% vs 20%, mainly due to GVHD.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Busulfan/therapeutic use , Hematopoietic Stem Cell Transplantation/adverse effects , Melphalan/therapeutic use , Myeloablative Agonists/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Transplantation Conditioning , Adult , Age Factors , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Busulfan/administration & dosage , Busulfan/adverse effects , Drug Therapy, Combination/adverse effects , Female , Follow-Up Studies , Graft vs Host Disease/epidemiology , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Humans , Incidence , Infusions, Intravenous , Male , Melphalan/adverse effects , Middle Aged , Myeloablative Agonists/administration & dosage , Myeloablative Agonists/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/prevention & control , Secondary Prevention , Survival Analysis , Texas , Transplantation Conditioning/adverse effects , Transplantation, Homologous , Young Adult
17.
Bone Marrow Transplant ; 48(5): 666-70, 2013 May.
Article in English | MEDLINE | ID: mdl-23085830

ABSTRACT

For patients with ALL who relapse following allo-SCT, only a second SCT provides a realistic chance for long-term disease remission. We retrospectively analyzed the outcomes of 31 patients with relapsed ALL after a prior allo-SCT, who received a second SCT (SCT2) at our center. With a median follow-up of 3 years, 1- and 3-year PFS was 23 and 11% and 1- and 3 year OS rates were 23 and 11%. Twelve patients (39%) were transplanted with active disease, of whom 75% attained a CR. We found a significant relationship between the time to treatment failure following first allograft (SCT1) and PFS following SCT2 (P=0.02, hazard ratio=0.93/month). In summary, a second transplant remains a potential treatment option for achieving response in a highly refractory patient population. While long-term survival is limited, a significant proportion of patients remains disease-free for up to 1 year following SCT2, providing a window of time to administer preventive interventions. Notably, our four long-term survivors received novel therapies with their second transplant underscoring the need for a fundamental change in the methods for SCT2 to improve outcome.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/surgery , Adolescent , Adult , Child , Female , Humans , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recurrence , Retrospective Studies , Treatment Outcome , Young Adult
18.
Biol Blood Marrow Transplant ; 18(12): 1819-26, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22750645

ABSTRACT

We investigated the safety and early disease control data for i.v. busulfan (Bu) in combination with clofarabine (Clo) in patients with acute lymphoblastic leukemia undergoing allogeneic hematopoietic stem cell transplantation (SCT). Fifty-one patients (median age, 36 years; range, 20-64 years) received a matched sibling (n = 24), syngeneic (n = 2), or matched unrelated donor transplant (n = 25) for acute lymphoblastic leukemia in first complete remission (n = 30), second complete remission (n = 13), or active disease (n = 8). More than one-half of the patients had a high-risk cytogenetic profile, as defined by the presence of t(9;22) (n = 17), t(4;11) (n = 3), or complex cytogenetics (n = 7). Clo 40 mg/m(2) was given once daily, with each dose followed by pharmacokinetically dosed Bu infused over 3 hours daily for 4 days, followed by hematopoietic SCT 2 days later. The Bu dose was based on drug clearance, as determined by the patient's response to a 32-mg/m(2) Bu test dose given 48 hours before the high-dose regimen. The target daily area under the receiver-operating characteristic curve was 5500 µM/min for patients age <60 years and 4000 µM/min for those age ≥60 years. The regimen was well tolerated, with a 100-day nonrelapse mortality rate of 6%. With a median follow-up of 14 months among surviving patients (range, 6-28 months), the 1-year overall survival, disease-free survival, and nonrelapse mortality rates were 67% (95% confidence interval [CI], 55%-83%), 54% (95% CI, 41%-71%), and 32% (95% CI, 16%-45%), respectively. For patients undergoing SCT in first remission, these respective rates were 74%, 64%, and 25%. Our data indicate that the combination of Clo and Bu provides effective disease control while maintaining a favorable safety profile.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/surgery , Adenine Nucleotides/administration & dosage , Adenine Nucleotides/adverse effects , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Arabinonucleosides/administration & dosage , Arabinonucleosides/adverse effects , Busulfan/administration & dosage , Busulfan/adverse effects , Clofarabine , Female , Humans , Male , Middle Aged , Prospective Studies , Transplantation Conditioning/methods , Young Adult
19.
Oncogene ; 31(37): 4085-94, 2012 Sep 13.
Article in English | MEDLINE | ID: mdl-22249254

ABSTRACT

The myelodysplastic syndromes (MDSs) comprise a group of disorders characterized by multistage progression from cytopenias to acute myeloid leukemia (AML). They display exaggerated apoptosis in early stages, but lose this behavior during evolution to AML. The molecular basis for loss of apoptosis is unknown. To investigate this critical event, we analyzed phosphatidylinositol (PI) 3'kinase signaling, implicated as a critical pathway of cell survival control in epithelial and hematological malignancies. PI 3'kinase activates Akt through its production of 3' phosphoinositides. In turn, the phosphoinositides are dephosphorylated by two lipid phosphatases, PTEN and SHIP-1, in myeloid cells. We studied primary MDS-enriched bone marrow cells and bone marrow sections by western blotting, immunohistochemistry, immunocytochemistry and quantitative PCR for components of the SHIP/PTEN/PI 3'kinase signaling circuit. We reported constitutively activated Akt, variable levels of PTEN and uniformly decreased SHIP-1 expression in MDS progenitor cells. Overexpression of SHIP-1, but not the phosphatase-deficient form, inhibited myeloid leukemic growth. Levels of microRNA (miR)-210 and miR-155 transcripts, which target SHIP-1, were increased in CD34(+) MDS cells compared with their normal counterparts. Direct binding of miR-210 to the 3' untranslated region of SHIP-1 was confirmed by luciferase reporter assay. Transfection of a myeloid cell line with miR-210 resulted in loss of SHIP-1 protein expression. These data suggest that miR-155 and miR-210/SHIP-1/Akt pathways could serve as clinical biomarkers for disease progression, and that miR-155 and miR-210 might serve as novel therapeutic targets in MDS.


Subject(s)
MicroRNAs/metabolism , Myelodysplastic Syndromes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Apoptosis/genetics , Bone Marrow Cells/metabolism , Cell Line, Tumor , Humans , Inositol Polyphosphate 5-Phosphatases , Leukemia, Myeloid, Acute/metabolism , Myelodysplastic Syndromes/genetics , Myeloid Cells/metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/deficiency , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...