Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
3.
J Clin Oncol ; 35(15): 1678-1685, 2017 May 20.
Article in English | MEDLINE | ID: mdl-28368672

ABSTRACT

Purpose Higher doses of the anthracycline daunorubicin during induction therapy for acute myeloid leukemia (AML) have been shown to improve remission rates and survival. We hypothesized that improvements in outcomes in adult AML may be further achieved by increased anthracycline dose during consolidation therapy. Patients and Methods Patients with AML in complete remission after induction therapy were randomly assigned to receive two cycles of consolidation therapy with cytarabine 100 mg/m2 daily for 5 days, etoposide 75 mg/m2 daily for 5 days, and idarubicin 9 mg/m2 daily for either 2 or 3 days (standard and intensive arms, respectively). The primary end point was leukemia-free survival (LFS). Results Two hundred ninety-three patients 16 to 60 years of age, excluding those with core binding factor AML and acute promyelocytic leukemia, were randomly assigned to treatment groups (146 to the standard arm and 147 to the intensive arm). Both groups were balanced for age, karyotypic risk, and FLT3-internal tandem duplication and NPM1 gene mutations. One hundred twenty patients in the standard arm (82%) and 95 patients in the intensive arm (65%) completed planned consolidation ( P < .001). Durations of severe neutropenia and thrombocytopenia were prolonged in the intensive arm, but there were no differences in serious nonhematological toxicities. With a median follow-up of 5.3 years (range, 0.6 to 9.9 years), there was a statistically significant improvement in LFS in the intensive arm compared with the standard arm (3-year LFS, 47% [95% CI, 40% to 56%] v 35% [95% CI, 28% to 44%]; P = .045). At 5 years, the overall survival rate was 57% in the intensive arm and 47% in the standard arm ( P = .092). There was no evidence of selective benefit of intensive consolidation within the cytogenetic or FLT3-internal tandem duplication and NPM1 gene mutation subgroups. Conclusion An increased cumulative dose of idarubicin during consolidation therapy for adult AML resulted in improved LFS, without increased nonhematologic toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Adolescent , Adult , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Consolidation Chemotherapy , Cytarabine/administration & dosage , Cytarabine/adverse effects , Disease-Free Survival , Dose-Response Relationship, Drug , Etoposide/administration & dosage , Etoposide/adverse effects , Female , Humans , Idarubicin/administration & dosage , Idarubicin/adverse effects , Male , Middle Aged , Nucleophosmin , Survival Rate , Young Adult
4.
Transplantation ; 101(11): 2695-2704, 2017 11.
Article in English | MEDLINE | ID: mdl-28319565

ABSTRACT

BACKGROUND: Most clinical allogeneic hemopoietic cell transplants (alloHCT) are now performed using reduced-intensity conditioning (RIC) instead of myeloablative conditioning (MAC); however, the biology underlying this treatment remains incompletely understood. METHODS: We investigated a murine model of major histocompatibility complex-matched multiple minor histocompatibility antigen-mismatched alloHCT using bone marrow (BM) cells and splenocytes from B6 (H-2) donor mice transplanted into BALB.B (H-2) recipients after RIC with fludarabine of 100 mg/kg per day for 5 days, cyclophosphamide of 60 mg/kg per day for 2 days, and total body irradiation (TBI). RESULTS: The lowest TBI dose capable of achieving complete donor chimerism in this mouse strain combination was 325 cGy given as a single fraction. Mice that underwent RIC had a reduced incidence and delayed onset of graft-versus-host disease (GVHD) and significantly prolonged survival compared with MAC-transplanted recipients (TBI of 850 cGy plus cyclophosphamide of 60 mg/kg per day for 2 days). Compared with syngeneic controls, RIC mice with GVHD showed evidence of BM suppression, have anemia, reduced BM cellularity, and showed profound reduction in BM B cell lymphopoiesis associated with damage to the endosteal BM niche. This was associated with an increase in BM CD8 effector T cells in RIC mice and elevated blood and BM plasma levels of T helper1 cytokines. Increasing doses of splenocytes resulted in increased incidence of GVHD in RIC mice. CONCLUSIONS: We demonstrate that the BM is a major target organ of GVHD in an informative clinically relevant RIC mouse major histocompatibility complex-matched alloHCT model by a process that seems to be driven by CD8 effector T cells.


Subject(s)
Bone Marrow Diseases/prevention & control , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Histocompatibility , Major Histocompatibility Complex , Transplantation Conditioning/methods , Animals , Bone Marrow Diseases/blood , Bone Marrow Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Cyclophosphamide/administration & dosage , Disease Models, Animal , Drug Administration Schedule , Female , Graft vs Host Disease/blood , Graft vs Host Disease/immunology , Mice, Inbred BALB C , Myeloablative Agonists/administration & dosage , Spleen/immunology , Time Factors , Transplantation Chimera/immunology , Transplantation Conditioning/adverse effects , Transplantation, Homologous , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Whole-Body Irradiation/adverse effects
5.
J Immunol ; 197(12): 4613-4625, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27837105

ABSTRACT

CD83 is a member of the Ig gene superfamily, first identified in activated lymphocytes. Since then, CD83 has become an important marker for defining activated human dendritic cells (DC). Several potential CD83 mRNA isoforms have been described, including a soluble form detected in human serum, which may have an immunosuppressive function. To further understand the biology of CD83, we examined its expression in different human immune cell types before and after activation using a panel of mouse and human anti-human CD83 mAb. The mouse anti-human CD83 mAbs, HB15a and HB15e, and the human anti-human CD83 mAb, 3C12C, were selected to examine cytoplasmic and surface CD83 expression, based on their different binding characteristics. Glycosylation of CD83, the CD83 mRNA isoforms, and soluble CD83 released differed among blood DC, monocytes, and monocyte-derived DC, and other immune cell types. A small T cell population expressing surface CD83 was identified upon T cell stimulation and during allogeneic MLR. This subpopulation appeared specifically during viral Ag challenge. We did not observe human CD83 on unstimulated human natural regulatory T cells (Treg), in contrast to reports describing expression of CD83 on mouse Treg. CD83 expression was increased on CD4+, CD8+ T, and Treg cells in association with clinical acute graft-versus-host disease in allogeneic hematopoietic cell transplant recipients. The differential expression and function of CD83 on human immune cells reveal potential new roles for this molecule as a target of therapeutic manipulation in transplantation, inflammation, and autoimmune diseases.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Immunoglobulins/metabolism , Membrane Glycoproteins/metabolism , Monocytes/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Antigens, CD/genetics , Antigens, Viral/immunology , Cells, Cultured , Glycosylation , Humans , Immunoglobulins/genetics , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mice , RNA Isoforms/genetics , RNA, Messenger/genetics , Transplantation, Homologous , CD83 Antigen
6.
Oncoimmunology ; 5(6): e1168555, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27471645

ABSTRACT

There are numerous transcriptional, proteomic and functional differences between monocyte-derived dendritic cells (Mo-DC) and primary blood dendritic cells (BDC). The CMRF-56 monoclonal antibody (mAb) recognizes a cell surface marker, which is upregulated on BDC following overnight culture. Given its unique ability to select a heterogeneous population of BDC, we engineered a human chimeric (h)CMRF-56 IgG4 mAb to isolate primary BDC for potential therapeutic vaccination. The ability to select multiple primary BDC subsets from patients and load them with in vitro transcribed (IVT) mRNA encoding tumor antigen might circumvent the issues limiting the efficacy of Mo-DC. After optimizing and validating the purification of hCMRF-56(+) BDC, we showed that transfection of hCMRF-56(+) BDC with mRNA resulted in efficient mRNA translation and antigen presentation by myeloid BDC subsets, while preserving superior DC functions compared to Mo-DC. Immune selected and transfected hCMRF-56(+) BDC migrated very efficiently in vitro and as effectively as cytokine matured Mo-DC in vivo. Compared to Mo-DC, hCMRF-56(+) BDC transfected with influenza matrix protein M1 displayed superior MHC peptide presentation and generated potent antigen specific CD8(+) T-cell recall responses, while Wilms tumor 1 (WT1) transfected CMRF-56(+) BDC generated effective primary autologous cytotoxic T-cell responses. The ability of the combined DC subsets within hCMRF-56(+) BDC to present mRNA delivered tumor antigens merits phase I evaluation as a reproducible generic platform for the next generation of active DC immune therapies.

7.
Contemp Clin Trials Commun ; 4: 9-13, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-29736466

ABSTRACT

An intensive induction regimen, consisting of idarubicin and high dose cytarabine, was assessed in 19 adult patients, median age 44 years, with newly diagnosed precursor-B acute lymphoblastic leukemia (ALL). Patients achieving a complete response (CR) were given an attenuated consolidation course. The primary endpoints were induction death rate and incidence of serious non-hematological toxicity. Grades 3-4 diarrhoea occurred in 47% of patients during induction. Two patients (11%) died during induction therapy, and 2 were withdrawn due to resistant disease or prolonged marrow hypoplasia. Fifteen patients achieved CR (79%), but levels of minimal residual disease (MRD) after induction were comparable with those previously observed using a modified pediatric protocol. Overall survival at 5 years was 36.8% while leukemia-free survival was 44.1%. An intensive AML protocol used in adults with ALL resulted in substantial toxicity and provided similar levels of cytoreduction to conventional ALL protocols, without improving long-term outcomes.

8.
Biol Blood Marrow Transplant ; 21(5): 941-4, 2015 May.
Article in English | MEDLINE | ID: mdl-25636379

ABSTRACT

High-dose cyclophosphamide given early after allogeneic hemopoietic cell transplantation has been shown to be effective prophylaxis against graft-versus-host disease (GVHD) in the setting of HLA-matched myeloablative bone marrow grafts, allowing avoidance of long-term immunosuppression with calcineurin inhibitors in some patients. Whether this approach is feasible using granulocyte colony-stimulating factor (G-CSF)-mobilized peripheral blood stem cell grafts is unknown. We conducted an exploratory phase 2 trial of cyclophosphamide given at 50 mg/kg i.v. on days 3 and 4 after transplantation as sole GVHD prophylaxis in recipients of G-CSF-mobilized peripheral blood stem cell grafts from HLA-matched related or unrelated donors after reduced-intensity conditioning therapy with fludarabine, carmustine, and melphalan. Five patients, ages 52 to 67 years, with high-risk hematologic malignancies were enrolled. Four of the 5 developed severe acute GVHD of grades 3 to 4, requiring treatment with methylprednisolone and cyclosporine; 3 were steroid refractory and were given salvage therapy. One of these 4 patients died of hepatic GVHD, one died of sepsis, and 2 survived. We conclude that post-transplantation cyclophosphamide is inadequate as sole GVHD prophylaxis in the context of peripheral blood reduced-intensity conditioning transplantations from HLA-matched donors. This trial is registered at ACTRN12613001154796.


Subject(s)
Cyclophosphamide/administration & dosage , Graft vs Host Disease/mortality , HLA Antigens , Hematologic Neoplasms , Peripheral Blood Stem Cell Transplantation , Transplantation Conditioning , Acute Disease , Aged , Female , Graft vs Host Disease/etiology , Hematologic Neoplasms/mortality , Hematologic Neoplasms/therapy , Histocompatibility Testing , Humans , Male , Middle Aged , Prospective Studies , Severity of Illness Index
9.
Leuk Lymphoma ; 56(5): 1406-15, 2015 May.
Article in English | MEDLINE | ID: mdl-25248882

ABSTRACT

Acute myeloid leukemia (AML) blasts express high levels of interlekin-3 (IL-3) receptor-α (CD123). CSL360 is a recombinant, chimeric immunoglobulin G1 (IgG1), anti-CD123 monoclonal antibody (MoAb) that neutralizes IL-3 and demonstrates anti-leukemic activity in vitro. This phase 1 study assessed safety, pharmacokinetics and bioactivity of weekly intravenous CSL360 for 12 weeks in 40 patients with advanced AML across five dose levels (0.1-10.0 mg/kg). Other than mild infusion reactions, CSL360 was well tolerated. The maximal tolerated dose was not reached. The half-life was 4.9 days, and the area under the curve (AUC) and maximum concentration (Cmax) increased proportionally with dose. Doses ≥ 3.0 mg/kg resulted in complete saturation and down-regulation of CD123 and abolition of ex vivo proliferative responsiveness to IL-3, indicating adequate blockade of IL-3 signaling. Two patients responded, with one remaining in complete remission after 17 doses. CSL360 bound CD123 specifically, but did not induce anti-leukemic activity in most patients. While safe, MoAb blockade of CD123 function is insufficient as a therapeutic strategy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Leukemic , Humans , Interleukin-3/metabolism , Interleukin-3 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-3 Receptor alpha Subunit/genetics , Interleukin-3 Receptor alpha Subunit/metabolism , Male , Middle Aged , Recurrence , Treatment Outcome , Young Adult
10.
Oncotarget ; 5(21): 10460-72, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25361005

ABSTRACT

The major regulators of human acute lymphoblastic leukemia (ALL) cell growth and survival mediate their effects through the phosphoinositide 3-kinase (PI-3K)/mammalian target of rapamycin (mTOR) pathway. We have shown that the mTOR inhibitor everolimus extended survival in a non-obese diabetic/severe combined immune-deficient (NOD/SCID) mouse xenograft model of human ALL. Since PI-3K has mTOR dependent and independent functions we examined the effect of the dual PI-3K/mTOR inhibitors BEZ235 and BGT226. These agents inhibited the proliferation of ALL cell lines with a three log greater potency than everolimus. However, the induction of cell death differed, with BGT226 being cytotoxic in the low micromolar range while a two log higher concentration of BEZ235 was required to produce the same effect. While all three agents extended the survival of NOD/SCID mice engrafted with human ALL, the responses of individual xenografts varied. Although differential phosphorylation of AKT on Ser(473) and Thr(308) in response to everolimus exposure was observed, this did not entirely explain the different in vivo responses to the drugs. Our data suggests that while dual PI-3K/mTOR inhibitors may improve therapeutic outcomes for a subset of ALL patients, patient selection will be important, with some patients likely to respond better to single mTOR inhibition.


Subject(s)
Imidazoles/administration & dosage , Phosphoinositide-3 Kinase Inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Quinolines/administration & dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Everolimus , Female , Humans , Imidazoles/pharmacology , In Vitro Techniques , Mice , Mice, SCID , Oncogene Protein v-akt/metabolism , Phosphorylation/drug effects , Precision Medicine , Quinolines/pharmacology , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Xenograft Model Antitumor Assays
11.
Br J Haematol ; 167(5): 618-25, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25142189

ABSTRACT

Gastrointestinal toxicity, including oral mucositis, is a frequent complication of intensive combination chemotherapy for acute myeloid leukaemia (AML) and contributes substantially to treatment-related mortality. We conducted a placebo-controlled randomized trial to evaluate the efficacy of palifermin (keratinocyte growth factor), given at 60 µg/kg per daily IV for 3 d before and after chemotherapy, for mucosal protection in adult patients with previously untreated AML receiving induction therapy with idarubicin, high-dose cytarabine and etoposide. Among 155 randomized patients, there was no statistically significant difference in the rate of grade 3 and 4 oral mucositis (primary study endpoint) between the two treatment arms (three in palifermin arm (4%), 8 in placebo arm (10%; P = 0·21); however, when considering the severity of oral mucositis (World Health Organization grade 0-4), there was evidence of reduced rates of higher grades of oral mucositis in the palifermin arm (P = 0·0007, test for trend). There was a statistically significantly lower rate of grades 3 and 4 gastrointestinal adverse events in the palifermin arm (21% vs. 44% in placebo arm; P = 0·003), mainly due to a reduction in severe diarrhoea (8% palifermin, 26% placebo; P = 0·01). Palifermin has activity as a mucosal protectant in AML patients receiving intensive chemotherapy. This trial is registered at ACTRN012605000095662.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Fibroblast Growth Factor 7/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Stomatitis/chemically induced , Stomatitis/prevention & control , Adolescent , Adult , Cytarabine/administration & dosage , Cytarabine/adverse effects , Etoposide/administration & dosage , Etoposide/adverse effects , Humans , Idarubicin/administration & dosage , Idarubicin/adverse effects , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Stomatitis/pathology
12.
Cytokine Growth Factor Rev ; 25(4): 355-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25131807

ABSTRACT

G-CSF was among the first cytokines to be identified and rapidly transitioned into clinical medicine. Initially used to promote the production of neutrophils in patients with chemotherapy-induced neutropenia it helped to revolutionize the delivery of cancer therapy. Its ability to mobilize hematopoietic stem cells from the bone marrow into the blood was subsequently exploited, changing the face of hematopoietic stem cell transplantation. Today the knowledge gained in unraveling the mechanisms of stem cell mobilization by G-CSF is being explored as a means to increase chemosensitivity in hematological malignancies. This review provides a brief history of G-CSF and then focuses on recent advances in our understanding of G-CSF-induced stem cell mobilization and the potential clinical application of this knowledge in chemo-sensitization.


Subject(s)
Granulocyte Colony-Stimulating Factor/therapeutic use , Hematologic Neoplasms/drug therapy , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/metabolism , Neutrophil Activation/drug effects , Bone Marrow Cells/metabolism , Chemokine CXCL12/metabolism , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Humans , Neutropenia/drug therapy , Neutrophils/immunology , Receptors, CXCR4/metabolism
13.
PLoS One ; 9(7): e103383, 2014.
Article in English | MEDLINE | ID: mdl-25061812

ABSTRACT

Resistance to apoptosis remains a significant problem in drug resistance and treatment failure in malignant disease. NO-aspirin is a novel drug that has efficacy against a number of solid tumours, and can inhibit Wnt signaling, and although we have shown Wnt signaling to be important for acute lymphoblastic leukemia (ALL) cell proliferation and survival inhibition of Wnt signaling does not appear to be involved in the induction of ALL cell death. Treatment of B lineage ALL cell lines and patient ALL cells with NO-aspirin induced rapid apoptotic cell death mediated via the extrinsic death pathway. Apoptosis was dependent on caspase-10 in association with the formation of the death-inducing signaling complex (DISC) incorporating pro-caspase-10 and tumor necrosis factor receptor 1 (TNF-R1). There was no measurable increase in TNF-R1 or TNF-α in response to NO-aspirin, suggesting that the process was ligand-independent. Consistent with this, expression of silencer of death domain (SODD) was reduced following NO-aspirin exposure and lentiviral mediated shRNA knockdown of SODD suppressed expansion of transduced cells confirming the importance of SODD for ALL cell survival. Considering that SODD and caspase-10 are frequently over-expressed in ALL, interfering with these proteins may provide a new strategy for the treatment of this and potentially other cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Caspase 10/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Adaptor Proteins, Signal Transducing/genetics , Caspase 10/genetics , Cells, Cultured , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Gene Silencing , Humans , Jurkat Cells
14.
PLoS One ; 9(7): e102494, 2014.
Article in English | MEDLINE | ID: mdl-25014496

ABSTRACT

Increasingly, anti-cancer medications are being reported to induce cell death mechanisms other than apoptosis. Activating alternate death mechanisms introduces the potential to kill cells that have defects in their apoptotic machinery, as is commonly observed in cancer cells, including in hematological malignancies. We, and others, have previously reported that the mTOR inhibitor everolimus has pre-clinical efficacy and induces caspase-independent cell death in acute lymphoblastic leukemia cells. Furthermore, everolimus is currently in clinical trial for acute lymphoblastic leukemia. Here we characterize the death mechanism activated by everolimus in acute lymphoblastic leukemia cells. We find that cell death is caspase-independent and lacks the morphology associated with apoptosis. Although mitochondrial depolarization is an early event, permeabilization of the outer mitochondrial membrane only occurs after cell death has occurred. While morphological and biochemical evidence shows that autophagy is clearly present it is not responsible for the observed cell death. There are a number of features consistent with paraptosis including morphology, caspase-independence, and the requirement for new protein synthesis. However in contrast to some reports of paraptosis, the activation of JNK signaling was not required for everolimus-induced cell death. Overall in acute lymphoblastic leukemia cells everolimus induces a cell death that resembles paraptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Caspases/genetics , Caspases/metabolism , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , Child , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Everolimus , Humans , Mitochondria/drug effects , Mitochondrial Membranes/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
15.
Cancer Res ; 74(10): 2803-15, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24686171

ABSTRACT

Sphingosine kinase 2 (SK2) may have utility as a prognostic marker in inflammatory diseases such as cancer in which it has been rationalized as a candidate therapeutic target. Here, we show that SK2 has an oncogenic role in acute lymphoblastic leukemia (ALL) by influencing expression of MYC. Genetic ablation of SK2 impaired leukemia development in a mouse model of ALL and pharmacologic inhibition extended survival in mouse xenograft models of human disease. SK2 attenuation in both the settings reduced MYC expression in leukemic cells, with reduced levels of acetylated histone H3 within the MYC gene associated with reduced levels of MYC protein and expression of MYC-regulated genes. Our results demonstrated that SK2 regulates MYC, which has a pivotal role in hematologic malignancies, providing a preclinical proof of concept for this pathway as a broad-based therapeutic target in this setting.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Acetylation , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Caspases/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Leukemic , Genes, myc , HEK293 Cells , Histones/metabolism , Humans , K562 Cells , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/biosynthesis , Pyridines/pharmacology , Xenograft Model Antitumor Assays
16.
Transplantation ; 96(8): 753-62, 2013 Oct 27.
Article in English | MEDLINE | ID: mdl-23903010

ABSTRACT

BACKGROUND: Dendritic cells (DC) are important in the development of acute graft-versus-host disease (GVHD) after allogeneic hemopoietic cell transplantation (alloHCT). The trafficking of immature DC from blood to GVHD target organs is likely to be regulated by chemokine receptors. METHODS: We performed flow cytometry to document the expression of chemokine receptors on circulating DC and correlated the findings after alloHCT with occurrence of acute GVHD. RESULTS: In normal individuals, plasmacytoid DC (pDC) expressed high levels of CCR5, whereas the major CD16 myeloid DC subpopulation lacked CCR5. However, its expression on CD16 cells was induced by culture in allogeneic mixed lymphocyte reaction supernatant, an effect largely mediated by interferon-γ. CCR5 was expressed on a significant proportion of CD16 DC in 42 alloHCT patients, whereas it was down-regulated on pDC. The maximum percentage of CCR5CD16 DC, at any time after transplantation, correlated with acute GVHD, whereas the minimum CCR5 on pDC showed a similar correlation. Before developing signs of GVHD, the maximum percentage CCR5CD16 DC was higher in patients with GVHD grades II to IV than in GVHD grades 0 and I, whereas the minimum percentage CCR5 on pDC was lower in GVHD grades II to IV than in GVHD grades 0 and I. CCR5 levels more than 20.5% on CD16 myeloid DC and less than 22.6% on CD123 pDC correlated with subsequent GVHD grades II to IV with high sensitivities and specificities. CONCLUSIONS: These observations may reflect DC activation and altered homing during the alloimmune response and could allow early diagnosis and therapeutic intervention before the clinical diagnosis of GVHD.


Subject(s)
Dendritic Cells/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Postoperative Complications/immunology , Receptors, CCR5/immunology , Acute Disease , Adolescent , Adult , Aged , Dendritic Cells/metabolism , Female , Flow Cytometry , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Graft vs Host Disease/metabolism , Humans , Male , Middle Aged , Postoperative Complications/metabolism , Receptors, CCR5/metabolism , Receptors, IgG/immunology , Receptors, IgG/metabolism , Transplantation, Homologous , Young Adult
17.
Lancet Infect Dis ; 13(6): 519-28, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23639612

ABSTRACT

BACKGROUND: Empirical treatment with antifungal drugs is often used in haematology patients at high risk of invasive aspergillosis. We compared a standard diagnostic strategy (culture and histology) with a rapid biomarker-based diagnostic strategy (aspergillus galactomannan and PCR) for directing the use of antifungal treatment in this group of patients. METHODS: In this open-label, parallel-group, randomised controlled trial, eligible patients were adults undergoing allogeneic stem-cell transplantation or chemotherapy for acute leukaemia, with no history of invasive fungal disease. Enrolled patients were randomly assigned (1:1) by a computer-generated schedule to follow either a standard diagnostic strategy (based on culture and histology) or a biomarker-based diagnostic strategy (aspergillus galactomannan and PCR) to direct treatment with antifungal drugs. Patients, were followed up for 26 weeks or until death. Masking of the use of different diagnostic tests was not possible for patients, treating physicians, or investigators. The primary endpoint was empirical treatment with antifungal drugs in the 26 weeks after enrolment (for the biomarker-based diagnostic strategy, a single postive galactomannan or PCR result was deemed insufficient to confirm invasive aspergillosis, so treatment in this context was classified as empirical). This outcome was assessed by an independent data review committee from which the study allocations were masked. Analyses were by intention to treat and included all enrolled patients. This study is registered with ClinicalTrial.gov, number NCT00163722. FINDINGS: 240 eligible patients were recruited from six Australian centres between Sept 30, 2005, and Nov 19, 2009. 122 were assigned the standard diagnostic strategy and 118 the biomarker-based diagnostic strategy. 39 patients (32%) in the standard diagnosis group and 18 (15%) in the biomarker diagnosis group received empirical antifungal treatment (difference 17%, 95% CI 4-26; p=0·002). The numbers of patients who had hepatotoxic and nephrotoxic effects did not differ significantly between the standard diagnosis and biomarker diagnosis groups (hepatotoxic effects: 21 [17%] vs 12 [10%], p=0·11; nephrotoxic effects: 52 [43%] vs 60 [51%], p=0·20). INTERPRETATION: Use of aspergillus galactomannan and PCR to direct treatment reduced use of empirical antifungal treatment. This approach is an effective strategy for the management of invasive aspergillosis in high-risk haematology patients. FUNDING: Australian National Health and Medical Research Council, Cancer Council New South Wales, Pfizer, Merck, Gilead Sciences.


Subject(s)
Antifungal Agents/therapeutic use , Aspergillosis/drug therapy , Aspergillosis/metabolism , Aspergillus/isolation & purification , Leukemia, Lymphoid/microbiology , Mannans/metabolism , Polymerase Chain Reaction/methods , Adult , Aspergillosis/diagnosis , Aspergillosis/pathology , Aspergillus/pathogenicity , Biomarkers/metabolism , Female , Follow-Up Studies , Galactose/analogs & derivatives , Humans , Leukemia, Lymphoid/drug therapy , Leukemia, Lymphoid/surgery , Male , Middle Aged , Stem Cell Transplantation , Transplantation, Homologous
18.
Br J Haematol ; 161(5): 623-638, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23521541

ABSTRACT

The sphingosine kinases (SphKs) have relatively recently been implicated in contributing to malignant cellular processes with particular interest in the oncogenic properties of SPHK1. Whilst SPHK1 has received considerable attention as a putative oncoprotein, SPHK2 has been much more difficult to study, with often conflicting data surrounding its role in cancer. Initial studies focused on non-haemopoietic malignancies, however a growing body of literature on the role of sphingolipid metabolism in haemopoietic malignancies is now emerging. This review provides an overview of the current state of knowledge of the SphKs and the bioactive lipid sphingosine 1-phosphate (S1P), the product of the reaction they catalyse. It then reviews the current literature regarding the roles of S1P and the SphKs in haemopoietic malignancies and discusses the compounds currently available that modulate sphingolipid metabolism and their potential and shortcomings as therapeutic agents for the treatment of haematological malignancies.


Subject(s)
Hematologic Neoplasms/enzymology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Antineoplastic Agents/therapeutic use , Drug Design , Drug Resistance, Neoplasm , Enzyme Inhibitors/therapeutic use , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Hematopoiesis/physiology , Humans , Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Sphingosine/analogs & derivatives , Sphingosine/metabolism
19.
Leuk Lymphoma ; 54(8): 1671-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23286335

ABSTRACT

This population registry-based study followed all cases of myeloma diagnosed in New South Wales, Australia, during 2002-2005 and compared survival outcomes of those who proceeded to autologous hematopoietic cell transplant (HCT) with those who did not. Data available consisted of demographic details and survival, and did not include disease details or treatment type or response. Of 708 patients, 270 (38%) had a HCT. The 5-year overall survival (OS) of HCT recipients was significantly better than for those who did not proceed to HCT (62% vs. 54%, p = 0.003). HCT was a significant favorable risk factor for OS, while age over 60 was an adverse risk factor. However, for patients alive at 1 year from diagnosis, there was no significant difference in survival between HCT and non-HCT patients, suggesting that worse disease biology and/or coexisting morbidities were likely to be major reasons for the poorer outcome for non-HCT patients.


Subject(s)
Hematopoietic Stem Cell Transplantation , Multiple Myeloma/therapy , Adult , Aged , Cause of Death , Female , Humans , Male , Middle Aged , Multiple Myeloma/mortality , Risk Factors , Transplantation, Autologous , Treatment Outcome
20.
Exp Hematol ; 41(3): 293-302.e1, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23178377

ABSTRACT

The CXCR4 antagonist Plerixafor (AMD3100) induces the rapid mobilization of hematopoietic stem and progenitor cells into the blood in mice and humans. AMD3100 similarly induces the mobilization of human acute lymphoblastic leukemia (ALL) cells into the blood in mice. In this study, the temporal response of pre-B ALL cells to AMD3100 was compared with that of normal hematopoietic progenitor cells (HPC) using an NOD/SCID xenograft model of ALL and BALB/c mice, respectively. ALL cells remained in the circulation up to 6 hours after AMD3100 administration, by which time normal HPCs were no longer detectable. AMD3100 also increased the proportion of actively cycling ALL cells in the peripheral blood. Together, these data suggest that ALL cells are more sensitive to the effects of bone marrow disruption than normal progenitors. Using the NOD/SCID xenograft model, we demonstrated that AMD3100 increased the efficacy of the cell cycle specific drug vincristine, resulting in reduced disease levels in the blood and spleens of animals over 3 weeks and extended the survival of NOD/SCID mice with ALL. These data demonstrate that mobilizing agents can increase the therapeutic effect of cell cycle dependent chemotherapeutic agents.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/drug effects , Heterocyclic Compounds/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Vincristine/pharmacology , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Benzylamines , Cell Cycle/drug effects , Cells, Cultured , Chemokine CXCL12/metabolism , Chemokine CXCL12/pharmacology , Chemotaxis/drug effects , Cyclams , Drug Synergism , Flow Cytometry , Hematopoiesis/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Precursor Cell Lymphoblastic Leukemia-Lymphoma/blood , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, CXCR4/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...