Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 19(4): 938-48, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23287564

ABSTRACT

PURPOSE: The cytidine analogs 5-azacytidine and decitabine, used to treat myelodysplastic syndromes (MDS), produce a molecular epigenetic effect, depletion of DNA-methyltransferase 1 (DNMT1). This action is S-phase dependent. Hence, genetic factors that decrease the half-lives of these drugs could impact efficacy. Documentation of such impact, and elucidation of underlying mechanisms, could lead to improved clinical application. EXPERIMENTAL DESIGN: Cytidine deaminase (CDA) rapidly inactivates 5-azacytidine/decitabine. The effect of CDA SNP A79C and gender on CDA expression, enzyme activity, and drug pharmacokinetics/pharmacodynamics was examined in mice and humans, and the impact on overall survival (OS) was evaluated in 5-azacytidine/decitabine-treated patients with MDS (n = 90) and cytarabine-treated patients with acute myeloid leukemia (AML) (n = 76). RESULTS: By high-performance liquid chromatography (HPLC), plasma CDA activity was decreased as expected in individuals with the SNP A79C. Interestingly and significantly, there was an even larger decrease in females than in males. Explaining this decrease, liver CDA expression was significantly lower in female versus male mice. As expected, decitabine plasma levels, measured by mass spectrometry, were significantly higher in females. In mathematical modeling, the detrimental impact of shorter drug half-life (e.g., in males) was greater in low compared with high S-phase fraction disease (e.g., MDS vs. AML), because in high S-phase fraction disease, even a short exposure treats a major portion of cells. Accordingly, in multivariate analysis, OS was significantly worse in male versus female patients with MDS treated with 5-azacytidine/decitabine. CONCLUSIONS: Increased CDA expression/activity in males contributes to decreased cytidine analog half-life and likely contributes to worse outcomes with 5-azacytidine or decitabine therapy.


Subject(s)
Cytidine Deaminase/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/drug therapy , Animals , Azacitidine/analogs & derivatives , Cytarabine/administration & dosage , Cytidine/administration & dosage , Cytidine/analogs & derivatives , Cytidine Deaminase/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Decitabine , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mice , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Polymorphism, Single Nucleotide , Survival Analysis , Treatment Outcome
2.
Blood ; 119(5): 1240-7, 2012 Feb 02.
Article in English | MEDLINE | ID: mdl-22160381

ABSTRACT

The deoxycytidine analog decitabine (DAC) can deplete DNA methyl-transferase 1 (DNMT1) and thereby modify cellular epigenetics, gene expression, and differentiation. However, a barrier to efficacious and accessible DNMT1-targeted therapy is cytidine deaminase, an enzyme highly expressed in the intestine and liver that rapidly metabolizes DAC into inactive uridine counterparts, severely limiting exposure time and oral bioavailability. In the present study, the effects of tetrahydrouridine (THU), a competitive inhibitor of cytidine deaminase, on the pharmacokinetics and pharmacodynamics of oral DAC were evaluated in mice and nonhuman primates. Oral administration of THU before oral DAC extended DAC absorption time and widened the concentration-time profile, increasing the exposure time for S-phase-specific depletion of DNMT1 without the high peak DAC levels that can cause DNA damage and cytotoxicity. THU also decreased interindividual variability in pharmacokinetics seen with DAC alone. One potential clinical application of DNMT1-targeted therapy is to increase fetal hemoglobin and treat hemoglobinopathy. Oral THU-DAC at a dose that would produce peak DAC concentrations of less than 0.2µM administered 2×/wk for 8 weeks to nonhuman primates was not myelotoxic, hypomethylated DNA in the γ-globin gene promoter, and produced large cumulative increases in fetal hemoglobin. Combining oral THU with oral DAC changes DAC pharmacology in a manner that may facilitate accessible noncytotoxic DNMT1-targeted therapy.


Subject(s)
Azacitidine/analogs & derivatives , Tetrahydrouridine/pharmacology , Administration, Oral , Animals , Antimetabolites/pharmacology , Antimetabolites, Antineoplastic/adverse effects , Antimetabolites, Antineoplastic/metabolism , Antimetabolites, Antineoplastic/pharmacokinetics , Area Under Curve , Azacitidine/administration & dosage , Azacitidine/adverse effects , Azacitidine/metabolism , Azacitidine/pharmacokinetics , Biological Availability , DNA Damage/drug effects , DNA Methylation/drug effects , Decitabine , Drug Interactions , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Inactivation, Metabolic , Injections, Intravenous , Injections, Subcutaneous , Mice , Papio anubis
3.
Blood ; 118(26): 6845-8, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22067383

ABSTRACT

IL-15 promotes activation and maintenance of natural killer (NK) and CD8(+) T effector memory (T(EM)) cells, making it a potential immunotherapeutic agent for the treatment of cancer and immunodeficiency states. Here we report the immunologic effects of 3 different IL-15 dosing strategies in Rhesus macaques. IL-15 at a dose of 20 µg/kg/d administered by continuous intravenous infusion for 10 days resulted in a massive (100-fold) expansion of CD8(+) T(EM) cells in the peripheral blood. In contrast, the administration of 20-40 µg/kg/d of IL-15 by subcutaneous injection resulted in a more modest (10-fold) expansion of CD8(+) T(EM) cells. NK expansion was similar in both the continuous intravenous and daily subcutaneous treatment groups. The observation that IL-15 administered by continuous intravenous infusion is able to induce markedly greater expansions of CD8(+) T(EM) cells than the same dose administered by other routes may have important implications for clinical development of this cytokine.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Immunologic Memory/drug effects , Interleukin-15/pharmacology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Drug , Humans , Immunologic Memory/immunology , Infusions, Intravenous , Injections, Subcutaneous , Interleukin-15/administration & dosage , Interleukin-15/pharmacokinetics , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Macaca mulatta
4.
Am J Physiol Gastrointest Liver Physiol ; 292(2): G647-56, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17053162

ABSTRACT

Previous studies utilizing an ex vivo porcine model of intestinal ischemic injury demonstrated that prostaglandin (PG)E(2) stimulates repair of mucosal barrier function via a mechanism involving Cl(-) secretion and reductions in paracellular permeability. Further experiments revealed that the signaling mechanism for PGE(2)-induced mucosal recovery was mediated via type-2 Cl(-) channels (ClC-2). Therefore, the objective of the present study was to directly investigate the role of ClC-2 in mucosal repair by evaluating mucosal recovery in ischemia-injured intestinal mucosa treated with the selective ClC-2 agonist lubiprostone. Ischemia-injured porcine ileal mucosa was mounted in Ussing chambers, and short-circuit current (I(sc)) and transepithelial electrical resistance (TER) were measured in response to lubiprostone. Application of 0.01-1 microM lubiprostone to ischemia-injured mucosa induced concentration-dependent increases in TER, with 1 microM lubiprostone stimulating a twofold increase in TER (DeltaTER = 26 Omega.cm(2); P < 0.01). However, lubiprostone (1 microM) stimulated higher elevations in TER despite lower I(sc) responses compared with the nonselective secretory agonist PGE(2) (1 microM). Furthermore, lubiprostone significantly (P < 0.05) reduced mucosal-to-serosal fluxes of (3)H-labeled mannitol to levels comparable to those of normal control tissues and restored occludin localization to tight junctions. Activation of ClC-2 with the selective agonist lubiprostone stimulated elevations in TER and reductions in mannitol flux in ischemia-injured intestine associated with structural changes in tight junctions. Prostones such as lubiprostone may provide a selective and novel pharmacological mechanism of accelerating recovery of acutely injured intestine compared with the nonselective action of prostaglandins such as PGE(2).


Subject(s)
Alprostadil/analogs & derivatives , Colon/drug effects , Fatty Acids/pharmacology , Ileum/drug effects , Intestinal Mucosa/drug effects , Alprostadil/pharmacology , Alprostadil/therapeutic use , Animals , Bumetanide/pharmacology , CLC-2 Chloride Channels , Chloride Channel Agonists , Chloride Channels/antagonists & inhibitors , Chlorides/pharmacology , Colon/blood supply , Colon/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Dinoprostone/pharmacology , Electric Impedance , Electrophysiology , Fatty Acids/therapeutic use , Female , Ileum/blood supply , Ileum/physiopathology , Indomethacin/pharmacology , Intestinal Mucosa/pathology , Intestinal Mucosa/physiopathology , Ischemia/metabolism , Ischemia/physiopathology , Lubiprostone , Male , Mannitol/metabolism , Membrane Proteins/metabolism , Occludin , Permeability , Recovery of Function/drug effects , Sus scrofa , Urea/analogs & derivatives , Urea/pharmacology , Zinc Compounds/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...