Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Hum Gene Ther ; 30(7): 865-881, 2019 07.
Article in English | MEDLINE | ID: mdl-30808234

ABSTRACT

Alport syndrome is a genetic disease caused by mutations in type IV collagen and is characterized by progressive kidney disease. The Col4α3-/- mouse model recapitulates the main features of human Alport syndrome. Previously, it was reported that kidney microRNA-21 (miR-21) expression is significantly increased in Col4α3-/- mice, and administration of anti-miR-21 oligonucleotides (anti-miR-21) attenuates kidney disease progression in Col4α3-/- mice, indicating that miR-21 is a viable therapeutic target for Alport syndrome. However, the expression pattern of miR-21 in the kidneys of patients with human Alport syndrome has not been evaluated. Paraffin-embedded kidney specimens were obtained from 27 patients with Alport syndrome and from 10 normal controls. They were evaluated for miR-21 expression and for in situ hybridization and mRNA expression by quantitative polymerase chain reaction. In addition, anti-miR-21 was administrated to Col4α3-/- mice at different stages of disease, and changes in proteinuria, kidney function, and survival were monitored. Transcriptomic analysis of mouse kidney was conducted using RNA sequencing. miR-21 expression was significantly elevated in kidney specimens from patients with Alport syndrome compared to normal controls. Elevated renal miR-21 expression positively correlated with 24 h urine protein, serum blood urea nitrogen, serum creatinine, and severity of kidney pathology. On histological evaluation, high levels of miR-21 were localized to damaged tubular epithelial cells and glomeruli. Kidney specimens from both humans and mice with Alport syndrome exhibited abnormal expression of genes involved in kidney injury, fibrosis, inflammation, mitochondrial function, and lipid metabolism. Administration of anti-miR-21 to Alport mice resulted in slowing of kidney function decline, partial reversal of abnormal gene expression associated with disease pathology, and improved survival. Increased levels of miR-21 in human Alport kidney samples showed a correlation with kidney disease severity measured by proteinuria, biomarkers of kidney function, and kidney histopathology scores. These human data, combined with the finding that a reduction of miR-21 in Col4α3-/- mice improves kidney phenotype and survival, support miR-21 as a viable therapeutic target for the treatment of Alport syndrome.


Subject(s)
Gene Expression Regulation , Genetic Predisposition to Disease , MicroRNAs/genetics , Nephritis, Hereditary/genetics , Adolescent , Animals , Autoantigens , Biomarkers , Biopsy , Child , Collagen Type IV/deficiency , Disease Models, Animal , Female , Fibrosis , Gene Expression Profiling , Genetic Association Studies , Humans , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Knockout , Nephritis, Hereditary/diagnosis , Nephritis, Hereditary/metabolism , Severity of Illness Index
2.
Kidney Int ; 94(2): 303-314, 2018 08.
Article in English | MEDLINE | ID: mdl-29759420

ABSTRACT

Lysyl oxidase like-2 (LOXL2) is an amine oxidase with both intracellular and extracellular functions. Extracellularly, LOXL2 promotes collagen and elastin crosslinking, whereas intracellularly, LOXL2 has been reported to modify histone H3, stabilize SNAIL, and reduce cell polarity. Although LOXL2 promotes liver and lung fibrosis, little is known regarding its role in renal fibrosis. Here we determine whether LOXL2 influences kidney disease in COL4A3 (-/-) Alport mice. These mice were treated with a small molecule inhibitor selective for LOXL2 or with vehicle and assessed for glomerular sclerosis and fibrosis, albuminuria, blood urea nitrogen, lifespan, pro-fibrotic gene expression and ultrastructure of the glomerular basement membrane. Laminin α2 deposition in the glomerular basement membrane and mesangial filopodial invasion of the glomerular capillaries were also assessed. LOXL2 inhibition significantly reduced interstitial fibrosis and mRNA expression of MMP-2, MMP-9, TGF-ß1, and TNF-α. LOXL2 inhibitor treatment also reduced glomerulosclerosis, expression of MMP-10, MMP-12, and MCP-1 mRNA in glomeruli, and decreased albuminuria and blood urea nitrogen. Mesangial filopodial invasion of the capillary tufts was blunted, as was laminin α2 deposition in the glomerular basement membrane, and glomerular basement membrane ultrastructure was normalized. There was no effect on lifespan. Thus, LOXL2 plays an important role in promoting both glomerular and interstitial pathogenesis associated with Alport syndrome in mice. Other etiologies of chronic kidney disease are implicated with our observations.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Enzyme Inhibitors/therapeutic use , Glomerular Basement Membrane/pathology , Glomerular Mesangium/pathology , Nephritis, Hereditary/pathology , Amino Acid Oxidoreductases/antagonists & inhibitors , Amino Acid Oxidoreductases/genetics , Animals , Autoantigens/genetics , Collagen Type IV/genetics , Disease Models, Animal , Disease Progression , Enzyme Inhibitors/pharmacology , Fibrosis , Glomerular Basement Membrane/metabolism , Glomerular Mesangium/metabolism , Humans , Laminin/metabolism , Mice , Nephritis, Hereditary/drug therapy , Nephritis, Hereditary/genetics , RNA, Messenger/metabolism , Up-Regulation
3.
J Med Chem ; 60(21): 9040-9052, 2017 11 09.
Article in English | MEDLINE | ID: mdl-29035567

ABSTRACT

AMP-activated protein kinase (AMPK) plays an essential role as a cellular energy sensor and master regulator of metabolism in eukaryotes. Dysregulated lipid and carbohydrate metabolism resulting from insulin resistance leads to hyperglycemia, the hallmark of type 2 diabetes mellitus (T2DM). While pharmacological activation of AMPK is anticipated to improve these parameters, the discovery of selective, direct activators has proven challenging. We now describe a hit-to-lead effort resulting in the discovery of a potent and selective class of benzimidazole-based direct AMPK activators, exemplified by 5-((5-([1,1'-biphenyl]-4-yl)-6-chloro-1H-benzo[d]imidazol-2-yl)oxy)-2-methylbenzoic acid, 42 (MK-3903). Compound 42 exhibited robust target engagement in mouse liver following oral dosing, leading to improved lipid metabolism and insulin sensitization in mice.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Benzimidazoles/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Administration, Oral , Animals , Benzimidazoles/administration & dosage , Benzimidazoles/therapeutic use , Drug Discovery , Insulin Resistance , Lipid Metabolism/drug effects , Mice
4.
J Med Chem ; 60(10): 4403-4423, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28471663

ABSTRACT

LOXL2 catalyzes the oxidative deamination of ε-amines of lysine and hydroxylysine residues within collagen and elastin, generating reactive aldehydes (allysine). Condensation with other allysines or lysines drives the formation of inter- and intramolecular cross-linkages, a process critical for the remodeling of the ECM. Dysregulation of this process can lead to fibrosis, and LOXL2 is known to be upregulated in fibrotic tissue. Small-molecules that directly inhibit LOXL2 catalytic activity represent a useful option for the treatment of fibrosis. Herein, we describe optimization of an initial hit 2, resulting in identification of racemic-trans-(3-((4-(aminomethyl)-6-(trifluoromethyl)pyridin-2-yl)oxy)phenyl)(3-fluoro-4-hydroxypyrrolidin-1-yl)methanone 28, a potent irreversible inhibitor of LOXL2 that is highly selective over LOX and other amine oxidases. Oral administration of 28 significantly reduced fibrosis in a 14-day mouse lung bleomycin model. The (R,R)-enantiomer 43 (PAT-1251) was selected as the clinical compound which has progressed into healthy volunteer Phase 1 trials, making it the "first-in-class" small-molecule LOXL2 inhibitor to enter clinical development.


Subject(s)
Amino Acid Oxidoreductases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Administration, Oral , Amino Acid Oxidoreductases/metabolism , Animals , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Fibrosis , Halogenation , Humans , Lung/drug effects , Lung/enzymology , Lung/pathology , Lung Diseases/drug therapy , Lung Diseases/enzymology , Lung Diseases/pathology , Male , Methylation , Mice, Inbred C57BL , Models, Molecular , Pyridines/administration & dosage , Pyridines/therapeutic use , Structure-Activity Relationship
5.
Nucleic Acids Res ; 44(2): e13, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26384419

ABSTRACT

Anti-miRNA (anti-miR) oligonucleotide drugs are being developed to inhibit overactive miRNAs linked to disease. To help facilitate the transition from concept to clinic, new research tools are required. Here we report a novel method--miRNA Polysome Shift Assay (miPSA)--for direct measurement of miRNA engagement by anti-miR, which is more robust than conventional pharmacodynamics using downstream target gene derepression. The method takes advantage of size differences between active and inhibited miRNA complexes. Active miRNAs bind target mRNAs in high molecular weight polysome complexes, while inhibited miRNAs are sterically blocked by anti-miRs from forming this interaction. These two states can be assessed by fractionating tissue or cell lysates using differential ultracentrifugation through sucrose gradients. Accordingly, anti-miR treatment causes a specific shift of cognate miRNA from heavy to light density fractions. The magnitude of this shift is dose-responsive and maintains a linear relationship with downstream target gene derepression while providing a substantially higher dynamic window for aiding drug discovery. In contrast, we found that the commonly used 'RT-interference' approach, which assumes that inhibited miRNA is undetectable by RT-qPCR, can yield unreliable results that poorly reflect the binding stoichiometry of anti-miR to miRNA. We also demonstrate that the miPSA has additional utility in assessing anti-miR cross-reactivity with miRNAs sharing similar seed sequences.


Subject(s)
Biological Assay , Gene Expression Regulation , MicroRNAs/antagonists & inhibitors , Polyribosomes/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Line , Centrifugation, Density Gradient , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fructose-Bisphosphate Aldolase/antagonists & inhibitors , Fructose-Bisphosphate Aldolase/genetics , Fructose-Bisphosphate Aldolase/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Oligonucleotides/genetics , Oligonucleotides/metabolism , Polyribosomes/genetics , Reverse Transcriptase Polymerase Chain Reaction
6.
J Clin Invest ; 125(1): 141-56, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25415439

ABSTRACT

MicroRNA-21 (miR-21) contributes to the pathogenesis of fibrogenic diseases in multiple organs, including the kidneys, potentially by silencing metabolic pathways that are critical for cellular ATP generation, ROS production, and inflammatory signaling. Here, we developed highly specific oligonucleotides that distribute to the kidney and inhibit miR-21 function when administered subcutaneously and evaluated the therapeutic potential of these anti-miR-21 oligonucleotides in chronic kidney disease. In a murine model of Alport nephropathy, miR-21 silencing did not produce any adverse effects and resulted in substantially milder kidney disease, with minimal albuminuria and dysfunction, compared with vehicle-treated mice. miR-21 silencing dramatically improved survival of Alport mice and reduced histological end points, including glomerulosclerosis, interstitial fibrosis, tubular injury, and inflammation. Anti-miR-21 enhanced PPARα/retinoid X receptor (PPARα/RXR) activity and downstream signaling pathways in glomerular, tubular, and interstitial cells. Moreover, miR-21 silencing enhanced mitochondrial function, which reduced mitochondrial ROS production and thus preserved tubular functions. Inhibition of miR-21 was protective against TGF-ß-induced fibrogenesis and inflammation in glomerular and interstitial cells, likely as the result of enhanced PPARα/RXR activity and improved mitochondrial function. Together, these results demonstrate that inhibition of miR-21 represents a potential therapeutic strategy for chronic kidney diseases including Alport nephropathy.


Subject(s)
MicroRNAs/genetics , Nephritis, Hereditary/therapy , Oligoribonucleotides, Antisense/genetics , Animals , Autoantigens/genetics , Collagen Type IV/deficiency , Collagen Type IV/genetics , Disease Progression , Fibrosis/metabolism , Kidney/metabolism , Kidney/pathology , Metabolic Networks and Pathways/genetics , Mice, 129 Strain , MicroRNAs/metabolism , Nephritis, Hereditary/metabolism , Nephritis, Hereditary/pathology , Reactive Oxygen Species/metabolism , Transcriptome , Up-Regulation
7.
Cell Metab ; 20(4): 626-38, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25295787

ABSTRACT

Macroautophagy (hereafter autophagy) is the major pathway by which macromolecules and organelles are degraded. Autophagy is regulated by the mTOR signaling pathway-the focal point for integration of metabolic information, with mTORC1 playing a central role in balancing biosynthesis and catabolism. Of the various inputs to mTORC1, the amino acid sensing pathway is among the most potent. Based upon transcriptome analysis of neurons subjected to nutrient deprivation, we identified let-7 microRNA as capable of promoting neuronal autophagy. We found that let-7 activates autophagy by coordinately downregulating the amino acid sensing pathway to prevent mTORC1 activation. Let-7 induced autophagy in the brain to eliminate protein aggregates, establishing its physiological relevance for in vivo autophagy modulation. Moreover, peripheral delivery of let-7 anti-miR repressed autophagy in muscle and white fat, suggesting that let-7 autophagy regulation extends beyond CNS. Hence, let-7 plays a central role in nutrient homeostasis and proteostasis regulation in higher organisms.


Subject(s)
Amino Acids/metabolism , Autophagy , MicroRNAs/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adipose Tissue, White/metabolism , Animals , Base Sequence , Brain/metabolism , Cells, Cultured , HEK293 Cells , Humans , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/antagonists & inhibitors , Monomeric GTP-Binding Proteins/antagonists & inhibitors , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Muscle, Skeletal/metabolism , Neurons/cytology , Neurons/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Sequence Alignment , Signal Transduction
8.
Sci Transl Med ; 4(121): 121ra18, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22344686

ABSTRACT

Scarring of the kidney is a major public health concern, directly promoting loss of kidney function. To understand the role of microRNA (miRNA) in the progression of kidney scarring in response to injury, we investigated changes in miRNA expression in two kidney fibrosis models and identified 24 commonly up-regulated miRNAs. Among them, miR-21 was highly elevated in both animal models and in human transplanted kidneys with nephropathy. Deletion of miR-21 in mice resulted in no overt abnormality. However, miR-21(-/-) mice suffered far less interstitial fibrosis in response to kidney injury, a phenotype duplicated in wild-type mice treated with anti-miR-21 oligonucleotides. Global derepression of miR-21 target mRNAs was readily detectable in miR-21(-/-) kidneys after injury. Analysis of gene expression profiles up-regulated in the absence of miR-21 identified groups of genes involved in metabolic pathways, including the lipid metabolism pathway regulated by peroxisome proliferator-activated receptor-α (Pparα), a direct miR-21 target. Overexpression of Pparα prevented ureteral obstruction-induced injury and fibrosis. Pparα deficiency abrogated the antifibrotic effect of anti-miR-21 oligonucleotides. miR-21 also regulated the redox metabolic pathway. The mitochondrial inhibitor of reactive oxygen species generation Mpv17l was repressed by miR-21, correlating closely with enhanced oxidative kidney damage. These studies demonstrate that miR-21 contributes to fibrogenesis and epithelial injury in the kidney in two mouse models and is a candidate target for antifibrotic therapies.


Subject(s)
Gene Silencing , Kidney/pathology , MicroRNAs/physiology , Animals , Fibrosis , Humans , Kidney/metabolism , Mice , Mice, Knockout , Up-Regulation
9.
ACS Med Chem Lett ; 1(9): 478-82, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-24900234

ABSTRACT

AMP-activated protein kinase (AMPK) is a heterotrimeric kinase that regulates cellular energy metabolism by affecting energy-consuming pathways such as de novo lipid biosynthesis and glucose production as well as energy-producing pathways such as lipid oxidation and glucose uptake. Accordingly, compounds that activate AMPK represent potential drug candidates for the treatment of hyperlipidemia and type 2 diabetes. Screening of a proprietary library of AMP mimetics identified the phosphonic acid 2 that bears little structural resemblance to AMP but is capable of activating AMPK with high potency (EC50 = 6 nM vs AMP EC50 = 6 µM) and specificity. Phosphonate prodrugs of 2 inhibited de novo lipogenesis in cellular and animal models of hyperlipidemia.

10.
Cancer Chemother Pharmacol ; 64(5): 981-91, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19283354

ABSTRACT

PURPOSE: Hepatocellular carcinoma (HCC) is a life-threatening condition with only one drug treatment regimen approved for use. Oncolytic nucleosides are minimally effective against HCC putatively because of their inability to achieve cytotoxic levels of the active metabolite [nucleoside triphosphate (NTP)] in tumor cells at doses that are well tolerated. The aim of our studies was to explore the utility of CYP3A-activated prodrugs of cytarabine and fludarabine monophosphate for the treatment of HCC. METHODS: Prodrugs of cytarabine and fludarabine monophosphates were evaluated for their ability to safely achieve NTP levels in the liver of normal mice that are cytotoxic to hepatoma cells. RESULTS: While therapeutic levels of NTPs are achieved in the livers of normal rodents after administration of the prodrugs, only MB07133 achieved these levels without exhibiting signs of liver toxicity or myelosuppression. CONCLUSIONS: As the levels of araCTP achieved in the liver at therapeutic doses are only toxic to proliferating cells (such as those in HCC tumors), but not the non-proliferative adjacent tissue, MB07133 treatment has the potential to be both efficacious and well tolerated in HCC patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/enzymology , Cytochrome P-450 CYP3A/biosynthesis , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Nucleosides/administration & dosage , Nucleosides/therapeutic use , Animals , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents/adverse effects , Carcinoma, Hepatocellular/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Cytarabine/administration & dosage , Cytarabine/therapeutic use , Cytochrome P-450 CYP3A/genetics , Drug Delivery Systems , Infusions, Intravenous , Liver/drug effects , Liver Neoplasms/genetics , Male , Mice , Nucleosides/adverse effects , Prodrugs , Thymidine/metabolism , Tissue Distribution , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Vidarabine/therapeutic use
11.
J Med Chem ; 49(26): 7711-20, 2006 Dec 28.
Article in English | MEDLINE | ID: mdl-17181153

ABSTRACT

Cytotoxic nucleosides have proven to be ineffective for the treatment of hepatocellular carcinoma (HCC) due, in part, to their inadequate conversion to their active nucleoside triphosphates (NTP) in the liver tumor and high conversion in other tissues. These characteristics lead to poor efficacy, high toxicity, and a drug class associated with an unacceptable therapeutic index. Cyclic 1-aryl-1,3-propanyl phosphate prodrugs selectively release the monophosphate of a nucleoside (NMP) into CYP3A4-expressing cells, such as hepatocytes, while leaving the prodrug intact in plasma and extrahepatic tissues. This prodrug strategy was applied to the monophosphate of the well-known cytotoxic nucleoside cytosine-1-beta-D-arabinofuranoside (cytarabine, araC). Compound 19S (MB07133), in mice, achieves good liver targeting compared to araC, generating >19-fold higher cytarabine triphosphate (araCTP) levels in the liver than levels of araC in the plasma and >12-fold higher araCTP levels in the liver than in the bone marrow, representing a >120-fold and >28-fold improvement, respectively, over araC administration.


Subject(s)
Antineoplastic Agents/pharmacology , Arabinonucleotides/chemical synthesis , Carcinoma, Hepatocellular/drug therapy , Cytidine Monophosphate/analogs & derivatives , Liver/drug effects , Prodrugs/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Arabinofuranosylcytosine Triphosphate/blood , Arabinonucleotides/pharmacokinetics , Arabinonucleotides/pharmacology , Bone Marrow/drug effects , Bone Marrow/metabolism , Chromatography, High Pressure Liquid , Cytidine Monophosphate/chemical synthesis , Cytidine Monophosphate/pharmacokinetics , Cytidine Monophosphate/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Liver Neoplasms/drug therapy , Male , Mice , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Rats , Tissue Distribution
12.
J Pharmacol Exp Ther ; 312(2): 554-60, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15340017

ABSTRACT

Targeting drugs to specific organs, tissues, or cells is an attractive strategy for enhancing drug efficacy and reducing side effects. Drug carriers such as antibodies, natural and manmade polymers, and labeled liposomes are capable of targeting drugs to blood vessels of individual tissues but often fail to deliver drugs to extravascular sites. An alternative strategy is to use low molecular weight prodrugs that distribute throughout the body but cleave intracellularly to the active drug by an organ-specific enzyme. Here we show that a series of phosphate and phosphonate prodrugs, called HepDirect prodrugs, results in liver-targeted drug delivery following a cytochrome P450-catalyzed oxidative cleavage reaction inside hepatocytes. Liver targeting was demonstrated in rodents for MB06866 [(2R,4S)-9-[2-[4-(3-chlorophenyl)-2-oxo-1,3,2-dioxaphosphorinan-2-yl]methoxyethyl]adenine (remofovir)], a Hep-Direct prodrug of the nucleotide analog adefovir (PMEA), and MB07133 [(2R,4S)-4-amino-1-[5-O-[2-oxo-4-(4-pyridyl)-1,3,2-dioxaphosphorinan-2-yl]-beta-d-arabinofuranosyl]-2(1H)-pyrimidinone], a HepDirect prodrug of cytarabine (araC) 5'-monophosphate. Liver targeting led to higher levels of the biologically active form of PMEA and araC in the liver and to lower levels in the most toxicologically sensitive organs. Liver targeting also confined production of the prodrug byproduct, an aryl vinyl ketone, to hepatocytes. Glutathione within the hepatocytes rapidly reacted with the byproduct to form a glutathione conjugate. No byproduct-related toxicity was observed in hepatocytes or animals treated with HepDirect prodrugs. A 5-day safety study in mice demonstrated the toxicological benefits of liver targeting. These findings suggest that HepDirect prodrugs represent a potential strategy for targeting drugs to the liver and achieving more effective therapies against chronic liver diseases such as hepatitis B, hepatitis C, and hepatocellular carcinoma.


Subject(s)
Adenine/analogs & derivatives , Adenine/pharmacology , Cytarabine/analogs & derivatives , Cytarabine/pharmacology , Drug Delivery Systems , Liver/drug effects , Organophosphorus Compounds/pharmacology , Prodrugs/pharmacology , Acetaminophen/toxicity , Adenine/administration & dosage , Adenine/pharmacokinetics , Adenine/toxicity , Analgesics, Non-Narcotic/toxicity , Animals , Biotransformation , Bone Marrow/metabolism , Cell Separation , Cytarabine/administration & dosage , Cytarabine/pharmacokinetics , Cytarabine/toxicity , Glutathione/metabolism , Hepatocytes/drug effects , In Vitro Techniques , Inactivation, Metabolic , Male , Mice , Organophosphonates/administration & dosage , Organophosphonates/pharmacokinetics , Organophosphonates/toxicity , Organophosphorus Compounds/pharmacokinetics , Rats , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...