Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 78(6): 1549-1560, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29343523

ABSTRACT

Acute lymphoblastic leukemia (ALL) is the most common type of pediatric cancer, although about 4 of every 10 cases occur in adults. The enzyme drug l-asparaginase serves as a cornerstone of ALL therapy and exploits the asparagine dependency of ALL cells. In addition to hydrolyzing the amino acid l-asparagine, all FDA-approved l-asparaginases also have significant l-glutaminase coactivity. Since several reports suggest that l-glutamine depletion correlates with many of the side effects of these drugs, enzyme variants with reduced l-glutaminase coactivity might be clinically beneficial if their antileukemic activity would be preserved. Here we show that novel low l-glutaminase variants developed on the backbone of the FDA-approved Erwinia chrysanthemi l-asparaginase were highly efficacious against both T- and B-cell ALL, while displaying reduced acute toxicity features. These results support the development of a new generation of safer l-asparaginases without l-glutaminase activity for the treatment of human ALL.Significance: A new l-asparaginase-based therapy is less toxic compared with FDA-approved high l-glutaminase enzymes Cancer Res; 78(6); 1549-60. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Asparaginase/pharmacology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Recombinant Proteins/metabolism , Animals , Asparaginase/genetics , Asparaginase/metabolism , Asparaginase/pharmacokinetics , Cell Line, Tumor , Female , Glutaminase/metabolism , Glutamine/blood , Humans , Male , Mice, Inbred C57BL , Mice, SCID , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Toxicity Tests, Acute , Xenograft Model Antitumor Assays/methods
2.
PLoS One ; 12(2): e0171703, 2017.
Article in English | MEDLINE | ID: mdl-28207794

ABSTRACT

We evaluated the mitigating effects of fibroblast growth factor 4 and 7 (FGF4 and FGF7, respectively) in comparison with long acting protected graft copolymer (PGC)-formulated FGF4 and 7 (PF4 and PF7, respectively) administered to C57BL/6J mice a day after exposure to LD50/30 (15.7 Gy) partial body irradiation (PBI) which targeted the gastrointestinal (GI) system. The PGC that we developed increased the bioavailability of FGF4 and FGF7 by 5- and 250-fold compared to without PGC, respectively, and also sustained a 24 hr presence in the blood after a single subcutaneous administration. The dose levels tested for mitigating effects on radiation injury were 3 mg/kg for the PF4 and PF7 and 1.5 mg each for their combination (PF4/7). Amifostine administered prior to PBI was used as a positive control. The PF4, PF7, or PF4/7 mitigated the radiation lethality in mice. The mitigating effect of PF4 and PF7 was similar to the positive control and PF7 was better than other mitigators tested. The plasma citrulline levels and hematology parameters were early markers of recovery and survival. GI permeability function appeared to be a late or full recovery indicator. The villus length and crypt number correlated with plasma citrulline level, indicating that it can act as a surrogate marker for these histology evaluations. The IL-18 concentrations in jejunum as early as day 4 and TPO levels in colon on day 10 following PBI showed statistically significant changes in irradiated versus non-irradiated mice which makes them potential biomarkers of radiation exposure. Other colon and jejunum cytokine levels are potentially useful but require larger numbers of samples than in the present study before their full utility can be realized.


Subject(s)
Acute Radiation Syndrome/drug therapy , Fibroblast Growth Factors/therapeutic use , Gastrointestinal Tract/drug effects , Radiation Injuries, Experimental/therapy , Animals , Cell Proliferation/drug effects , Drug Delivery Systems/methods , Female , Fibroblast Growth Factor 4/adverse effects , Fibroblast Growth Factor 4/therapeutic use , Fibroblast Growth Factor 7/adverse effects , Fibroblast Growth Factor 7/therapeutic use , Fibroblast Growth Factors/adverse effects , Fibroblasts/drug effects , Gastrointestinal Tract/pathology , Gastrointestinal Tract/radiation effects , Kaplan-Meier Estimate , Lethal Dose 50 , Male , Mice, Inbred C57BL , Polylysine/chemistry , Polymers/chemistry
3.
Drug Chem Toxicol ; 36(3): 284-95, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22947079

ABSTRACT

SHetA2 is a heteroarotinoid that has shown selective inhibition of cancer cell growth and an induction of apoptosis without activation of nuclear retinoic acid receptors. In the rat study, SHetA2 was administered in 1% aqueous methylcellulose/0.2% Tween 80 by oral gavage at 0, 100, 500, and 2,000 mg/kg/day for 28 days. The high-dose administration induced decreased activity in male rats, decreased body-weight gains and food consumption, and changes in organ weights. The major metabolite of SHetA2 in rat plasma was monohydroxy SHetA2, which was considerably higher than the parent compound after oral and intravenous administration. Pharmacokinetic analysis showed extremely low (<1%) systemic bioavailability of SHetA2 for all doses tested. The dose of 2,000 mg/kg/day was considered as the lowest observed adverse effect level. The no observed adverse effect level (NOAEL) was 500 mg/kg/day. In the dog study, no toxicity of SHetA2 in 30% aqueous Solutol(®) HS 15 was observed in any tested dose groups (0, 100, 400, and 1,500 mg/kg/day). The major metabolite of SHetA2 in dog plasma was also monohydroxy SHetA2, which was equal to or lower than the parent compound after oral administration. SHetA2 levels in dog plasma were notably higher, when compared to levels in rat plasma. However, exposure was not dose proportional, as exemplified by a lack of proportional increase in maximum concentration or area under the plasma concentration-time curve with increasing dose. The NOAEL was not established and was considered to be above 1,500 mg/kg/day.


Subject(s)
Anticarcinogenic Agents/pharmacokinetics , Anticarcinogenic Agents/toxicity , Chromans/pharmacokinetics , Chromans/toxicity , Thiones/pharmacokinetics , Thiones/toxicity , Administration, Oral , Adrenal Glands/drug effects , Adrenal Glands/pathology , Animals , Anticarcinogenic Agents/administration & dosage , Area Under Curve , Chromans/administration & dosage , Dogs , Eating/drug effects , Female , Male , Motor Activity/drug effects , No-Observed-Adverse-Effect Level , Organ Size/drug effects , Prostate/drug effects , Prostate/pathology , Rats , Rats, Sprague-Dawley , Species Specificity , Thiones/administration & dosage , Toxicity Tests , Weight Gain/drug effects
4.
Chem Biol Interact ; 197(1): 16-22, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22450444

ABSTRACT

2-Chloro-5-nitro-N-phenylbenzamide (GW9662), a potent irreversible PPAR-γ antagonist, has shown promise as a cancer chemopreventive agent and is undergoing preclinical evaluations. Studies were initiated to assess its bacterial mutagenicity and pharmacokinetic profile in two animal species prior to subchronic oral toxicity evaluations and the results are reported here. GW9662 was mutagenic in both TA98 and TA100 bacterial strains with and without metabolic activation but was negative in the nitroreductase-deficient strains (TA98NR and TA100NR) also with and without metabolic activation, indicating that GW9662 mutagenicity is dependent on nitroreduction. The mutagenic activity was predominantly via a base-substitution mechanism. Following oral dosing in rats and dogs, the parent compound, GW9662, was virtually absent from plasma samples, but there was chromatographic evidence for the presence of metabolites in the plasma as a result of oral dosing. Metabolite identification studies showed that an amine metabolite ACPB (5-amino-2-chloro-N-phenylbenzamide), a product of nitro reduction, was the predominant species exhibiting large and persistent plasma levels. Thus systemic circulation of GW9662 has been attained largely in the form of its reduced metabolite, probably a product of gut bacterial metabolism. GW9662 was detectable in plasma of rats and dogs after intravenous dose albeit at low concentrations. Pharmacokinetic analysis following intravenous dosing in rats showed a rapid clearance and an extensive tissue distribution which could have accounted for the very low plasma levels. Of note, the amine metabolite was absent following intravenous dosing in both rats and dogs, confirming it being a product of presystemic metabolism. The potential utility of GW9662 as a chemopreventive agent, especially as an Estrogen Receptor-α (ER-α) inducer in an otherwise ER-α negative breast tissue, is of great interest. However, the results shown here suggest that additional animal toxicological and bioavailability studies are required to establish a role of GW9662 as a chemopreventive agent.


Subject(s)
Anilides/metabolism , Anilides/pharmacokinetics , Mutagens/metabolism , Mutagens/pharmacokinetics , Nitroreductases/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/enzymology , Amines/metabolism , Amines/pharmacokinetics , Animals , Biological Availability , Biotransformation , Dogs , Male , PPAR gamma/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Salmonella typhimurium/genetics
5.
Drug Chem Toxicol ; 34(3): 300-10, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21649485

ABSTRACT

9-cis-UAB30 is a potential chemopreventative agent that has been shown to be effective on many different types of tumors. The safety and toxicity of 9-cis-UAB30 had not been previously established. These studies were conducted to evaluate the potential toxicity and pharmacokinetics in a rodent and a nonrodent species for the purpose of investigational new drug submission. Oral gavage administration of 9-cis-UAB30 at the doses 0, 3, 15, and 100 mg/kg/day to CD® rats for 28 days showed a dose-dependent (although not dose-proportional) increase in plasma drug levels in week 4. The liver was the target organ for toxicity of 9-cis-UAB30. Hepatomegaly along with increases in serum aspartate-aminotransferase and alkaline-phosphatase levels were seen in rats. Moderate hypoalbuminemia and hyperglobulinemia resulted in a decreased albumin/globulin ratio. Histopathology revealed hepatocellular change consistent with hepatic glycogen deposition. Toxicity studies in dogs did not show treatment-related toxicity at doses as high as 100 mg/kg/day (highest dose tested) administered by capsules for 28 days. No effects on the central nervous system (functional observational battery in rats) or cardiovascular function (safety pharmacology study in telemeterized dogs) were seen. The no observed adverse effect level (NOAEL) in the rat studies was 3 mg/kg/day; however, the adverse effects seen in rats receiving 15 mg/kg/day (the least observed adverse effect level) was a slight, but statistically significant, elevation in fibrinogen and decrease in prothrombin time, which may be a sign of some tendency for increased blood coagulation. The NOAEL in the dog study was at least 100 mg/kg/day.


Subject(s)
Anticarcinogenic Agents/toxicity , Drug-Related Side Effects and Adverse Reactions/chemically induced , Fatty Acids, Unsaturated/toxicity , Naphthalenes/toxicity , Administration, Oral , Animals , Anticarcinogenic Agents/pharmacokinetics , Dogs , Dose-Response Relationship, Drug , Drug-Related Side Effects and Adverse Reactions/blood , Drug-Related Side Effects and Adverse Reactions/pathology , Fatty Acids, Unsaturated/pharmacokinetics , Female , Male , Naphthalenes/pharmacokinetics , No-Observed-Adverse-Effect Level , Organ Size/drug effects , Organ Specificity , Rats , Species Specificity , Toxicity Tests, Chronic
6.
Toxicology ; 273(1-3): 19-28, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20430063

ABSTRACT

2,2,5,7,8-Pentamethyl-6-chromanol (PMCol) was administered by gavage in rats for 28 days at dose levels of 0, 100, 500, and 2000mg/kg/day. PMCol administration induced decreases in body weight gains and food consumption, hepatotoxicity (increased TBILI, ALB, ALT, TP; increased relative liver weights; increased T4 and TSH), nephrotoxicity (increased BUN and BUN/CREAT, histopathology lesions), effect on lipid metabolism (increased CHOL), anemia, increase in WBC counts (total and differential), coagulation (FBGN upward arrow and PT downward arrow) and hyperkeratosis of the nonglandular stomach in the 2000mg/kg/day dose group (in one or both sexes). In the 500mg/kg/day dose group, toxicity was seen to a lesser extent. In the 100mg/kg/day dose group, only increased CHOL (females) was observed. To assess the toxicity of PMCol in male dogs it was administered orally by capsule administration for 28 days at dose levels of 0, 50, 200 and 800mg/kg/day (four male dogs/dose group). PMCol treatment at 800mg/kg/day resulted in pronounced toxicity to the male dogs. Target organs of toxicity were liver and thymus. Treatment at 200mg/kg/day resulted in toxicity consistent with slight adverse effect on the liver only. The results of the safety pharmacology study indicate that doses of 0, 50, 200 and 800mg/kg administered orally did not have an effect on the QT interval, blood pressures and body temperatures following dosing over a 24-h recording period. Under the conditions of this study, the no-observed-adverse effect level (NOAEL) for daily oral administration of PMCol by gavage for 28 days to male rats was 100mg/kg/day and 50mg/kg in male dogs. In female rats, the NOAEL was not established due to statistically significant and biologically meaningful increases in CHOL level seen in the 100mg/kg/day dose group. The results of these studies indicated that administration of PMCol at higher dose levels resulted in severe toxicity in dogs and moderate toxicity in rats, however, administration at lower levels is considered to be less likely to result in toxicity following 28 days of exposure. Sex-related differences were seen in rats. Male rats appeared to have greater sensitivity to nephrotoxicity, while female animals had a greater incidence of hepatoxicity and changes in hematological parameters evaluated, especially at a dose of 500mg/kg/day, which correlated to the higher plasma drug levels in female rats. It appeared that dogs were generally more sensitive than rats to oral administration of PMCol. Further examination of the potential toxic effects of PMCol in longer term studies is required prior to understanding the full risks of PMCol administration as a chemopreventative agent.


Subject(s)
Anticarcinogenic Agents/toxicity , Chromans/toxicity , Administration, Oral , Animals , Anticarcinogenic Agents/administration & dosage , Chromans/administration & dosage , Dogs , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Male , Organ Size/drug effects , Rats , Toxicity Tests
7.
Chem Biol Interact ; 179(2-3): 263-72, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19135037

ABSTRACT

Several compounds, including 2,10-dicarbethoxy-6-methoxy-5,7-dihydroindolo[2,3-b]carbazole (SR13668), (2E,4E,6Z,8E)-8-(3',4'-dihydro-1'(2'H)-napthalen-1'-ylidene)-3,7-dimethyl-2,4,6-octatrienoic acid (9-cis-UAB30), and 2,2,5,7,8-pentamethyl-6-chromanol (PMCol), were selected as promising chemopreventive agents and have entered preclinical trials for cancer prevention. The potential for adverse drug events resulting from interactions with other administered drugs, food components, or food additives presents an important question. Among the most important drug-drug interactions (DDI) is the potential of a new chemical entity (NCE) to induce cytochrome P450 enzymes (P450). Drug induction of P450 enzymes can lead to adverse drug interactions by increasing the metabolism of other drugs that are substrates for the induced isoform. Currently, sandwich cultured primary human hepatocytes are the standard for predicting human P450 enzyme induction in vitro as these cells retain the ability to respond to prototypical P450 inducers with the same specificity and potency exhibited in vivo. Therefore, a select panel of inducible P450 target genes (CYP1A2, CYP2B6, and CYP3A4) and their induction activity (measured by LC-MS/MS of respective marker substrate metabolites) were monitored in cultured hepatocytes following treatment with SR13668, 9-cis-UAB30, or PMCol to predict clinically significant drug-induced expression. The concentration ranges of the NCE used were selected to maximize the clinical relevance of these results. All responses were evaluated according to major prototypical P450 inducers (i.e., 3-methylcholanthrene, 3-MC; phenobarbital, PB; rifampicin, RIF) and increases > or = 40% of the respective positive control(s) were considered an indication of demonstrable induction. Herein, we report that there is low potential for DDI with SR13668 and PMCol due to enzyme induction of CYP1A2, CYP2B6, and CYP3A4 expression at the concentrations examined. Similarly, the study results suggested that 9-cis-UAB30 has low potential to induce CYP1A2 and CYP3A4 expression at the concentrations examined. However, 9-cis-UAB30 was shown to significantly induce CYP2B6 enzyme activity at 10 microM suggesting the potential for DDI as a result.


Subject(s)
Carbazoles/pharmacology , Chromans/pharmacology , Cytochrome P-450 Enzyme System/biosynthesis , Fatty Acids, Unsaturated/pharmacology , Hepatocytes/drug effects , Hepatocytes/enzymology , Naphthalenes/pharmacology , Cells, Cultured , Cytochrome P-450 Enzyme System/genetics , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Hepatocytes/cytology , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...