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

Database
Type of study
Language
Affiliation country
Publication year range
1.
Cancer Prev Res (Phila) ; 11(1): 27-37, 2018 01.
Article in English | MEDLINE | ID: mdl-29074535

ABSTRACT

Flaxseed consumption is associated with reduced oxidative stress and inflammation in lung injury models and has shown anticancer effects for breast and prostate tissues. However, the chemopreventive potential of flaxseed remains unexplored for lung cancer. In this study, we investigated the effect of flaxseed on tobacco smoke carcinogen (NNK)-induced lung tumorigenesis in an A/J mouse model. Mice exposed to NNK were fed a control diet or a 10% flaxseed-supplemented diet for 26 weeks. Flaxseed-fed mice showed reduced lung tumor incidence (78%) and multiplicity, with an average of 2.7 ± 2.3 surface lung tumor nodules and 1.0 ± 0.9 H&E cross-section nodules per lung compared with the control group, which had 100% tumor incidence and an average of 10.2 ± 5.7 surface lung tumor nodules and 3.9 ± 2.6 H&E cross-section nodules per lung. Furthermore, flaxseed-fed mice had a lower incidence of adenocarcinomas compared with control-fed mice. Western blotting performed on normal lung tissues showed flaxseed suppressed phosphorylation (activation) of p-AKT, p-ERK, and p-JNK kinases. RNA-Seq data obtained from normal lung and lung tumors of control and flaxseed-fed mice suggested that flaxseed intake resulted in differential expression of genes involved in inflammation-mediated cytokine signaling (IL1, 6, 8, 9, and 12α), xenobiotic metabolism (several CYPs, GSTs, and UGTs), and signaling pathways (AKT and MAPK) involved in tumor cell proliferation. Together, our results indicate that dietary flaxseed supplementation may be an effective chemoprevention strategy for chemically induced lung carcinogenesis by altering signaling pathways, inflammation, and oxidative stress. Cancer Prev Res; 11(1); 27-37. ©2017 AACR.


Subject(s)
Carcinogenesis/drug effects , Carcinogens/toxicity , Cytokines/metabolism , Flax/chemistry , Inflammation Mediators/metabolism , Lung Neoplasms/prevention & control , Plant Extracts/pharmacology , Animals , Anticarcinogenic Agents/pharmacology , Benzo(a)pyrene/toxicity , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cytochrome P-450 CYP1A1/metabolism , Cytokines/genetics , Glucuronosyltransferase/metabolism , Glutathione Transferase/metabolism , High-Throughput Nucleotide Sequencing , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/metabolism , Male , Metabolic Detoxication, Phase II , Mice , Mice, Inbred A , Nitrosamines/toxicity , Seeds/chemistry
2.
Target Oncol ; 12(3): 323-332, 2017 06.
Article in English | MEDLINE | ID: mdl-28357727

ABSTRACT

BACKGROUND: The combination of everolimus and the imidazoquinoline derivative, BEZ235 (dactolisib), a dual PI3K/mTOR inhibitor, demonstrated synergy in a preclinical model. OBJECTIVE: To establish clinical feasibility, a phase Ib dose-escalation trial investigating safety and pharmacokinetics of this combination in patients with advanced tumors was performed. PATIENTS AND METHODS: BEZ235 was orally administered daily in escalating doses of 200, 400, and 800 mg along with everolimus at 2.5 mg daily in 28-day cycles. Nineteen patients were enrolled. Adverse events and tumor responses were evaluated using CTCAE v4.0 and RECIST 1.1, respectively. Pharmacokinetic analyses were performed. RESULTS: Common toxicities observed included fatigue, diarrhea, nausea, mucositis, and elevated liver enzymes. No confirmed responses were observed. BEZ235 pharmacokinetics exhibited dose-proportional increases in Cmax and AUC0-24 over the three doses, with high inter-individual variability. Non-compartmental and population pharmacokinetic-based simulations indicated significant increases in everolimus Cmax and AUC0-24 on day 28 and decreased clearance to 13.41 L/hr. CONCLUSIONS: The combination of BEZ235 and everolimus demonstrated limited efficacy and tolerance. BEZ235 systemic exposure increased in a dose-proportional manner while oral bioavailability was quite low, which may be related to gastrointestinal-specific toxicity. The changes in steady-state pharmacokinetics of everolimus with BEZ235 highlight potential drug-drug interactions when these two drugs are administered together. Clinicaltrials.gov: NCT01508104.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Everolimus/therapeutic use , Imidazoles/therapeutic use , Neoplasms/drug therapy , Quinolines/therapeutic use , Administration, Oral , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Drug Evaluation, Preclinical , Drug Synergism , Everolimus/adverse effects , Female , Humans , Imidazoles/adverse effects , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Quinolines/adverse effects , TOR Serine-Threonine Kinases/metabolism , Tumor Lysis Syndrome/etiology
3.
J Pharm Sci ; 98(1): 362-77, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18563832

ABSTRACT

The disposition of three 4-aminoquinoline leads, namely isoquine (ISO), des-ethyl isoquine (DEI) and N-tert-butyl isoquine (NTBI), were studied in a range of in vivo and in vitro assays to assist in selecting an appropriate candidate for further development. Analogous to amodiaquine (ADQ), ISO undergoes oxidative N-dealkylation to form DEI in vivo. Blood clearance of DEI was as much as 10-fold lower than that of ISO in animals and after oral administration, metabolite exposure exceeded that of parent by as much as 14-fold. Replacement of the N-ethyl with an N-tert-butyl substituent substantially reduced N-dealkylation as blood clearance of NTBI was approximately 2 to 3-fold lower than DEI in mouse, rat, dog and monkey. Mean NTBI oral bioavailability was generally higher than the other leads (>/=68%). Blood cell association was substantial for NTBI, particularly in dog and monkey, where blood to plasma concentration ratios >4 were observed. Human plasma protein binding was similar for NTBI, DEI, and des-ethyl amodiaquine (DEA). Allometric scaling predicted human blood clearance (CL) for NTBI to be low ( approximately 12% liver blood flow). All the 4-aminoquinolines inhibited recombinant human cytochrome P450 2D6 with similar potency; DEI also inhibited 1A2. On balance, NTBI appeared the most promising lead to progress towards full development.


Subject(s)
Aminoquinolines/pharmacokinetics , Antimalarials/pharmacokinetics , Aminoquinolines/blood , Aminoquinolines/metabolism , Animals , Antimalarials/blood , Antimalarials/metabolism , Biological Availability , Dogs , Drug Evaluation, Preclinical/methods , Female , Humans , Macaca fascicularis , Male , Mice , Rats , Rats, Sprague-Dawley , Species Specificity
4.
J Pharm Sci ; 95(12): 2657-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16892205

ABSTRACT

GW844520 is a potent and selective inhibitor of the cytochrome bc1 complex of mitochondrial electron transport in P. falciparum, the parasite primarily responsible for the mortality associated with malaria worldwide. GW844520 is fully active against the parasite including resistance isolates, showing no cross resistance with agents in use. To evaluate full potential of this development candidate, we conducted drug metabolism and pharmacokinetic studies of this novel anti-malarial. GW844520 had low blood clearance of about 0.5-4% of hepatic blood flow and a steady-state volume of distribution of 2-4 times total body water in mouse, rat, dog, and monkey. Oral bioavailability was high (51-100%). Consistent with the in vivo data, GW844520 had low intrinsic clearance in liver microsomes and hepatocytes of animal and human origin, high passive cellular permeability and was not a P-glycoprotein substrate. GW844520 did not associate appreciably with blood cells but was highly bound to plasma proteins (>99%) in all species. GW844520 was a substrate and inhibitor of human CYP2D6 but not of CYP1A2, 2C9, 2C19, and 3A4. This conjunctive analysis supports continued evaluation of this compound in definitive pre-IND studies and exemplifies our strategy supporting the discovery of novel agents to treat diseases of the developing world.


Subject(s)
Antimalarials/pharmacokinetics , Pyridones/pharmacokinetics , Animals , Antimalarials/blood , Biological Transport , Biotransformation , Cell Line , Cell Membrane Permeability , Cytochrome P-450 Enzyme Inhibitors , Dogs , Drug Evaluation, Preclinical , Electron Transport Complex III/antagonists & inhibitors , Female , Hepatocytes/metabolism , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred Strains , Microsomes, Liver/metabolism , Protein Binding , Pyridones/blood , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL