Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Biomed Chromatogr ; 25(5): 628-634, 2011 May.
Article in English | MEDLINE | ID: mdl-20845374

ABSTRACT

We herein describe the development and validation of an HPLC method for the quantitation of 7-(benzylamino)-1,3,4,8-tetrahydropyrrolo [4,3,2-de]quinolin-8(1H)-one (BA-TPQ), a newly synthesized iminoquinone anticancer agent. BA-TPQ was extracted from plasma and tissue samples by first precipitating proteins with acetonitrile followed by a liquid-liquid extraction with ethyl acetate. Chromatographic separation was carried out using a gradient flow rate on a Zorbax SB C(18) column, and the effluent was monitored by UV detection at 346 nm. The method was found to be precise, accurate, and specific, with a linear range of 3.91-1955.0 ng/mL in plasma, 19.55-1955.0 ng/mL in spleen, brain, and liver homogenates and 19.55-3910.0 ng/mL in heart, lung and kidney homogenates. The method was stable under all relevant conditions. Using this method, we also carried out an initial study determining plasma pharmacokinetics and tissue distribution of BA-TPQ in mice following intravenous administration. In summary, this simple and sensitive HPLC method can be used in future preclinical and clinical studies of BA-TPQ.


Subject(s)
Chromatography, High Pressure Liquid/methods , Pyrroles/analysis , Pyrroles/pharmacokinetics , Quinolones/analysis , Quinolones/pharmacokinetics , Animals , Antineoplastic Agents/analysis , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Least-Squares Analysis , Male , Mice , Pyrroles/blood , Quinolones/blood , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
2.
Mar Drugs ; 8(7): 2129-41, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20714427

ABSTRACT

Marine natural products and their synthetic derivatives represent a major source of novel candidate anti-cancer compounds. We have recently tested the anti-cancer activity of more than forty novel compounds based on an iminoquinone makaluvamine scaffold, and have found that many of the compounds exert potent cytotoxic activity against human cancer cell lines. One of the most potent compounds, BA-TPQ [(11,12),7-(benzylamino)-1,3,4,8-tetrahydropyrrolo[4,3,2-de]quinolin-8(1H)-one], was active against a variety of human cancer cell lines, and inhibited the growth of breast and prostate xenograft tumors in mice. However, there was some toxicity noted in the mice following administration of the compound. In order to further the development of BA-TPQ, and in a search for potential sites of accumulation that might underlie the observed toxicity of the compound, we accomplished preclinical pharmacological studies of the compound. We herein report the in vitro and in vivo pharmacological properties of BA-TPQ, including its stability in plasma, plasma protein binding, metabolism by S9 enzymes, and plasma and tissue distribution. We believe these studies will be useful for further investigations, and may be useful for other investigators examining the use of similar compounds for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Pyrroles/pharmacology , Quinolones/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Neoplasms/pathology , Protein Binding , Pyrroles/pharmacokinetics , Pyrroles/toxicity , Quinolones/pharmacokinetics , Quinolones/toxicity , Tissue Distribution , Xenograft Model Antitumor Assays
3.
Invest New Drugs ; 28(3): 234-41, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19274441

ABSTRACT

We recently synthesized a series of novel makaluvamine compounds, and found that the most potent was FBA-TPQ. The effects of FBA-TPQ on human (LNCaP and PC3) and murine (TRAMP C1) prostate cancer cells were evaluated. Potential mechanisms of action of the compound were also determined. FBA-TPQ exhibited dose-dependent cytotoxicity in the low micromolar range, inhibited proliferation, caused cell cycle arrest, and induced apoptosis in prostate cancer cell lines. The compound also decreased the expression of the androgen receptor and PSA. The results presented herein support the further development of FBA-TPQ as a novel agent for prostate cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Pyrroles/therapeutic use , Quinolones/therapeutic use , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Prostate-Specific Antigen/metabolism , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Quinolones/chemical synthesis , Quinolones/pharmacology , Receptors, Androgen/metabolism
4.
Anticancer Agents Med Chem ; 9(8): 882-903, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19538162

ABSTRACT

The MDM2 oncogene is overexpressed in various human cancers. Its expression correlates with the phenotypes of high-grade, late-stage, and more resistant tumors. The auto-regulatory loop between MDM2 and the tumor suppressor p53 has long been considered the epitome of a rational target for cancer therapy. As such, many novel agents have been generated to interfere with the interaction of the two proteins, which results in the activation of p53. Among these agents are several small molecule inhibitors synthesized based upon the crystal structures of the MDM2-p53 complex. With use of high-throughput screening, several specific and effective agents for inhibition of the protein-protein interaction were discovered. Recent investigations, however, have demonstrated that many proteins regulate the MDM2-p53 interaction, and that MDM2 may have p53-independent oncogenic functions. In order for novel MDM2 inhibitors to be translated to the clinic, it is necessary to obtain a better understanding of the regulation of MDM2 and of the MDM2-p53 interaction. In particular, the implications of various interactions between certain regulator(s) and MDM2/p53 under different circumstances need to be elucidated to determine which pathway(s) represent the best targets for therapy. Targeting both MDM2 itself and regulators of MDM2 and the MDM2-p53 interaction, or use of MDM2 inhibitors in combination with conventional treatments, may improve prospects for tumor eradication.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Proto-Oncogene Proteins c-mdm2/genetics , Cell Cycle , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oncogenes , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism
5.
Med Chem ; 5(3): 227-36, 2009 May.
Article in English | MEDLINE | ID: mdl-19442212

ABSTRACT

The high mortality rate and lack of effective therapies make lung cancer an ideal target for novel therapeutic agents. The present study was designed to implement a novel chemical synthesis pathway and to determine the biological activities of synthetic makaluvamine analogs in human lung cancer. Seventeen compounds were synthesized and purified, and their chemical structures were elucidated on the basis of physicochemical constants and NMR spectra. Their in vitro activity was determined in human lung cancer cell lines. Based on initial screens, compound Ic was found to be the most potent, and was therefore used as a model for further studies in lung cancer cells. Ic induced both apoptosis and S-phase cell cycle arrest. Furthermore, it activated p53 and induced cleavage of PARP and caspases 8 and 9. Our preclinical data indicate that the makaluvamine analogs are potential therapeutic agents against lung cancer, providing a basis for further development of Ic (and perhaps other analogs) as a novel anti-cancer agent.


Subject(s)
Alkaloids/chemical synthesis , Alkaloids/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Lung Neoplasms/drug therapy , Quinolones/chemical synthesis , Quinolones/pharmacology , Alkaloids/chemistry , Alkaloids/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/pathology , Quinolones/chemistry , Quinolones/therapeutic use
6.
Mol Cell Pharmacol ; 1(1): 29-43, 2009.
Article in English | MEDLINE | ID: mdl-20333321

ABSTRACT

Inflammation is closely linked to cancer, and many anti-cancer agents are also used to treat inflammatory diseases, such as rheumatoid arthritis. Moreover, chronic inflammation increases the risk for various cancers, indicating that eliminating inflammation may represent a valid strategy for cancer prevention and therapy. This article explores the relationship between inflammation and cancer with an emphasis on epidemiological evidence, summarizes the current use of anti-inflammatory agents for cancer prevention and therapy, and describes the mechanisms underlying the anti-cancer effects of anti-inflammatory agents. Since monotherapy is generally insufficient for treating cancer, the combined use of anti-inflammatory agents and conventional cancer therapy is also a focal point in discussion. In addition, we also briefly describe future directions that should be explored for anti-cancer anti-inflammatory agents.

SELECTION OF CITATIONS
SEARCH DETAIL
...