Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Chem Res Toxicol ; 34(9): 2145-2156, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34472326

ABSTRACT

Cytochrome P450 enzymes (CYPs) play an important role in bioactivating or detoxifying polycyclic aromatic hydrocarbons (PAHs), common environmental contaminants. While it is widely accepted that exposure to PAHs induces CYPs, effectively increasing rates of xenobiotic metabolism, dose- and time-response patterns of CYP induction are not well-known. In order to better understand dose- and time-response relationships of individual CYPs following induction, we exposed B6129SF1/J mice to single or repeated doses (2-180 µmol/kg/d) of benzo[a]pyrene (BaP) or Supermix-10, a mixture of the top 10 most abundant PAHs found at the Portland Harbor Superfund Site. In hepatic microsomes from exposed mice, we measured amounts of active CYPs using activity-based protein profiling and total CYP expression using global proteomics. We observed rapid Cyp1a1 induction after 6 h at the lowest PAH exposures and broad induction of many CYPs after 3 daily PAH doses at 72 h following the first dose. Using samples displaying Cyp1a1 induction, we observed significantly higher metabolic affinity for BaP metabolism (Km reduced 3-fold), 3-fold higher intrinsic clearance, but no changes to the Vmax. Mice dosed with the highest PAH exposures exhibited 1.7-5-fold higher intrinsic clearance rates for BaP compared to controls and higher Vmax values indicating greater amounts of enzymes capable of metabolizing BaP. This study demonstrates exposure to PAHs found at superfund sites induces enzymes in dose- and time-dependent patterns in mice. Accounting for specific changes in enzyme profiles, relative rates of PAH bioactivation and detoxification, and resulting risk will help translate internal dosimetry of animal models to humans and improve risk assessments of PAHs at superfund sites.


Subject(s)
Benzo(a)pyrene/metabolism , Cytochrome P-450 Enzyme System/metabolism , Liver/metabolism , Animals , Female , Liver/enzymology , Mice , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Proteome/metabolism , Proteomics
2.
Chem Res Toxicol ; 32(6): 1259-1267, 2019 06 17.
Article in English | MEDLINE | ID: mdl-30938511

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are ubiquitous environmental contaminants generated from combustion of carbon-based matter. Upon ingestion, these molecules can be bioactivated by cytochrome P450 monooxygenases to oxidized toxic metabolites. Some of these metabolites are potent carcinogens that can form irreversible adducts with DNA and other biological macromolecules. Conjugative enzymes, such as glutathione S-transferases or UDP-glucuronosyltransferases, are responsible for the detoxification and/or facilitate the elimination of these carcinogens. While responses to PAH exposures have been extensively studied for the bioactivating cytochrome P450 enzymes, much less is known regarding the response of glutathione S-transferases in mammalian systems. In this study, we investigated the expression and activity responses of murine hepatic glutathione S-transferases to benzo[ a]pyrene exposure using global proteomics and activity-based protein profiling for chemoproteomics, respectively. Using this approach, we identified several enzymes exhibiting increased activity including GSTA2, M1, M2, M4, M6, and P1. The activity of one GST enzyme, GSTA4, was found to be downregulated with increasing B[ a]P dose. Activity responses of several of these enzymes were identified as being expression-independent when comparing global and activity-based data sets, possibly alluding to as of yet unknown regulatory post-translational mechanisms.


Subject(s)
Benzo(a)pyrene/pharmacology , Glutathione Transferase/metabolism , Animals , Benzo(a)pyrene/chemistry , Enzyme Induction/drug effects , Female , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred Strains , Molecular Probes/chemistry , Molecular Structure , Proteomics , RNA, Messenger/drug effects , RNA, Messenger/metabolism
3.
Toxicol Lett ; 269: 23-32, 2017 Mar 05.
Article in English | MEDLINE | ID: mdl-28119020

ABSTRACT

Polycyclic aromatic hydrocarbons (PAHs) are contaminants that are ubiquitously found in the environment, produced through combustion of organic matter or petrochemicals, and many of which are procarcinogens. The prototypic PAH, benzo[a]pyrene (B[a]P) and the highly carcinogenic dibenzo[def,p]chrysene (DBC) are metabolically activated by isoforms of the P450 enzyme superfamily producing benzo[a]pyrene-7,8-dihydrodiol (B[a]P diol), dibenzo[def,p]chrysene-11,12 diol (DBC diol). Each of these diols can be further metabolized by cytochrome P450 enzymes to highly reactive diol-epoxide metabolites that readily react with DNA or by phase II conjugation facilitating excretion. To complement prior in vitro metabolism studies with parent B[a]P and DBC, both phase I metabolism and phase II glucuronidation of B[a]P diol and DBC diol were measured in hepatic microsomes from female B6129SF1/J mice, male Sprague-Dawley rats, and female humans. Metabolic parameters, including intrinsic clearance and Michaelis-Menten kinetics were calculated from substrate depletion data. Mice and rats demonstrated similar B[a]P diol phase I metabolic rates. Compared to rodents, human phase I metabolism of B[a]P diol demonstrated lower overall metabolic capacity, lower intrinsic clearance at higher substrate concentrations (>0.14µM), and higher intrinsic clearance at lower substrate concentrations (<0.07µM). Rates of DBC diol metabolism did not saturate in mice or humans and were highest overall in mice. Higher affinity constants and lower capacities were observed for DBC diol glucuronidation compared to B[a]P diol glucuronidation; however, intrinsic clearance values for these compounds were consistent within each species. Kinetic parameters reported here will be used to extend physiologically based pharmacokinetic (PBPK) models to include the disposition of B[a]P and DBC metabolites in animal models and humans to support future human health risk assessments.


Subject(s)
Chrysenes/metabolism , Dihydroxydihydrobenzopyrenes/metabolism , Microsomes, Liver/drug effects , Animals , Benzo(a)pyrene/metabolism , Carcinogens/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Female , Male , Mice , Microsomes, Liver/metabolism , Polycyclic Aromatic Hydrocarbons/metabolism , Rats , Rats, Sprague-Dawley , Risk Assessment , Toxicity Tests
4.
Methods Mol Biol ; 1406: 271-87, 2016.
Article in English | MEDLINE | ID: mdl-26820963

ABSTRACT

The study of pharmacokinetics (PK) and pharmacodynamics (PD) in cancer drug discovery and development is often paired and described in reciprocal terms, where PK is the analysis of the change in drug concentration with time and PD is the analysis of the biological effects of the drug at various concentrations over different time courses. While PK is defined by how a compound is absorbed, distributed, metabolized, and eliminated, PD refers to the measure of a compound's ability to interact with its intended target, leading to a biologic effect. Recent advances in anti-breast cancer drug discovery have resulted in several new drugs, but there is still a high attrition rate during clinical development. One reason for this failure is attributed to inappropriate correlation between the PK and PD parameters and subsequent extrapolation to human subjects. In this chapter, we describe the protocols of PK and PD studies in breast cancer models to assess the efficacy of an anti-breast cancer compound, noting the types and endpoints employed, and explain why it is important to link PK and PD in order to establish and evaluate dose/concentration-response relationships and subsequently describe and predict the effect-time courses for a given drug dose.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/pathology , Drug Discovery/methods , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/metabolism , Biological Transport , Cell Line, Tumor , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/metabolism , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Feces/chemistry , Female , Humans , Mice
5.
Med Res Rev ; 35(6): 1220-67, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26227500

ABSTRACT

Polycomb group (PcG) proteins are transcriptional repressors that regulate several crucial developmental and physiological processes in the cell. More recently, they have been found to play important roles in human carcinogenesis and cancer development and progression. The deregulation and dysfunction of PcG proteins often lead to blocking or inappropriate activation of developmental pathways, enhancing cellular proliferation, inhibiting apoptosis, and increasing the cancer stem cell population. Genetic and molecular investigations of PcG proteins have long been focused on their PcG functions. However, PcG proteins have recently been shown to exert non-classical-Pc-functions, contributing to the regulation of diverse cellular functions. We and others have demonstrated that PcG proteins regulate the expression and function of several oncogenes and tumor suppressor genes in a PcG-independent manner, and PcG proteins are associated with the survival of patients with cancer. In this review, we summarize the recent advances in the research on PcG proteins, including both the Pc-repressive and non-classical-Pc-functions. We specifically focus on the mechanisms by which PcG proteins play roles in cancer initiation, development, and progression. Finally, we discuss the potential value of PcG proteins as molecular biomarkers for the diagnosis and prognosis of cancer, and as molecular targets for cancer therapy.


Subject(s)
Neoplasms/physiopathology , Polycomb-Group Proteins/physiology , Gene Expression Regulation/physiology , Humans , Polycomb-Group Proteins/genetics , Transcription, Genetic/physiology
6.
Chin J Nat Med ; 13(7): 554-60, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26233847

ABSTRACT

We have recently designed and synthesized several novel iminoquinone anticancer agents that have entered preclinical development for the treatment of human cancers. Herein we developed and validated a quantitative HPLC-MS/MS analytical method for one of the lead novel anticancer makaluvamine analog, TCBA-TPQ, and conducted a pharmacokinetic study in laboratory rats. Our results indicated that the HPLC-MS/MS method was precise, accurate, and specific. Using this method, we carried out in vitro and in vivo evaluations of the pharmacological properties of TCBA-TPQ and plasma pharmacokinetics in rats. Our results provide a basis for future preclinical and clinical development of this promising anticancer marine analog.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Pyrroles/pharmacokinetics , Quinolones/pharmacokinetics , Tandem Mass Spectrometry/methods , Animals , Antineoplastic Agents/blood , Pyrroles/blood , Quinolones/blood , Rats, Sprague-Dawley
7.
Oncotarget ; 6(25): 21379-94, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26041888

ABSTRACT

The Mouse Double Minute 2 (MDM2) oncogene plays a critical role in cancer development and progression through p53-dependent and p53-independent mechanisms. Both natural and synthetic MDM2 inhibitors have been shown anticancer activity against several human cancers. We have recently identified a novel ginsenoside, 25-OCH3-PPD (GS25), one of the most active anticancer ginsenosides discovered thus far, and have demonstrated its MDM2 inhibition and anticancer activity in various human cancer models, including prostate cancer. However, the oral bioavailability of GS25 is limited, which hampers its further development as an oral anticancer agent. The present study was designed to develop a novel nanoparticle formulation for oral delivery of GS25. After GS25 was successfully encapsulated into PEG-PLGA nanoparticles (GS25NP) and its physicochemical properties were characterized, the efficiency of MDM2 targeting, anticancer efficacy, pharmacokinetics, and safety were evaluated in in vitro and in vivo models of human prostate cancer. Our results indicated that, compared with the unencapsulated GS25, GS25NP demonstrated better MDM2 inhibition, improved oral bioavailability and enhanced in vitro and in vivo activities. In conclusion, the validated nano-formulation for GS25 oral delivery improves its molecular targeting, oral bioavailability and anticancer efficacy, providing a basis for further development of GS25 as a novel agent for cancer therapy and prevention.


Subject(s)
Antineoplastic Agents/administration & dosage , Ginsenosides/administration & dosage , Nanoparticles/chemistry , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Caco-2 Cells , Cell Line, Tumor , Drug Carriers , Drug Delivery Systems , Ginsenosides/pharmacology , Humans , Male , Mice , Nanomedicine , Neoplasm Transplantation , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tissue Distribution , Tumor Suppressor Protein p53/metabolism
8.
Biomed Chromatogr ; 29(5): 654-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25294254

ABSTRACT

There is an increasing interest in targeting the MDM2 oncogene for cancer therapy. SP-141, a novel designed small molecule MDM2 inhibitor, exerts excellent in vitro and in vivo anticancer activity. To facilitate the preclinical development of this candidate anticancer agent, we have developed an HPLC method for the quantitative analysis of SP-141. The method was validated to be precise, accurate, and specific, with a linear range of 16.2-32,400 ng/mL in plasma, 16.2-6480 ng/mL in homogenates of brain, heart, liver, kidneys, lungs, muscle and tumor, and 32.4-6480 ng/mL in spleen homogenates. The lower limit of quantification was 16.2 ng/mL in plasma and all the tissue homogenates, except for spleen homogenates, where it was 32.4 ng/mL. The intra- and inter-assay precisions (coefficient of variation) were between 0.86 and 13.39%, and accuracies (relative errors) ranged from -8.50 to 13.92%. The relative recoveries were 85.6-113.38%. SP-141 was stable in mouse plasma, modestly plasma bound and metabolized by S9 microsomal enzymes. We performed an initial pharmacokinetic study in tumor-bearing nude mice, demonstrating that SP-141 has a short half-life in plasma and wide tissue distribution. In summary, this HPLC method can be used in future preclinical and clinical investigations of SP-141.


Subject(s)
Antineoplastic Agents/blood , Chromatography, High Pressure Liquid/methods , Indoles/blood , Neoplasms/drug therapy , Pyridines/blood , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Female , Half-Life , Humans , Indoles/administration & dosage , Indoles/pharmacokinetics , Mice , Mice, Nude , Neoplasms/blood , Pyridines/administration & dosage , Pyridines/pharmacokinetics , Tissue Distribution
9.
Med Res Rev ; 35(2): 225-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25164622

ABSTRACT

Ribosomes are essential components of the protein synthesis machinery. The process of ribosome biogenesis is well organized and tightly regulated. Recent studies have shown that ribosomal proteins (RPs) have extraribosomal functions that are involved in cell proliferation, differentiation, apoptosis, DNA repair, and other cellular processes. The dysfunction of RPs has been linked to the development and progression of hematological, metabolic, and cardiovascular diseases and cancer. Perturbation of ribosome biogenesis results in ribosomal stress, which triggers activation of the p53 signaling pathway through RPs-MDM2 interactions, resulting in p53-dependent cell cycle arrest and apoptosis. RPs also regulate cellular functions through p53-independent mechanisms. We herein review the recent advances in several forefronts of RP research, including the understanding of their biological features and roles in regulating cellular functions, maintaining cell homeostasis, and their involvement in the pathogenesis of human diseases. We also highlight the translational potential of this research for the identification of molecular biomarkers, and in the discovery and development of novel treatments for human diseases.


Subject(s)
Disease , Ribosomal Proteins/metabolism , Cell Nucleolus/metabolism , Humans , Mitochondria/metabolism , Ribosomes/metabolism , Stress, Physiological
10.
Curr Med Chem ; 22(2): 264-89, 2015.
Article in English | MEDLINE | ID: mdl-25386819

ABSTRACT

The activation of nuclear factor-kappaB (NFκB), a proinflammatory transcription factor, is a commonly observed phenomenon in breast cancer. It facilitates the development of a hormone-independent, invasive, high-grade, and late-stage tumor phenotype. Moreover, the commonly used cancer chemotherapy and radiotherapy approaches activate NFκB, leading to the development of invasive breast cancers that show resistance to chemotherapy, radiotherapy, and endocrine therapy. Inhibition of NFκB results in an increase in the sensitivity of cancer cells to the apoptotic effects of chemotherapeutic agents and radiation and restoring hormone sensitivity, which is correlated with increased disease-free survival in patients with breast cancer. In this review article, we focus on the role of the NFκB signaling pathways in the development and progression of breast cancer and the validity of NFκB as a potential target for breast cancer prevention and therapy. We also discuss the recent findings that NFκB may have tumor suppressing activity in certain cancer types. Finally, this review also covers the state-of-the-art development of NFκB inhibitors for cancer therapy and prevention, the challenges in targeting validation, and pharmacology and toxicology evaluations of these agents from the bench to the bedside.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/therapy , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Female , Humans , Inflammatory Breast Neoplasms/drug therapy , Inflammatory Breast Neoplasms/metabolism , Inflammatory Breast Neoplasms/prevention & control , Inflammatory Breast Neoplasms/therapy , Molecular Targeted Therapy , Signal Transduction
11.
Nat Commun ; 5: 5086, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25271708

ABSTRACT

A requirement for Mouse Double Minute 2 (MDM2) oncogene activation has been suggested to be associated with cancer progression and metastasis, including breast cancer. To date, most MDM2 inhibitors have been designed to block the MDM2-p53-binding interphase, and have low or no efficacy against advanced breast cancer with mutant or deficient p53. Here we use a high-throughput screening and computer-aided, structure-based rational drug design, and identify a lead compound, SP-141, which can directly bind to MDM2, inhibit MDM2 expression and induce its autoubiquitination and proteasomal degradation. SP-141 has strong in vitro and in vivo antibreast cancer activity, with no apparent host toxicity. While further investigation is needed, our data indicate that SP-141 is a novel targeted therapeutic agent that may especially benefit patients with advanced disease.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Indoles/administration & dosage , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyridines/administration & dosage , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Drug Design , Female , Humans , Mice , Mice, Nude , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
12.
Oncotarget ; 5(22): 11604-19, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25344099

ABSTRACT

RYBP is a member of the polycomb group (PcG) proteins that typically act as transcriptional repressors via epigenetic modification of chromatin. The present study was designed to investigate the role of RYBP in HCC progression, chemosensitivity, and patient survival, and to explore the underlying molecular mechanism(s). In this study we investigated the expression of RYBP in 400 pairs of human HCC tissues and matched noncancerous samples. The effects of RYBP on HCC tumor growth and metastasis and chemosensitivity were determined both in vitro and in vivo. We herein demonstrate that the RYBP expression in HCC tissue samples was significantly lower than that in matched noncancerous liver tissues. Clinically, the low expression of RYBP was an independent predictor of a poor prognosis in patients with HCC. In in vitro HCC models, enforced RYBP expression inhibited cell growth and invasion, induced apoptosis, and increased the chemosensitivity of the cells, while RYBP knockdown led to the opposite effects. Furthermore, RYBP expression was induced by cisplatin, and adenovirus-mediated RYBP expression inhibited HCC tumor growth and sensitized HCC to conventional chemotherapy in vivo. Our results demonstrate that reactivating RYBP in cancer cells may provide an effective and safe therapeutic approach to HCC therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , Liver Neoplasms/metabolism , Adenoviridae/genetics , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Cisplatin/chemistry , Combined Modality Therapy , Disease Progression , Disease-Free Survival , Down-Regulation , Female , Hep G2 Cells , Humans , Liver Neoplasms/drug therapy , Mice , Mice, Nude , Multivariate Analysis , Neoplasm Transplantation , Prognosis , Repressor Proteins , Tissue Array Analysis , Treatment Outcome
13.
Article in English | MEDLINE | ID: mdl-25195025

ABSTRACT

In the present study, a specific and sensitive liquid chromatography-triple quadrupole mass spectrometry method was developed and validated for the determination of SP-141, a novel pyrido[b]indole anticancer agent. After a liquid-liquid extraction with n-hexane-dichloromethane-2-propanol (20:10:1, v/v/v) mixture, the analyte was separated on a Kinetex C18 column (50×2.1mm, 2.6µm) with mobile phases comprising of water (0.1% formic acid, v/v) and acetonitrile (0.1% formic acid, v/v) at a flow rate of 0.4mL/min. The test compound (SP-141) and the internal standard (SP-157) were analyzed in the multiple reaction-monitoring mode using the mass transitions m/z 325.1 → 282.0. The method was linear in the concentration range of 0.648-162ng/mL with coefficients of determination (R(2)) of 0.999 in mouse plasma. The lower limit of quantification was 0.648ng/mL. The intra- and inter-day assay precisions (coefficient of variation, %CV) were less than 4.2% and accuracies (relative error, %RE) ranged from -6.1% to 2.1%. The extraction recoveries were between 97.1 and 103.1% and the relative matrix effect was minimal. In addition, SP-141 was found to be stable in the plasma after three freeze-thaw cycles, at 37°C and 4°C for 24h, and at -80°C for 4 weeks. It was also stable in the stock solution at room temperature for 24h and after preparation in the autosampler for 36h. The validated method was successfully applied to an initial pharmacokinetic study of SP-141 in CD-1 mice following intraperitoneal and intravenous administrations.


Subject(s)
Antineoplastic Agents/blood , Chromatography, High Pressure Liquid/methods , Indoles/blood , Pyridines/blood , Tandem Mass Spectrometry/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Indoles/chemistry , Indoles/pharmacokinetics , Linear Models , Mice , Pyridines/chemistry , Pyridines/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
14.
Biochim Biophys Acta ; 1846(2): 297-311, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25072963

ABSTRACT

The NFAT signaling pathway regulates various aspects of cellular functions; NFAT acts as a calcium sensor, integrating calcium signaling with other pathways involved in development and growth, immune response, and inflammatory response. The NFAT family of transcription factors regulates diverse cellular functions such as cell survival, proliferation, migration, invasion, and angiogenesis. The NFAT isoforms are constitutively activated and overexpressed in several cancer types wherein they transactivate downstream targets that play important roles in cancer development and progression. Though the NFAT family has been conclusively proved to be pivotal in cancer progression, the different isoforms play distinct roles in different cellular contexts. In this review, our discussion is focused on the mechanisms that drive the activation of various NFAT isoforms in cancer. Additionally, we analyze the potential of NFAT as a valid target for cancer prevention and therapy.


Subject(s)
NFATC Transcription Factors/physiology , Neoplasms/pathology , Animals , Cell Proliferation , Cell Transformation, Neoplastic , Disease Progression , Homeostasis , Humans , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/genetics , Neoplasm Metastasis , Neoplasms/drug therapy , Protein Isoforms , Signal Transduction , Tumor Microenvironment
15.
Eur J Med Chem ; 68: 473-481, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24044895

ABSTRACT

Four new sesquiterpenoid dimers (lineariifolianoids E-H, 1-4), five new sesquiterpenoids (5-9), and seven known sesquiterpenoids (10-16) were isolated from the aerial parts of Inula lineariifolia Turcz. Their structures were determined by spectroscopic data analysis and X-ray diffraction studies. The compounds were then evaluated for their in vitro cytotoxicity against two human breast cancer cell lines (MCF-7 and MDA-MB-231) and one normal breast cell line (MCF-10A). Lineariifolianoid E (1) showed IC50 values of 1.56 µM and 2.75 µM against MCF-7 and MDA-MB-231, respectively. However, lineariifolianoid E demonstrated low toxicity to MCF-10A cells, which indicated a selective cytotoxicity for tumor cells. Further studies also presented that lineariifolianoid E had significant, dose-dependent effects on cell cycle progression and apoptosis in breast cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Inula/chemistry , Plant Components, Aerial/chemistry , Plant Extracts/pharmacology , Terpenes/pharmacology , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Female , Humans , Molecular Structure , Terpenes/chemistry , Terpenes/isolation & purification
16.
J Biomed Res ; 27(4): 254-71, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23885265

ABSTRACT

The p53 tumor suppressor is a key transcription factor regulating cellular pathways such as DNA repair, cell cycle, apoptosis, angiogenesis, and senescence. It acts as an important defense mechanism against cancer onset and progression, and is negatively regulated by interaction with the oncoprotein MDM2. In human cancers, the TP53 gene is frequently mutated or deleted, or the wild-type p53 function is inhibited by high levels of MDM2, leading to downregulation of tumor suppressive p53 pathways. Thus, the inhibition of MDM2-p53 interaction presents an appealing therapeutic strategy for the treatment of cancer. However, recent studies have revealed the MDM2-p53 interaction to be more complex involving multiple levels of regulation by numerous cellular proteins and epigenetic mechanisms, making it imperative to reexamine this intricate interplay from a holistic viewpoint. This review aims to highlight the multifaceted network of molecules regulating the MDM2-p53 axis to better understand the pathway and exploit it for anticancer therapy.

17.
Curr Cancer Drug Targets ; 13(5): 519-41, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23597193

ABSTRACT

MicroRNAs (miRNAs) are endogenous small non-coding RNAs that regulate gene expression by binding to the 3´untranslated region of target mRNA, resulting in posttranscriptional gene silencing via mRNA degradation or translation inhibition. miRNAs are involved in many biological processes including carcinogenesis. They can act as oncogenes or tumor suppressors and their aberrant expressions are intimately linked with cancer development and progression. Therefore, miRNAs have been utilized as potential biomarkers for cancer diagnosis, prognosis, as well as cancer therapeutic targets. Recently, it has been demonstrated that dietary and natural chemopreventive agents exert their anticancer activities through the regulation of one or more miRNAs. In addition to expounding the latest findings of miRNAs in cancer, this review also discusses the recent efforts on the translational research of miRNAs, with an emphasis on natural products in the treatment of cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Biological Products/therapeutic use , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Molecular Targeted Therapy , Neoplasms/prevention & control , Phytotherapy , Animals , Biomarkers, Tumor/antagonists & inhibitors , Humans , Neoplasms/genetics , RNA Interference
18.
Curr Cancer Drug Targets ; 13(5): 596-609, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23597194

ABSTRACT

Natural products with biodiversity and chemical variations present a rich source for the discovery and development of new therapeutic and preventive drugs. Bioactive components derived from natural medicines including traditional Chinese medicine have been widely used for the screening of effective and safe anticancer drugs. Meanwhile, the investigation on mechanism of action (MOA) of natural bioactive components has a critical role in identifying and validating new molecular targets of those anticancer agents. Considering the high complexity of pharmacodynamic (PD) and pharmacokinetic (PK) characteristics of natural product anticancer agents, there are several major challenges in understanding mechanisms of action in vitro and in vivo for these agents. The recent rapid progress made in molecular and cell biology, genetics and genomics, and translational medicine, preclinical investigations provides an impetus for a better understanding of mechanisms of action and structure-activity relationships (SAR) of natural products. In addition, the simultaneous evaluation of PD-PK characterizations would allow a full assessment of the safety, efficacy, and indication of natural product anticancer drugs in various regimens and in various clinical settings. In this review, we provide a brief summary for recent advances in translational pharmacology, focusing on target validation and PK-PD, MOA, and SAR. Several examples for clinically used agents, and cancer preventive agents and therapeutic agents under preclinical and clinical development are used to illustrate the importance of such translational research and challenges we are facing.


Subject(s)
Antineoplastic Agents/therapeutic use , Biological Products/therapeutic use , Biomarkers, Tumor/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Phytotherapy , Animals , Humans , Neoplasms/metabolism , Translational Research, Biomedical
19.
PLoS One ; 7(9): e44883, 2012.
Article in English | MEDLINE | ID: mdl-23028659

ABSTRACT

The purpose of the present study was to determine the in vitro and in vivo anti-cancer activity and pharmacological properties of 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide, KCN1. In the present study, we investigated the in vitro activity of KCN1 on cell proliferation and cell cycle distribution of pancreatic cancer cells, using the MTT and BrdUrd assays, and flow cytometry. The in vivo anti-cancer effects of KCN1 were evaluated in two distinct xenograft models of pancreatic cancer. We also developed an HPLC method for the quantitation of the compound, and examined its stability in mouse plasma, plasma protein binding, and degradation by mouse S9 microsomal enzymes. Furthermore, we examined the pharmacokinetics of KCN1 following intravenous or intraperitoneal injection in mice. Results showed that, in a dose-dependent manner, KCN1 inhibited cell growth and induced cell cycle arrest in human pancreatic cancer cells in vitro, and showed in vivo anticancer efficacy in mice bearing Panc-1 or Mia Paca-2 tumor xenografts. The HPLC method provided linear detection of KCN1 in all of the matrices in the range from 0.1 to 100 µM, and had a lower limit of detection of 0.085 µM in mouse plasma. KCN1 was very stable in mouse plasma, extensively plasma bound, and metabolized by S9 microsomal enzymes. The pharmacokinetic studies indicated that KCN1 could be detected in all of the tissues examined, most for at least 24 h. In conclusion, our preclinical data indicate that KCN1 is a potential therapeutic agent for pancreatic cancer, providing a basis for its future development.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Pancreatic Neoplasms/pathology , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Benzopyrans/blood , Benzopyrans/metabolism , Benzopyrans/pharmacokinetics , Biological Availability , Blood Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Stability , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Half-Life , Humans , Mice , Reproducibility of Results , Sulfonamides/blood , Sulfonamides/metabolism , Sulfonamides/pharmacokinetics , Temperature , Xenograft Model Antitumor Assays
20.
PLoS One ; 7(7): e41586, 2012.
Article in English | MEDLINE | ID: mdl-22911819

ABSTRACT

Although ginseng and related herbs have a long history of utility for various health benefits, their application in cancer therapy and underlying mechanisms of action are not fully understood. Our recent work has shown that 20(S)-25-methoxyl-dammarane-3ß, 12ß, 20-triol (25-OCH(3)-PPD), a newly identified ginsenoside from Panax notoginseng, exerts activities against a variety of cancer cells in vitro and in vivo. This study was designed to investigate its anti-breast cancer activity and the underlying mechanisms of action. We observed that 25-OCH(3)-PPD decreased the survival of breast cancer cells by induction of apoptosis and G1 phase arrest and inhibited the growth of breast cancer xenografts in vivo. We further demonstrated that, in a dose- and time-dependent manner, 25-OCH(3)-PPD inhibited MDM2 expression at both transcriptional and post-translational levels in human breast cancer cells with various p53 statuses (wild type and mutant). Moreover, 25-OCH(3)-PPD inhibited in vitro cell migration, reduced the expression of epithelial-to-mesenchymal transition (EMT) markers, and prevented in vivo metastasis of breast cancer. In summary, 25-OCH(3)-PPD is a potential therapeutic and anti-metastatic agent for human breast cancer through down-regulating MDM2. Further preclinical and clinical development of this agent is warranted.


Subject(s)
Biological Products/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Down-Regulation , Ginsenosides/therapeutic use , Proto-Oncogene Proteins c-mdm2/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biological Products/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Ginsenosides/pharmacology , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Protein Stability/drug effects , Proto-Oncogene Proteins c-mdm2/metabolism , Tetracycline/pharmacology , Time Factors , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...