Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nutrients ; 11(2)2019 Feb 15.
Article in English | MEDLINE | ID: mdl-30781364

ABSTRACT

Dysregulation of epidermal growth factor receptor (EGFR)/human epidermal growth factor-2 (HER2) family is a hallmark of aggressive breast cancer. Small-molecule tyrosine kinase inhibitors are among the most effective cancer targeted treatments. (-)-Oleocanthal (OC) is a naturally occurring phenolic secoiridoid lead from extra-virgin olive oil with documented anti-cancer activities via targeting mesenchymal epithelial transition factor (c-Met). Dysregulation of c-Met promotes aggressiveness to breast cancer-targeted therapies. Lapatinib (LP) is an FDA-approved dual EGFR/HER2 inhibitor for HER2-amplified breast cancer. HER2-Positive tumor cells can escape targeted therapies like LP effects by overexpressing c-Met. Combined OC-LP treatment is hypothesized to be mechanistically synergistic against HER2-overexpressing breast cancer. Combined sub-effective treatments of OC-LP resulted in synergistic anti-proliferative effects against the HER2-positive BT-474 and SK-BR-3 breast cancer cell lines, compared to OC or LP monotherapy. Antibody array and Western blot analysis showed that combined OC-LP treatment significantly inhibited EGFR, HER2, and c-Met receptor activation, as well as multiple downstream signaling proteins, compared to individual OC or LP treatment. OC-LP Combination significantly inhibited invasion and migration of breast cancer cells through reduced activation of focal adhesion kinase (FAK) and paxillin. Combined treatment of OC-10 mg/kg with LP-12.5 mg/kg suppressed more than 90% of BT-474 tumor cells growth in a nude mouse xenograft model, compared to individual OC or LP treatment. Activated c-Met, EGFR, HER2, and protein kinase B (AKT) were significantly suppressed in combination-treated mice tumors, compared to OC or LP monotherapy. This study reveals the OC future potential as combination therapy to sensitize HER2-overexpressing breast cancers and significantly reduce required doses of targeted HER family therapeutics.


Subject(s)
Aldehydes/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Lapatinib/administration & dosage , Phenols/administration & dosage , Receptor, ErbB-2/drug effects , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cyclopentane Monoterpenes , Drug Therapy, Combination , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/drug effects , Female , Humans , Mice , Receptor, ErbB-2/metabolism
2.
J Nat Prod ; 80(4): 932-952, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28245124

ABSTRACT

(+)-Usnic acid (1) is a common bioactive lichen-derived secondary metabolite with a characteristic dibenzofuran scaffold. It displayed low micromolar antiproliferative activity levels and, notably, induced autophagy in a panel of diverse breast cancer cell lines, suggesting the mechanistic (formerly "mammalian") target of rapamycin (mTOR) as a potential macromolecular target. The cellular autophagic markers were significantly upregulated due to the inhibition of mTOR downstream effectors. Additionally, 1 showed an optimal binding pose at the mTOR kinase pocket aided by multiple interactions to critical amino acids. Rationally designed benzylidene analogues of 1 displayed excellent fitting into a targeted deep hydrophobic pocket at the core of the kinase cleft, through stacking with the phenolic side chain of the Tyr2225 residue. Several potent analogues were generated, including 52, that exhibited potent (nM concentrations) antiproliferative, antimigratory, and anti-invasive activities against cells from multiple breast cancer clonal lines, without affecting the nontumorigenic MCF-10A mammary epithelial cells. Analogue 52 also exhibited potent mTOR inhibition and autophagy induction. Furthermore, 52 showed potent in vivo antitumor activity in two athymic nude mice breast cancer xenograft models. Collectively, usnic acid and analogues are potential lead mTOR inhibitors appropriate for future use to control breast malignancies.


Subject(s)
Benzofurans/chemical synthesis , Benzofurans/pharmacology , Benzylidene Compounds/pharmacology , Breast Neoplasms/drug therapy , Sirolimus/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Benzofurans/chemistry , Benzylidene Compounds/chemistry , Cell Proliferation/drug effects , Crystallography, X-Ray , Disease Models, Animal , Female , Humans , Lichens/chemistry , Mice , Mice, Nude , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Kinase Inhibitors/chemistry , TOR Serine-Threonine Kinases/drug effects , Xenograft Model Antitumor Assays
3.
Biochem Pharmacol ; 126: 51-68, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27940262

ABSTRACT

Epidermal growth factor receptor (EGFR) is a key player in proliferation and metastasis of various cancers. Discovery of novel EGFR inhibitors is still an urgent clinical oncology unmet need. Pachycladins are eunicellin-based diterpenoids isolated from the soft coral Cladiella pachycladous species. This study evaluated the anticancer activity of pachycladins A-E against diverse breast and cervical cancer cells. Pachycladin A (1) potently inhibited the proliferation of multiple cancer cell lines, without being cytotoxic to non-cancerous cells. The antiproliferative activity of 1 is mediated through cytostatic mechanisms rather than inducing apoptosis, as evidenced by lack of TUNEL response. Additionally, 1 arrested cell cycle in either G1 or G2/M phase, according to the cancer type, which induced caspase-dependent and independent apoptosis only after prolonged treatment. Meanwhile, 1 potently decreased microvessel formation and endothelial cell migration, suggesting its potential antiangiogenic activity. Different kinase profiling platforms revealed the exquisite potency and selectivity of 1 towards EGFR, even compared to other members of the EGFR family. In cancer cells, the antiproliferative activity of 1 was associated with suppression of EGFR activation and its downstream effectors. Interestingly, 1 significantly inhibited the drug-resistant T790M EGFR mutant, which is believed to be an attractive feature of EGFR inhibitors. Docking studies characterized the structural determinants required for efficient wild and mutant EGFR inhibition. Overlay studies of 1 with known EGFR inhibitors provided future guidance to chemically improve its binding affinity. Together, the anticancer activity of 1 is mediated by direct effects on tumor growth and angiogenesis, selectively via deactivating EGFR signaling, providing an excellent scaffold to control EGF-dependent cancers.


Subject(s)
Anthozoa/chemistry , Antineoplastic Agents/pharmacology , Diterpenes/pharmacology , ErbB Receptors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Breast Neoplasms , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Diterpenes/chemistry , Drug Screening Assays, Antitumor , ErbB Receptors/genetics , Female , Humans , Models, Molecular , Mutation , Neovascularization, Physiologic/drug effects , Protein Binding , Signal Transduction , Structure-Activity Relationship , Uterine Cervical Neoplasms
4.
Oncotarget ; 7(41): 67551-67573, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27563824

ABSTRACT

Hodgkin lymphoma (HL) is a lymphoid malignancy that is typically derived from germinal-center B cells. EBV infection, mutations in NF-κB pathway genes, and genetic susceptibility are known risk factors for developing HL. CD30 and NF-κB have been identified as potential biomarkers in pediatric HL patients, and these molecules may represent therapeutic targets. Although current risk adapted and response based treatment approaches yield overall survival rates of >95%, treatment of relapse or refractory patients remains challenging. Targeted HL therapy with the antibody-drug conjugate Brentuximab vedotin (Bv) has proven to be superior to conventional salvage chemotherapy and clinical trials are being conducted to incorporate Bv into frontline therapy that substitutes Bv for alkylating agents to minimize secondary malignancies. The appearance of secondary malignancies has been a concern in pediatric HL, as these patients are at highest risk among all childhood cancer survivors. The risk of developing secondary leukemia following childhood HL treatment is 10.4 to 174.8 times greater than the risk in the general pediatric population and the prognosis is significantly poorer than the other hematological malignancies with a mortality rate of nearly 100%. Therefore, identifying clinically valuable biomarkers is of utmost importance to stratify and select patients who may or may not need intensive regimens to maintain optimal balance between maximal survival rates and averting late effects. Here we discuss epidemiology, risk factors, staging, molecular and genetic prognostic biomarkers, treatment for low and high-risk patients, and the late occurrence of secondary malignancies in pediatric HL.


Subject(s)
Hodgkin Disease , Adolescent , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Child , Child, Preschool , Clinical Trials as Topic , Female , Humans , Male , Translational Research, Biomedical
5.
Eur J Med Chem ; 118: 299-315, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27258622

ABSTRACT

Dysregulated receptor tyrosine kinase c-Met and its ligand HGF is valid and attractive molecular target for therapeutic blockade in cancer. Inspired by the chemical structure of the naturally occurring olive secoiridoid (-)-oleocanthal (1) and its documented anticancer activity against c-Met-dependent malignancies, a previous study reported tyrosol sinapate (4) as a c-Met inhibitor hit. This study reports additional semisynthetic optimization and SAR of 4 to improve its selective activity against c-Met-dependent breast cancer by increasing its capacity to inhibit c-Met phosphorylation. Forty-three compounds (5-47) were synthesized, among which the novel analog homovanillyl sinapate (HVS-16) was distinguished for its remarkable activity. HVS-16 substantially impaired c-Met-mediated proliferation, migration, and invasion across human breast cancer cell lines in two- and three-dimensional culture systems, while similar treatment doses were found to have effect neither on the non-tumorigenic human mammary epithelial cell growth nor on the c-Met independent breast cancer cell viability. HVS-16 showed a dose-dependent inhibition of ligand-mediated c-Met activation in human breast cancer cells. Docking studies revealed that HVS-16 fits very well inside c-Met crystal structures, satisfying critical interactions at the ATP binding site. This study identified important structural pharmacophoric features in HVS-16 and correlated its postulated binding pose with c-Met kinase assay data that would guide future olive secoiridoid bioisostere lead design. Results presented herein suggest HVS-16 as a promising c-Met inhibitor validated hit with potential to control invasive breast malignancies with aberrant c-Met activity.

6.
Oncotarget ; 7(22): 32247-73, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27086914

ABSTRACT

The hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (c-Met) signaling axis has gained considerable attention as an attractive molecular target for therapeutic blockade of cancer. Inspired by the chemical structure of S (-)-oleocanthal, a natural secoiridoid from extra-virgin olive oil with documented anticancer activity against c-Met-dependent malignancies, the research presented herein reports on the discovery of the novel olive-derived homovanillyl sinapate (HVS) as a promising c-Met inhibitor. HVS was distinguished for its remarkable potency against wild-type c-Met and its oncogenic variant in cell-free assays and confirmed by in silico docking studies. Furthermore, HVS substantially impaired the c-Met-mediated growth across a broad spectrum of breast cancer cells, while similar treatment doses had no effect on the non-tumorigenic mammary epithelial cell growth. In addition, HVS caused a dose-dependent inhibition of HGF-induced, but not epidermal growth factor (EGF)-induced, cell scattering in addition to HGF-mediated migration, invasion, and 3-dimensional (3D) proliferation of tumor cell spheroids. HVS treatment effects were mediated via inhibition of ligand-mediated c-Met activation and its downstream mitogenic signaling and blocking molecular mediators involved in cellular motility across different cellular contexts. An interesting feature of HVS is its good selectivity for c-Met and Abelson murine leukemia viral oncogene homolog 1 (ABL1) when profiled against a panel of kinases. Docking studies revealed interactions likely to impart high dual affinity for both ABL1 and c-Met kinases. HVS markedly reduced tumor growth, showed excellent pharmacodynamics, and suppressed cell proliferation and microvessel density in an orthotopic model of triple negative breast cancer. Collectively, the present findings suggested that the oleocanthal-based HVS is a promising c-Met inhibitor lead entity with excellent therapeutic potential to control malignancies with aberrant c-Met activity.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Coumaric Acids/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Binding Sites , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Catalysis , Cell Movement/drug effects , Cell Proliferation/drug effects , Coumaric Acids/chemistry , Coumaric Acids/metabolism , Dose-Response Relationship, Drug , Female , Humans , MCF-7 Cells , Male , Mice, Nude , Molecular Docking Simulation , Molecular Structure , Neoplasm Invasiveness , Phosphorylation , Point Mutation , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Binding , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/drug effects , Spheroids, Cellular , Structure-Activity Relationship , Time Factors , Xenograft Model Antitumor Assays
7.
Oncotarget ; 7(28): 44735-44762, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27007053

ABSTRACT

Fibroblast growth factor (FGF) signaling is essential for normal and cancer biology. Mammalian FGF family members participate in multiple signaling pathways by binding to heparan sulfate and FGF receptors (FGFR) with varying affinities. FGF2 is the prototype member of the FGF family and interacts with its receptor to mediate receptor dimerization, phosphorylation, and activation of signaling pathways, such as Ras-MAPK and PI3K pathways. Excessive mitogenic signaling through the FGF/FGFR axis may induce carcinogenic effects by promoting cancer progression and increasing the angiogenic potential, which can lead to metastatic tumor phenotypes. Dysregulated FGF/FGFR signaling is associated with aggressive cancer phenotypes, enhanced chemotherapy resistance and poor clinical outcomes. In vitro experimental settings have indicated that extracellular FGF2 affects proliferation, drug sensitivity, and apoptosis of cancer cells. Therapeutically targeting FGF2 and FGFR has been extensively assessed in multiple preclinical studies and numerous drugs and treatment options have been tested in clinical trials. Diagnostic assays are used to quantify FGF2, FGFRs, and downstream signaling molecules to better select a target patient population for higher efficacy of cancer therapies. This review focuses on the prognostic significance of FGF2 in cancer with emphasis on therapeutic intervention strategies for solid and hematological malignancies.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Hematologic Neoplasms/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Precision Medicine/methods , Animals , Antineoplastic Agents/therapeutic use , Fibroblast Growth Factor 2/antagonists & inhibitors , Hematologic Neoplasms/drug therapy , Humans , Molecular Targeted Therapy/methods , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction/drug effects
8.
Phytother Res ; 30(4): 557-66, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26744260

ABSTRACT

Breast cancer is a major health problem affecting the female population worldwide. The triple-negative breast cancers (TNBCs) are characterized by malignant phenotypes, worse patient outcomes, poorest prognosis, and highest mortality rates. The proto-oncogenic receptor tyrosine kinase c-Met is usually dysregulated in TNBCs, contributing to their oncogenesis, tumor progression, and aggressive cellular invasiveness that is strongly linked to tumor metastasis. Therefore, c-Met is proposed as a promising candidate target for the control of TNBCs. Lichens-derived metabolites are characterized by their structural diversity, complexity, and novelty. The chemical space of lichen-derived metabolites has been extensively investigated, albeit their biological space is still not fully explored. The anticancer-guided fractionation of Usnea strigosa (Ach.) lichen extract led to the identification of the depsidone-derived norstictic acid as a novel bioactive hit against breast cancer cell lines. Norstictic acid significantly suppressed the TNBC MDA-MB-231 cell proliferation, migration, and invasion, with minimal toxicity to non-tumorigenic MCF-10A mammary epithelial cells. Molecular modeling, Z'-LYTE biochemical kinase assay and Western blot analysis identified c-Met as a potential macromolecular target. Norstictic acid treatment significantly suppressed MDA-MB-231/GFP tumor growth of a breast cancer xenograft model in athymic nude mice. Lichen-derived natural products are promising resources to discover novel c-Met inhibitors useful to control TNBCs.


Subject(s)
Lactones/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Salicylates/pharmacology , Triple Negative Breast Neoplasms/pathology , Usnea/chemistry , Animals , Cell Line, Tumor/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Female , Humans , Mice , Mice, Nude , Molecular Docking Simulation , Neoplasm Invasiveness , Proto-Oncogene Proteins c-met/metabolism , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
9.
Phytother Res ; 29(11): 1776-82, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26248874

ABSTRACT

The established anticancer and neuroprotective properties of oleocanthal combined with the reported role of mammalian target of rapamycin (mTOR) in cancer and Alzheimer's disease development encouraged us to examine the possibility that oleocanthal inhibits mTOR. To validate this hypothesis, we docked oleocanthal into the adenosine triphosphate binding pocket of a close mTOR protein homologue, namely, PI3K-γ. Apparently, oleocanthal shared nine out of ten critical binding interactions with a potent dual PIK3-γ/mTOR natural inhibitor. Subsequent experimental validation indicated that oleocanthal indeed inhibited the enzymatic activity of mTOR with an IC50 value of 708 nM. Oleocanthal inhibits the growth of several breast cancer cell lines at low micromolar concentration in a dose-dependent manner. Oleocanthal treatment caused a marked downregulation of phosphorylated mTOR in metastatic breast cancer cell line (MDA-MB-231). These results strongly indicate that mTOR inhibition is at least one of the factors of the reported anticancer and neuroprotective properties of oleocanthal.


Subject(s)
Aldehydes/pharmacology , Models, Molecular , Olive Oil , Phenols/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cyclopentane Monoterpenes , Female , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Sirolimus
10.
Oncotarget ; 6(30): 28693-715, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26293675

ABSTRACT

Syndecan-1 (SDC1, CD138) is a key cell surface adhesion molecule essential for maintaining cell morphology and interaction with the surrounding microenvironment. Deregulation of SDC1 contributes to cancer progression by promoting cell proliferation, metastasis, invasion and angiogenesis, and is associated with relapse through chemoresistance. SDC1 expression level is also associated with responses to chemotherapy and with prognosis in multiple solid and hematological cancers, including multiple myeloma and Hodgkin lymphoma. At the tissue level, the expression levels of SDC1 and the released extracellular domain of SDC1 correlate with tumor malignancy, phenotype, and metastatic potential for both solid and hematological tumors in a tissue-specific manner. The SDC1 expression profile varies among cancer types, but the differential expression signatures between normal and cancer cells in epithelial and stromal compartments are directly associated with aggressiveness of tumors and patient's clinical outcome and survival. Therefore, relevant biomarkers of SDC signaling may be useful for selecting patients that would most likely respond to a particular therapy at the time of diagnosis or perhaps for predicting relapse. In addition, the reciprocal expression signature of SDC between tumor epithelial and stromal compartments may have synergistic value for patient selection and the prediction of clinical outcome.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Syndecan-1/metabolism , Animals , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Prognosis , Signal Transduction , Syndecan-1/genetics , Tumor Microenvironment
11.
Mar Drugs ; 13(1): 288-311, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25580621

ABSTRACT

Receptor tyrosine kinases are key regulators of cellular growth and proliferation. Dysregulations of receptor tyrosine kinases in cancer cells may promote tumorigenesis by multiple mechanisms including enhanced cell survival and inhibition of cell death. Araguspongines represent a group of macrocyclic oxaquinolizidine alkaloids isolated from the marine sponge Xestospongia species. This study evaluated the anticancer activity of the known oxaquinolizidine alkaloids araguspongines A, C, K and L, and xestospongin B against breast cancer cells. Araguspongine C inhibited the proliferation of multiple breast cancer cell lines in vitro in a dose-dependent manner. Interestingly, araguspongine C-induced autophagic cell death in HER2-overexpressing BT-474 breast cancer cells was characterized by vacuole formation and upregulation of autophagy markers including LC3A/B, Atg3, Atg7, and Atg16L. Araguspongine C-induced autophagy was associated with suppression of c-Met and HER2 receptor tyrosine kinase activation. Further in-silico docking studies and cell-free Z-LYTE assays indicated the potential of direct interaction between araguspongine C and the receptor tyrosine kinases c-Met and HER2 at their kinase domains. Remarkably, araguspongine C treatment resulted in the suppression of PI3K/Akt/mTOR signaling cascade in breast cancer cells undergoing autophagy. Induction of autophagic death in BT-474 cells was also associated with decreased levels of inositol 1,4,5-trisphosphate receptor upon treatment with effective concentration of araguspongine C. In conclusion, results of this study are the first to reveal the potential of araguspongine C as an inhibitor to receptor tyrosine kinases resulting in the induction of autophagic cell death in breast cancer cells.


Subject(s)
Alkaloids/therapeutic use , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Breast Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinolizines/therapeutic use , Receptor, ErbB-2/antagonists & inhibitors , Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Female , Humans , Quinolizines/pharmacology , Signal Transduction/drug effects
12.
Chem Biol Drug Des ; 85(2): 231-43, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24954090

ABSTRACT

Bioassay-guided fractionation of Terminalia bentzoe L. leaves methanol extract identified the known triterpene oleanolic acid (1) as its major breast cancer cell migration inhibitor. Further chemical optimization afforded five new (9-12 and 15) and seven known (4-8, 13, and 14) semisynthetic analogues. All compounds were tested for their ability to inhibit human breast cancer MDA-MB-231 cells migration, proliferation, and invasion. The results revealed that 3-O-[N-(3'-chlorobenzenesulfonyl)-carbamoyl]-oleanolic acid (11) and 3-O-[N-(5'-fluorobenzenesulfonyl)-carbamoyl]-oleanolic acid (12) were the most active hits at low µM concentration. Western blot analysis indicated the activity of 1, 11, and 12 might be related, at least in part, to the suppression of Brk/Paxillin/Rac1 signaling pathway. Pharmacophore modeling study was conducted to better understand the common structural binding epitopes important for the antimigratory activity. The sulfonyl carbamoyl moiety with an optimal bulkiness electron-deficient phenyl ring is associated with improved activity. This study is the first to discover the antimigratory and anti-invasive activities of oleanolic acid and analogues through targeting the Brk/Paxillin/Rac1 axis.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Neoplasm Invasiveness/prevention & control , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Signal Transduction/drug effects , Breast/drug effects , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Drug Discovery , Female , Humans , Models, Molecular , Neoplasm Invasiveness/pathology , Neoplasm Proteins/metabolism , Paxillin/metabolism , Protein-Tyrosine Kinases/metabolism , Terminalia/chemistry , rac1 GTP-Binding Protein/metabolism
13.
Eur J Pharmacol ; 740: 209-17, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-25046839

ABSTRACT

Oleanolic acid (OA), a pentacyclic triterpene acid widely distributed in food and traditional herbal remedies, exhibits diverse therapeutic effects. OA has been subjected to various chemical modifications to optimize its anticancer effect. Among other analogs, 3-O-[N-(p-fluorobenzenesulfonyl)-carbamoyl]-oleanolic acid (PFOA) was semisynthesized from OA. This study evaluates the cytotoxic effects of PFOA on MDA-MB-231, MCF-7, BT-474, and T-47D human breast cancer cells. Acute treatment of PFOA inhibited breast cancer cell viability in a dose-dependent manner. Treatment of PFOA at cytotoxic doses significantly induced apoptosis in cancer cells as shown by flow cytometry analysis. Activation of apoptosis in MCF-7 and BT-474 cells seemed to be initiated through induction of Fas ligand, which resulted in activation of caspase-8 and PARP-1, whereas apoptosis in MDA-MB-231 cells was initiated by the activation of caspase-9, caspase-3 and PARP-1. The mechanism of apoptosis induction in T-47D involves activation of PARP-1. PFOA decreased the expression of EGFR, HER-2, MET and ERα in human breast cancer cell lines. These findings suggest that PFOA inhibits cell growth, activates apoptosis, and decreases the expression of key proteins involved in progression of breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Oleanolic Acid/analogs & derivatives , Sulfonamides/pharmacology , Apoptosis/drug effects , Breast Neoplasms , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , ErbB Receptors/metabolism , Estrogen Receptor alpha/metabolism , Female , Humans , Oleanolic Acid/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Receptor, ErbB-2/metabolism
14.
Org Biomol Chem ; 12(28): 5295-303, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-24927150

ABSTRACT

Breast and prostate cancers are among the most common cancers worldwide with devastating statistics for the metastatic, chemotherapy- and radiotherapy-resistant phenotypes. Novel therapies interfering with new and/or multiple pathways involved in the pathology of cancer are urgently needed. Preliminary results showed that the marine natural product Z-4-hydroxyphenylmethylene hydantoin (PMH, ) and its 4-ethylthio-analog (SEth, ) promoted tight junction formation and showed anti-invasive and anti-migratory activities in vitro against metastatic prostate cancer cells and inhibited tumor growth and micrometastases in distant organs in orthotopic and transgenic mice models. This study focuses on the design and synthesis of second-generation PMHs with enhanced antitumor activities. A series of substituted benzaldehydes was selected based on earlier SAR studies and reacted with hydantoin to yield 11 new compounds . Compounds were evaluated for their antiproliferative, antimigratory and anti-invasive properties in vitro against the human mammary and prostate cancer cell lines MDA-MB-231 and PC-3, respectively. A Western blot analysis of the most active analog showed its ability to suppress the expression of the total levels of c-Met and FAK, with subsequent reduction of their phosphorylated (activated) levels in MDA-MB-231 cells. In addition, also inhibited Brk, paxillin and Rac1 phosphorylation. was formulated using hydroxypropyl ß-cyclodextrin (HPCD) to improve its solubility and was further evaluated in a nude mice xenograft model using MDA-MB-231/GFP cells. PMH reduced breast tumor growth and suppressed Ki-67, CD31, p-Brk and p-FAK expression in tumor samples. Thus, is a potential lead for the control of invasive breast malignancies.


Subject(s)
Antineoplastic Agents/chemical synthesis , Biological Products/chemistry , Breast Neoplasms/drug therapy , Focal Adhesion Kinase 1/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Hydantoins/chemistry , Neoplasm Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzaldehydes/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Female , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Humans , Hydantoins/pharmacology , Male , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Paxillin/genetics , Paxillin/metabolism , Prostate/drug effects , Prostate/metabolism , Prostate/pathology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Xenograft Model Antitumor Assays , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
15.
PLoS One ; 9(5): e97622, 2014.
Article in English | MEDLINE | ID: mdl-24849787

ABSTRACT

Dysregulation of the Hepatocyte growth factor (HGF)/c-Met signaling axis upregulates diverse tumor cell functions, including cell proliferation, survival, scattering and motility, epithelial-to-mesenchymal transition (EMT), angiogenesis, invasion, and metastasis. (-)-Oleocanthal is a naturally occurring secoiridoid from extra-virgin olive oil, which showed antiproliferative and antimigratory activity against different cancer cell lines. The aim of this study was to characterize the intracellular mechanisms involved in mediating the anticancer effects of (-)-oleocanthal treatment and the potential involvement of c-Met receptor signaling components in breast cancer. Results showed that (-)-oleocanthal inhibits the growth of human breast cancer cell lines MDA-MB-231, MCF-7 and BT-474 while similar treatment doses were found to have no effect on normal human MCF10A cell growth. In addition, (-)-oleocanthal treatment caused a dose-dependent inhibition of HGF-induced cell migration, invasion and G1/S cell cycle progression in breast cancer cell lines. Moreover, (-)-oleocanthal treatment effects were found to be mediated via inhibition of HGF-induced c-Met activation and its downstream mitogenic signaling pathways. This growth inhibitory effect is associated with blockade of EMT and reduction in cellular motility. Further results from in vivo studies showed that (-)-oleocanthal treatment suppressed tumor cell growth in an orthotopic model of breast cancer in athymic nude mice. Collectively, the findings of this study suggest that (-)-oleocanthal is a promising dietary supplement lead with potential for therapeutic use to control malignancies with aberrant c-Met activity.


Subject(s)
Aldehydes/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Olea/chemistry , Phenols/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclopentane Monoterpenes , Epithelial-Mesenchymal Transition/drug effects , Female , Hepatocyte Growth Factor/pharmacology , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
16.
Mar Drugs ; 12(4): 2282-304, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24736807

ABSTRACT

Sipholenol A is a natural sipholane triterpenoid isolated from the Red Sea sponge, Callyspongia siphonella. Previous studies showed the antimigratory and antiproliferative activities of the semisynthetic sipholenol A esters against breast cancer cell lines. This study investigated the effects of sipholenol A-4-O-3',4'-dichlorobenzoate (SPA) on the growth, migration and invasion of diverse human breast cancer cells. Results showed that SPA inhibited the growth of the human breast cancer cells, MDA-MB-231, MCF-7, BT-474 and T-47D, in a dose-dependent manner. Immunofluorescent analysis showed that SPA significantly reduced Ki-67-positive cells in MDA-MB-231 cells. Flow cytometry and Western blot analyses revealed that SPA treatment suppressed MDA-MB-231 cell growth by inducing cell cycle arrest at the G1 phase. In addition, SPA suppressed breast cancer cell migration, invasion and decreased Brk and FAK activation in a dose-dependent manner. Molecular docking study suggested a perfect fitting at the FAK's FERM domain, inhibiting the main autophosphorylation site, Y397, which was further confirmed by Western blot analysis. Most known small molecule FAK inhibitors target the kinase domain, creating several off-target side effects. The in vivo studies showed that SPA treatment suppressed breast tumor growth and Ki-67, CD31, p-Brk and p-FAK expression in orthotopic breast cancer in nude mice. In conclusion, SPA inhibited the growth, invasion and migration of breast cancer cells possibly via deactivating Brk and FAK signaling, suggesting good potential for therapeutic use to control invasive breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Callyspongia/chemistry , Triterpenes/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Blotting, Western , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Dose-Response Relationship, Drug , Female , Flow Cytometry , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Mice , Mice, Nude , Molecular Docking Simulation , Neoplasm Invasiveness/prevention & control , Signal Transduction/drug effects , Triterpenes/chemistry , Triterpenes/isolation & purification
17.
Methods Mol Biol ; 1139: 259-68, 2014.
Article in English | MEDLINE | ID: mdl-24619686

ABSTRACT

Major histocompatibility complex (MHC) Class II-positive, invariant chain (Ii)-suppressed tumor cells induce both T helper and cytotoxic T lymphocytes' responses. Genetically controlled immunotherapy could be utilized for prophylactic vaccination of tumor-free individuals who are at high risk of developing tumor and can be therapeutic for treating established tumors that are nonresponsive to existing therapies. In this chapter, we provide practical methods to create a potent in vivo tumor cell vaccine by inducing MHC Class II and Ii using MHC Class II transactivator (CIITA) or interferon-gamma (IFN-γ) and subsequently inhibiting Ii by antisense oligonucleotides. We also describe the development of an adenoviral vector.


Subject(s)
Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Transformation, Neoplastic/genetics , Oligonucleotides, Antisense/genetics , Adenoviridae/genetics , Animals , Cell Line, Tumor , DNA, Recombinant/genetics , Electroporation , Enzyme Activation , Genetic Vectors/genetics , HLA-D Antigens/genetics , Humans , Interferon-gamma/genetics , Mice , Nuclear Proteins/genetics , Ribonuclease H/metabolism , Trans-Activators/genetics , Transfection , Vaccination
18.
Eur J Med Chem ; 73: 310-24, 2014 Feb 12.
Article in English | MEDLINE | ID: mdl-24487236

ABSTRACT

Sipholenol A, a triterpene isolated from the Red Sea sponge Callyspongia siphonella, was previously shown to reverse multidrug resistance in P-glycoprotein-overexpressing cancer cells. Moreover, sipholanes showed promising in vitro inhibitory effects against the invasion and migration of the metastatic human breast cancer cell line MDA-MB-231. The breast tumor kinase (Brk), a mediator of cancer cell phenotypes important for proliferation, survival, and migration, was proposed as a potential target. This study reports additional semisynthetic optimization of sipholenol A esters to improve the breast cancer antimigratory and antiproliferative activities as well as Brk phosphorylation inhibition. Fifteen new sipholenol A analogs (25-39) were semisynthesized. Sipholenol A 4ß-4',5'-dichlorobenzoate ester (29) was the most potent, with an IC50 value of 1.3 µM in the migration assay. The level of Brk phosphorylation inhibition of 29 was assessed using the Z'-LYTE™ kinase assay and Western blot analysis. Active analogs showed no toxicity on the non-tumorigenic epithelial breast cell line MCF10A at doses equal to their IC50 values or higher in migration and proliferation assays, suggesting their selectivity towards malignant cells. Pharmacophore modeling and 3D-QSAR studies were conducted to identify important pharmacophoric features and correlate 3D-chemical structure with activity. These studies provided the evidence for future design of novel antimigratory compounds based on a simplified sipholane structure possessing rings A and B (perhydrobenzoxepine) connected to substituted aromatic esters, with the elimination of rings C and D ([5,3,0]bicyclodecane system). This will enable the future synthesis of the new active entities feasibly and cost-effectively. These results demonstrate the potential of marine natural products for the discovery of novel scaffolds for the control and management of metastatic breast cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cell Movement/drug effects , Cell Proliferation/drug effects , Neoplasm Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Triterpenes/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Drug Design , Epithelial Cells/drug effects , Female , Humans , Models, Molecular , Molecular Structure , Phosphorylation , Quantitative Structure-Activity Relationship , Triterpenes/chemistry , Triterpenes/pharmacology , Triterpenes/toxicity
19.
Biofactors ; 40(1): 49-58, 2014.
Article in English | MEDLINE | ID: mdl-23804535

ABSTRACT

Vitamin E is a generic term that refers to a family of compounds that is further divided into two subgroups called tocopherols and tocotrienols. Although all natural forms of vitamin E display potent antioxidant activity, tocotrienols are significantly more potent than tocopherols in inhibiting tumor cell growth and viability, and anticancer activity of tocotrienols is mediated independently of their antioxidant activity. In addition, the anticancer effects of tocotrienols are observed using treatment doses that have little or no effect on normal cell function or viability. This review will summarize experimental studies that have identified the intracellular mechanism mediating the anticancer effects of tocotrienols. Evidence is also provided showing that combined treatment of tocotrienol with other cancer chemotherapies can result in a synergistic inhibition in cancer cell growth and viability. Taken together, these findings strongly indicate that tocotrienols may provide significant health benefits in the prevention and/or treatment of cancer when used either alone as monotherapy or in combination with other anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Tocotrienols/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/prevention & control , Cell Proliferation/drug effects , Drug Synergism , Female , Humans , Tocotrienols/therapeutic use
20.
Eur J Med Chem ; 59: 329-41, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23274571

ABSTRACT

The vitamin E family members γ- and δ-tocotrienols (2 and 3, respectively) are known natural products with documented anticancer activities. Redox-silent structural modifications, such as esterification, etherification and carbamoylation, of 2 and 3 significantly enhanced their anticancer activities. However, hit-to-lead optimization of tocotrienols and their analogs was yet to be reported at the outset of the project described herein. Subjecting the chroman ring of 2 and 3 to the electrophilic substitution reactions, namely, Mannich and Lederer-Manasse procedures, afforded 42 new products. These included the 3,4-dihydro-1,3-oxazines 3-29 and 35-44, Mannich bases 30-31, and the hydroxymethyl analogs 32-34. Of these, the δ-tocotrienol analogs 8, 11, 18, 24, 25, 27, and 40 inhibited the proliferation of the highly metastatic +SA mammary epithelial cancer cell line, with IC(50) values in the nanomolar (nM) range. In NCI's 60 human tumor cell line panel, 8, 17, 38, and 40 showed antiproliferative activity, with nM GI(50) values. The δ-tocotrienol analogs 10 and 38 inhibited the migration of the highly metastatic human breast cancer cell line MDA-MB-231 with IC(50) values of 1.3 and 1.5 µM, respectively, in the wound-healing assay. A dose of 0.5 mg/day for 14 days of one of the active analogs, 30, significantly slowed the growth of +SA mammary tumors in the syngeneic BALB/c mouse model, compared to the vehicle- and the parent γ-tocotrienol-treated control groups. Electrophilic substitution reactions promoted tocotrienols to lead level and can enable their future use to control metastatic breast malignancies.


Subject(s)
Tocotrienols/chemistry , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Mice , Mice, Inbred BALB C , Plant Oils/chemistry , Tocotrienols/chemical synthesis , Tocotrienols/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...