Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Int J Cell Biol ; 2024: 2699572, 2024.
Article in English | MEDLINE | ID: mdl-38352698

ABSTRACT

Prolonged low-dose administration (PLDA) of several FDA-approved drugs for noncancer conditions or dietary compounds is associated with a lower incidence of specific types of cancers and with the lower formation of metastasis. However, the underlying mechanism is unknown; there is a discrepancy between the concentration of drugs needed to kill cancer cells in vitro and the actual serum levels (10 and >1000 times lower) found in patients. In this study, we evaluated the hypothesis that clonogenicity may be the target of PLDA. We compared the effect of nigericin (NIG) and menadione (MEN) on the human A549 and H460 lung and MCF-7 and MDA-MB-231 breast cancer cell lines using routine MTT and colony forming assays (CFA). The ability of both NIG and MEN to eliminate 100% of cancer cells was at least 2-10 times more potent in CFA compared to MTT assays. Our results revealed the existence of a short post-reattachment window of time when cancer cells growing at low density are more sensitive to PLDA of specific drugs likely by targeting clonogenic rather than proliferation pathways. This short ultrasensitive window of time (SUSWoT) was cell- and drug-type specific: the SUSWoT for NIG was present in H460, A549, and MDA-MB-231 cells but not evident in MCF-7 cells. Conversely, a similar SUSWoT for MEN was present in MCF-7, MDA-MD-231, and A549 cells but not evident in H460 cells. Our findings partially explain the decreased incidence of specific types of cancer by PLDA of FDA-approved drugs (or dietary compounds) for noncancer conditions.

2.
Cancer Biother Radiopharm ; 37(5): 364-371, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35076279

ABSTRACT

Background: Mold brachytherapy using high-energy ß--emitting radioisotopes is a promising treatment modality for skin cancers and keloids. Simple methodologies for consistent and stable incorporation of radionuclides into the matrix are desired for preparation of therapeutic sources. Methods: The authors report a facile strategy for the stable incorporation of Yttrium-90 (90Y) into amidoxime-functionalized polyacrylonitrile-polyvinylidene fluoride (PAN-PVDF) membranes. The strategy consisted of surface modification of PAN-PVDF membranes by reaction with hydroxylamine, characterization of the functionalized membranes, and optimization of experimental variables for maximum loading of 90Y onto the membranes. Quality control tests essential for confirming the suitability of the 90Y therapeutic sources for human application, such as uniformity of activity distribution, absence of leaching of activity, and estimation of surface contamination, were performed. Theoretical calculations to estimate the dose imparted by the 90Y therapeutic sources at varying depths of tissue were also carried out to predict the possible therapeutic outcome of treatment. Results: A facile method for large-scale preparation of 90Y-based mold brachytherapy sources could be established. Conclusions: The source fabrication methodology standardized in this work could be tailored for fabrication of custom-made 90Y sources for individualized treatment of superficial tumors, Bowen's disease, and keloids.


Subject(s)
Brachytherapy , Keloid , Skin Neoplasms , Brachytherapy/methods , Humans , Keloid/drug therapy , Radiometry/methods , Yttrium Radioisotopes/therapeutic use
3.
World J Stem Cells ; 13(7): 934-943, 2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34367485

ABSTRACT

The classical cancer stem cell (CSCs) theory proposed the existence of a rare but constant subpopulation of CSCs. In this model cancer cells are organized hierarchically and are responsible for tumor resistance and tumor relapse. Thus, eliminating CSCs will eventually lead to cure of cancer. This simplistic model has been challenged by experimental data. In 2010 we proposed a novel and controversial alternative model of CSC biology (the Stemness Phenotype Model, SPM). The SPM proposed a non-hierarchical model of cancer biology in which there is no specific subpopulation of CSCs in tumors. Instead, cancer cells are highly plastic in term of stemness and CSCs and non-CSCs can interconvert into each other depending on the microenvironment. This model predicts the existence of cancer cells ranging from a pure CSC phenotype to pure non-CSC phenotype and that survival of a single cell can originate a new tumor. During the past 10 years, a plethora of experimental evidence in a variety of cancer types has shown that cancer cells are indeed extremely plastic and able to interconvert into cells with different stemness phenotype. In this review we will (1) briefly describe the cumulative evidence from our laboratory and others supporting the SPM; (2) the implications of the SPM in translational oncology; and (3) discuss potential strategies to develop more effective therapeutic regimens for cancer treatment.

4.
BMJ Case Rep ; 12(7)2019 Jul 08.
Article in English | MEDLINE | ID: mdl-31289153

ABSTRACT

Sarcomatoid carcinoma is a rare clinical entity, especially when presenting in the nasopharynx. We describe the first documented case of nasopharyngeal sarcomatoid carcinoma with intracranial extension in a 59-year-old Caucasian man presenting with severe bifrontal headache and diplopia, secondary to left abducens nerve palsy. We highlight some of the major diagnostic challenges and describe its unusual histological appearance. We outline the importance of a multidisciplinary approach to his management, which includes input from the medicine, neurosurgery, Ear, Nose and Throat (ENT), pathology, radiology, oncology and respiratory teams. In the context of limited evidence, we then describe the rationale to proceed with induction chemotherapy followed by concurrent chemoradiotherapy. Although there was a partial response to treatment, it was not sufficient enough to allow subsequent surgical clearance. The plan going forward is to palliate with chemotherapy as and when the disease progresses.


Subject(s)
Chemoradiotherapy/methods , Induction Chemotherapy/methods , Nasopharyngeal Carcinoma/pathology , Sarcoma/pathology , Abducens Nerve Diseases/complications , Abducens Nerve Diseases/etiology , Diplopia/etiology , Headache/diagnosis , Headache/etiology , Humans , Male , Middle Aged , Nasopharyngeal Carcinoma/diagnostic imaging , Nasopharyngeal Carcinoma/therapy , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness , Patient Care Team/standards , Sarcoma/therapy , Tomography, X-Ray Computed , Watchful Waiting
5.
Stem Cells Int ; 2019: 6254269, 2019.
Article in English | MEDLINE | ID: mdl-30918522

ABSTRACT

The biguanides metformin (MET) and to a lesser extent buformin (BUF) have recently been shown to exert anticancer effects. In particular, MET targets cancer stem cells (CSCs) in a variety of cancer types but these compounds have not been extensively tested for combination therapy. In this study, we investigated in vitro the anticancer activity of MET and BUF alone or in combination with 2-deoxy-D-glucose (2-DG) and WZB-117 (WZB), which are a glycolysis and a GLUT-1 inhibitor, respectively, in H460 human lung cancer cells growing under three different culture conditions with varying degrees of stemness: (1) routine culture conditions (RCCs), (2) floating lung tumorspheres (LTSs) that are enriched for stem-like cancer cells, and (3) adherent cells under prolonged periods (8-12 days) of serum starvation (PPSS). These cells are highly resistant to conventional anticancer drugs such as paclitaxel, hydroxyurea, and colchicine and display an increased level of stemness markers. As single agents, MET, BUF, 2-DG, and WZB-117 potently inhibited the viability of cells growing under RCCs. Both MET and BUF showed a strong synergistic effect when used in combination with 2-DG. A weak potentiation was observed when used with WZB-117. Under RCCs, H460 cells were more sensitive to MET and BUF and WZB-117 compared to nontumorigenic Beas-2B cells. While LTSs were less sensitive to each single drug, both MET and BUF in combination with 2-DG showed a strong synergistic effect and reduced cell viability to similar levels compared to the parental H460 cells. Adherent cells growing under PPSS were also less sensitive to each single drug, and MET and BUF showed a strong synergistic effect on cell viability in combination with 2-DG. Overall, our data demonstrates that the combination of BGs with either 2-DG or WZB-117 has "broad-spectrum" anticancer activities targeting cells growing under a variety of cell culture conditions with varying degrees of stemness. These properties may be useful to overcome the chemoresistance due to intratumoral heterogeneity found in lung cancer.

6.
J Cell Physiol ; 234(4): 4445-4453, 2019 04.
Article in English | MEDLINE | ID: mdl-30191978

ABSTRACT

Three-dimensional (3D) culture systems such as floating spheroids (FSs) and floating tumorspheres (FTs) are widely used as tumor models of chemoresistance. FTs are considered to be enriched in cancer stem-like cells (CS-LCs). In this study, we used cancer cell lines (lung H460, prostate LnCAP, and breast MCF-7) able to form FSs under anchorage-independent conditions and compared with cell lines (prostate PC3 and breast MDA-MB-231) that cannot form FSs under similar conditions. Independent of their ability to form FTs all cell lines growing under anchorage-independent conditions become highly resistant to obatoclax, colchicine, and hydroxyurea. We used anti-E-cadherin antibody (that blocked the formation of FSs) and demonstrated that floating LnCAP cells showed similar chemoresistance regardless of the formation of spheroids. Our results demonstrate that the development of chemoresistance is not because of the formation of a complex 3D structure and/or enrichment of CS-LCs but is likely the result of cell detachment per se and their ability to survive under anchorage-independent conditions. We propose that FSs and FTs could be useful models to study chemoresistance of cancer cells associated with cell detachment (e.g., circulating tumor cells) but they may not be representative of other types of chemoresistance that arise in vivo in attached cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Adhesion , Drug Resistance, Neoplasm , Neoplasms/drug therapy , Antibodies/pharmacology , Antigens, CD/metabolism , Cadherins/antagonists & inhibitors , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Culture Techniques , Cell Survival/drug effects , Colchicine/pharmacology , Culture Media, Serum-Free , Female , Humans , Hydroxyurea/pharmacology , Indoles , MCF-7 Cells , Male , Neoplasms/metabolism , Neoplasms/pathology , PC-3 Cells , Phenotype , Pyrroles/pharmacology , Spheroids, Cellular
7.
Cancers (Basel) ; 10(10)2018 Sep 27.
Article in English | MEDLINE | ID: mdl-30262730

ABSTRACT

Ion homeostasis is extremely important for the survival of both normal as well as neoplastic cells. The altered ion homeostasis found in cancer cells prompted the investigation of several ionophores as potential anticancer agents. Few ionophores, such as Salinomycin, Nigericin and Obatoclax, have demonstrated potent anticancer activities against cancer stem-like cells that are considered highly resistant to chemotherapy and responsible for tumor relapse. The preclinical success of these compounds in in vitro and in vivo models have not been translated into clinical trials. At present, phase I/II clinical trials demonstrated limited benefit of Obatoclax alone or in combination with other anticancer drugs. However, future development in targeted drug delivery may be useful to improve the efficacy of these compounds. Alternatively, these compounds may be used as leading molecules for the development of less toxic derivatives.

8.
Proteomics Clin Appl ; 12(3): e1700086, 2018 05.
Article in English | MEDLINE | ID: mdl-29283216

ABSTRACT

PURPOSE: Poor molecular characterization of idiopathic pulmonary fibrosis (IPF) has led to insufficient understanding of the pathogenesis of the disease, resulting in lack of effective therapies and poor prognosis. Particularly, the role of lipid imbalance due to impaired lipid metabolism in the pathogenesis of IPF has been poorly studied. EXPERIMENTAL DESIGN: The authors have used shotgun lipidomics in a bleomycin (BLM) mouse model of pulmonary fibrosis with vascular endothelial growth factor (VEGF)-inhibitor CBO-P11 as a therapeutic measure, to identify a comprehensive set of lipids that contribute to the pathogenesis of pulmonary fibrosis. RESULTS: The authors report that attenuation of BLM-induced fibrotic response with CBO-P11 cotreatment is accompanied by a decrease in total lipid content and specific downregulation of lipids, which are upregulated in response to BLM treatment. CONCLUSION AND CLINICAL RELEVANCE: Dysregulated lipids identified in this study hold the potential of being future biomarkers for IPF.


Subject(s)
Bleomycin/adverse effects , Computational Biology , Endothelial Growth Factors/pharmacology , Lipid Metabolism , Peptides, Cyclic/pharmacology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Endothelial Growth Factors/therapeutic use , Fatty Acids/biosynthesis , Lipid Metabolism/drug effects , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Peptides, Cyclic/therapeutic use , Phospholipids/metabolism , Pulmonary Fibrosis/drug therapy , Up-Regulation/drug effects
9.
Pharmacol Rep ; 69(4): 788-797, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28605700

ABSTRACT

BACKGROUND: Resveratrol is a dietary compound that has been widely reported for its anticancer activities. However, successful extrapolation of its effects to pre-clinical studies is met with limited success due to inadequate bioavailability. We investigated the potential of combination therapy to improve the efficacy of resveratrol in a more physiologically relevant dose range. METHODS: The effect of resveratrol on canonical Wnt signaling was evaluated by Western blotting. Wnt modulators HLY78 (activator) and salinomycin (inhibitor) were evaluated in combination with resveratrol for their effect on breast cancer cell viability (MTT assay), cell cycle progression and apoptosis (Western blotting). Bliss independency model was used to evaluate combinatorial effects of resveratrol-salinomycin combination. RESULTS: Resveratrol downregulated canonical Wnt signaling proteins in treated breast cancer cells (MCF-7, MDA-MB-231 and MDA-MB-468) in the dose range of 50-200µM, which also affected cellular viability. However, at very low doses (0-50µM), resveratrol exhibited no cellular toxicity. Co-treatment with salinomycin significantly potentiated the anti-cancer effects of resveratrol, whereas HLY78 co-treatment had minimal effect. Bliss independency model revealed that Wnt inhibition synergistically potentiates the effects of resveratrol in MCF-7 and BT474 cells. Significantly downregulated canonical Wnt signaling proteins and marker of epithelial-mesenchymal transition (EMT), vimentin were observed in cells treated with resveratrol-salinomycin combination. Cell cycle arrest, caspase activation and apoptosis induction in cells treated with resveratrol-salinomycin combination further confirmed the efficacy of the combination. CONCLUSION: We report a novel resveratrol-salinomycin combination for targeting ER-positive breast cancer cells and present evidence for successful pre-clinical implementation of resveratrol.


Subject(s)
Breast Neoplasms/drug therapy , Pyrans/administration & dosage , Pyrans/therapeutic use , Receptors, Estrogen/metabolism , Stilbenes/administration & dosage , Stilbenes/therapeutic use , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Drug Therapy, Combination , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Resveratrol , Signal Transduction , Wnt Proteins/genetics , Wnt Proteins/metabolism
10.
Tumour Biol ; 39(6): 1010428317705331, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28618929

ABSTRACT

While there are targeted treatments for triple positive breast cancers, lack of specific biomarkers for triple-negative breast cancers (TNBC) has hindered the development of therapies for this subset of cancers. In this study, we evaluated the anticancer properties of cardiac glycoside Digitoxin (Dtx) and its synthetic analog MonoD on breast cancer cell lines MCF-7 (estrogen receptor-positive breast cancer) and MDA-MB-468 (triple-negative breast cancer). Both cardiac glycosides, at concentrations within the therapeutic range, increased the fraction of cells in the G0/G1 phase of the cell cycle, decreased viability, and inhibited the migration of MCF-7 and MDA-MB-468 cells. Both cardiac glycosides increased production of superoxide and induced apoptosis in both cell types. Reduced protein levels of nuclear factor kappa B and IkappaB kinase-beta were found in cardiac glycoside-treated cells, indicating that the cellular effects of these compounds are mediated via nuclear factor kappa B pathway. This study demonstrates the cytotoxic potential of digitoxin, and more importantly its synthetic analog MonoD, in the treatment of triple-positive breast cancer and more importantly the aggressive triple-negative breast cancer. Collectively, this study provides a basis for the reevaluation of cardiac glycosides in the treatment of breast cancer and more importantly reveals their potential in the treatment of triple-negative breast cancers.


Subject(s)
Digitoxin/administration & dosage , Receptors, Estrogen/genetics , Triple Negative Breast Neoplasms/drug therapy , Animals , Carcinogenesis/genetics , Cardiac Glycosides/genetics , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Digitoxin/analogs & derivatives , Female , Humans , MCF-7 Cells , Mice , NF-kappa B/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
11.
J Cell Biochem ; 118(11): 3834-3845, 2017 11.
Article in English | MEDLINE | ID: mdl-28387458

ABSTRACT

Prostate cancer (PCa) is one of the leading causes of cancer-related deaths in men worldwide. Fatty acid synthase (FASN) is reported to be overexpressed in several cancers including PCa, and this has led to clinical cancer treatments that utilize various FASN inhibitors such as the anti-obesity drug, Orlistat. However, pharmacological limitations have impeded the progress in cancer treatments expected thus far with FASN inhibition. In this study, we investigated a novel therapeutic combination to enhance the toxic potential of Orlistat in three different PCa cell-lines (DU145, PC3, and LNCaP). We show that Orlistat and 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) (AMP-activated protein kinase [AMPK] activator) co-treatment induces significant downregulation of two key fatty acid synthesis regulatory proteins (FASN, Sterol regulatory element-binding protein 1 [SREBP-1c]) as compared to control and Orlistat alone. Orlistat and AICAR co-treatment induced a significant decrease in cell viability and proliferation, and a significant increase in apoptosis in all three PCa cell-lines. Apoptosis induction was preceded by a marked increase in reactive oxygen species (ROS) production followed by G0/G1 cell cycle arrest and activation of pro-apoptotic caspases. We also observed a significant decrease in migration potential and VEGF expression in Orlistat and AICAR co-treated samples in all three PCa cell-lines. Compound C (AMPK inhibitor) negatively affected some of the enhanced anti-cancer effects observed with Orlistat treatment. We conclude that AICAR co-treatment potentiates the anti-proliferative effects of Orlistat at a low dose (100 µM), and this combination has the potential to be a viable and effective therapeutic option in PCa treatment. J. Cell. Biochem. 118: 3834-3845, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Aminoimidazole Carboxamide/analogs & derivatives , Apoptosis/drug effects , Cell Movement/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Lactones/pharmacology , Prostatic Neoplasms/drug therapy , Resting Phase, Cell Cycle/drug effects , Ribonucleotides/metabolism , Aminoimidazole Carboxamide/metabolism , Cell Line, Tumor , Humans , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Orlistat , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Reactive Oxygen Species/metabolism
12.
Cell Death Discov ; 3: 17009, 2017.
Article in English | MEDLINE | ID: mdl-28250972

ABSTRACT

Breast cancer is second most prevalent cancer in women, and the second only to lung cancer in cancer-related deaths. It is a heterogeneous disease and has several subtypes based on the presence or absence of hormone receptors and/or human epidermal growth factor receptor 2 (HER2). Hormone receptor-positive and HER2-enriched cancers can be targeted using hormone and HER2-targeting therapies such as trastuzumab or lapatinib. However, triple-negative breast cancers (TNBCs) do not express any of the receptors and therefore are resistant to most targeted therapies, and cytotoxic chemotherapies are the only viable option available for the treatment of TNBCs. Recently, cardiac glycosides (CGs) have emerged as potential anticancer agents that impart their antiproliferative effect by targeting multiple pathways. In this study our aim was to evaluate anticancer effects of two naturally occurring CGs, Convallatoxin (CT) and Peruvoside (PS), on ER+ and TNBCs cells. CT and PS demonstrated dose- and time-dependent cytotoxic effect on MCF-7 cells, which was further supported by loss of colony formation on drug treatment. CT and PS arrested MCF-7 cells in the G0/G1 phase and reduced the viability of MCF-7-derived mammospheres (MMs). Interestingly, while CT and PS imparted cell death in TNBCs cells from both Caucasians (MDA-MB-231 cells) and African Americans (MDA-MB-468 cells) in a dose- and time-dependent manner, the drugs were much more potent in MDA-MB-468 as compared with TNBC MDA-MB-231 cells. Both drugs significantly inhibited migration and invasion of both MCF-7 and MDA-MB-468 cells. An assessment of intracellular pathways indicated that both drugs were able to modulate several key cellular pathways such as EMT, cell cycle, proliferation and cell death in both cell types. Our data suggest a promising role for CGs in breast cancer treatment specifically in targeting TNBCs derived from African Americans, and provides impetus for further investigation of the anticancer potential of this class of drugs.

13.
Tumour Biol ; 39(3): 1010428317694310, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28351327

ABSTRACT

Multiple factors including tumor heterogeneity and intrinsic or acquired resistance have been associated with drug resistance in lung cancer. Increased stemness and the plasticity of cancer cells have been identified as important mechanisms of resistance; therefore, treatments targeting cancer cells independent of stemness phenotype would be much more effective in treating lung cancer. In this article, we have characterized the anticancer effects of the antibiotic Nigericin in cells displaying varying degrees of stemness and resistance to anticancer drugs, arising from (1) routine culture conditions, (2) prolonged periods of serum starvation. These cells are highly resistant to conventional anticancer drugs such as Paclitaxel, Hydroxyurea, Colchicine, Obatoclax, Wortmannin, and LY294002, and the multidrug-resistant phenotype of cells growing under prolonged periods of serum starvation is likely the result of extensive rewiring of signaling pathways, and (3) lung tumorspheres that are enriched for cancer stem-like cells. We found that Nigericin potently inhibited the viability of cells growing under routine culture conditions, prolonged periods of serum starvation, and lung tumorspheres. In addition, we found that Nigericin downregulated the expression of key proteins in the Wnt canonical signaling pathway such as LRP6, Wnt5a/b, and ß-catenin, but promotes ß-catenin translocation into the nucleus. The antitumor effects of Nigericin were potentiated by the Wnt activator HLY78 and by therapeutic levels of the US Food and Drug Administration-approved drug Digitoxin and its novel synthetic analog MonoD. We believe that Nigericin may be used in a co-therapy model in combination with other novel chemotherapeutic agents in order to achieve potent inhibition of cancers that display varying degrees of stemness, potentially leading to sustained anticancer effects.


Subject(s)
Benzodioxoles/administration & dosage , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , Nigericin/administration & dosage , Phenanthridines/administration & dosage , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Multiple/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Wnt Signaling Pathway/drug effects
14.
J Cell Physiol ; 232(9): 2280-2286, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27925198

ABSTRACT

In cancer cells, the reversible nature of the stemness status in terms of chemoresistance has been poorly characterized. In this study, we have simulated one cycle of environmental conditions to study such reversibility by first generating floating tumorspheres (FTs) from lung and breast cancer cells by culturing them in serum-free media without the addition of any external mitogenic stimulation, and subsequently (after 2 weeks) re-incubating them back in serum-containing media to simulate routine culture conditions (RCCs). We found that cancer cells are extremely plastic: cells grown under RCCs become multidrug-resistant when grown as FTs, but upon re-incubation under RCCs quickly re-attach and lose the acquired resistance. These phenotypic changes are accompanied by concomitant changes in the expression of key proteins associated with multiple pathways important for chemoresistance, survival, and stemness maintenance. Therefore, our strategy provides an excellent experimental model to study environmental factors that modulate the plasticity of cancer cells. J. Cell. Physiol. 232: 2280-2286, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Cell Plasticity/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion , Cell Proliferation , Culture Media, Serum-Free/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MCF-7 Cells , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phenotype , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Spheroids, Cellular , Time Factors
15.
J Cell Physiol ; 232(9): 2497-2507, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27662422

ABSTRACT

Lung cancer is a leading cause of cancer-related death in the United States. Although several drugs have been developed that target individual biomarkers, their success has been limited due to intrinsic or acquired resistance for the specific targets of such drugs. A more effective approach is to target multiple pathways that dictate cancer progression. Cardiac glycosides demonstrate such multimodal effects on cancer cell survival, and our aim was to evaluate the effect of two naturally occurring monosaccaridic cardiac glycosides-Convallatoxin and Peruvoside on lung cancer cells. Although both drugs had significant anti-proliferative effects on H460 and Calu-3 lung cancer cells, Convallatoxin demonstrated twofold higher activity as compared to Peruvoside using both viability and colony forming assays, suggesting a role for the aglycone region in dictating drug potency. The tumor suppressor p53 was found to be important for action of both drugs-p53-underexpressing cells were less sensitive as compared to p53-positive H460 cells. Further, assessment of p53-underexpressing H460 cells showed that drugs were able to arrest cells in the G0/G1 phase of the cell cycle in a dose-dependent manner. Both drugs significantly inhibited migration and invasion of cancer cells and decreased the viability of floating tumorspheres. An assessment of intracellular pathways indicated that both drugs were able to modulate proteins that are involved in apoptosis, autophagy, cell cycle, proliferation, and EMT. Our data suggest, a promising role for cardiac glycosides in lung cancer treatment, and provides impetus for further investigation of the anti-cancer potential of this class of drugs. J. Cell. Physiol. 232: 2497-2507, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cardenolides/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Strophanthins/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasm Invasiveness , Spheroids, Cellular , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Ear Nose Throat J ; 95(6): E12-6, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27304446

ABSTRACT

Vascular tumors arising in the temporal bone represent 0.7% of all lesions in this area. Hemangiomas confined to the tympanic membrane are exceptionally rare. We report a new case of tympanic membrane hemangioma that arose in 59-year-old man who presented with a 2-month history of constant right-sided pulsatile tinnitus and associated neck discomfort. The lesion and a cuff of healthy tympanic membrane were excised, and the resulting defect was repaired with a temporalis fascia graft. At 3 months, the neotympanum was well healed and the patient's symptoms had resolved. We also review the limited number of previously published cases of tympanic membrane hemangioma, and we discuss the presentation and management of these lesions. Some tympanic hemangiomas are asymptomatic and others present as hearing loss, tinnitus, otalgia, and/or otorrhea. Pure-tone audiometry and high-resolution computed tomography of the temporal bones are essential to determine the extent of the lesion. An excisional biopsy is recommended because it is curative and it provides histologic confirmation of the diagnosis. Excision requires elevation of a tympanomeatal flap, en bloc resection, and reconstruction of the tympanic membrane defect. Small lesions can be excised via a permeatal or endaural approach, while larger lesions require a postauricular approach.


Subject(s)
Ear Neoplasms/diagnostic imaging , Hemangioma/diagnostic imaging , Tympanic Membrane/diagnostic imaging , Ear Neoplasms/pathology , Ear Neoplasms/surgery , Hemangioma/pathology , Hemangioma/surgery , Humans , Male , Middle Aged , Otoscopy , Tomography, X-Ray Computed , Tympanic Membrane/pathology , Tympanic Membrane/surgery
17.
Stem Cells Int ; 2016: 5603135, 2016.
Article in English | MEDLINE | ID: mdl-26880969

ABSTRACT

Like with most solid tumors, the presence of a subpopulation of cancer stem cells (CSCs) or cancer stem-like cells (CS-LCs) has been associated with chemoresistance and tumor relapse in lung cancer cells. In the absence of serum, CSCs/CS-LCs have the ability to grow as lung tumorspheres (LTSs), and this system is routinely used for isolation and characterization of putative CSCs/CS-LCs. Methods to isolate LTSs are usually performed in serum-free media supplemented with specific additives such as epidermal growth factor and basic fibroblast growth factor. In this study, we report the generation of LTSs without the addition of any external mitogenic stimulation. LTSs generated in this manner demonstrated several traits usually associated with increased stemness such as elevated expression of the stemness-associated marker Sox2 and increased chemoresistance to conventional anticancer drugs. In addition, we report that the FDA-approved drug Digitoxin, at concentration close to its therapeutic level, decreased the viability of LTSs and downregulated Sox2 independent of the PI3K/AKT pathway. The potential use of LTSs generated without the addition of any external mitogenic stimulation to study the role of specific factor(s) associated with stemness properties is also discussed.

18.
J Cell Physiol ; 231(4): 817-28, 2016 04.
Article in English | MEDLINE | ID: mdl-26264876

ABSTRACT

We have synthesized a novel derivative of Digitoxin, termed "MonoD", which demonstrates cytotoxic effects in lung cancer cells with much higher potency as compared to Digitoxin. Our data show that within 1 h of MonoD treatment, H460 cells showed increased oxidative stress, increased formation of autophagic vacuoles, and increased expression of pro-autophagic markers Beclin-1 and LC3-II. Cells pretreated with MnTBAP, a superoxide scavenger not only lowered superoxide production, but also had lower levels of LC3-II and Beclin-1. Prolonged treatment with MonoD-induced apoptosis in lung cancer cells. We investigated MonoD-dependent regulation of Akt and Bcl2, proteins that are known regulators of both autophagy and apoptosis. Molecular and pharmacologic inhibitors of Bcl2 and Akt, when combined with MonoD, led to higher expression of LC3-II and Beclin-1 as compared to MonoD alone, suggesting a repressive effect for these proteins in MonoD-dependent autophagy. Pretreatment of cells with an autophagy inhibitor repressed the apoptotic potential of MonoD, confirming that early autophagic flux is important to drive apoptosis. Therapeutic entities such as MonoD that target multiple pathways such as autophagy and apoptosis may prove advantageous over current therapies that have unimodal basis for action and may drive sustained tumor regression, which is highly desirable. J. Cell. Physiol. 231: 817-828, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Digitoxin/analogs & derivatives , Digitoxin/pharmacology , Lung Neoplasms/pathology , Biomarkers/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Digoxigenin/analogs & derivatives , Digoxigenin/pharmacology , Humans , Lung Neoplasms/metabolism , Models, Biological , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Superoxides/metabolism , Vacuoles/drug effects , Vacuoles/metabolism
19.
Proteomics ; 16(1): 33-46, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26425798

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with a life expectancy of less than 5 years post diagnosis for most patients. Poor molecular characterization of IPF has led to insufficient understanding of the pathogenesis of the disease, resulting in lack of effective therapies. In this study, we have integrated a label-free LC-MS based approach with systems biology to identify signaling pathways and regulatory nodes within protein interaction networks that govern phenotypic changes that may lead to IPF. Ingenuity Pathway Analysis of proteins modulated in response to bleomycin treatment identified PI3K/Akt and Wnt signaling as the most significant profibrotic pathways. Similar analysis of proteins modulated in response to vascular endothelial growth factor (VEGF) inhibitor (CBO-P11) treatment identified natural killer cell signaling and PTEN signaling as the most significant antifibrotic pathways. Mechanistic/mammalian target of rapamycin (mTOR) and extracellular signal-regulated kinase (ERK) were identified to be key mediators of pro- and antifibrotic response, where bleomycin (BLM) treatment resulted in increased expression and VEGF inhibitor treatment attenuated expression of mTOR and ERK. Using a BLM mouse model of pulmonary fibrosis and VEGF inhibitor CBO-P11 as a therapeutic measure, we identified a comprehensive set of signaling pathways and proteins that contribute to the pathogenesis of pulmonary fibrosis that can be targeted for therapy against this fatal disease.


Subject(s)
Bleomycin , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Protein Interaction Maps , Signal Transduction , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Adherens Junctions/metabolism , Animals , Cell Line , Endothelial Growth Factors/pharmacology , Humans , Mice , Mice, Inbred C57BL , PTEN Phosphohydrolase/metabolism , Peptides, Cyclic/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Maps/drug effects , Protein Serine-Threonine Kinases/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Wnt Signaling Pathway/drug effects
20.
Oncol Rep ; 35(2): 878-86, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26573786

ABSTRACT

Despite significant advances in the understanding of lung cancer biology, the prognosis of cancer patients remains poor. Part of the failure of anticancer therapy is due to intratumoral heterogeneity in these patients that limits the efficacy of single agents. Therefore, there is an urgent need for new anticancer drugs or drug combination regimens that possess increased activity against all cellular subtypes found within the tumor. In this study, we evaluated the in vitro antiproliferative activity of the cardiac glycosides (CGs) digitoxin and its synthetic analog MonoD on H460 lung cancer cells grown under different culture conditions. The CGs were tested alone in H460 cells under routine culture as well as in cells growing under short (24-72 h) and prolonged serum starvation (7 days) in order to evaluate the activity of drugs on cancer cells under varied degrees of proliferation. Our results showed that both CGs, and MonoD in particular, have potent antiproliferative activity at clinically relevant concentrations against cells in all the tested culture conditions. In contrast, paclitaxel, hydroxyurea and colchicine were only active in cells growing in routine culture conditions, and relatively inactive in serum-starved conditions. Importantly, both CGs were able to potentiate the effect of clinically relevant concentrations of hydroxyurea or paclitaxel in serum-starved conditions. When paclitaxel was used in combination with CGs, the highest antiproliferative effect was obtained when paclitaxel was administered first, followed by either digitoxin or MonoD. Our results indicate that CGs have potential clinical applications in translational oncology especially in combination with other drugs, and warrants further investigation of CGs in more advanced preclinical models of lung cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Digitoxin/pharmacology , Hexoses/pharmacology , Lung Neoplasms/pathology , Cell Line, Tumor , Drug Synergism , Humans , Hydroxyurea/pharmacology , Inhibitory Concentration 50 , Paclitaxel/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...