Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1869(6): 166702, 2023 08.
Article in English | MEDLINE | ID: mdl-37044238

ABSTRACT

Chemoresistance is a primary cause of breast cancer treatment failure, and protein-protein interactions significantly contribute to chemoresistance during different stages of breast cancer progression. In pursuit of novel biomarkers and relevant protein-protein interactions occurring during the emergence of breast cancer chemoresistance, we used a computational predictive biological (CPB) approach. CPB identified associations of adhesion molecules with proteins connected with different breast cancer proteins associated with chemoresistance. This approach identified an association of Integrin ß1 (ITGB1) with chemoresistance and breast cancer stem cell markers. ITGB1 activated the Focal Adhesion Kinase (FAK) pathway promoting invasion, migration, and chemoresistance in breast cancer by upregulating Erk phosphorylation. FAK also activated Wnt/Sox2 signaling, which enhanced self-renewal in breast cancer. Activation of the FAK pathway by ITGB1 represents a novel mechanism linked to breast cancer chemoresistance, which may lead to novel therapies capable of blocking breast cancer progression by intervening in ITGB1-regulated signaling pathways.


Subject(s)
Breast Neoplasms , Integrin beta1 , Female , Humans , Biomarkers , Breast Neoplasms/drug therapy , Cell Line, Tumor , Drug Resistance, Neoplasm , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Integrin beta1/metabolism
2.
Front Oncol ; 12: 896633, 2022.
Article in English | MEDLINE | ID: mdl-35928861

ABSTRACT

Despite the advancement in research methodologies and technologies for cancer research, there is a high rate of anti-cancer drug attrition. In this review, we discuss different conventional and modern approaches in cancer research and how human-centric models can improve on the voids conferred by more traditional animal-centric models, thereby offering a more reliable platform for drug discovery. Advanced three-dimensional cell culture methodologies, along with in silico computational analysis form the core of human-centric cancer research. This can provide a holistic understanding of the research problems and help design specific and accurate experiments that could lead to the development of better cancer therapeutics. Here, we propose a new human-centric research roadmap that promises to provide a better platform for cancer research and drug discovery.

3.
Biochim Biophys Acta Mol Cell Res ; 1866(6): 1004-1018, 2019 06.
Article in English | MEDLINE | ID: mdl-30878502

ABSTRACT

Estrogen receptor (ER) antagonist, tamoxifen has been universally used for the treatment of the ER-positive breast cancer; however, the inevitable emergence of resistance to tamoxifen obstructs the successful treatment of this cancer. So, there is an immediate requirement for the search of a novel therapeutic target for treatment of this cancer. Acquired tamoxifen-resistant breast cancer cell lines MCF-7 (MCF-7/TAM-R) and T47D (T47D/TAM-R) showed higher apoptotic resistance accompanied by induction of pro-survival autophagy compared to their parental cells. Besides, tamoxifen resistance was associated with reduced production of ATP and with overexpression of glycolytic pathways, leading to induced autophagy to meet the energy demand. Further, our study revealed that LDHA; one of the key molecules of glycolysis in association with Beclin-1 induced pro-survival autophagy in tamoxifen-resistant breast cancer. Mechanistically, pharmacological and genetic inhibition of LDHA reduced the pro-survival autophagy, with the restoration of apoptosis and reverting back the EMT like phenomena noticed in tamoxifen-resistant breast cancer. In total, targeting LDHA opened a novel strategy to interrupt autophagy and tamoxifen resistance in breast cancer.


Subject(s)
Beclin-1/genetics , Breast Neoplasms/genetics , Drug Resistance, Neoplasm , L-Lactate Dehydrogenase/genetics , Tamoxifen/pharmacology , Autophagy , Cell Line, Tumor , Cell Survival , Epithelial-Mesenchymal Transition , Female , Glycolysis , Humans , MCF-7 Cells , Proteasome Endopeptidase Complex/metabolism
4.
Mol Pharm ; 16(1): 24-40, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30513203

ABSTRACT

The effective delivery of target-specific siRNA to the brain by exploiting the exosomes derived from dendritic cells renders the paradigm shift for the prospective use of nanosized exosomes as a delivery system. Although the in vivo targeting strategies by other nanovesicles like liposomes exist, still this novel exosome-based delivery approach holds an inclusive dominance of in vivo security and reduced immunogenicity. Achieving promising exosome-based delivery strategies warrants more desirable exploration of their biology. Over the years, the invention of novel production, characterization, targeting strategies, and cargo loading techniques of exosome improved its ability to reach clinics. Essentially, exosome-based delivery of therapeutics assures to conquer the major hurdles, like delivery of cargos across impermeable biological barriers, like the blood-brain barrier, biocompatibility, increased solubility, metabolic stability, improved circulation time, target specific delivery, and pharmacokinetics, and thereby enhanced the efficacy of loaded therapeutic agents. In this article, we cover the current status of exosome as a delivery vehicle for therapeutics and the challenges that need to be overcome, and we also discuss future perspectives of this exciting field of research to transform it from bench to clinical reality.


Subject(s)
Drug Delivery Systems/methods , Exosomes/metabolism , Animals , Blood-Brain Barrier/metabolism , Humans , Models, Theoretical , RNA, Small Interfering/metabolism
5.
Oncogene ; 37(33): 4546-4561, 2018 08.
Article in English | MEDLINE | ID: mdl-29743594

ABSTRACT

Although there is a strong correlation between multinucleated cells (MNCs) and cancer chemo-resistance in variety of cancers, our understanding of how multinucleated cells modulate the tumor micro-environment is limited. We captured multinucleated cells from triple-negative chemo-resistant breast cancers cells in a time frame, where they do not proliferate but rather significantly regulate their micro-environment. We show that oxidatively stressed MNCs induce chemo-resistance in vitro and in vivo by secreting VEGF and MIF. These factors act through the RAS/MAPK pathway to induce chemo-resistance by upregulating anti-apoptotic proteins. In MNCs, elevated reactive oxygen species (ROS) stabilizes HIF-1α contributing to increase production of VEGF and MIF. Together the data indicate, that the ROS-HIF-1α signaling axis is very crucial in regulation of chemo-resistance by MNCs. Targeting ROS-HIF-1α in future may help to abrogate drug resistance in breast cancer.


Subject(s)
Drug Resistance, Neoplasm/physiology , Reactive Oxygen Species/metabolism , Triple Negative Breast Neoplasms/metabolism , Breast/metabolism , Cell Line, Tumor , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Signal Transduction/physiology , Tumor Microenvironment/physiology , Vascular Endothelial Growth Factor A/metabolism
6.
Sci Rep ; 8(1): 783, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29335481

ABSTRACT

Determination of cancer aggressiveness is mainly assessed in tissues by looking at the grade of cancer. There is a lack of specific method to determine aggressiveness of cancer cells in vitro. In our present work, we have proposed a bio-impedance based non-invasive method to differentiate aggressive property of two breast cancer cell lines. Real-time impedance analysis of MCF-7 (less aggressive) and MDA-MB-231 cells (more aggressive) demonstrated unique growth pattern. Detailed slope-analysis of impedance curves at different growth phases showed that MDA-MB-231 had higher proliferation rate and intrinsic resistance to cell death, when allowed to grow in nutrient and space limiting conditions. This intrinsic nature of death resistance of MDA-MB-231 was due to modulation and elongation of filopodia, which was also observed during scanning electron microscopy. Results were also similar when validated by cell cycle analysis. Additionally, wavelet based analysis was used to demonstrate that MCF-7 had lesser micromotion based cellular activity, when compared with MDA-MB-231. Combined together, we hypothesize that analysis of growth rate, death resistance and cellular energy, through bioimpedance based analysis can be used to determine and compare aggressiveness of multiple cancer cell lines. This further opens avenues for extrapolation of present work to human tumor tissue samples.


Subject(s)
Cell Proliferation , Electric Impedance , Apoptosis , Cell Cycle Checkpoints , Cell Line, Tumor , Humans , MCF-7 Cells , Microscopy, Electron, Scanning
7.
Cell Signal ; 35: 24-36, 2017 07.
Article in English | MEDLINE | ID: mdl-28347875

ABSTRACT

Tumor angiogenesis and invasion are deregulated biological processes that drive multistage transformation of tumors from a benign to a life-threatening malignant state activating multiple signaling pathways including MD-2/TLR4/NF-κB. Development of potential inhibitors of this signaling is emerging area for discovery of novel cancer therapeutics. In the current investigation, we identified Iturin A (A lipopeptide molecule from Bacillus megaterium) as a potent inhibitor of angiogenesis and cancer invasion by various in vitro and in vivo methods. Iturin A was found to suppress VEGF, a powerful inducer of angiogenesis and key player in tumor invasion, as confirmed by ELISA, western blot and real time PCR. Iturin A inhibited endothelial tube arrangement, blood capillary formation, endothelial sprouting and vascular growth inside the matrigel. In addition, Iturin A inhibited MMP-2/9 expression in MDA-MB-231 and HUVEC cells. Cancer invasion, migration and colony forming ability were significantly hampered by Iturin A. Expressions of MD-2/TLR4 and its downstream MyD88, IKK-α and NF-κB were also reduced in treated MDA-MB-231 and HUVEC cells. Western blot and immunofluorescence study showed that nuclear accumulation of NF-κB was hampered by Iturin A. MD-2 siRNA or plasmid further confirmed the efficacy of Iturin A by suppressing MD-2/TLR4 signaling pathway. The in silico docking study showed that the Iturin A interacted well with the MD-2 in MD-2/TLR4 receptor complex. Conclusively, inhibition of MD-2/TLR4 complex with Iturin A offered strategic advancement in cancer therapy.


Subject(s)
Lymphocyte Antigen 96/genetics , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Peptides, Cyclic/administration & dosage , Toll-Like Receptor 4/genetics , Bacillus megaterium/chemistry , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Human Umbilical Vein Endothelial Cells , Humans , I-kappa B Kinase/genetics , Lymphocyte Antigen 96/chemistry , Myeloid Differentiation Factor 88/genetics , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Peptides, Cyclic/chemistry , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Toll-Like Receptor 4/chemistry
8.
Biochim Biophys Acta Gen Subj ; 1861(1 Pt A): 3039-3052, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27721046

ABSTRACT

BACKGROUND: Gold nanorods, by virtue of surface plasmon resonance, convert incident light energy (NIR) into heat energy which induces hyperthermia. We designed unique, multifunctional, gold nanorod embedded block copolymer micelle loaded with GW627368X for targeted drug delivery and photothermal therapy. METHODS: Glutathione responsive diblock co-polymer was synthesized by RAFT process forming self-assembled micelle on gold nanorods prepared by seed mediated method and GW627368X was loaded on to the reduction responsive gold nanorod embedded micelle. Photothermal therapy was administered using cwNIR laser (808nm; 4W/cm2). Efficacy of nanoformulated GW627368X, photothermal therapy and combination of both were evaluated in vitro and in vivo. RESULTS: In response to photothermal treatment, cells undergo regulated, patterned cell death by necroptosis. Combining GW627368X with photothermal treatment using single nanoparticle enhanced therapeutic outcome. In addition, these nanoparticles are effective X-ray CT contrast agents, thus, can help in monitoring treatment. CONCLUSION: Reduction responsive nanorod embedded micelle containing folic acid and lipoic acid when treated on cervical cancer cells or tumour bearing mice, aggregate in and around cancer cells. Due to high glutathione concentration, micelles degrade releasing drug which binds surface receptors inducing apoptosis. When incident with 808nm cwNIR lasers, gold nanorods bring about photothermal effect leading to hyperthermic cell death by necroptosis. Combination of the two modalities enhances therapeutic efficacy by inducing both forms of cell death. GENERAL SIGNIFICANCE: Our proposed treatment strategy achieves photothermal therapy and targeted drug delivery simultaneously. It can prove useful in overcoming general toxicities associated with chemotherapeutics and intrinsic/acquired resistance to chemo and radiotherapy.


Subject(s)
Drug Delivery Systems/methods , Gold/chemistry , Hyperthermia, Induced , Micelles , Nanotubes/chemistry , Neoplasms/therapy , Phototherapy , Polymers/chemistry , Animals , Biocompatible Materials/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Contrast Media/chemistry , Drug Liberation , Endocytosis/drug effects , Humans , Inhibitory Concentration 50 , Isoindoles/pharmacology , Mice , Nanotubes/ultrastructure , Polymers/chemical synthesis , Spectrophotometry, Ultraviolet , Spectroscopy, Near-Infrared , Sulfonamides/pharmacology , X-Rays
9.
Tumour Biol ; 36(12): 9829-37, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26159854

ABSTRACT

Despite the recent advances in diagnostic and therapeutic strategies, oral squamous cell carcinoma (OSCC) remains a major health burden. Protein biomarker discovery for early detection will help to improve patient survival rate in OSCC. Mass spectrometry-based proteomics has emerged as an excellent approach for detection of protein biomarkers in various types of cancers. In the current study, we have used 4-Plex isobaric tags for relative and absolute quantitation (iTRAQ)-based shotgun quantitative proteomic approach to identify proteins that are differentially expressed in cancerous tissues compared to normal tissues. The high-resolution mass spectrometric analysis resulted in identifying 2,074 proteins, among which 288 proteins were differentially expressed. Further, it was noticed that 162 proteins were upregulated, while 125 proteins were downregulated in OSCC-derived cancer tissue samples as compared to the adjacent normal tissues. We identified some of the known molecules which were reported earlier in OSCC such as MMP-9 (8.4-fold), ZNF142 (5.6-fold), and S100A7 (3.5-fold). Apart from this, we have also identified some novel signature proteins which have not been reported earlier in OSCC including ras-related protein Rab-2A isoform, RAB2A (4.6-fold), and peroxiredoxin-1, PRDX1 (2.2-fold). The immunohistochemistry-based validation using tissue microarray slides in OSCC revealed overexpression of the RAB2A and PRDX1 gene in 80 and 68 % of the tested clinical cases, respectively. This study will not only serve as a resource of candidate biomarkers but will contribute towards the existing knowledge on the role of the candidate molecules towards disease progression and therapeutic potential.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/genetics , Mouth Neoplasms/genetics , Peroxiredoxins/biosynthesis , rab GTP-Binding Proteins/biosynthesis , Aged , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Mouth Neoplasms/pathology , Peroxiredoxins/genetics , Proteome/genetics , Proteomics , Tandem Mass Spectrometry , rab GTP-Binding Proteins/genetics
10.
Cancer Biol Ther ; 16(6): 922-32, 2015.
Article in English | MEDLINE | ID: mdl-25894216

ABSTRACT

Prostaglandin E2, the major COX-2 product, acts via 4 functionally distinct prostanoid receptors, EP(1-4). PGE-2, through its receptors, feeds back to positively increase COX-2 expression augmenting its own synthesis thereby driving angiogenesis, while suppressing apoptosis and innate immunity. In addition to the well characterized PGE2/EP4/cAMP/PKA/CREB, EP4 activation increases GSK3 phosphorylation via PI3K and Akt consequently reducing ß-catenin phosphorylation. EP4 induces angiogenesis by enhancing VEGF production via ERK activation. These effects of EP4 are asserted either directly or via EGFR transactivation depending on the type of cancer. In view of the safety concerns regarding long term use of COX-2 inhibitors and to find more effective alternatives, we evaluated the potential of EP4 prostanoid receptor as a target for treating cancer progression using a highly selective EP4 antagonist, 4-(4,9-diethoxy-1,3-dihydro-1-oxo-2H-benz[f]isoindol-2-yl)-N-(phenylsulfonyl)-benzeneacetamide. Oral administration of GW627368X showed significant tumor regression characterized by tumor reduction and induction of apoptosis. Reduction in prostaglandin E2 synthesis also led to reduced level of VEGF in plasma. Regulation of multiple pathways downstream of EP4 was evident by down regulation of COX-2, p-Akt, p-MAPK and p-EGFR. Considering wide distribution of the EP4 prostanoid receptor in major organs and the array of physiological processes it contributes to, the safety profile of the drug was analyzed. No major organ toxicity, immunosupression, behavioral change or change in blood parameters attributable to the drug was observed. The results assert the significance of EP4 prostanoid receptor as a therapeutic target as well as the safety of EP4 blockade by GW627368X.


Subject(s)
Antineoplastic Agents/pharmacology , Dinoprostone/antagonists & inhibitors , Isoindoles/pharmacology , Sarcoma/metabolism , Sulfonamides/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclooxygenase 2/metabolism , Disease Models, Animal , Humans , Isoindoles/administration & dosage , MAP Kinase Signaling System , Mice , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Sarcoma/blood , Sarcoma/drug therapy , Sarcoma/pathology , Sulfonamides/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Article in English | MEDLINE | ID: mdl-26764722

ABSTRACT

The paper presents a study to differentiate normal and cancerous cells using label-free bioimpedance signal measured by electric cell-substrate impedance sensing. The real-time-measured bioimpedance data of human breast cancer cells and human epithelial normal cells employs fluctuations of impedance value due to cellular micromotions resulting from dynamic structural rearrangement of membrane protrusions under nonagitated condition. Here, a wavelet-based multiscale quantitative analysis technique has been applied to analyze the fluctuations in bioimpedance. The study demonstrates a method to classify cancerous and normal cells from the signature of their impedance fluctuations. The fluctuations associated with cellular micromotion are quantified in terms of cellular energy, cellular power dissipation, and cellular moments. The cellular energy and power dissipation are found higher for cancerous cells associated with higher micromotions in cancer cells. The initial study suggests that proposed wavelet-based quantitative technique promises to be an effective method to analyze real-time bioimpedance signal for distinguishing cancer and normal cells.


Subject(s)
Breast Neoplasms/pathology , Cell Separation/instrumentation , Epithelial Cells/cytology , Wavelet Analysis , Cell Proliferation , Electric Impedance , Humans , MCF-7 Cells , Time Factors
12.
J Biol Chem ; 289(37): 25468-73, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25053418

ABSTRACT

Effects of glucose on the susceptibility of antifungal agents were investigated against Candida spp. Increasing the concentration of glucose decreased the activity of antifungal agents; voriconazole was the most affected drugs followed by amphotericin B. No significant change has been observed for anidulafungin. Biophysical interactions between antifungal agents with glucose molecules were investigated using isothermal titration calorimetry, Fourier transform infrared, and (1)H NMR. Glucose has a higher affinity to bind with voriconazole by hydrogen bonding and decrease the susceptibility of antifungal agents during chemotherapy. In addition to confirming the results observed in vitro, theoretical docking studies demonstrated that voriconazole presented three important hydrogen bonds and amphotericin B presented two hydrogen bonds that stabilized the glucose. In vivo results also suggest that the physiologically relevant higher glucose level in the bloodstream of diabetes mellitus mice might interact with the available selective agents during antifungal therapy, thus decreasing glucose activity by complex formation. Thus, proper selection of drugs for diabetes mellitus patients is important to control infectious diseases.


Subject(s)
Antifungal Agents/chemistry , Candida/drug effects , Drug Resistance, Fungal/drug effects , Glucose/metabolism , Animals , Antifungal Agents/administration & dosage , Biophysical Phenomena , Calorimetry , Candida/metabolism , Glucose/chemistry , Humans , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Mice , Spectroscopy, Fourier Transform Infrared
13.
Life Sci ; 93(21): 783-90, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24044882

ABSTRACT

AIM: Thymoquinone (TQ), the predominant bioactive constituent of black seed oil (Nigella Sativa), has been shown to possess antineoplastic activity against multifarious tumors. However, the meticulous mechanism of TQ on Akt mediated survival pathway is still unrevealed in breast cancer. Here, we investigated TQ's mechanism of action against PI3K/Akt signaling and its downstream targets by modulating proteins translational machinery, leading to apoptosis in cancer cells. MAIN METHODS: MDA-MB-468 and T-47D cells were treated with TQ and evaluated for its anticancer activity through phase distribution and western blot. Modulatory effects of TQ on Akt were affirmed through kinase and drug potential studies. KEY FINDINGS: Studies revealed G1 phase arrest till 24h incubation with TQ while extended exposure showed phase shift to subG1 indicating apoptosis, supported by suppression of cyclin D1, cyclin E and cyclin dependent kinase inhibitor p27 expression. Immunoblot and membrane potential studies revealed mitochondrial impairment behind apoptotic process with upregulation of Bax, cytoplasmic cytochrome c and procaspase-3, PARP cleavage along with Bcl-2, Bcl-xL and survivin downregulation. Moreover, we construed the rationale behind mitochondrial dysfunction by examining the phosphorylation status of PDK1, PTEN, Akt, c-raf, GSK-3ß and Bad in TQ treated cells, thus ratifying the involvement of Akt in apoptosis. Further, the consequential effect of Akt inhibition by TQ is proven by translational repression through deregulated phosphorylation of 4E-BP1, eIF4E, S6R and p70S6K. SIGNIFICANCE: Our observations for the first time may provide a new insight for the development of novel therapies for Akt overexpressed breast cancer by TQ.


Subject(s)
Apoptosis/drug effects , Benzoquinones/pharmacology , Breast Neoplasms/pathology , Cyclin D1/biosynthesis , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Molecular Targeted Therapy , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
14.
BMC Cancer ; 13: 273, 2013 Jun 03.
Article in English | MEDLINE | ID: mdl-23731702

ABSTRACT

BACKGROUND: Tamoxifen (TAM) is widely used in the chemotherapy of breast cancer and as a preventive agent against recurrence after surgery. However, extended TAM administration for breast cancer induces increased VEGF levels in patients, promoting new blood vessel formation and thereby limiting its efficacy. Celecoxib (CXB), a selective COX-2 inhibitor, suppresses VEGF gene expression by targeting the VEGF promoter responsible for its inhibitory effect. For this study, we had selected CXB as non-steroidal anti-inflammatory drug in combination with TAM for suppressing VEGF expression and simultaneously reducing doses of both the drugs. METHODS: The effects of CXB combined with TAM were examined in two human breast cancer cell lines in culture, MCF7 and MDA-MB-231. Assays of proliferation, apoptosis, angiogenesis, metastasis, cell cycle distribution, and receptor signaling were performed. RESULTS: Here, we elucidated how the combination of TAM and CXB at nontoxic doses exerts anti-angiogenic effects by specifically targeting VEGF/VEGFR2 autocrine signaling through ROS generation. At the molecular level, TAM-CXB suppresses VHL-mediated HIF-1α activation, responsible for expression of COX-2, MMP-2 and VEGF. Besides low VEGF levels, TAM-CXB also suppresses VEGFR2 expression, confirmed through quantifying secreted VEGF levels, luciferase and RT-PCR studies. Interestingly, we observed that TAM-CXB was effective in blocking VEGFR2 promoter induced expression and further 2 fold decrease in VEGF levels was observed in combination than TAM alone in both cell lines. Secondly, TAM-CXB regulated VEGFR2 inhibits Src expression, responsible for tumor progression and metastasis. FACS and in vivo enzymatic studies showed significant increase in the reactive oxygen species upon TAM-CXB treatment. CONCLUSIONS: Taken together, our experimental results indicate that this additive combination shows promising outcome in anti-metastatic and apoptotic studies. In a line, our preclinical studies evidenced that this additive combination of TAM and CXB is a potential drug candidate for treatment of breast tumors expressing high levels of VEGF and VEGFR2. This ingenious combination might be a better tailored clinical regimen than TAM alone for breast cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autocrine Communication/drug effects , Breast Neoplasms/metabolism , Neovascularization, Pathologic , Receptors, Vascular Endothelial Growth Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Blotting, Western , Celecoxib , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Neovascularization, Pathologic/chemically induced , Pyrazoles/administration & dosage , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sulfonamides/administration & dosage , Tamoxifen/administration & dosage , Tamoxifen/adverse effects , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...