Introduction Conclusions

Slides:



Advertisements
Similar presentations
Isosteviol derivatives induced apoptosis in Human lung cancer via targeting MEK/MAPK pathway: An in vitro and in vivo study Ahmed M Malki 1,,PhD Stephen.
Advertisements

第三章 Survivin siRNA nano particles are capable of inhibiting liver cancer cell growth both in vitro and in vivo Suoqin Tang,MD, Kuiyao Qu,MD, Yi Zhang,MD.
Date of download: 9/17/2016 Copyright © 2016 American Medical Association. All rights reserved. From: Differential Responses of Human Papillary Thyroid.
Volume 49, Issue 1, Pages (January 2006)
Volume 16, Issue 12, Pages (December 2014)
Copyright © 2007 American Medical Association. All rights reserved.
Generation of Novel Thyroid Cancer Stem-Like Cell Clones
Introduction Conclusions
Enhanced Sensitivity to Sunitinib by Inhibition of Akt1 Expression in Human Castration- resistant Prostate Cancer PC3 Cells Both In Vitro and In Vivo 
Vascular Endothelial Growth Factor and Basic Fibroblast Growth Factor Induce Expression of CXCR4 on Human Endothelial Cells  Rosalba Salcedo, Ken Wasserman,
Cell Physiol Biochem 2013;32: DOI: /
A Novel Cinnamide YLT26 Induces Breast Cancer Cells Apoptosis via ROS-Mitochondrial Apoptotic Pathway in Vitro and Inhibits.
Corresponding author:
Cantharidin Inhibits the Growth of Triple-Negative Breast Cancer Cells by Suppressing Autophagy and Inducing Apoptosis in Vitro and in.
Fig. 1. APG increased the sensitivity of BEL-7402/ADM cells to ADM
Ke Xu, Ph.D. Putuo Hospital and Cancer Institute,
Saikosaponin-D Enhances Radiosensitivity of Hepatoma Cells under Hypoxic Conditions by Inhibiting Hypoxia-Inducible Factor-1α Cell Physiol Biochem 2014;33:37-51.
Figure 1. Herbacetin binds to AKT1/2 and suppresses each respective kinase activity. The effect of herbacetin on (A) PI3K/AKT and (B) MAPK signaling pathway.
Corresponding author:
MicroRNA-101 Inhibits Growth, Proliferation and Migration and Induces Apoptosis of Breast Cancer Cells by Targeting Sex-Determining Region Y-Box 2 Cell.
Fig. 6. Knockdown of TCF-4 inhibits TCTP-induced glioma cell proliferation in vitro and in vivo. (A) TCF-4 was knocked down in U-251/TCTP and H-4/TCTP.
Inhibitory effect of (-)-epigallocatechin-3-gallate and bleomycin on human pancreatic cancer MiaPaca-2 cell growth Bimonte S1, Leongito M1, Barbieri.
The Autophagy Inhibitor Chloroquine Overcomes the Innate Resistance of Wild-Type EGFR Non-Small-Cell Lung Cancer Cells to Erlotinib  Yiyu Zou, PhD, Yi-He.
Fig. 4. Effect of FTY720 on brain tumor stem cell (BTSC) invasiveness
Fig. 4. TR3-dependent induction of ATF3 by DIM-C-pPhOCH 3 in human pancreatic cancer cells. ( A ) Induction of ATF3 by DIM-C-pPhOCH 3
Figure 1. Herbacetin binds to AKT1/2 and suppresses each respective kinase activity. The effect of herbacetin on (A) PI3K/AKT and (B) MAPK signaling pathway.
Cyclin-Dependent Kinase 2 Promotes Tumor Proliferation and Induces Radio Resistance in Glioblastoma  Jia Wang, Tong Yang, Gaofeng Xu, Hao Liu, Chunying.
Volume 49, Issue 1, Pages (January 2006)
Yunguang Sun, PhD, Luigi Moretti, MD, Nicholas J
Volume 140, Issue 2, Pages e2 (February 2011)
Up-Regulation of RFC3 Promotes Triple Negative Breast Cancer Metastasis and is Associated With Poor Prognosis Via EMT  Zhen-Yu He, San-Gang Wu, Fang Peng,
IFN-γ Induces Gastric Cancer Cell Proliferation and Metastasis Through Upregulation of Integrin β3-Mediated NF-κB Signaling  Yuan-Hua Xu, Zheng-Li Li,
MDH2 Stimulated by Estrogen-GPR30 Pathway Down-Regulated PTEN Expression Promoting the Proliferation and Invasion of Cells in Endometrial Cancer  Yan.
Hydrogen Sulfide Demonstrates Promising Antitumor Efficacy in Gastric Carcinoma by Targeting MGAT5  Rui Wang, Qilin Fan, Junjie Zhang, Xunan Zhang, Yuqi.
Volume 191, Issue 1, Pages (January 2014)
Cyclooxygenase-2 Overexpression in Human Basal Cell Carcinoma Cell Line Increases Antiapoptosis, Angiogenesis, and Tumorigenesis  Jeng-Wei Tjiu, Yi-Hua.
Human endothelial cells express CCR2 and respond to MCP-1: direct role of MCP-1 in angiogenesis and tumor progression by Rosalba Salcedo, Maria Lourdes.
Cell Physiol Biochem 2016;39: DOI: /
The Zinc Ionophore PCI-5002 Radiosensitizes Non-small Cell Lung Cancer Cells by Enhancing Autophagic Cell Death  Kwang Woon Kim, PhD, Christina K. Speirs,
by Jun Yuan, David B. Lovejoy, and Des R. Richardson
Regulation of Human Melanoma Growth and Metastasis by AGE–AGE Receptor Interactions  Riichiro Abe, Tadamichi Shimizu, Hiroshi Sugawara, Hirokazu Watanabe,
Volume 11, Issue 2, Pages (February 2005)
Antisense Oligonucleotides Targeting Y-Box Binding Protein-1 Inhibit Tumor Angiogenesis by Downregulating Bcl-xL-VEGFR2/-Tie Axes  Kiyoko Setoguchi, Lin.
a b MCF-7 TR2 MCF-7 TR2 (Fold change) MTT Assay , (Fold change)
Toll-Like Receptor 5 Engagement Modulates Tumor Development and Growth in a Mouse Xenograft Model of Human Colon Cancer  Sang Hoon Rhee, Eunok Im, Charalabos.
Volume 71, Issue 8, Pages (April 2007)
Combining the Multitargeted Tyrosine Kinase Inhibitor Vandetanib with the Antiestrogen Fulvestrant Enhances Its Antitumor Effect in Non-small Cell Lung.
The Autophagy Inhibitor Chloroquine Overcomes the Innate Resistance of Wild-Type EGFR Non-Small-Cell Lung Cancer Cells to Erlotinib  Yiyu Zou, PhD, Yi-He.
Transdifferentiation of Melanoma Cells by the Reprogramming Factors Attenuates Malignant Nature In Vitro and In Vivo  Mikiro Takaishi, Shigetoshi Sano 
Volume 130, Issue 3, Pages (September 2013)
Volume 152, Issue 1, Pages (January 2019)
Inhibiting MDM2-p53 Interaction Suppresses Tumor Growth in Patient-Derived Non– Small Cell Lung Cancer Xenograft Models  Josephine Hai, PhD, Shingo Sakashita,
Inhibition of KLF4 by Statins Reverses Adriamycin-Induced Metastasis and Cancer Stemness in Osteosarcoma Cells  Yangling Li, Miao Xian, Bo Yang, Meidan.
Ryang Hwa Lee, Nara Yoon, John C. Reneau, Darwin J. Prockop 
Einar K. Rofstad, Bjørn A. Graff  Journal of Investigative Dermatology 
The HIV protease and PI3K/Akt inhibitor nelfinavir does not improve the curative effect of fractionated irradiation in PC-3 prostate cancer in vitro and.
Apelin Expression in Human Non-small Cell Lung Cancer: Role in Angiogenesis and Prognosis  Judit Berta, MS, Istvan Kenessey, MD, Judit Dobos, PhD, Jozsef.
Volume 21, Issue 5, Pages (May 2013)
miR-124 Inhibits Lung Tumorigenesis Induced by K-ras Mutation and NNK
Molecular Therapy - Nucleic Acids
Molecular Therapy - Nucleic Acids
TROP-2 exhibits tumor suppressive functions in cervical cancer by dual inhibition of IGF-1R and ALK signaling  Sarah T.K. Sin, Yan Li, Ming Liu, Stephanie.
Marta Vilalta, Marjan Rafat, Amato J. Giaccia, Edward E. Graves 
Inhibition of PAX3 by TGF-β Modulates Melanocyte Viability
Bcl-2 and bcl-xL Antisense Oligonucleotides Induce Apoptosis in Melanoma Cells of Different Clinical Stages  Robert A. Olie, Christoph Hafner, Renzo Küttel,
Fig. 6 RUNX/CBFB interaction inhibitor, Ro5-3335, significantly decreases mouse neurofibroma growth in vivo. RUNX/CBFB interaction inhibitor, Ro5-3335,
Antitumor activity of MLN8237 against TNBC patient-derived tumor xenografts (PDTX) in vivo. Antitumor activity of MLN8237 against TNBC patient-derived.
Antitumor effects of celastrol in vitro and in vivo.
VC KX-01 Total Src p-Y416 Src Supplementary Figure S1. KX-01 at low dose inhibited phosphorylation of Src in MDA-MB-231 xenografts.
Targeting GAPLINC decreased CD44 expression and tumor growth in vivo.
Presentation transcript:

Introduction Conclusions Morphine Promotes Tumor Angiogenesis and Increases Breast Cancer Progression. Bimonte S. 1; Barbieri A. 2; Cascella M. 1 ; Rea D.1; Palma G. 1; Luciano A.1; Izzo F.3; Arra C.2; Cuomo A.1 1Division of Anesthesia and Pain Medicine - IRCCS “ Fondazione G. Pascale ”, Napoli, Italia; 2 S.S.D. Sperimentazione Animale- IRCCS “ Fondazione G. Pascale”, Napoli, Italia. 3Division of Abdominal Surgical Oncology, Hepatobiliary Unit. Corresponding author: s.bimonte@istitutotumori.na.it Introduction   Background: Morphine is considered a highly potent analgesic agent used to relieve suffering of patients with cancer. Several in vitro and in vivo studies showed that morphine also modulates angiogenesis and regulates tumor cell growth. Unfortunately, the results obtained by these studies are still contradictory. Material and methods: In order to dissect the role of morphine in cancer cell growth and angiogenesis we performed in vitro studies on ER-negative human breast carcinoma cells, MDA.MB231 and in vivo studies on heterotopic mouse model of human triple negative breast cancer, TNBC. Results: We demonstrated that morphine in vitro enhanced the proliferation and inhibited the apoptosis of MDA.MB231 cells. In vivo studies performed on xenograft mouse model of TNBC revealed that tumors of mice treated with morphine were larger than those observed in other groups. Moreover, morphine was able to enhance the neo-angiogenesis. Conclusions: Our data showed that morphine at clinical relevant doses promotes angiogenesis and increases breast cancer progression. Figure 2. Morphine promotes the tumor growth in TBNC mouse model. (a) Morphine promotes tumor growth in breast tumor xenograft model. Breast tumor growth in mice treated with vehicle (•) and morphine (▪). Tumor volumes increased after 28 days of morphine treatment until 35 days (P < 0.05) as compared with control (vehicle-treated). Each point represents the mean of five separate experiments. (b-c) Measurements of fluorescence per second depicting microvessel tumor (FITC-DEXTRANE) using MacroFluo images showed that morphine enhances the angiogenesis in tumor of mice (b) with respect to controls (c). Figure 1: Morphine stimulates proliferation in MDA.MB231 cell lines. MDA.MB231 cells were incubated in medium containing (a) medium control, (b) 1 μm morphine, (c) 10 μm morphine, and (d) 100 μm morphine. Cell migration rates were quantitatively assessed by counting the number of cells in the denuded area at 0, 24, and 48 h after wound induction. At 48 h after wound induction, there were clearly more cells in the denuded area of morphine treated cells than untreated cells. (e) MTT assay results show an enhancement of proliferation in breast cancer cells treated with morphine with respect to control cells. Data are representative of three independent experiments ( value < 0.05). (f-g) Western blot showing that morphine reduces the expression of p53 in MDA.MB231 cells treated with morphine (lanes 2, 3, and 4) with respect to controls (lane 1) in dose dependent manner. β-Actin was used as loading control. Figure 3. Morphine promotes angiogenesis formation in heterotopic mouse model of breast cancer. Immunohistochemically analysis for PECAM-1showed the enhancement of PECAM-1 expression in morphine treated group (b), compared to controls (a). Arrows indicate positive staining of micro-vessel staining. Conclusions We demonstrated that morphine in vitro enhanced the proliferation and inhibited the apoptosis of MDA.MB231 cells. In vivo studies performed on xenograft mouse model of TNBC revealed that tumors of mice treated with morphine were larger than those observed in other groups. Moreover, morphine was able to enhance the neo-angiogenesis. Our data showed that morphine at clinical relevant doses promotes angiogenesis and increases breast cancer progression