62 results on '"Kshipra M. Gharpure"'
Search Results
2. FABP4 as a key determinant of metastatic potential of ovarian cancer
- Author
-
Kshipra M. Gharpure, Sunila Pradeep, Marta Sans, Rajesha Rupaimoole, Cristina Ivan, Sherry Y. Wu, Emine Bayraktar, Archana S. Nagaraja, Lingegowda S. Mangala, Xinna Zhang, Monika Haemmerle, Wei Hu, Cristian Rodriguez-Aguayo, Michael McGuire, Celia Sze Ling Mak, Xiuhui Chen, Michelle A. Tran, Alejandro Villar-Prados, Guillermo Armaiz Pena, Ragini Kondetimmanahalli, Ryan Nini, Pranavi Koppula, Prahlad Ram, Jinsong Liu, Gabriel Lopez-Berestein, Keith Baggerly, Livia S. Eberlin, and Anil K. Sood
- Subjects
Science - Abstract
In ovarian cancer, metastatic phenotype may impact surgical outcomes. Here, the authors show miR-409-3p regulates FABP4 which can increase metastatic potential of ovarian cancer, and treatment with DOPC nanoliposomes containing either miR-409--3p mimic or FABP4 siRNA inhibits tumor progression in mice.
- Published
- 2018
- Full Text
- View/download PDF
3. Differential Effects of EGFL6 on Tumor versus Wound Angiogenesis
- Author
-
Kyunghee Noh, Lingegowda S. Mangala, Hee-Dong Han, Ningyan Zhang, Sunila Pradeep, Sherry Y. Wu, Shaolin Ma, Edna Mora, Rajesha Rupaimoole, Dahai Jiang, Yunfei Wen, Mian M.K. Shahzad, Yasmin Lyons, MinSoon Cho, Wei Hu, Archana S. Nagaraja, Monika Haemmerle, Celia S.L. Mak, Xiuhui Chen, Kshipra M. Gharpure, Hui Deng, Wei Xiong, Charles V. Kingsley, Jinsong Liu, Nicholas Jennings, Michael J. Birrer, Richard R. Bouchard, Gabriel Lopez-Berestein, Robert L. Coleman, Zhiqiang An, and Anil K. Sood
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Angiogenesis inhibitors are important for cancer therapy, but clinically approved anti-angiogenic agents have shown only modest efficacy and can compromise wound healing. This necessitates the development of novel anti-angiogenesis therapies. Here, we show significantly increased EGFL6 expression in tumor versus wound or normal endothelial cells. Using a series of in vitro and in vivo studies with orthotopic and genetically engineered mouse models, we demonstrate the mechanisms by which EGFL6 stimulates tumor angiogenesis. In contrast to its antagonistic effects on tumor angiogenesis, EGFL6 blockage did not affect normal wound healing. These findings have significant implications for development of anti-angiogenesis therapies. : Noh et al. identify EGFL6 as an angiogenic target that is selectively present in tumor endothelial cells in a hypoxic tumor microenvironment. EGFL6 blockade exerts robust anti-angiogenic and anti-tumor effects without affecting wound healing. These findings suggest an important approach for effectively targeting tumor angiogenesis. Keywords: tumor endothelial cells, ovarian cancer, chitosan nanoparticles, tumor vasculature, wound healing
- Published
- 2017
- Full Text
- View/download PDF
4. Platelets reduce anoikis and promote metastasis by activating YAP1 signaling
- Author
-
Monika Haemmerle, Morgan L. Taylor, Tony Gutschner, Sunila Pradeep, Min Soon Cho, Jianting Sheng, Yasmin M. Lyons, Archana S. Nagaraja, Robert L. Dood, Yunfei Wen, Lingegowda S. Mangala, Jean M. Hansen, Rajesha Rupaimoole, Kshipra M. Gharpure, Cristian Rodriguez-Aguayo, Sun Young Yim, Ju-Seog Lee, Cristina Ivan, Wei Hu, Gabriel Lopez-Berestein, Stephen T. Wong, Beth Y. Karlan, Douglas A. Levine, Jinsong Liu, Vahid Afshar-Kharghan, and Anil K. Sood
- Subjects
Science - Abstract
Platelets have been associated with increased tumor growth and metastasis but the mechanistic details of this interaction are still unclear. Here the authors show that platelets improve anoikis resistance of cancer cells and increase metastasis by activating Yap through a RhoA/MYPT-PP1 pathway.
- Published
- 2017
- Full Text
- View/download PDF
5. Author Correction: Hypoxia-mediated downregulation of miRNA biogenesis promotes tumour progression
- Author
-
Rajesha Rupaimoole, Sherry Y. Wu, Sunila Pradeep, Cristina Ivan, Chad V. Pecot, Kshipra M. Gharpure, Archana S. Nagaraja, Guillermo N. Armaiz-Pena, Michael McGuire, Behrouz Zand, Heather J. Dalton, Justyna Filant, Justin Bottsford Miller, Chunhua Lu, Nouara C. Sadaoui, Lingegowda S. Mangala, Morgan Taylor, Twan van den Beucken, Elizabeth Koch, Cristian Rodriguez-Aguayo, Li Huang, Menashe Bar-Eli, Bradly G. Wouters, Milan Radovich, Mircea Ivan, George A. Calin, Wei Zhang, Gabriel Lopez-Berestein, and Anil K. Sood
- Subjects
Science - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
6. A miR-192-EGR1-HOXB9 regulatory network controls the angiogenic switch in cancer
- Author
-
Sherry Y. Wu, Rajesha Rupaimoole, Fangrong Shen, Sunila Pradeep, Chad V. Pecot, Cristina Ivan, Archana S. Nagaraja, Kshipra M. Gharpure, Elizabeth Pham, Hiroto Hatakeyama, Michael H. McGuire, Monika Haemmerle, Viviana Vidal-Anaya, Courtney Olsen, Cristian Rodriguez-Aguayo, Justyna Filant, Ehsan A. Ehsanipour, Shelley M. Herbrich, Sourindra N. Maiti, Li Huang, Ji Hoon Kim, Xinna Zhang, Hee-Dong Han, Guillermo N. Armaiz-Pena, Elena G. Seviour, Sue Tucker, Min Zhang, Da Yang, Laurence J. N. Cooper, Rouba Ali-Fehmi, Menashe Bar-Eli, Ju-Seog Lee, Prahlad T. Ram, Keith A. Baggerly, Gabriel Lopez-Berestein, Mien-Chie Hung, and Anil K. Sood
- Subjects
Science - Abstract
The formation of blood vessels in tumours, angiogenesis, is a promising target for therapy. Here, the authors show that microRNA192 has anti-angiogenic functions and negatively regulates EGR1 and HOXB9, and that delivery of this microRNA to tumours in vivocan reduce angiogenesis and tumour growth.
- Published
- 2016
- Full Text
- View/download PDF
7. Long Noncoding RNA Ceruloplasmin Promotes Cancer Growth by Altering Glycolysis
- Author
-
Rajesha Rupaimoole, Jaehyuk Lee, Monika Haemmerle, Hui Ling, Rebecca A. Previs, Sunila Pradeep, Sherry Y. Wu, Cristina Ivan, Manuela Ferracin, Jennifer B. Dennison, Niki M. Zacharias Millward, Archana S. Nagaraja, Kshipra M. Gharpure, Michael McGuire, Nidhin Sam, Guillermo N. Armaiz-Pena, Nouara C. Sadaoui, Cristian Rodriguez-Aguayo, George A. Calin, Ronny I. Drapkin, Jeffery Kovacs, Gordon B. Mills, Wei Zhang, Gabriel Lopez-Berestein, Pratip K. Bhattacharya, and Anil K. Sood
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Long noncoding RNAs (lncRNAs) significantly influence the development and regulation of genome expression in cells. Here, we demonstrate the role of lncRNA ceruloplasmin (NRCP) in cancer metabolism and elucidate functional effects leading to increased tumor progression. NRCP was highly upregulated in ovarian tumors, and knockdown of NRCP resulted in significantly increased apoptosis, decreased cell proliferation, and decreased glycolysis compared with control cancer cells. In an orthotopic mouse model of ovarian cancer, siNRCP delivered via a liposomal carrier significantly reduced tumor growth compared with control treatment. We identified NRCP as an intermediate binding partner between STAT1 and RNA polymerase II, leading to increased expression of downstream target genes such as glucose-6-phosphate isomerase. Collectively, we report a previously unrecognized role of the lncRNA NRCP in modulating cancer metabolism. As demonstrated, DOPC nanoparticle-incorporated siRNA-mediated silencing of this lncRNA in vivo provides therapeutic avenue toward modulating lncRNAs in cancer.
- Published
- 2015
- Full Text
- View/download PDF
8. Table S6 from PRKRA/PACT Expression Promotes Chemoresistance of Mucinous Ovarian Cancer
- Author
-
Anil K. Sood, Gabriel Lopez-Berestein, Kwong K. Wong, Michael Frumovitz, Koji Matsuo, Ju-Seog Lee, Cristina Ivan, Geoffrey Bartholomeusz, Johnathon Rose, Young Gyu Eun, Cristian Rodriguez-Aguayo, Takashi Mitamura, Lingegowda S. Mangala, Archana S. Nagaraja, Kshipra M. Gharpure, Yasmin Lyons, Jean M. Hansen, Wei Hu, Kyunghee Noh, Emine Bayraktar, Sunila Pradeep, Rajesha Rupaimoole, Sherry Y. Wu, Michael McGuire, and Takeshi Hisamatsu
- Abstract
Nanostring miRNA data
- Published
- 2023
- Full Text
- View/download PDF
9. Data from PRKRA/PACT Expression Promotes Chemoresistance of Mucinous Ovarian Cancer
- Author
-
Anil K. Sood, Gabriel Lopez-Berestein, Kwong K. Wong, Michael Frumovitz, Koji Matsuo, Ju-Seog Lee, Cristina Ivan, Geoffrey Bartholomeusz, Johnathon Rose, Young Gyu Eun, Cristian Rodriguez-Aguayo, Takashi Mitamura, Lingegowda S. Mangala, Archana S. Nagaraja, Kshipra M. Gharpure, Yasmin Lyons, Jean M. Hansen, Wei Hu, Kyunghee Noh, Emine Bayraktar, Sunila Pradeep, Rajesha Rupaimoole, Sherry Y. Wu, Michael McGuire, and Takeshi Hisamatsu
- Abstract
For mucinous ovarian cancer (MOC), standard platinum-based therapy is largely ineffective. We sought to identify possible mechanisms of oxaliplatin resistance of MOC and develop strategies to overcome this resistance. A kinome-based siRNA library screen was carried out using human MOC cells to identify novel targets to enhance the efficacy of chemotherapy. In vitro and in vivo validations of antitumor effects were performed using mouse MOC models. Specifically, the role of PRKRA/PACT in oxaliplatin resistance was interrogated. We focused on PRKRA, a known activator of PKR kinase, and its encoded protein PACT because it was one of the five most significantly downregulated genes in the siRNA screen. In orthotopic mouse models of MOC, we observed a significant antitumor effect of PRKRA siRNA plus oxaliplatin. In addition, expression of miR-515-3p was regulated by PACT–Dicer interaction, and miR-515-3p increased the sensitivity of MOC to oxaliplatin. Mechanistically, miR-515-3p regulated chemosensitivity, in part, by targeting AXL. The PRKRA/PACT axis represents an important therapeutic target in MOC to enhance sensitivity to oxaliplatin.
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Metronomic Docetaxel in PRINT Nanoparticles and EZH2 Silencing Have Synergistic Antitumor Effect in Ovarian Cancer
- Author
-
Anil K. Sood, Joseph M. DeSimone, Gabriel Lopez-Berestein, Mary E. Napier, J. Christopher Luft, Sherry Y. Wu, Tojan B. Rahhal, Guillermo N. Armaiz-Pena, Rajesha Rupaimoole, Hee-Dong Han, Selanere L. Mangala, Sunila Pradeep, Takahito Miyake, Charles J. Bowerman, Kevin S. Chu, and Kshipra M. Gharpure
- Abstract
The purpose of this study was to investigate the antitumor effects of a combination of metronomic doses of a novel delivery vehicle, PLGA-PRINT nanoparticles containing docetaxel, and antiangiogenic mEZH2 siRNA incorporated into chitosan nanoparticles. In vivo dose-finding studies and therapeutic experiments were conducted in well-established orthotopic mouse models of epithelial ovarian cancer. Antitumor effects were determined on the basis of reduction in mean tumor weight and number of metastatic tumor nodules in the animals. The tumor tissues from these in vivo studies were stained to evaluate the proliferation index (Ki67), apoptosis index (cleaved caspase 3), and microvessel density (CD31). The lowest dose of metronomic regimen (0.5 mg/kg) resulted in significant reduction in tumor growth. The combination of PLGA-PRINT-docetaxel and CH-mEZH2 siRNA showed significant antitumor effects in the HeyA8 and SKOV3ip1 tumor models (P < 0.05). Individual as well as combination therapies showed significant antiangiogenic, antiproliferative, and proapoptotic effects, and combination therapy had additive effects. Metronomic delivery of PLGA-PRINT-docetaxel combined with CH-mEZH2 siRNA has significant antitumor activity in preclinical models of ovarian cancer. Mol Cancer Ther; 13(7); 1750–7. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
11. Data from Therapeutic Silencing of KRAS Using Systemically Delivered siRNAs
- Author
-
Anil K. Sood, Lee M. Ellis, Gabriel Lopez-Berestein, Scott Kopetz, Heather J. Dalton, Behrouz Zand, Vianey Gonzalez-Villasana, Trent A. Waugh, Kshipra M. Gharpure, Maria Pia Morelli, Archana S. Nagaraja, Cristian Rodriguez-Aguayo, Salma Azam, Anshumaan Maharaj, Rajat Bhattacharya, Takeshi Hisamatsu, Rajesha Rupaimoole, Justyna Filant, Seth Bellister, Sherry Y. Wu, and Chad V. Pecot
- Abstract
Despite being among the most common oncogenes in human cancer, to date, there are no effective clinical options for inhibiting KRAS activity. We investigated whether systemically delivered KRAS siRNAs have therapeutic potential in KRAS-mutated cancer models. We identified KRAS siRNA sequences with notable potency in knocking down KRAS expression. Using lung and colon adenocarcinoma cell lines, we assessed antiproliferative effects of KRAS silencing in vitro. For in vivo experiments, we used a nanoliposomal delivery platform, DOPC, for systemic delivery of siRNAs. Various lung and colon cancer models were used to determine efficacy of systemic KRAS siRNA based on tumor growth, development of metastasis, and downstream signaling. KRAS siRNA sequences induced >90% knockdown of KRAS expression, significantly reducing viability in mutant cell lines. In the lung cancer model, KRAS siRNA treatment demonstrated significant reductions in primary tumor growth and distant metastatic disease, while the addition of CDDP was not additive. Significant reductions in Ki-67 indices were seen in all treatment groups, whereas significant increases in caspase-3 activity were only seen in the CDDP treatment groups. In the colon cancer model, KRAS siRNA reduced tumor KRAS and pERK expression. KRAS siRNAs significantly reduced HCP1 subcutaneous tumor growth, as well as outgrowth of liver metastases. Our studies demonstrate a proof-of-concept approach to therapeutic KRAS targeting using nanoparticle delivery of siRNA. This study highlights the potential translational impact of therapeutic RNA interference, which may have broad applications in oncology, especially for traditional “undruggable” targets. Mol Cancer Ther; 13(12); 2876–85. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figures 1 - 2 from Therapeutic Silencing of KRAS Using Systemically Delivered siRNAs
- Author
-
Anil K. Sood, Lee M. Ellis, Gabriel Lopez-Berestein, Scott Kopetz, Heather J. Dalton, Behrouz Zand, Vianey Gonzalez-Villasana, Trent A. Waugh, Kshipra M. Gharpure, Maria Pia Morelli, Archana S. Nagaraja, Cristian Rodriguez-Aguayo, Salma Azam, Anshumaan Maharaj, Rajat Bhattacharya, Takeshi Hisamatsu, Rajesha Rupaimoole, Justyna Filant, Seth Bellister, Sherry Y. Wu, and Chad V. Pecot
- Abstract
Supplementary figure 1 provides a schematic of DOPC:siRNA nanoliposomal preparation, and Supplementary figure 2 shows tumor volumetric measurements of the A549 orthotopic lung cancer therapeutic experiment described in the main figure 2.
- Published
- 2023
- Full Text
- View/download PDF
13. Table S1, Table S2, Table S3, Table S5 from PRKRA/PACT Expression Promotes Chemoresistance of Mucinous Ovarian Cancer
- Author
-
Anil K. Sood, Gabriel Lopez-Berestein, Kwong K. Wong, Michael Frumovitz, Koji Matsuo, Ju-Seog Lee, Cristina Ivan, Geoffrey Bartholomeusz, Johnathon Rose, Young Gyu Eun, Cristian Rodriguez-Aguayo, Takashi Mitamura, Lingegowda S. Mangala, Archana S. Nagaraja, Kshipra M. Gharpure, Yasmin Lyons, Jean M. Hansen, Wei Hu, Kyunghee Noh, Emine Bayraktar, Sunila Pradeep, Rajesha Rupaimoole, Sherry Y. Wu, Michael McGuire, and Takeshi Hisamatsu
- Abstract
Table S1 shows antibody concentrations. Table S2 shows siRNA sequences. Table S3 shows qPCR primers sequences. Table S5 shows IC50 values of oxaliplatin.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Table 1 from Metronomic Docetaxel in PRINT Nanoparticles and EZH2 Silencing Have Synergistic Antitumor Effect in Ovarian Cancer
- Author
-
Anil K. Sood, Joseph M. DeSimone, Gabriel Lopez-Berestein, Mary E. Napier, J. Christopher Luft, Sherry Y. Wu, Tojan B. Rahhal, Guillermo N. Armaiz-Pena, Rajesha Rupaimoole, Hee-Dong Han, Selanere L. Mangala, Sunila Pradeep, Takahito Miyake, Charles J. Bowerman, Kevin S. Chu, and Kshipra M. Gharpure
- Abstract
PDF - 312K, Storage stability of PLGA-PRINT nanoparticles.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Materials and Methods and Supplementary Figure Legends from Metronomic Docetaxel in PRINT Nanoparticles and EZH2 Silencing Have Synergistic Antitumor Effect in Ovarian Cancer
- Author
-
Anil K. Sood, Joseph M. DeSimone, Gabriel Lopez-Berestein, Mary E. Napier, J. Christopher Luft, Sherry Y. Wu, Tojan B. Rahhal, Guillermo N. Armaiz-Pena, Rajesha Rupaimoole, Hee-Dong Han, Selanere L. Mangala, Sunila Pradeep, Takahito Miyake, Charles J. Bowerman, Kevin S. Chu, and Kshipra M. Gharpure
- Abstract
PDF - 78K, Supplementary methods and legends for supplementary figures.
- Published
- 2023
- Full Text
- View/download PDF
16. Data from Sustained Adrenergic Signaling Promotes Intratumoral Innervation through BDNF Induction
- Author
-
Anil K. Sood, Mariella De Biasi, Susan K. Lutgendorf, Steve W. Cole, Gabriel Lopez-Berestein, Frank C. Marini, Stephen T.C. Wong, Prahlad T. Ram, Erika L. Spaeth, Vasudha Sehgal, Cristian Rodriguez-Aguayo, Lingegowda S. Mangala, Yu Kang, Myrthala Moreno-Smith, Justin Bottsford-Miller, Wei Hu, Morgan Taylor, Heather J. Dalton, Behrouz Zand, Hee Dong Han, Xiaoyun Xu, Sunila Pradeep, Sherry Y. Wu, Rebecca A. Previs, Rajesha Rupaimoole, Kshipra M. Gharpure, Monika Haemmerle, Danielle M. Herder, Merve Ozcan, Robert Dood, Tatiana Ortiz, Nouara C. Sadaoui, Archana S. Nagaraja, Guillermo N. Armaiz-Pena, and Julie K. Allen
- Abstract
Mounting clinical and preclinical evidence supports a key role for sustained adrenergic signaling in the tumor microenvironment as a driver of tumor growth and progression. However, the mechanisms by which adrenergic neurotransmitters are delivered to the tumor microenvironment are not well understood. Here we present evidence for a feed-forward loop whereby adrenergic signaling leads to increased tumoral innervation. In response to catecholamines, tumor cells produced brain-derived neurotrophic factor (BDNF) in an ADRB3/cAMP/Epac/JNK-dependent manner. Elevated BDNF levels in the tumor microenvironment increased innervation by signaling through host neurotrophic receptor tyrosine kinase 2 receptors. In patients with cancer, high tumor nerve counts were significantly associated with increased BDNF and norepinephrine levels and decreased overall survival. Collectively, these data describe a novel pathway for tumor innervation, with resultant biological and clinical implications.Significance: Sustained adrenergic signaling promotes tumor growth and metastasis through BDNF-mediated tumoral innervation. Cancer Res; 78(12); 3233–42. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplementary data from Adrenergic Stimulation of DUSP1 Impairs Chemotherapy Response in Ovarian Cancer
- Author
-
Anil K. Sood, Steven W. Cole, Susan K. Lutgendorf, Gabriel Lopez-Berestein, Vasudha Sehgal, Prahlad Ram, Cristina Ivan, Cristian Rodriguez-Aguayo, Jean M. Hansen, Rebecca A. Previs, Kshipra M. Gharpure, Tao Liu, Rajesha Rupaimoole, Wei Hu, Piotr L. Dorniak, Guillermo N. Armaiz-Pena, Archana S. Nagaraja, and Yu Kang
- Abstract
Supplementary Figure 1 Functional genomic analysis of HeyA8 and SKOV3ip ovarian cancer cells 4 h after exposure to norepinephrine and compared to control cells. Supplementary Figure 2 A, Western blot analysis of ADRB1, ADRB2, ADRB3, and DUSP1 expression in 10 epithelial ovarian cancer cell lines and one breast cancer cell line (MDA-231), using non-transformed ovarian epithelium cells (HIO-180) as control. B, DUSP1 mRNA levels of cells exposed to norepinephrine 10 μM for 4 h. Supplementary Figure 3 A, DUSP1 silencing. Two commercially available siRNAs that directly target the DUSP1 gene were transfected to norepinephrine-treated HeyA8 cells. B, DUSP1 overexpression. SKOV3ip1 cells were transiently transfected with a DUSP1-myc/DDK-tagged expression vector (Origene). Supplementary Figure 4 SKOV3ip1 cells were treated with paclitaxel alone (10nM) or in combination with norepinephrine (NE; 10μM) or vascular endothelial growth factor (VEGF; 10ng/ml or 50ng/ml). NE or VEGF was administered 30 minutes prior to paclitaxel exposure.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Movie 2 from Sustained Adrenergic Signaling Promotes Intratumoral Innervation through BDNF Induction
- Author
-
Anil K. Sood, Mariella De Biasi, Susan K. Lutgendorf, Steve W. Cole, Gabriel Lopez-Berestein, Frank C. Marini, Stephen T.C. Wong, Prahlad T. Ram, Erika L. Spaeth, Vasudha Sehgal, Cristian Rodriguez-Aguayo, Lingegowda S. Mangala, Yu Kang, Myrthala Moreno-Smith, Justin Bottsford-Miller, Wei Hu, Morgan Taylor, Heather J. Dalton, Behrouz Zand, Hee Dong Han, Xiaoyun Xu, Sunila Pradeep, Sherry Y. Wu, Rebecca A. Previs, Rajesha Rupaimoole, Kshipra M. Gharpure, Monika Haemmerle, Danielle M. Herder, Merve Ozcan, Robert Dood, Tatiana Ortiz, Nouara C. Sadaoui, Archana S. Nagaraja, Guillermo N. Armaiz-Pena, and Julie K. Allen
- Abstract
CARS imaging of ex vivo ovarian cancer samples
- Published
- 2023
- Full Text
- View/download PDF
19. Figures S1-S6, Table S1-S3 from Sustained Adrenergic Signaling Promotes Intratumoral Innervation through BDNF Induction
- Author
-
Anil K. Sood, Mariella De Biasi, Susan K. Lutgendorf, Steve W. Cole, Gabriel Lopez-Berestein, Frank C. Marini, Stephen T.C. Wong, Prahlad T. Ram, Erika L. Spaeth, Vasudha Sehgal, Cristian Rodriguez-Aguayo, Lingegowda S. Mangala, Yu Kang, Myrthala Moreno-Smith, Justin Bottsford-Miller, Wei Hu, Morgan Taylor, Heather J. Dalton, Behrouz Zand, Hee Dong Han, Xiaoyun Xu, Sunila Pradeep, Sherry Y. Wu, Rebecca A. Previs, Rajesha Rupaimoole, Kshipra M. Gharpure, Monika Haemmerle, Danielle M. Herder, Merve Ozcan, Robert Dood, Tatiana Ortiz, Nouara C. Sadaoui, Archana S. Nagaraja, Guillermo N. Armaiz-Pena, and Julie K. Allen
- Abstract
Fig. S1. Sustained adrenergic signaling increases nerve counts in tumors. Fig. S2. Characterization of tumoral innervation. Fig. S3. NE induces BDNF expression. Fig. S4. NE-induced BDNF expression is mediated by ADRB3/Epac/Jnk. Fig. S5. BDNF increases nerve counts. Fig. S6. Adrenergic-mediated mTrkB activation leads to increased in vivo tumor nodule counts. Table S1. Alteration in pathways associated with neuronal growth and function after NE treatment (HeyA8 and SKOV3ip1 cells). Table S2. Association of Clinicopathologic variables with BDNF protein expression. Table S3. Association of Clinicopathologic variables with Nerve Counts.
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary figures S1-5, Tables S1-5 from XPO1/CRM1 Inhibition Causes Antitumor Effects by Mitochondrial Accumulation of eIF5A
- Author
-
Anil K. Sood, Robert L. Coleman, John E. Wiktorowicz, David H. Hawke, Dilara McCauley, Sharon Shacham, Tao Liu, Yu Kang, Takeshi Hisamatsu, Morgan L. Taylor, Rebecca A. Previs, Heather J. Dalton, Min Soon Cho, Wei Hu, Archana S. Nagaraja, Kshipra M. Gharpure, Yunfei Wen, Behrouz Zand, Rajesha Rupaimoole, Sherry Y. Wu, Sunila Pradeep, and Takahito Miyake
- Abstract
Supplementary Figures S1-5, Tables S1-5. Figure S1 The Cytotoxic Effect of KPT-185 is Tumor Suppressor-independent. Figure S2 Combination Index of KPT-185 with Chemotherapeutic Agents. Figure S3 1D Gel Analysis of Cytoplasmic Protein. Figure S4 Effects of Selinexor (KPT-330) in an Ovarian Cancer Mouse Model. Figure S5 The Efficacy of Selinexor (KPT-330) on Tumor Regression and Metastasis Table S1 p53 status and IC50 concentration of KPT-185 after 72 hours of incubation in human cancer cell lines Table S2 Combination index (CI) of KPT-185 with cytotoxic agents Table S3 Proteins showing altered mitochondrial localization after treatment with KPT-185 Table S4 1D Proteomic analysis of cytoplasmic proteins immunoprecipitated with eIF5A Table S5 siRNA Sequences
- Published
- 2023
- Full Text
- View/download PDF
21. Data from Adrenergic Stimulation of DUSP1 Impairs Chemotherapy Response in Ovarian Cancer
- Author
-
Anil K. Sood, Steven W. Cole, Susan K. Lutgendorf, Gabriel Lopez-Berestein, Vasudha Sehgal, Prahlad Ram, Cristina Ivan, Cristian Rodriguez-Aguayo, Jean M. Hansen, Rebecca A. Previs, Kshipra M. Gharpure, Tao Liu, Rajesha Rupaimoole, Wei Hu, Piotr L. Dorniak, Guillermo N. Armaiz-Pena, Archana S. Nagaraja, and Yu Kang
- Abstract
Purpose: Chronic adrenergic activation has been shown to associate with adverse clinical outcomes in cancer patients, but the underlying mechanisms are not well understood. The focus of the current study was to determine the functional and biologic effects of adrenergic pathways on response to chemotherapy in the context of ovarian cancer.Experimental Design: Increased DUSP1 production by sympathetic nervous system mediators (e.g., norepinephrine) was analyzed by real-time quantitative RT-PCR and by Western blotting. In vitro chemotherapy-induced cell apoptosis was examined by flow cytometry. For in vivo therapy, a well-characterized model of chronic stress was used.Results: Catecholamines significantly inhibited paclitaxel- and cisplatin-induced apoptosis in ovarian cancer cells. Genomic analyses of cells treated with norepinephrine identified DUSP1 as a potential mediator. DUSP1 overexpression resulted in reduced paclitaxel-induced apoptosis in ovarian cancer cells compared with control; conversely, DUSP1 gene silencing resulted in increased apoptosis compared with control cells. DUSP1 gene silencing in vivo significantly enhanced response to paclitaxel and increased apoptosis. In vitro analyses indicated that norepinephrine-induced DUSP1 gene expression was mediated through ADRB2 activation of cAMP–PLC–PKC–CREB signaling, which inhibits JNK-mediated phosphorylation of c-Jun and protects ovarian cancer cells from apoptosis. Moreover, analysis of The Cancer Genome Atlas data showed that increased DUSP1 expression was associated with decreased overall (P = 0.049) and progression-free (P = 0.0005) survival.Conclusions: These findings provide a new understanding of the mechanisms by which adrenergic pathways can impair response to chemotherapy and have implications for cancer management. Clin Cancer Res; 22(7); 1713–24. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Movie 1 from Sustained Adrenergic Signaling Promotes Intratumoral Innervation through BDNF Induction
- Author
-
Anil K. Sood, Mariella De Biasi, Susan K. Lutgendorf, Steve W. Cole, Gabriel Lopez-Berestein, Frank C. Marini, Stephen T.C. Wong, Prahlad T. Ram, Erika L. Spaeth, Vasudha Sehgal, Cristian Rodriguez-Aguayo, Lingegowda S. Mangala, Yu Kang, Myrthala Moreno-Smith, Justin Bottsford-Miller, Wei Hu, Morgan Taylor, Heather J. Dalton, Behrouz Zand, Hee Dong Han, Xiaoyun Xu, Sunila Pradeep, Sherry Y. Wu, Rebecca A. Previs, Rajesha Rupaimoole, Kshipra M. Gharpure, Monika Haemmerle, Danielle M. Herder, Merve Ozcan, Robert Dood, Tatiana Ortiz, Nouara C. Sadaoui, Archana S. Nagaraja, Guillermo N. Armaiz-Pena, and Julie K. Allen
- Abstract
CARS imaging of ex vivo ovarian cancer samples
- Published
- 2023
- Full Text
- View/download PDF
23. Adrenergic-mediated increases in INHBA drive CAF phenotype and collagens
- Author
-
Gabriel Lopez-Berestein, Lorenzo Cohen, Sunila Pradeep, Guillermo N. Armaiz-Pena, Cristian Rodriguez-Aguayo, Prahlad T. Ram, Samuel C. Mok, Lingegowda S. Mangala, Yasmin A. Lyons, Julie K. Allen, Cristina Ivan, Keith A. Baggerly, R.L. Dood, Jinsong Liu, Sherry Y. Wu, Yu Kang, Monika Haemmerle, Susan K. Lutgendorf, Steve W. Cole, Anil K. Sood, Kshipra M. Gharpure, Nouara C. Sadaoui, Ying Wang, Nicholas B. Jennings, and Archana S. Nagaraja
- Subjects
Expression of Concern ,0301 basic medicine ,Stromal cell ,Adrenergic ,Biology ,Metastasis ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,medicine ,030304 developmental biology ,0303 health sciences ,business.industry ,General Medicine ,medicine.disease ,Phenotype ,3. Good health ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,biology.protein ,Medicine ,ACTA2 ,Corrigendum ,business ,Ovarian cancer ,Research Article - Abstract
Adrenergic signaling is known to promote tumor growth and metastasis, but the effects on tumor stroma are not well understood. An unbiased bioinformatics approach analyzing tumor samples from patients with known biobehavioral profiles identified a prominent stromal signature associated with cancer-associated fibroblasts (CAFs) in those with a high biobehavioral risk profile (high Center for Epidemiologic Studies Depression Scale [CES-D] score and low social support). In several models of epithelial ovarian cancer, daily restraint stress resulted in significantly increased CAF activation and was abrogated by a nonspecific β-blocker. Adrenergic signaling-induced CAFs had significantly higher levels of collagen and extracellular matrix components than control tumors. Using a systems-based approach, we found INHBA production by cancer cells to induce CAFs. Ablating inhibin β A decreased CAF phenotype both in vitro and in vivo. In preclinical models of breast and colon cancers, there were increased CAFs and collagens following daily restraint stress. In an independent data set of renal cell carcinoma patients, there was an association between high depression (CES-D) scores and elevated expression of ACTA2, collagens, and inhibin β A. Collectively, our findings implicate adrenergic influences on tumor stroma as important drivers of CAFs and establish inhibin β A as an important regulator of the CAF phenotype in ovarian cancer.
- Published
- 2021
- Full Text
- View/download PDF
24. FABP4 as a key determinant of metastatic potential of ovarian cancer
- Author
-
Jinsong Liu, Guillermo N. Armaiz Pena, Kshipra M. Gharpure, Prahlad T. Ram, Anil K. Sood, Lingegowda S. Mangala, Monika Haemmerle, Rajesha Rupaimoole, Alejandro Villar-Prados, Sunila Pradeep, Archana S. Nagaraja, Xinna Zhang, Sherry Y. Wu, Pranavi Koppula, Wei Hu, Celia Sze Ling Mak, Michael McGuire, Xiuhui Chen, Cristina Ivan, Ryan Nini, Livia S. Eberlin, Keith A. Baggerly, Emine Bayraktar, Ragini Kondetimmanahalli, Michelle A. Tran, Cristian Rodriguez-Aguayo, Marta Sans, and Gabriel Lopez-Berestein
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Science ,Mice, Nude ,General Physics and Astronomy ,Disease ,Fatty Acid-Binding Proteins ,Article ,General Biochemistry, Genetics and Molecular Biology ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,lcsh:Science ,Ovarian Neoplasms ,Chemotherapy ,Multidisciplinary ,business.industry ,General Chemistry ,Hypoxia (medical) ,medicine.disease ,Debulking ,3. Good health ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,lcsh:Q ,Female ,medicine.symptom ,Ovarian cancer ,business - Abstract
The standard treatment for high-grade serous ovarian cancer is primary debulking surgery followed by chemotherapy. The extent of metastasis and invasive potential of lesions can influence the outcome of these primary surgeries. Here, we explored the underlying mechanisms that could increase metastatic potential in ovarian cancer. We discovered that FABP4 (fatty acid binding protein) can substantially increase the metastatic potential of cancer cells. We also found that miR-409-3p regulates FABP4 in ovarian cancer cells and that hypoxia decreases miR-409-3p levels. Treatment with DOPC nanoliposomes containing either miR-409-3p mimic or FABP4 siRNA inhibited tumor progression in mouse models. With RPPA and metabolite arrays, we found that FABP4 regulates pathways associated with metastasis and affects metabolic pathways in ovarian cancer cells. Collectively, these findings demonstrate that FABP4 is functionally responsible for aggressive patterns of disease that likely contribute to poor prognosis in ovarian cancer., In ovarian cancer, metastatic phenotype may impact surgical outcomes. Here, the authors show miR-409-3p regulates FABP4 which can increase metastatic potential of ovarian cancer, and treatment with DOPC nanoliposomes containing either miR-409--3p mimic or FABP4 siRNA inhibits tumor progression in mice.
- Published
- 2018
25. ADH1B promotes mesothelial clearance and ovarian cancer infiltration
- Author
-
Yunfei Wen, Kshipra M. Gharpure, Olivia D. Lara, Christopher J. LaFargue, Sunila Pradeep, Rajesha Rupaimoole, Lingegowda S. Mangala, Cristina Ivan, Wei Hu, Keith A. Baggerly, Anil K. Sood, Sherry Y. Wu, and Archana S. Nagaraja
- Subjects
0301 basic medicine ,Cell ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,medicine ,business.industry ,ECM degradation ,alcohol dehydrogenase ,medicine.disease ,Debulking ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,residual disease ,Ovarian cancer ,business ,mesothelial clearance ,Infiltration (medical) ,Extracellular Matrix Degradation ,Mesothelial Cell ,Research Paper - Abstract
Primary debulking surgery followed by adjuvant chemotherapy is the standard treatment for ovarian cancer. Residual disease after primary surgery is associated with poor patient outcome. Previously, we discovered ADH1B to be a molecular biomarker of residual disease. In the current study, we investigated the functional role of ADH1B in promoting ovarian cancer cell invasiveness and contributing to residual disease. We discovered that ADH1B overexpression leads to a more infiltrative cancer cell phenotype, promotes metastasis, increases the adhesion of cancer cells to mesothelial cells, and increases extracellular matrix degradation. Live cell imaging revealed that ADH1B-overexpressing cancer cells efficiently cleared the mesothelial cell layer compared to control cells. Moreover, gene array analysis revealed that ADH1B affects several pathways related to the migration and invasion of cancer cells. We also discovered that hypoxia increases ADH1B expression in ovarian cancer cells. Collectively, these findings indicate that ADH1B plays an important role in the pathways that promote ovarian cancer cell infiltration and may increase the likelihood of residual disease following surgery.
- Published
- 2018
26. Author Correction: Hypoxia-mediated downregulation of miRNA biogenesis promotes tumour progression
- Author
-
Mircea Ivan, Li Huang, Kshipra M. Gharpure, Nouara C. Sadaoui, Anil K. Sood, Cristina Ivan, Lingegowda S. Mangala, Behrouz Zand, Guillermo N. Armaiz-Pena, Elizabeth Koch, George A. Calin, Sherry Y. Wu, Morgan Taylor, Milan Radovich, Menashe Bar-Eli, Wei Zhang, Gabriel Lopez-Berestein, Justyna Filant, Rajesha Rupaimoole, Twan van den Beucken, Bradly G. Wouters, Sunila Pradeep, Michael McGuire, Cristian Rodriguez-Aguayo, Chad V. Pecot, Justin Bottsford Miller, Heather J. Dalton, Archana S. Nagaraja, and Chunhua Lu
- Subjects
Multidisciplinary ,business.industry ,Science ,General Physics and Astronomy ,General Chemistry ,Hypoxia (medical) ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Text mining ,Downregulation and upregulation ,medicine ,Cancer research ,lcsh:Q ,medicine.symptom ,lcsh:Science ,MiRNA biogenesis ,business - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
27. ADAMTS16 mutations sensitize ovarian cancer cells to platinum-based chemotherapy
- Author
-
Anil K. Sood, Kshipra M. Gharpure, Maya Yasukawa, Archana S. Nagaraja, Yuexin Liu, Limei Hu, Wei Zhang, David Cogdell, and Sunila Pradeep
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,endocrine system diseases ,medicine.medical_treatment ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Ovarian carcinoma ,BRCAness ,medicine ,platinum-based chemotherapy ,Cisplatin ,Chemotherapy ,Cell growth ,business.industry ,ADAMTS ,ADAMTS16 ,medicine.disease ,female genital diseases and pregnancy complications ,3. Good health ,chemosensitivity ,Serous fluid ,ovarian cancer ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Ovarian cancer ,business ,Research Paper ,medicine.drug - Abstract
Ovarian cancer is one of the most lethal malignant tumors in women. The prognosis of ovarian cancer patients depends, in part, on their response to platinum-based chemotherapy. Our recent analysis of genomics and clinical data from the Cancer Genome Atlas demonstrated that somatic mutations of ADAMTS 1, 6, 8, 9, 15, 16, 18 and L1 genes were associated with higher sensitivity to platinum and longer progression-free survival, overall survival, and platinum-free survival duration in 512 patients with high-grade serous ovarian carcinoma. Among the ADAMTS mutations, ADAMTS16 is the most commonly affected gene in ovarian cancer. However, the functional role of these mutations in ovarian cancer cells is largely unknown. We performed in vitro studies to compare the functional effects of the six identified ADAMTS missense mutations on the platinum sensitivity of ovarian cancer cells. We also used a well-characterized in vivo mouse model to evaluate the response of ovarian cancer cells with ADAMTS16 mutations to platinum-based therapy. Our results showed that exogenously expressed ADAMTS16 missense mutations inhibited cell growth or sensitized tumor cells to cisplatin and inhibited tumor growth in vivo. Orthotopic xenograft experiments showed that mice injected with ovarian cancer cells that exogenously expressed ADAMTS16 mutations had a better response to cisplatin treatment. Thus, these functional studies provide evidence that mutations of ADAMTS16 actively contribute to therapeutic response in ovarian cancer.
- Published
- 2016
- Full Text
- View/download PDF
28. PRKRA/PACT Expression Promotes Chemoresistance of Mucinous Ovarian Cancer
- Author
-
Yasmin A. Lyons, Anil K. Sood, Sunila Pradeep, Cristian Rodriguez-Aguayo, Takashi Mitamura, Wei Hu, Jean M. Hansen, Emine Bayraktar, Koji Matsuo, Kshipra M. Gharpure, Archana S. Nagaraja, Takeshi Hisamatsu, Geoffrey Bartholomeusz, Gabriel Lopez-Berestein, Cristina Ivan, Sherry Y. Wu, Young-Gyu Eun, Lingegowda S. Mangala, Johnathon L. Rose, Kwong Kwok Wong, Michael Frumovitz, Ju Seog Lee, Michael McGuire, Kyunghee Noh, and Rajesha Rupaimoole
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Kinase ,business.industry ,Activator (genetics) ,medicine.disease ,Pact ,Protein kinase R ,Article ,Oxaliplatin ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,In vivo ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Cancer research ,Kinome ,business ,Ovarian cancer ,medicine.drug - Abstract
For mucinous ovarian cancer (MOC), standard platinum-based therapy is largely ineffective. We sought to identify possible mechanisms of oxaliplatin resistance of MOC and develop strategies to overcome this resistance. A kinome-based siRNA library screen was carried out using human MOC cells to identify novel targets to enhance the efficacy of chemotherapy. In vitro and in vivo validations of antitumor effects were performed using mouse MOC models. Specifically, the role of PRKRA/PACT in oxaliplatin resistance was interrogated. We focused on PRKRA, a known activator of PKR kinase, and its encoded protein PACT because it was one of the five most significantly downregulated genes in the siRNA screen. In orthotopic mouse models of MOC, we observed a significant antitumor effect of PRKRA siRNA plus oxaliplatin. In addition, expression of miR-515-3p was regulated by PACT–Dicer interaction, and miR-515-3p increased the sensitivity of MOC to oxaliplatin. Mechanistically, miR-515-3p regulated chemosensitivity, in part, by targeting AXL. The PRKRA/PACT axis represents an important therapeutic target in MOC to enhance sensitivity to oxaliplatin.
- Published
- 2018
29. Improving vascular maturation using noncoding RNAs increases antitumor effect of chemotherapy
- Author
-
Cristina Ivan, Xinna Zhang, Sunila Pradeep, Dahai Jiang, Monika Haemmerle, Kshipra M. Gharpure, Takemi Tanaka, Sourindra Maiti, David G. Gorenstein, Rajesha Rupaimoole, Ganesh L.R. Lokesh, Sherry Y. Wu, Hongyu Wang, Archana S. Nagaraja, Gabriel Lopez-Berestein, Lingegowda S. Mangala, Anil K. Sood, Nataliya Bulayeva, Cristian Rodriguez-Aguayo, David E. Volk, Emine Bayraktar, Xin Li, Hyun Jin Choi, Xianbin Yang, Varatharasa Thiviyanathan, Recep Bayraktar, Laurence J.N. Cooper, Wei Hu, Kevin P. Rosenblatt, Li Li, Michael McGuire, Anoma Somasunderam, and Piotr L. Dorniak
- Subjects
0301 basic medicine ,Expression of Concern ,medicine.medical_treatment ,Antineoplastic Agents ,Transfection ,Neovascularization ,Antiangiogenesis Therapy ,03 medical and health sciences ,0302 clinical medicine ,Annexin ,Cell Line, Tumor ,Neoplasms ,microRNA ,Gene silencing ,Medicine ,Humans ,Chemotherapy ,Neovascularization, Pathologic ,business.industry ,Endothelial Cells ,General Medicine ,Hypoxia (medical) ,Aptamers, Nucleotide ,3. Good health ,MicroRNAs ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Nanoparticles ,medicine.symptom ,business ,Corrigendum ,Research Article - Abstract
Current antiangiogenesis therapy relies on inhibiting newly developed immature tumor blood vessels and starving tumor cells. This strategy has shown transient and modest efficacy. Here, we report a better approach to target cancer-associated endothelial cells (ECs), reverse permeability and leakiness of tumor blood vessels, and improve delivery of chemotherapeutic agents to the tumor. First, we identified deregulated microRNAs (miRs) from patient-derived cancer-associated ECs. Silencing these miRs led to decreased vascular permeability and increased maturation of blood vessels. Next, we screened a thioaptamer (TA) library to identify TAs selective for tumor-associated ECs. An annexin A2-targeted TA was identified and used for delivery of miR106b-5p and miR30c-5p inhibitors, resulting in vascular maturation and antitumor effects without inducing hypoxia. These findings could have implications for improving vascular-targeted therapy.
- Published
- 2018
30. Hypoxia-upregulated microRNA-630 targets Dicer, leading to increased tumor progression
- Author
-
Li Huang, Guillermo N. Armaiz-Pena, Sunila Pradeep, Da Yang, Menashe Bar-Eli, Anil K. Sood, Michael McGuire, Rebecca A. Previs, Chad V. Pecot, Archana S. Nagaraja, Wei Zhang, Sherry Y. Wu, Cristian Rodriguez-Aguayo, Gabriel Lopez-Berestein, Lingegowda S. Mangala, Kshipra M. Gharpure, Rajesha Rupaimoole, Cristina Ivan, and George A. Calin
- Subjects
Ribonuclease III ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Cancer Research ,Biology ,Bioinformatics ,Article ,Metastasis ,DEAD-box RNA Helicases ,Mice ,03 medical and health sciences ,Growth factor receptor ,Cell Line, Tumor ,Neoplasms ,Gene expression ,microRNA ,Genetics ,medicine ,Animals ,Humans ,Hypoxia ,Molecular Biology ,Drosha ,Ovarian Neoplasms ,Regulation of gene expression ,medicine.disease ,Cell Hypoxia ,3. Good health ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,miRNA-630 ,030104 developmental biology ,Tumor progression ,miRNAs ,Liposomes ,Disease Progression ,Phosphatidylcholines ,biology.protein ,Cancer research ,Female ,Dicer - Abstract
MicroRNAs (miRNAs) are small RNA molecules that affect cellular processes by controlling gene expression. Recent studies have shown that hypoxia downregulates Drosha and Dicer, key enzymes in miRNA biogenesis, causing a decreased pool of miRNAs in cancer and resulting in increased tumor growth and metastasis. Here we demonstrate a previously unrecognized mechanism by which hypoxia downregulates Dicer. We found that miR-630, which is upregulated under hypoxic conditions, targets and downregulates Dicer expression. In an orthotopic mouse model of ovarian cancer, delivery of miR-630 using 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) nanoliposomes resulted in increased tumor growth and metastasis, and decreased Dicer expression. Treatment with the combination of anti-miR-630 and anti-vascular endothelial growth factor antibody in mice resulted in rescue of Dicer expression and significantly decreased tumor growth and metastasis. These results indicate that targeting miR-630 is a promising approach to overcome Dicer deregulation in cancer. As demonstrated in the study, use of DOPC nanoliposomes for anti-miR delivery serves as a better alternative approach to cell line-based overexpression of sense or antisense miRNAs, while avoiding potential in vitro selection effects. Findings from this study provide a new understanding of miRNA biogenesis downregulation observed under hypoxia and suggest therapeutic avenues to target this dysregulation in cancer.
- Published
- 2016
- Full Text
- View/download PDF
31. Long Noncoding RNA Ceruloplasmin Promotes Cancer Growth by Altering Glycolysis
- Author
-
Nouara C. Sadaoui, Gabriel Lopez-Berestein, Jaehyuk Lee, Jennifer B. Dennison, Cristina Ivan, Rebecca A. Previs, Manuela Ferracin, Cristian Rodriguez-Aguayo, Monika Haemmerle, Pratip K. Bhattacharya, Kshipra M. Gharpure, Anil K. Sood, Rajesha Rupaimoole, Jeffery Kovacs, Sunila Pradeep, Wei Zhang, Sherry Y. Wu, Nidhin Sam, Michael McGuire, Ronny Drapkin, Hui Ling, Niki Zacharias Millward, Archana S. Nagaraja, Gordon B. Mills, Guillermo N. Armaiz-Pena, George A. Calin, Rupaimoole, Rajesha, Lee, Jaehyuk, Haemmerle, Monika, Ling, Hui, Previs, Rebecca A., Pradeep, Sunila, Wu, Sherry Y., Ivan, Cristina, Ferracin, Manuela, Dennison, Jennifer B., Millward, Niki M. Zacharia, Nagaraja, Archana S., Gharpure, Kshipra M., Mcguire, Michael, Sam, Nidhin, Armaiz-Pena, Guillermo N., Sadaoui, Nouara C., Rodriguez-Aguayo, Cristian, Calin, George A., Drapkin, Ronny I., Kovacs, Jeffery, Mills, Gordon B., Zhang, Wei, Lopez-Berestein, Gabriel, Bhattacharya, Pratip K., and Sood, Anil K.
- Subjects
Transplantation, Heterologous ,Mice, Nude ,Apoptosis ,Kaplan-Meier Estimate ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,RNA interference ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,RNA, Small Interfering ,lcsh:QH301-705.5 ,Cell Proliferation ,Ovarian Neoplasms ,Gene knockdown ,Glucose-6-Phosphate Isomerase ,Ceruloplasmin ,RNA ,Cancer ,medicine.disease ,Long non-coding RNA ,3. Good health ,STAT1 Transcription Factor ,lcsh:Biology (General) ,Tumor progression ,Cancer cell ,Disease Progression ,Cancer research ,lncRNAs, cancer metabolism ,Female ,RNA Interference ,RNA, Long Noncoding ,RNA Polymerase II ,Glycolysis - Abstract
SummaryLong noncoding RNAs (lncRNAs) significantly influence the development and regulation of genome expression in cells. Here, we demonstrate the role of lncRNA ceruloplasmin (NRCP) in cancer metabolism and elucidate functional effects leading to increased tumor progression. NRCP was highly upregulated in ovarian tumors, and knockdown of NRCP resulted in significantly increased apoptosis, decreased cell proliferation, and decreased glycolysis compared with control cancer cells. In an orthotopic mouse model of ovarian cancer, siNRCP delivered via a liposomal carrier significantly reduced tumor growth compared with control treatment. We identified NRCP as an intermediate binding partner between STAT1 and RNA polymerase II, leading to increased expression of downstream target genes such as glucose-6-phosphate isomerase. Collectively, we report a previously unrecognized role of the lncRNA NRCP in modulating cancer metabolism. As demonstrated, DOPC nanoparticle-incorporated siRNA-mediated silencing of this lncRNA in vivo provides therapeutic avenue toward modulating lncRNAs in cancer.
- Published
- 2015
- Full Text
- View/download PDF
32. Erythropoietin Stimulates Tumor Growth via EphB4
- Author
-
Shyon Haghpeykar, Jean M. Hansen, Rebecca L. Stone, Chiyi Xiong, Alpa M. Nick, Sunila Pradeep, Kshipra M. Gharpure, Gabriel Lopez-Berestein, Guillermo N. Armaiz-Pena, Mien Chie Hung, Chun Li, Edna M. Mora, Cristian Rodriguez-Aguayo, Sherry Y. Wu, Archana S. Nagaraja, Martin Stein, Hee Dong Han, Rebecca A. Previs, Anil K. Sood, Blake W. Goodman, Kyunghee Noh, Masato Nishimura, Robert L. Coleman, Armin Schneider, Min Soon Cho, Chad V. Pecot, Rajesha Rupaimoole, Jie Huang, Lingegowda S. Mangala, Bulent Ozpolat, Yunfei Wen, Jinsong Liu, Pablo E. Vivas-Mejia, Stephan Brock, Padmavathi Jaladurgam, John E. Ladbury, Diana L. Urbauer, Koji Matsuo, Heather J. Dalton, Carola Krüger, Christopher G. Danes, David B. Jackson, Loren J. Stagg, Wei Hu, Behrouz Zand, and S. Neslihan Alpay
- Subjects
Adult ,STAT3 Transcription Factor ,Cancer Research ,Blotting, Western ,Receptor, EphB4 ,Mice, Nude ,Breast Neoplasms ,Kaplan-Meier Estimate ,Article ,Young Adult ,Breast cancer ,Mediator ,Cell Line, Tumor ,hemic and lymphatic diseases ,Receptors, Erythropoietin ,medicine ,Animals ,Humans ,STAT3 ,Erythropoietin ,Aged ,Aged, 80 and over ,Ovarian Neoplasms ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Cancer ,Cell Biology ,Middle Aged ,medicine.disease ,Recombinant Proteins ,Erythropoietin receptor ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,Oncology ,Tumor progression ,Immunology ,Cancer cell ,Disease Progression ,MCF-7 Cells ,biology.protein ,Cancer research ,Female ,Protein Binding ,medicine.drug - Abstract
SummaryWhile recombinant human erythropoietin (rhEpo) has been widely used to treat anemia in cancer patients, concerns about its adverse effects on patient survival have emerged. A lack of correlation between expression of the canonical EpoR and rhEpo’s effects on cancer cells prompted us to consider the existence of an alternative Epo receptor. Here, we identified EphB4 as an Epo receptor that triggers downstream signaling via STAT3 and promotes rhEpo-induced tumor growth and progression. In human ovarian and breast cancer samples, expression of EphB4 rather than the canonical EpoR correlated with decreased disease-specific survival in rhEpo-treated patients. These results identify EphB4 as a critical mediator of erythropoietin-induced tumor progression and further provide clinically significant dimension to the biology of erythropoietin.
- Published
- 2015
- Full Text
- View/download PDF
33. Sustained Adrenergic Signaling Promotes Intratumoral Innervation Through Bdnf Induction
- Author
-
Archana S. Nagaraja, Monika Haemmerle, Morgan Taylor, Danielle M. Herder, Stephen T. C. Wong, Merve Ozcan, Nouara C. Sadaoui, Mariella De Biasi, Frank C. Marini, Prahlad T. Ram, Lingegowda S. Mangala, Heather J. Dalton, Steve W. Cole, Wei Hu, Kshipra M. Gharpure, Susan K. Lutgendorf, Hee Dong Han, Anil K. Sood, Vasudha Sehgal, Sherry Y. Wu, Sunila Pradeep, Rebecca A. Previs, Yu Kang, Rajesha Rupaimoole, Erika L. Spaeth, Xiaoyun Xu, Myrthala Moreno-Smith, R.L. Dood, Behrouz Zand, Cristian Rodriguez-Aguayo, Tatiana Ortiz, Julie K. Allen, Guillermo N. Armaiz-Pena, Gabriel Lopez-Berestein, Justin Bottsford-Miller, and Temel Onkoloji
- Subjects
0301 basic medicine ,Cancer Research ,Adrenergic ,Receptor tyrosine kinase ,Mice ,Norepinephrine ,03 medical and health sciences ,Neurotrophic factors ,Cell Line, Tumor ,Neoplasms ,Cyclic AMP ,Tumor Microenvironment ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Receptor, trkB ,Medicine ,Peripheral Nerves ,Receptor ,Intratumoral innervation ,Feedback, Physiological ,Brain-derived neurotrophic factor ,Tumor microenvironment ,Membrane Glycoproteins ,biology ,business.industry ,Brain-Derived Neurotrophic Factor ,Bdnf induction ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Oncology ,Adrenergic signaling ,Receptors, Adrenergic, beta-3 ,biology.protein ,Cancer research ,Female ,Biyokimya. Hücre biyolojisi. Hücre genetiği ,Signal transduction ,business ,Signal Transduction ,Neurotrophin - Abstract
Mounting clinical and preclinical evidence supports a key role for sustained adrenergic signaling in the tumor microenvironment as a driver of tumor growth and progression. However, the mechanisms by which adrenergic neurotransmitters are delivered to the tumor microenvironment are not well understood. Here we present evidence for a feed-forward loop whereby adrenergic signaling leads to increased tumoral innervation. In response to catecholamines, tumor cells produced brain-derived neurotrophic factor (BDNF) in an ADRB3/cAMP/Epac/JNK-dependent manner. Elevated BDNF levels in the tumor microenvironment increased innervation by signaling through host neurotrophic receptor tyrosine kinase 2 receptors. In patients with cancer, high tumor nerve counts were significantly associated with increased BDNF and norepinephrine levels and decreased overall survival. Collectively, these data describe a novel pathway for tumor innervation, with resultant biological and clinical implications. Significance: Sustained adrenergic signaling promotes tumor growth and metastasis through BDNF-mediated tumoral innervation. Cancer Res; 78(12); 3233–42. ©2018 AACR.
- Published
- 2018
34. List of Contributors
- Author
-
Santanu Adhikary, Carlos Baliñas, Shakri Banerjee, Sharmila A. Bapat, Rahul Bhome, Liberalis D. Boila, Vincenzo Bonnici, Debojit Bose, Laura Boyero, Jayprokas Chakrabarti, Damayanti Chakravarti, JiaJia Chan, Alain Chariot, Shankha S. Chatterjee, Taiping Chen, Pierre Close, Isabel F. Coira, Marta Cuadros, Juan Cui, Chandrima Das, Shaoli Das, Simone de Brot, Ng Desi, Nadya Dimitrova, Rachel L. Dittmar, Katherine Emo, Sayak Ganguli, Kshipra M. Gharpure, Suman Ghosal, Rosalba Giugno, Damjan Glavacˇ, Frank Grützner, Sohini Gupta, Agnik Halder, Nina Hauptman, Antonio Herrera, Louise House, Changzhi Huang, Shenglin Huang, Reuben Jacob, Victoria James, Daša Jevšinek Skok, Ragini Kondetimmanahalli, Eunice Lee, Sebastian A. Leidel, Yan Li, Yuan Li, Gabriel Lopez-Berestein, Jose A. Lupiáñez, Joel Martín-Padrón, Pedro P. Medina, Alex Mirnezami, Sanga Mitra, Nigel P. Mongan, Shravanti Mukherjee, Suchismita Panda, Paola Peinado, Alfredo Pulvirenti, Fernando J. Reyes-Zurita, Maria I. Rodriguez, Susanta Roychoudhury, Siddhartha Roy, Eva E. Rufino-Palomares, Catrin S. Rutland, Emre Sayan, Flavia Scoyni, Amitava Sengupta, Sharmila Sengupta, Subrata Sen, Sweta Sharma Saha, Anjali Shiras, Yuming Shi, Sayantani Sinha, Ondrej Slaby, Anil K. Sood, Kamila Souckova, Yvonne Tay, Yu Wang, Zhiguo Wang, Sherry Y. Wu, Tianqi Yang, Eda Yildirim, and Zhengzhou Ying
- Published
- 2018
- Full Text
- View/download PDF
35. Platelets reduce anoikis and promote metastasis by activating YAP1 signaling
- Author
-
Tony Gutschner, Cristian Rodriguez-Aguayo, Min Soon Cho, Sunila Pradeep, Anil K. Sood, Jinsong Liu, Yunfei Wen, Ju Seog Lee, Vahid Afshar-Kharghan, Gabriel Lopez-Berestein, Beth Y. Karlan, Stephen T. C. Wong, R.L. Dood, Cristina Ivan, Sun Young Yim, Rajesha Rupaimoole, Douglas A. Levine, Kshipra M. Gharpure, Lingegowda S. Mangala, Yasmin M. Lyons, Wei Hu, Monika Haemmerle, Morgan Taylor, Archana S. Nagaraja, Jianting Sheng, and Jean M. Hansen
- Subjects
0301 basic medicine ,RHOA ,Nude ,General Physics and Astronomy ,Inbred C57BL ,Metastasis ,Mice ,0302 clinical medicine ,2.1 Biological and endogenous factors ,Anoikis ,Platelet ,Aetiology ,Neoplasm Metastasis ,Cancer ,Ovarian Neoplasms ,YAP1 ,Heterologous ,Tumor ,Multidisciplinary ,biology ,Chemistry ,Adaptor Proteins ,3. Good health ,030220 oncology & carcinogenesis ,RNA Interference ,Female ,Signal transduction ,Signal Transduction ,Blood Platelets ,Science ,Transplantation, Heterologous ,Mice, Nude ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,03 medical and health sciences ,Rare Diseases ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Adaptor Proteins, Signal Transducing ,Transplantation ,Tumor microenvironment ,Gene Expression Profiling ,Signal Transducing ,YAP-Signaling Proteins ,General Chemistry ,Phosphoproteins ,medicine.disease ,Coculture Techniques ,Mice, Inbred C57BL ,030104 developmental biology ,Cancer cell ,Immunology ,biology.protein ,Cancer research ,Transcription Factors - Abstract
Thrombocytosis is present in more than 30% of patients with solid malignancies and correlates with worsened patient survival. Tumor cell interaction with various cellular components of the tumor microenvironment including platelets is crucial for tumor growth and metastasis. Although it is known that platelets can infiltrate into tumor tissue, secrete pro-angiogenic and pro-tumorigenic factors and thereby increase tumor growth, the precise molecular interactions between platelets and metastatic cancer cells are not well understood. Here we demonstrate that platelets induce resistance to anoikis in vitro and are critical for metastasis in vivo. We further show that platelets activate RhoA-MYPT1-PP1-mediated YAP1 dephosphorylation and promote its nuclear translocation which induces a pro-survival gene expression signature and inhibits apoptosis. Reduction of YAP1 in cancer cells in vivo protects against thrombocytosis-induced increase in metastasis. Collectively, our results indicate that cancer cells depend on platelets to avoid anoikis and succeed in the metastatic process., Platelets have been associated with increased tumor growth and metastasis but the mechanistic details of this interaction are still unclear. Here the authors show that platelets improve anoikis resistance of cancer cells and increase metastasis by activating Yap through a RhoA/MYPT-PP1 pathway.
- Published
- 2017
- Full Text
- View/download PDF
36. Metabolic Markers and Statistical Prediction of Serous Ovarian Cancer Aggressiveness by Ambient Ionization Mass Spectrometry Imaging
- Author
-
Anil K. Sood, Jonathan H. Young, R.L. Dood, Livia S. Eberlin, Li Liang, Marta Sans, Jinsong Liu, Robert Tibshirani, Kshipra M. Gharpure, and Jialing Zhang
- Subjects
0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Spectrometry, Mass, Electrospray Ionization ,Serous carcinoma ,Biology ,Carcinoma, Ovarian Epithelial ,Mass spectrometry imaging ,Article ,03 medical and health sciences ,0302 clinical medicine ,Stroma ,medicine ,Carcinoma ,Biomarkers, Tumor ,Humans ,Neoplasms, Glandular and Epithelial ,Cystadenocarcinoma ,Ambient ionization ,Ovarian Neoplasms ,Cancer ,medicine.disease ,Cystadenocarcinoma, Serous ,Serous fluid ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Female - Abstract
Ovarian high-grade serous carcinoma (HGSC) results in the highest mortality among gynecological cancers, developing rapidly and aggressively. Dissimilarly, serous borderline ovarian tumors (BOT) can progress into low-grade serous carcinomas and have relatively indolent clinical behavior. The underlying biological differences between HGSC and BOT call for accurate diagnostic methodologies and tailored treatment options, and identification of molecular markers of aggressiveness could provide valuable biochemical insights and improve disease management. Here, we used desorption electrospray ionization (DESI) mass spectrometry (MS) to image and chemically characterize the metabolic profiles of HGSC, BOT, and normal ovarian tissue samples. DESI-MS imaging enabled clear visualization of fine papillary branches in serous BOT and allowed for characterization of spatial features of tumor heterogeneity such as adjacent necrosis and stroma in HGSC. Predictive markers of cancer aggressiveness were identified, including various free fatty acids, metabolites, and complex lipids such as ceramides, glycerophosphoglycerols, cardiolipins, and glycerophosphocholines. Classification models built from a total of 89,826 individual pixels, acquired in positive and negative ion modes from 78 different tissue samples, enabled diagnosis and prediction of HGSC and all tumor samples in comparison with normal tissues, with overall agreements of 96.4% and 96.2%, respectively. HGSC and BOT discrimination was achieved with an overall accuracy of 93.0%. Interestingly, our classification model allowed identification of three BOT samples presenting unusual histologic features that could be associated with the development of low-grade carcinomas. Our results suggest DESI-MS as a powerful approach for rapid serous ovarian cancer diagnosis based on altered metabolic signatures. Cancer Res; 77(11); 2903–13. ©2017 AACR.
- Published
- 2017
37. Role of YAP1 as a Marker of Sensitivity to Dual AKT and P70S6K Inhibition in Ovarian and Uterine Malignancies
- Author
-
Kshipra M. Gharpure, Cristina Ivan, Anil K. Sood, Sarah L. Linesch, Kyunghee Noh, Jie Huang, Rebecca A. Previs, Monika Haemmerle, Rajesha Rupaimoole, Lingegowda S. Mangala, Michael J. Wagner, Michael McGuire, Guillermo N. Armaiz-Pena, Sunila Pradeep, Heather J. Dalton, Sherry Y. Wu, Archana S. Nagaraja, Yasmin A. Lyons, Piotr L. Dorniak, Justyna Filant, and Jean M. Hansen
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,Angiogenesis ,Uterus ,AKT1 ,Angiogenesis Inhibitors ,Apoptosis ,Kaplan-Meier Estimate ,chemistry.chemical_compound ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Ovarian Neoplasms ,Ribosomal Protein S6 Kinases, 70-kDa ,Articles ,Tumor Burden ,Bevacizumab ,medicine.anatomical_structure ,Paclitaxel ,030220 oncology & carcinogenesis ,Uterine Neoplasms ,Female ,medicine.drug ,medicine.medical_specialty ,Mice, Nude ,Biology ,Inhibitory Concentration 50 ,03 medical and health sciences ,Uterine cancer ,Cell Line, Tumor ,Internal medicine ,Biomarkers, Tumor ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Cancer ,YAP-Signaling Proteins ,Phosphoproteins ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Endocrinology ,chemistry ,Ovarian cancer ,Proto-Oncogene Proteins c-akt ,Transcription Factors - Abstract
Background The PI3K/AKT/P70S6K pathway is an attractive therapeutic target in ovarian and uterine malignancies because of its high rate of deregulation and key roles in tumor growth. Here, we examined the biological effects of MSC2363318A, which is a novel inhibitor of AKT1, AKT3, and P70S6K. Methods Orthotopic murine models of ovarian and uterine cancer were utilized to study the effect of MSC2363318A on survival and regression. For each cell line, 10 mice were treated in each of the experimental arms tested. Moreover, in vitro experiments in 21 cell lines (MTT, immunoblot analysis, plasmid transfection, reverse phase protein array [RPPA]) were carried out to characterize underlying mechanisms and potential biomarkers of response. All statistical tests were two-sided. Results MSC2363318A decreased tumor growth and metastases in multiple murine orthotopic models of ovarian (SKOV3ip1, HeyA8, and Igrov1) and uterine (Hec1a) cancer by reducing proliferation and angiogenesis and increasing cell death. Statistically significant prolonged overall survival was achieved with combination MSC2363318A and paclitaxel in the SKUT2 (endometrioid) uterine cancer mouse model ( P
- Published
- 2017
- Full Text
- View/download PDF
38. Metronomic Docetaxel in PRINT Nanoparticles and EZH2 Silencing Have Synergistic Antitumor Effect in Ovarian Cancer
- Author
-
J. Christopher Luft, Takahito Miyake, Sunila Pradeep, Kevin S. Chu, Mary E. Napier, Hee Dong Han, Kshipra M. Gharpure, Sherry Y. Wu, Charles J. Bowerman, Gabriel Lopez-Berestein, Guillermo N. Armaiz-Pena, Joseph M. DeSimone, Rajesha Rupaimoole, Anil K. Sood, Selanere Mangala, and Tojan B. Rahhal
- Subjects
Cancer Research ,Proliferation index ,Combination therapy ,Mice, Nude ,Antineoplastic Agents ,Caspase 3 ,Docetaxel ,Pharmacology ,Biology ,Article ,Mice ,In vivo ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Gene silencing ,Enhancer of Zeste Homolog 2 Protein ,Gene Silencing ,RNA, Small Interfering ,Randomized Controlled Trials as Topic ,Ovarian Neoplasms ,Polycomb Repressive Complex 2 ,medicine.disease ,Oncology ,Apoptosis ,Administration, Metronomic ,Nanoparticles ,Female ,Taxoids ,Ovarian cancer ,medicine.drug - Abstract
The purpose of this study was to investigate the antitumor effects of a combination of metronomic doses of a novel delivery vehicle, PLGA-PRINT nanoparticles containing docetaxel, and antiangiogenic mEZH2 siRNA incorporated into chitosan nanoparticles. In vivo dose-finding studies and therapeutic experiments were conducted in well-established orthotopic mouse models of epithelial ovarian cancer. Antitumor effects were determined on the basis of reduction in mean tumor weight and number of metastatic tumor nodules in the animals. The tumor tissues from these in vivo studies were stained to evaluate the proliferation index (Ki67), apoptosis index (cleaved caspase 3), and microvessel density (CD31). The lowest dose of metronomic regimen (0.5 mg/kg) resulted in significant reduction in tumor growth. The combination of PLGA-PRINT-docetaxel and CH-mEZH2 siRNA showed significant antitumor effects in the HeyA8 and SKOV3ip1 tumor models (P < 0.05). Individual as well as combination therapies showed significant antiangiogenic, antiproliferative, and proapoptotic effects, and combination therapy had additive effects. Metronomic delivery of PLGA-PRINT-docetaxel combined with CH-mEZH2 siRNA has significant antitumor activity in preclinical models of ovarian cancer. Mol Cancer Ther; 13(7); 1750–7. ©2014 AACR.
- Published
- 2014
- Full Text
- View/download PDF
39. Differential Effects of EGFL6 on Tumor versus Wound Angiogenesis
- Author
-
Yasmin A. Lyons, Sunila Pradeep, Sherry Y. Wu, Archana S. Nagaraja, Wei Hu, Robert L. Coleman, Dahai Jiang, Kyunghee Noh, Kshipra M. Gharpure, Mian M.K. Shahzad, Ningyan Zhang, Celia S.L. Mak, Rajesha Rupaimoole, Wei Xiong, Edna M. Mora, Lingegowda S. Mangala, Yunfei Wen, Zhiqiang An, Nicholas B. Jennings, Min Soon Cho, Monika Haemmerle, Xiuhui Chen, Shaolin Ma, Hee Dong Han, Richard R. Bouchard, Michael J. Birrer, Jinsong Liu, Gabriel Lopez-Berestein, Hui Deng, Anil K. Sood, and Charles V. Kingsley
- Subjects
0301 basic medicine ,Tumor angiogenesis ,Integrins ,Angiogenesis ,Blotting, Western ,Cancer therapy ,In Vitro Techniques ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Cell Movement ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,Phosphorylation ,lcsh:QH301-705.5 ,Glycoproteins ,Mice, Knockout ,Chitosan ,Wound Healing ,Neovascularization, Pathologic ,business.industry ,Normal wound healing ,Calcium-Binding Proteins ,Twist-Related Protein 1 ,Differential effects ,Receptor, TIE-2 ,3. Good health ,Neoplasm Proteins ,030104 developmental biology ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,Genetically Engineered Mouse ,Cancer research ,Nanoparticles ,Female ,business ,Wound healing ,Peptides ,Cell Adhesion Molecules ,Proto-Oncogene Proteins c-akt - Abstract
Summary: Angiogenesis inhibitors are important for cancer therapy, but clinically approved anti-angiogenic agents have shown only modest efficacy and can compromise wound healing. This necessitates the development of novel anti-angiogenesis therapies. Here, we show significantly increased EGFL6 expression in tumor versus wound or normal endothelial cells. Using a series of in vitro and in vivo studies with orthotopic and genetically engineered mouse models, we demonstrate the mechanisms by which EGFL6 stimulates tumor angiogenesis. In contrast to its antagonistic effects on tumor angiogenesis, EGFL6 blockage did not affect normal wound healing. These findings have significant implications for development of anti-angiogenesis therapies. : Noh et al. identify EGFL6 as an angiogenic target that is selectively present in tumor endothelial cells in a hypoxic tumor microenvironment. EGFL6 blockade exerts robust anti-angiogenic and anti-tumor effects without affecting wound healing. These findings suggest an important approach for effectively targeting tumor angiogenesis. Keywords: tumor endothelial cells, ovarian cancer, chitosan nanoparticles, tumor vasculature, wound healing
- Published
- 2016
40. A miR-192-EGR1-HOXB9 regulatory network controls the angiogenic switch in cancer
- Author
-
Cristian Rodriguez-Aguayo, Gabriel Lopez-Berestein, Fangrong Shen, Shelley M. Herbrich, Kshipra M. Gharpure, Justyna Filant, Prahlad T. Ram, Viviana Vidal-Anaya, Elena G. Seviour, Guillermo N. Armaiz-Pena, Ehsan A. Ehsanipour, Anil K. Sood, Monika Haemmerle, Sourindra Maiti, Rajesha Rupaimoole, Min Zhang, Chad V. Pecot, Sunila Pradeep, Laurence J.N. Cooper, Ji Hoon Kim, Courtney Olsen, Hiroto Hatakeyama, Susan L. Tucker, Archana S. Nagaraja, Hee Dong Han, Elizabeth Pham, Da Yang, Sherry Y. Wu, Rouba Ali-Fehmi, Keith A. Baggerly, Xinna Zhang, Cristina Ivan, Menashe Bar-Eli, Mien Chie Hung, Li Huang, Ju Seog Lee, and Michael McGuire
- Subjects
0301 basic medicine ,Angiogenic Switch ,Angiogenesis ,Science ,Regulator ,Down-Regulation ,General Physics and Astronomy ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Neovascularization ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,microRNA ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,Early Growth Response Protein 1 ,Homeodomain Proteins ,Ovarian Neoplasms ,Multidisciplinary ,Neovascularization, Pathologic ,Cancer ,Genetic Therapy ,General Chemistry ,medicine.disease ,Kidney Neoplasms ,Tumor Burden ,3. Good health ,MicroRNAs ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Immunology ,Phosphatidylcholines ,Cancer research ,Female ,Growth inhibition ,medicine.symptom - Abstract
A deeper mechanistic understanding of tumour angiogenesis regulation is needed to improve current anti-angiogenic therapies. Here we present evidence from systems-based miRNA analyses of large-scale patient data sets along with in vitro and in vivo experiments that miR-192 is a key regulator of angiogenesis. The potent anti-angiogenic effect of miR-192 stems from its ability to globally downregulate angiogenic pathways in cancer cells through regulation of EGR1 and HOXB9. Low miR-192 expression in human tumours is predictive of poor clinical outcome in several cancer types. Using 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) nanoliposomes, we show that miR-192 delivery leads to inhibition of tumour angiogenesis in multiple ovarian and renal tumour models, resulting in tumour regression and growth inhibition. This anti-angiogenic and anti-tumour effect is more robust than that observed with an anti-VEGF antibody. Collectively, these data identify miR-192 as a central node in tumour angiogenesis and support the use of miR-192 in an anti-angiogenesis therapy., The formation of blood vessels in tumours, angiogenesis, is a promising target for therapy. Here, the authors show that microRNA192 has anti-angiogenic functions and negatively regulates EGR1 and HOXB9, and that delivery of this microRNA to tumours in vivo can reduce angiogenesis and tumour growth.
- Published
- 2016
- Full Text
- View/download PDF
41. Adrenergic stimulation of DUSP1 impairs chemotherapy response in ovarian cancer
- Author
-
Archana S. Nagaraja, Piotr L. Dorniak, Anil K. Sood, Kshipra M. Gharpure, Guillermo N. Armaiz-Pena, Cristina Ivan, Tao Liu, Gabriel Lopez-Berestein, Susan K. Lutgendorf, Jean M. Hansen, Cristian Rodriguez-Aguayo, Vasudha Sehgal, Prahlad T. Ram, Rebecca A. Previs, Steve W. Cole, Yu Kang, Wei Hu, and Rajesha Rupaimoole
- Subjects
0301 basic medicine ,Cancer Research ,Transcription, Genetic ,Proto-Oncogene Proteins c-jun ,medicine.medical_treatment ,Adrenergic ,Antineoplastic Agents ,Apoptosis ,Pharmacology ,Models, Biological ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Adrenergic Agents ,Catecholamines ,In vivo ,Stress, Physiological ,Cell Line, Tumor ,medicine ,Gene silencing ,Animals ,Humans ,Phosphorylation ,Promoter Regions, Genetic ,Regulation of gene expression ,Ovarian Neoplasms ,Chemotherapy ,business.industry ,JNK Mitogen-Activated Protein Kinases ,Dual Specificity Phosphatase 1 ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Paclitaxel ,chemistry ,030220 oncology & carcinogenesis ,Female ,Receptors, Adrenergic, beta-2 ,Ovarian cancer ,business ,Signal Transduction - Abstract
Purpose: Chronic adrenergic activation has been shown to associate with adverse clinical outcomes in cancer patients, but the underlying mechanisms are not well understood. The focus of the current study was to determine the functional and biologic effects of adrenergic pathways on response to chemotherapy in the context of ovarian cancer. Experimental Design: Increased DUSP1 production by sympathetic nervous system mediators (e.g., norepinephrine) was analyzed by real-time quantitative RT-PCR and by Western blotting. In vitro chemotherapy-induced cell apoptosis was examined by flow cytometry. For in vivo therapy, a well-characterized model of chronic stress was used. Results: Catecholamines significantly inhibited paclitaxel- and cisplatin-induced apoptosis in ovarian cancer cells. Genomic analyses of cells treated with norepinephrine identified DUSP1 as a potential mediator. DUSP1 overexpression resulted in reduced paclitaxel-induced apoptosis in ovarian cancer cells compared with control; conversely, DUSP1 gene silencing resulted in increased apoptosis compared with control cells. DUSP1 gene silencing in vivo significantly enhanced response to paclitaxel and increased apoptosis. In vitro analyses indicated that norepinephrine-induced DUSP1 gene expression was mediated through ADRB2 activation of cAMP–PLC–PKC–CREB signaling, which inhibits JNK-mediated phosphorylation of c-Jun and protects ovarian cancer cells from apoptosis. Moreover, analysis of The Cancer Genome Atlas data showed that increased DUSP1 expression was associated with decreased overall (P = 0.049) and progression-free (P = 0.0005) survival. Conclusions: These findings provide a new understanding of the mechanisms by which adrenergic pathways can impair response to chemotherapy and have implications for cancer management. Clin Cancer Res; 22(7); 1713–24. ©2015 AACR.
- Published
- 2015
42. FAK regulates platelet extravasation and tumor growth after antiangiogenic therapy withdrawal
- Author
-
Rajesha Rupaimoole, Rebecca L. Stone, Anil K. Sood, Monika Haemmerle, Chad V. Pecot, Hyun Jin Choi, Jean M. Hansen, Vahid Afshar-Kharghan, Lingegowda S. Mangala, Heather J. Dalton, Kshipra M. Gharpure, Sunila Pradeep, Sherry Y. Wu, Hee Dong Han, Alan R. Burns, Morgan Taylor, Behrouz Zand, Justin Bottsford-Miller, Min Soon Cho, Gabriel Lopez-Berestein, Archana S. Nagaraja, Guillermo N. Armaiz-Pena, Tony Gutschner, and Alpa M. Nick
- Subjects
0301 basic medicine ,Blood Platelets ,medicine.medical_specialty ,Indazoles ,Bevacizumab ,Antibodies, Neoplasm ,Neovascularization ,Pazopanib ,03 medical and health sciences ,Antiangiogenesis Therapy ,Mice ,0302 clinical medicine ,Internal medicine ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Tumor Microenvironment ,Animals ,Humans ,Mice, Knockout ,Ovarian Neoplasms ,Tumor microenvironment ,Sulfonamides ,Tumor hypoxia ,Neovascularization, Pathologic ,business.industry ,General Medicine ,medicine.disease ,Xenograft Model Antitumor Assays ,Extravasation ,Cell Hypoxia ,Neoplasm Proteins ,Adenosine Diphosphate ,030104 developmental biology ,Endocrinology ,Pyrimidines ,030220 oncology & carcinogenesis ,Focal Adhesion Kinase 1 ,Cancer research ,Female ,medicine.symptom ,Ovarian cancer ,business ,medicine.drug ,Research Article - Abstract
Recent studies in patients with ovarian cancer suggest that tumor growth may be accelerated following cessation of antiangiogenesis therapy; however, the underlying mechanisms are not well understood. In this study, we aimed to compare the effects of therapy withdrawal to those of continuous treatment with various antiangiogenic agents. Cessation of therapy with pazopanib, bevacizumab, and the human and murine anti-VEGF antibody B20 was associated with substantial tumor growth in mouse models of ovarian cancer. Increased tumor growth was accompanied by tumor hypoxia, increased tumor angiogenesis, and vascular leakage. Moreover, we found hypoxia-induced ADP production and platelet infiltration into tumors after withdrawal of antiangiogenic therapy, and lowering platelet counts markedly inhibited tumor rebound after withdrawal of antiangiogenic therapy. Focal adhesion kinase (FAK) in platelets regulated their migration into the tumor microenvironment, and FAK-deficient platelets completely prevented the rebound tumor growth. Additionally, combined therapy with a FAK inhibitor and the antiangiogenic agents pazopanib and bevacizumab reduced tumor growth and inhibited negative effects following withdrawal of antiangiogenic therapy. In summary, these results suggest that FAK may be a unique target in situations in which antiangiogenic agents are withdrawn, and dual targeting of FAK and VEGF could have therapeutic implications for ovarian cancer management.
- Published
- 2015
43. Sustained adrenergic signaling leads to increased metastasis in ovarian cancer via increased PGE2 synthesis
- Author
-
Susan K. Lutgendorf, Sherry Y. Wu, Yu Kang, Steve W. Cole, Nouara C. Sadaoui, Kshipra M. Gharpure, Cristian Rodriguez-Aguayo, Tan Lin, Behrouz Zand, Rebecca A. Previs, Jean M. Hansen, Piotr L. Dorniak, Guillermo N. Armaiz-Pena, Peiying Yang, Anil K. Sood, Gabriel Lopez-Berestein, Archana S. Nagaraja, Julie K. Allen, Cristina Ivan, Sunila Pradeep, and Rajesha Rupaimoole
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Inflammation ,Biology ,Dinoprostone ,Article ,Metastasis ,Norepinephrine (medication) ,03 medical and health sciences ,Mice ,Norepinephrine ,0302 clinical medicine ,Internal medicine ,Cell Line, Tumor ,Genetics ,medicine ,Gene silencing ,Animals ,Humans ,Gene Silencing ,Prostaglandin E2 ,Neoplasm Metastasis ,Molecular Biology ,Prostaglandin-E Synthases ,Ovarian Neoplasms ,medicine.disease ,3. Good health ,030104 developmental biology ,Endocrinology ,Cell culture ,Cyclooxygenase 2 ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,Signal transduction ,Ovarian cancer ,medicine.drug ,Signal Transduction - Abstract
Adrenergic stimulation adversely affects tumor growth and metastasis, but the underlying mechanisms are not well understood. Here, we uncovered a novel mechanism by which catecholamines induce inflammation by increasing prostaglandin E2 (PGE2) levels in ovarian cancer cells. Metabolic changes in tumors isolated from patients with depression and mice subjected to restraint stress showed elevated PGE2 levels. Increased metabolites and PTGS2 and PTGES protein levels were found in Skov3-ip1 and HeyA8 cells treated with norepinephrine, and these changes were shown to be mediated by ADRB2 receptor signaling. Silencing PTGS2 resulted in significantly decreased migration and invasion in ovarian cancer cells in the presence of norepinephrine and decreased tumor burden and metastasis in restraint stress orthotopic models. In human ovarian cancer samples, concurrent increased ADRB2, PTGS2 and PTGES expression was associated with reduced overall and progression-free patient survival. In conclusion, increased adrenergic stimulation results in increased PGE2 synthesis via ADRB2-Nf-kB-PTGS2 axis, which drives tumor growth and metastasis.
- Published
- 2015
44. XPO1/CRM1 Inhibition Causes Antitumor Effects by Mitochondrial Accumulation of eIF5A
- Author
-
David H. Hawke, John E. Wiktorowicz, Sharon Shacham, Archana S. Nagaraja, Morgan Taylor, Rebecca A. Previs, Kshipra M. Gharpure, Sherry Y. Wu, Dilara McCauley, Robert L. Coleman, Heather J. Dalton, Rajesha Rupaimoole, Behrouz Zand, Tao Liu, Yu Kang, Takeshi Hisamatsu, Anil K. Sood, Takahito Miyake, Sunila Pradeep, Yunfei Wen, Min Soon Cho, and Wei Hu
- Subjects
Proteomics ,Cancer Research ,Programmed cell death ,Active Transport, Cell Nucleus ,Fluorescent Antibody Technique ,Mice, Nude ,Receptors, Cytoplasmic and Nuclear ,Antineoplastic Agents ,Apoptosis ,Mitochondrion ,Biology ,Karyopherins ,Transfection ,Article ,Mice ,Peptide Initiation Factors ,Tandem Mass Spectrometry ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Electrophoresis, Gel, Two-Dimensional ,Enzyme Inhibitors ,RNA, Small Interfering ,Ovarian Neoplasms ,Cancer ,Mammary Neoplasms, Experimental ,RNA-Binding Proteins ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Mitochondria ,Oncology ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Cancer cell ,Cancer research ,Topotecan ,Female ,Signal transduction ,Ovarian cancer ,medicine.drug ,Chromatography, Liquid ,Signal Transduction - Abstract
Purpose: XPO1 inhibitors have shown promise for cancer treatment, and yet the underlying mechanisms for the antitumor effects are not well understood. In this study, we explored the usefulness of selective inhibitors of nuclear export (SINE) compounds that are specific inhibitors of XPO1. Experimental Design: We used proteomic analysis in XPO1 inhibitor–treated ovarian cancer cell lines and examined antitumor effects in ovarian and breast cancer mouse models. We also studied the effects of XPO1 inhibitor in combination with chemotherapeutic agents. Results: XPO1 inhibitor treatment substantially increased the percentage of apoptotic cells (60%) after 72 hours of incubation. XPO1 inhibitor promoted the accumulation of eIF5A in mitochondria, leading to cancer cell death. Topotecan showed the greatest synergistic effect with XPO1 inhibitor. XPO1 inhibitors prevented the translocation of IGF2BP1 from the nucleus to the cytoplasm, thereby permitting the localization of eIF5A in the mitochondria. This process was p53, RB, and FOXO independent. Significant antitumor effects were observed with XPO1 inhibitor monotherapy in orthotopic ovarian (P < 0.001) and breast (P < 0.001) cancer mouse models, with a further decrease in tumor burden observed in combination with topotecan or paclitaxel (P < 0.05). This mitochondrial accumulation of eIF5A was highly dependent on the cytoplasmic IGF2BP1 levels. Conclusions: We have unveiled a new understanding of the role of eIF5A and IGF2BP1 in XPO1 inhibitor–mediated cell death and support their clinical development for the treatment of ovarian and other cancers. Our data also ascertain the combinations of XPO1 inhibitors with specific chemotherapy drugs for therapeutic trials. Clin Cancer Res; 21(14); 3286–97. ©2015 AACR.
- Published
- 2015
45. Abstract 473: PRKRA/PACT expression promotes chemoresistance in mucinous ovarian cancer
- Author
-
Takeshi Hisamatsu, Jean M. Hansen, Gabriel Lopez-Berestein, Kyunghee Noh, Rajesha Rupaimoole, Justyna Filant, Cristina Ivan, Cristian Rodriguez-Aguayo, Geoffrey Bartholomeusz, Kwong Kwok Wong, Koji Matsuo, Kshipra M. Gharpure, Lingegowda S. Mangala, Archana S. Nagaraja, Anil K. Sood, Yasmin A. Lyons, Michael McGuire, Takashi Mitamura, Sunila Pradeep, Ju-Seong Lee, Michael Frumovitz, and Sherry Y. Wu
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Cancer ,Synthetic lethality ,medicine.disease ,Pact ,Oxaliplatin ,Internal medicine ,Cancer research ,Medicine ,business ,Ovarian cancer ,Chemotherapy resistance ,medicine.drug - Abstract
Purpose: To investigate the mechanisms of chemotherapy resistance and developing strategies to enhance therapeutic responses in mucinous ovarian cancer (MOC). Experimental design: We carried out a kinome-based siRNA screen using human MOC to identify novel targets to enhance the efficacy of chemotherapy in MOC cell lines. In vitro and in vivo validation studies were carried out using MOC models. We specifically interrogated the role of PRKRA in MOC based on our screen results. Results: Among the 939 genes in the screen, we focused on PRKRA/PACT because it was one of the top 5 target genes that exhibited the greatest extent of synthetic lethality in the target gene-siRNA plus oxaliplatin group relative to the target gene-siRNA group. The combination of oxaliplatin plus siPRKRA treatment resulted in significantly reduced cell viability compared with oxaliplatin plus control siRNA in RMUG-L-ip1 or RMUG-S-ip1 MOC cells (p Conclusion: The PRKRA/PACT axis represents an important therapeutic opportunity in MOC for enhancing oxaliplatin efficacy. Citation Format: Takeshi Hisamatsu, Michael McGuire, Sherry Y. Wu, Rajesha Rupaimoole, Sunila Pradeep, Kyunghee Noh, Justyna Filant, Jean M. Hansen, Yasmin Lyons, Kshipra M. Gharpure, Archana S. Nagaraja, Lingegowda S. Mangala, Takashi Mitamura, Cristian Rodriguez-Aguayo, Geoffrey A. Bartholomeusz, Cristina Ivan, Ju-Seong Lee, Koji Matsuo, Michael Frumovitz, Kwong K. Wong, Gabriel Lopez-Berestein, Anil K. Sood. PRKRA/PACT expression promotes chemoresistance in mucinous ovarian cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 473. doi:10.1158/1538-7445.AM2017-473
- Published
- 2017
- Full Text
- View/download PDF
46. Abstract 5377: Intragenic DNA methylation and increased gene expression: New directions from pan-cancer analysis
- Author
-
Keith A. Baggerly, Sherry Y. Wu, Rajesha Rupaimoole, Hui Yao, Michael McGuire, Ying Wang, Anil K. Sood, Gabriel Lopez-Berestein, Shelley M. Herbrich, and Kshipra M. Gharpure
- Subjects
Genetics ,Cancer Research ,Promoter ,Methylation ,Biology ,medicine.disease_cause ,Molecular biology ,Oncology ,DNA methylation ,medicine ,Illumina Methylation Assay ,Epigenetics ,Carcinogenesis ,Gene ,RNA-Directed DNA Methylation - Abstract
Purpose: While the impact of DNA methylation at the promoter region as a repressive influence is well-known, the vast majority of DNA methylation actually occurs outside of the promoter region. The function of intragenic DNA methylation remains incompletely understood. Here, we investigate how intragenic DNA methylation impacts gene expression, and how these modifications differ between normal and corresponding cancer tissues. Methods: A pan-cancer analysis of gene expression array data was integrated with Illumina 450k methylation array data across ten tumor types to produce correlation values between DNA methylation and gene expression at distinct loci on a genome-wide basis. The probes exhibiting a significant correlation between gene expression and DNA methylation were selected for further analysis. Results: We uncovered robust positive correlations between gene expression and 3’ methylation in 3,200 genes, with 590 genes in bladder cancer, and 256 genes in squamous cell lung carcinoma displaying a correlation coefficient of >0.5. Furthermore, we observed 44 genes with a correlation coefficient of >0.7 in at least two tissue types. The majority of genes with the strongest correlations are transcription factors known to play roles in differentiation and development, with an enrichment of zinc finger and homeobox-containing genes. Further analysis of these genes exhibit divergent 3’ methylation when comparing normal and tumor tissues. Importantly, the extent of 3’ methylation of these genes is associated with patient overall survival in 5 of the 10 tumor types analyzed, strongly suggesting that this process plays a role in cancer pathogenesis. Conclusion: DNA methylation of the 3’ region is a functionally and clinically relevant epigenetic modification, and may serve as a novel target for inhibiting tumorigenesis and tumor progression. Citation Format: Michael H. McGuire, Shelley Herbrich, Sherry Wu, Ying Wang, Rajesha Rupaimoole, Hui Yao, Kshipra Gharpure, Gabriel Lopez-Berestein, Keith Baggerly, Anil Sood. Intragenic DNA methylation and increased gene expression: New directions from pan-cancer analysis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5377. doi:10.1158/1538-7445.AM2017-5377
- Published
- 2017
- Full Text
- View/download PDF
47. Therapeutic Silencing of KRAS using Systemically Delivered siRNAs
- Author
-
Salma H. Azam, Scott Kopetz, Sherry Y. Wu, Heather J. Dalton, Archana S. Nagaraja, Rajat Bhattacharya, Rajesha Rupaimoole, Anil K. Sood, Anshumaan Maharaj, Chad V. Pecot, Trent A. Waugh, Cristian Rodriguez-Aguayo, Kshipra M. Gharpure, Vianey Gonzalez-Villasana, Takeshi Hisamatsu, Lee M. Ellis, Gabriel Lopez-Berestein, Justyna Filant, Seth Bellister, Maria Pia Morelli, and Behrouz Zand
- Subjects
Cancer Research ,Small interfering RNA ,endocrine system diseases ,Colorectal cancer ,Gene Expression ,Biology ,medicine.disease_cause ,Article ,Metastasis ,Mice ,Cell Line, Tumor ,Neoplasms ,medicine ,Gene silencing ,Animals ,Humans ,Gene Silencing ,RNA, Small Interfering ,Lung cancer ,neoplasms ,Gene Transfer Techniques ,Cancer ,medicine.disease ,Molecular biology ,Primary tumor ,Xenograft Model Antitumor Assays ,digestive system diseases ,respiratory tract diseases ,Disease Models, Animal ,Oncology ,Liposomes ,Cancer research ,ras Proteins ,Nanoparticles ,Female ,RNA Interference ,KRAS - Abstract
Despite being among the most common oncogenes in human cancer, to date, there are no effective clinical options for inhibiting KRAS activity. We investigated whether systemically delivered KRAS siRNAs have therapeutic potential in KRAS-mutated cancer models. We identified KRAS siRNA sequences with notable potency in knocking down KRAS expression. Using lung and colon adenocarcinoma cell lines, we assessed antiproliferative effects of KRAS silencing in vitro. For in vivo experiments, we used a nanoliposomal delivery platform, DOPC, for systemic delivery of siRNAs. Various lung and colon cancer models were used to determine efficacy of systemic KRAS siRNA based on tumor growth, development of metastasis, and downstream signaling. KRAS siRNA sequences induced >90% knockdown of KRAS expression, significantly reducing viability in mutant cell lines. In the lung cancer model, KRAS siRNA treatment demonstrated significant reductions in primary tumor growth and distant metastatic disease, while the addition of CDDP was not additive. Significant reductions in Ki-67 indices were seen in all treatment groups, whereas significant increases in caspase-3 activity were only seen in the CDDP treatment groups. In the colon cancer model, KRAS siRNA reduced tumor KRAS and pERK expression. KRAS siRNAs significantly reduced HCP1 subcutaneous tumor growth, as well as outgrowth of liver metastases. Our studies demonstrate a proof-of-concept approach to therapeutic KRAS targeting using nanoparticle delivery of siRNA. This study highlights the potential translational impact of therapeutic RNA interference, which may have broad applications in oncology, especially for traditional “undruggable” targets. Mol Cancer Ther; 13(12); 2876–85. ©2014 AACR.
- Published
- 2014
48. Clodronate inhibits tumor angiogenesis in mouse models of ovarian cancer
- Author
-
Sunila Pradeep, Archana S. Nagaraja, Hernan G. Vasquez, Jie Huang, Yunfei Wen, Nicole M Reusser, Anil K. Sood, Takahito Miyake, Kshipra M. Gharpure, Vianey Gonzalez-Villasana, Gabriel Lopez-Berestein, Rajesha Rupaimoole, Wei Hu, Nicholas B. Jennings, and Heather J. Dalton
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Angiogenesis ,Angiogenesis Inhibitors ,Biology ,Metastasis ,In vivo ,Cell Movement ,Cell Line, Tumor ,medicine ,Macrophage ,Animals ,Humans ,Pharmacology ,Ovarian Neoplasms ,Tumor microenvironment ,Bone Density Conservation Agents ,Macrophages ,Cancer ,Endothelial Cells ,medicine.disease ,Mice, Inbred C57BL ,Oncology ,Cancer research ,Molecular Medicine ,Clodronic acid ,Cytokines ,Female ,Clodronic Acid ,Ovarian cancer ,medicine.drug ,Research Paper - Abstract
Purpose Bisphosphonates have been shown to inhibit and deplete macrophages. The effects of bisphosphonates on other cell types in the tumor microenvironment have been insufficiently studied. Here, we sought to determine the effects of bisphosphonates on ovarian cancer angiogenesis and growth via their effect on the microenvironment, including macrophage, endothelial and tumor cell populations. Experimental Design Using in vitro and in vivo models, we examined the effects of clodronate on angiogenesis and macrophage density, and the overall effect of clodronate on tumor size and metastasis. Results Clodronate inhibited the secretion of pro-angiogenic cytokines by endothelial cells and macrophages, and decreased endothelial migration and capillary tube formation. In treated mice, clodronate significantly decreased tumor size, number of tumor nodules, number of tumor-associated macrophages and tumor capillary density. Conclusions Clodronate is a potent inhibitor of tumor angiogenesis. These results highlight clodronate as a potential therapeutic for cancer.
- Published
- 2014
49. Molecular biomarkers of residual disease after surgical debulking of high-grade serous ovarian cancer
- Author
-
Cristina Ivan, Bulent Ozpolat, Keith A. Baggerly, Sherry Y. Wu, Jamie Guenthoer, Erin K. Crane, Gabriel Lopez-Berestein, Wei Hu, Shelley M. Herbrich, Heather J. Dalton, Alpa M. Nick, Rajesha Rupaimoole, Robert L. Coleman, Susan L. Tucker, Kshipra M. Gharpure, Anil K. Sood, and Anna K. Unruh
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Neoplasm, Residual ,Disease ,Bioinformatics ,Residual ,Fatty Acid-Binding Proteins ,Real-Time Polymerase Chain Reaction ,Article ,Cohort Studies ,symbols.namesake ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,RNA, Messenger ,Fisher's exact test ,Ovarian Neoplasms ,business.industry ,Reverse Transcriptase Polymerase Chain Reaction ,Alcohol Dehydrogenase ,Cancer ,Cytoreduction Surgical Procedures ,Debulking ,medicine.disease ,Prognosis ,Cystadenocarcinoma, Serous ,Survival Rate ,Serous fluid ,Quartile ,Cohort ,symbols ,Female ,Neoplasm Grading ,business ,Follow-Up Studies - Abstract
Purpose: Residual disease following primary cytoreduction is associated with adverse overall survival in patients with epithelial ovarian cancer. Accurate identification of patients at high risk of residual disease has been elusive, lacking external validity and prompting many to undergo unnecessary surgical exploration. Our goal was to identify and validate molecular markers associated with high rates of residual disease. Methods: We interrogated two publicly available datasets from chemonaïve primary high-grade serous ovarian tumors for genes overexpressed in patients with residual disease and significant at a 10% false discovery rate (FDR) in both datasets. We selected genes with wide dynamic range for validation in an independent cohort using quantitative RT-PCR to assay gene expression, followed by blinded prediction of a patient subset at high risk for residual disease. Predictive success was evaluated using a one-sided Fisher exact test. Results: Forty-seven probe sets met the 10% FDR criterion in both datasets. These included FABP4 and ADH1B, which tracked tightly, showed dynamic ranges >16-fold and had high expression levels associated with increased incidence of residual disease. In the validation cohort (n = 139), FABP4 and ADH1B were again highly correlated. Using the top quartile of FABP4 PCR values as a prespecified threshold, we found 30 of 35 cases of residual disease in the predicted high-risk group (positive predictive value = 86%) and 54 of 104 among the remaining patients (P = 0.0002; OR, 5.5). Conclusion: High FABP4 and ADH1B expression is associated with significantly higher risk of residual disease in high-grade serous ovarian cancer. Patients with high tumoral levels of these genes may be candidates for neoadjuvant chemotherapy. Clin Cancer Res; 20(12); 3280–8. ©2014 AACR.
- Published
- 2014
50. Hypoxia-mediated downregulation of miRNA biogenesis promotes tumour progression
- Author
-
Guillermo N. Armaiz-Pena, Anil K. Sood, Behrouz Zand, Gabriel Lopez-Berestein, Cristian Rodriguez-Aguayo, Justyna Filant, Morgan Taylor, Sherry Y. Wu, Chad V. Pecot, Sunila Pradeep, Twan van den Beucken, Menashe Bar-Eli, Nouara C. Sadaoui, Bradly G. Wouters, Elizabeth Koch, George A. Calin, Wei Zhang, Kshipra M. Gharpure, Rajesha Rupaimoole, Milan Radovich, Lingegowda S. Mangala, Cristina Ivan, Archana S. Nagaraja, Chunhua Lu, Heather J. Dalton, Justin Bottsford Miller, Li Huang, Michael McGuire, Mircea Ivan, Radiotherapie, RS: GROW - Oncology, and RS: GROW - R1 - Prevention
- Subjects
Ribonuclease III ,Vascular Endothelial Growth Factor A ,Small interfering RNA ,Epithelial-Mesenchymal Transition ,Nude ,Mice, Nude ,General Physics and Astronomy ,Down-Regulation ,Bioinformatics ,Models, Biological ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,DEAD-box RNA Helicases ,Proto-Oncogene Protein c-ets-1 ,Mice ,ELK1 ,Downregulation and upregulation ,Models ,Cell Line, Tumor ,Neoplasms ,microRNA ,Animals ,Humans ,Epithelial–mesenchymal transition ,Author Correction ,Drosha ,Neoplastic ,Multidisciplinary ,Tumor ,biology ,General Chemistry ,Biological ,Cell Hypoxia ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Gene Expression Regulation ,Cancer cell ,biology.protein ,Cancer research ,Disease Progression ,Female ,Dicer - Abstract
Cancer-related deregulation of miRNA biogenesis has been suggested, but the underlying mechanisms remain elusive. Here we report a previously unrecognized effect of hypoxia in the downregulation of Drosha and Dicer in cancer cells that leads to dysregulation of miRNA biogenesis and increased tumour progression. We show that hypoxia-mediated downregulation of Drosha is dependent on ETS1/ELK1 transcription factors. Moreover, mature miRNA array and deep sequencing studies reveal altered miRNA maturation in cells under hypoxic conditions. At a functional level, this phenomenon results in increased cancer progression in vitro and in vivo, and data from patient samples are suggestive of miRNA biogenesis downregulation in hypoxic tumours. Rescue of Drosha by siRNAs targeting ETS1/ELK1 in vivo results in significant tumour regression. These findings provide a new link in the mechanistic understanding of global miRNA downregulation in the tumour microenvironment.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.