Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Murali Ravoori is active.

Publication


Featured researches published by Murali Ravoori.


Nature Medicine | 2006

Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma

Premal H. Thaker; Liz Y. Han; Aparna A. Kamat; Jesusa M.G. Arevalo; Rie Takahashi; Chunhua Lu; Nicholas B. Jennings; Guillermo N. Armaiz-Pena; James A. Bankson; Murali Ravoori; William M. Merritt; Yvonne G. Lin; Lingegowda S. Mangala; Tae Jin Kim; Robert L. Coleman; Charles N. Landen; Yang Li; Edward Felix; Angela Sanguino; Robert A. Newman; Mary Lloyd; David M. Gershenson; Vikas Kundra; Gabriel Lopez-Berestein; Susan K. Lutgendorf; Steven W. Cole; Anil K. Sood

Stress can alter immunological, neurochemical and endocrinological functions, but its role in cancer progression is not well understood. Here, we show that chronic behavioral stress results in higher levels of tissue catecholamines, greater tumor burden and more invasive growth of ovarian carcinoma cells in an orthotopic mouse model. These effects are mediated primarily through activation of the tumor cell cyclic AMP (cAMP)–protein kinase A (PKA) signaling pathway by the β2 adrenergic receptor (encoded by ADRB2). Tumors in stressed animals showed markedly increased vascularization and enhanced expression of VEGF, MMP2 and MMP9, and we found that angiogenic processes mediated the effects of stress on tumor growth in vivo. These data identify β-adrenergic activation of the cAMP–PKA signaling pathway as a major mechanism by which behavioral stress can enhance tumor angiogenesis in vivo and thereby promote malignant cell growth. These data also suggest that blocking ADRB-mediated angiogenesis could have therapeutic implications for the management of ovarian cancer.


Cancer Cell | 2010

Regulation of Tumor Angiogenesis by EZH2

Chunhua Lu; Hee Dong Han; Lingegowda S. Mangala; Rouba Ali-Fehmi; Christopher S. Newton; Laurent Ozbun; Guillermo N. Armaiz-Pena; Wei Hu; Rebecca L. Stone; Adnan R. Munkarah; Murali Ravoori; Mian M.K. Shahzad; Jeong Won Lee; Edna Mora; Robert R. Langley; Amy R. Carroll; Koji Matsuo; Whitney A. Spannuth; Rosemarie Schmandt; Nicholas B. Jennings; Blake W. Goodman; Robert B. Jaffe; Alpa M. Nick; Hye Sun Kim; Eylem Güven; Ya Huey Chen; Long Yuan Li; Ming Chuan Hsu; Robert L. Coleman; George A. Calin

Although VEGF-targeted therapies are showing promise, new angiogenesis targets are needed to make additional gains. Here, we show that increased Zeste homolog 2 (EZH2) expression in either tumor cells or in tumor vasculature is predictive of poor clinical outcome. The increase in endothelial EZH2 is a direct result of VEGF stimulation by a paracrine circuit that promotes angiogenesis by methylating and silencing vasohibin1 (vash1). Ezh2 silencing in the tumor-associated endothelial cells inhibited angiogenesis mediated by reactivation of VASH1, and reduced ovarian cancer growth, which is further enhanced in combination with ezh2 silencing in tumor cells. Collectively, these data support the potential for targeting ezh2 as an important therapeutic approach.


Nature Communications | 2013

Tumour angiogenesis regulation by the miR-200 family

Chad V. Pecot; Rajesha Rupaimoole; Da Yang; Rehan Akbani; Cristina Ivan; Chunhua Lu; Sherry Y. Wu; Hee Dong Han; Maitri Y. Shah; Cristian Rodriguez-Aguayo; Justin Bottsford-Miller; Yuexin Liu; Sang Bae Kim; Anna K. Unruh; Vianey Gonzalez-Villasana; Li Huang; Behrouz Zand; Myrthala Moreno-Smith; Lingegowda S. Mangala; Morgan Taylor; Heather J. Dalton; Vasudha Sehgal; Yunfei Wen; Yu Kang; Keith A. Baggerly; Ju Seog Lee; Prahlad T. Ram; Murali Ravoori; Vikas Kundra; Xinna Zhang

The miR-200 family is well known to inhibit the epithelial-mesenchymal transition, suggesting it may therapeutically inhibit metastatic biology. However, conflicting reports regarding the role of miR-200 in suppressing or promoting metastasis in different cancer types have left unanswered questions. Here we demonstrate a difference in clinical outcome based on miR-200s role in blocking tumour angiogenesis. We demonstrate that miR-200 inhibits angiogenesis through direct and indirect mechanisms by targeting interleukin-8 and CXCL1 secreted by the tumour endothelial and cancer cells. Using several experimental models, we demonstrate the therapeutic potential of miR-200 delivery in ovarian, lung, renal and basal-like breast cancers by inhibiting angiogenesis. Delivery of miR-200 members into the tumour endothelium resulted in marked reductions in metastasis and angiogenesis, and induced vascular normalization. The role of miR-200 in blocking cancer angiogenesis in a cancer-dependent context defines its utility as a potential therapeutic agent.


Molecular Cancer Therapeutics | 2011

Dual Inhibition of Tumor Energy Pathway by 2-Deoxyglucose and Metformin Is Effective against a Broad Spectrum of Preclinical Cancer Models

Jae Ho Cheong; Eun Sung Park; Jiyong Liang; Jennifer B. Dennison; Dimitra Tsavachidou; Catherine Nguyen-Charles; Kwai Wa Cheng; Hassan Hall; Dong Zhang; Yiling Lu; Murali Ravoori; Vikas Kundra; Jaffer A. Ajani; Ju Seog Lee; Waun Ki Hong; Gordon B. Mills

Tumor cell proliferation requires both growth signals and sufficient cellular bioenergetics. The AMP-activated protein kinase (AMPK) pathway seems dominant over the oncogenic signaling pathway suppressing cell proliferation. This study investigated the preclinical efficacy of targeting the tumor bioenergetic pathway using a glycolysis inhibitor 2-deoxyglucose (2DG) and AMPK agonists, AICAR and metformin. We evaluated the in vitro antitumor activity of 2DG, metformin or AICAR alone, and 2DG in combination either with metformin or AICAR. We examined in vivo efficacy using xenograft mouse models. 2DG alone was not sufficient to promote tumor cell death, reflecting the limited efficacy showed in clinical trials. A combined use of 2DG and AICAR also failed to induce cell death. However, 2DG and metformin led to significant cell death associated with decrease in cellular ATP, prolonged activation of AMPK, and sustained autophagy. Gene expression analysis and functional assays revealed that the selective AMPK agonist AICAR augments mitochondrial energy transduction (OXPHOS) whereas metformin compromises OXPHOS. Importantly, forced energy restoration with methyl pyruvate reversed the cell death induced by 2DG and metformin, suggesting a critical role of energetic deprivation in the underlying mechanism of cell death. The combination of 2DG and metformin inhibited tumor growth in mouse xenograft models. Deprivation of tumor bioenergetics by dual inhibition of energy pathways might be an effective novel therapeutic approach for a broad spectrum of human tumors. Mol Cancer Ther; 10(12); 2350–62. ©2011 AACR.


Clinical Cancer Research | 2007

Pharmacodynamic Markers of Perifosine Efficacy

Bryan T. Hennessy; Yiling Lu; Enrique Poradosu; Qianghua Yu; Shuangxing Yu; Hassan Hall; Mark S. Carey; Murali Ravoori; Ana M. Gonzalez-Angulo; Robert Birch; I. Craig Henderson; Vikas Kundra; Gordon B. Mills

Purpose: It is critical to develop methods to quantify the early pharmacodynamic effects of targeted therapeutics in vivo to make drug development more efficient and ensure biologically relevant dosing. Furthermore, an ability to identify patients likely to respond to targeted therapeutics would decrease the size, duration, and cost of clinical trials, resulting in more efficient translation to improved patient outcomes. Recent studies suggest that perifosine inhibits the phosphatidylinositol-3′-kinase (PI3K) pathway by preventing cell membrane recruitment of the AKT pleckstrin homology domain. Experimental Design: A novel functional proteomics technology, reverse phase protein array, was used to establish and quantify pharmacodynamic markers of perifosine efficacy. Results: Perifosine selectively prevents AKT recruitment to the membrane and blocks activation of downstream effectors. Perifosine inhibited breast, ovarian, and prostate cancer models. Growth inhibition was associated with apoptosis. Activation of AKT as a consequence of genomic aberrations predicted perifosine efficacy. In cell lines and xenografts, there was a highly statistically significant correlation between the degree of antitumor efficacy of different perifosine doses and quantified down-regulation of phosphorylation of AKT and of its downstream targets, particularly S6. Conclusions: Because of a strong correlation between proportional modulation of PI3K pathway biomarkers and quantified perifosine efficacy, it is likely that early measurement of such pharmacodynamic biomarkers with reverse phase protein array will optimize selection of responding patients and guide perifosine dosing. Furthermore, PI3K pathway activation status may allow baseline selection of patients most likely to respond to perifosine alone or in combination with other therapies.


Cancer Research | 2007

Antitumor and Antivascular Effects of AVE8062 in Ovarian Carcinoma

Tae Jin Kim; Murali Ravoori; Charles N. Landen; Aparna A. Kamat; Liz Y. Han; Chunhua Lu; Yvonne G. Lin; William M. Merritt; Nicholas B. Jennings; Whitney A. Spannuth; Robert R. Langley; David M. Gershenson; Robert L. Coleman; Vikas Kundra; Anil K. Sood

The purpose of this study was to examine the therapeutic efficacy and underlying mechanisms of action of a vascular-disrupting agent, AVE8062, and to determine its effects on tumor metabolic activity. The in vitro and in vivo effects of AVE8062 alone and in combination with docetaxel were tested in chemotherapy-sensitive and chemotherapy-resistant ovarian cancer models. Tumors were analyzed for necrosis, microvessel density, endothelial cell apoptosis, and proliferation following treatment. The effect of AVE8062 on tumor regression and metabolic activity was examined by magnetic resonance (MR) or by [18F]fluorodeoxyglucose ([18F]FDG) uptake by positron emission tomography (PET) with MR imaging, respectively. AVE8062 monotherapy was effective in inhibiting tumor growth in all models (range 43-51% versus control; P < 0.05). Combination therapy was even more effective in inhibiting tumor growth (range 76-90% compared with controls, P < 0.01). AVE8062 in combination with chemotherapy significantly prolonged survival in HeyA8-injected mice (P < 0.001) compared with other groups. AVE8062-based therapy resulted in rapid development of central tumor necrosis, decreased microvessel density, decreased proliferation, and induction of apoptosis of tumor-associated endothelial cells. MR imaging showed regression of established HeyA8 ovarian tumors and [18F]FDG PET with MR showed rapid decrease in metabolic activity after AVE8062 therapy. Combination of AVE8062 plus docetaxel results in potent inhibition of ovarian cancer growth. These results suggest that AVE8062 may be useful as a clinical therapeutic approach for ovarian cancer patients and that functional [18F]FDG PET imaging may predict clinical response before an anatomic reduction in tumor size.


PLOS ONE | 2009

New Dual Mode Gadolinium Nanoparticle Contrast Agent for Magnetic Resonance Imaging

Ketan B. Ghaghada; Murali Ravoori; Divya Sabapathy; James A. Bankson; Vikas Kundra; Ananth Annapragada

Background Liposomal-based gadolinium (Gd) nanoparticles have elicited significant interest for use as blood pool and molecular magnetic resonance imaging (MRI) contrast agents. Previous generations of liposomal MR agents contained gadolinium-chelates either within the interior of liposomes (core-encapsulated gadolinium liposomes) or presented on the surface of liposomes (surface-conjugated gadolinium liposomes). We hypothesized that a liposomal agent that contained both core-encapsulated gadolinium and surface-conjugated gadolinium, defined herein as dual-mode gadolinium (Dual-Gd) liposomes, would result in a significant improvement in nanoparticle-based T1 relaxivity over the previous generations of liposomal agents. In this study, we have developed and tested, both in vitro and in vivo, such a dual-mode liposomal-based gadolinium contrast agent. Methodology/Principal Findings Three types of liposomal agents were fabricated: core-encapsulated, surface-conjugated and dual-mode gadolinium liposomes. In vitro physico-chemical characterizations of the agents were performed to determine particle size and elemental composition. Gadolinium-based and nanoparticle-based T1 relaxivities of various agents were determined in bovine plasma. Subsequently, the agents were tested in vivo for contrast-enhanced magnetic resonance angiography (CE-MRA) studies. Characterization of the agents demonstrated the highest gadolinium atoms per nanoparticle for Dual-Gd liposomes. In vitro, surface-conjugated gadolinium liposomes demonstrated the highest T1 relaxivity on a gadolinium-basis. However, Dual-Gd liposomes demonstrated the highest T1 relaxivity on a nanoparticle-basis. In vivo, Dual-Gd liposomes resulted in the highest signal-to-noise ratio (SNR) and contrast-to-noise ratio in CE-MRA studies. Conclusions/Significance The dual-mode gadolinium liposomal contrast agent demonstrated higher particle-based T1 relaxivity, both in vitro and in vivo, compared to either the core-encapsulated or the surface-conjugated liposomal agent. The dual-mode gadolinium liposomes could enable reduced particle dose for use in CE-MRA and increased contrast sensitivity for use in molecular imaging.


Cancer Research | 2006

The 3p21.3 Tumor Suppressor NPRL2 Plays an Important Role in Cisplatin-Induced Resistance in Human Non–Small-Cell Lung Cancer Cells

Kentaro Ueda; Hiroyuki Kawashima; Shoichiro Ohtani; Wu Guo Deng; Murali Ravoori; Jim Bankson; Boning Gao; Luc Girard; John D. Minna; Jack A. Roth; Vikas Kundra; Lin Ji

NPRL2 is one of the novel candidate tumor suppressor genes identified in the human chromosome 3p21.3 region. The NPRL2 has shown potent tumor suppression activity in vitro and in vivo and has been suggested to be involved in DNA mismatch repair, cell cycle checkpoint signaling, and regulation of the apoptotic pathway. In this study, we analyzed the endogenous expression of the NPRL2 protein and the cellular response to cisplatin in 40 non-small-cell lung cancer cell lines and found that expression of NPRL2 was significantly and reciprocally correlated to cisplatin sensitivity, with a Spearman correlation coefficient of -0.677 (P < 0.00001). Exogenously introduced expression of NPRL2 by N-[1-(2,3-dioleoyloxyl)propyl]-NNN-trimethylammoniummethyl sulfate:cholesterol nanoparticle-mediated gene transfer significantly resensitized the response to cisplatin, yielding a 40% greater inhibition of tumor cell viability and resulting in a 2- to 3-fold increase in induction of apoptosis by activation of multiple caspases in NPRL2-transfected cells compared with untransfected cells at an equal dose of cisplatin. Furthermore, a systemic treatment with a combination of NPRL2 nanoparticles and cisplatin in a human H322 lung cancer orthotopic mouse model significantly enhanced the therapeutic efficacy of cisplatin and overcame cisplatin-induced resistance (P < 0.005). These findings implicate the potential of NPRL2 as a biomarker for predicting cisplatin response in lung cancer patients and as a molecular therapeutic agent for enhancing response and resensitizing nonresponders to cisplatin treatment.


Science Translational Medicine | 2014

Prostate cancer cell-stromal cell crosstalk via FGFR1 mediates antitumor activity of dovitinib in bone metastases.

Xinhai Wan; Paul G. Corn; Jun Yang; Nallasivam Palanisamy; Michael W. Starbuck; Elsa M. Li Ning Tapia; Amado J. Zurita; Ana Aparicio; Murali Ravoori; Elba Vazquez; Dan R. Robinson; Yi-Mi Wu; Xuhong Cao; Matthew K. Iyer; Wallace McKeehan; Vikas Kundra; Fen Wang; Patricia Troncoso; Arul M. Chinnaiyan; Christopher J. Logothetis; Nora M. Navone

Dovitinib is therapeutically active in a subset of patients with prostate cancer bone metastases, partly due to blockade of FGFR-mediated stromal-epithelial interactions in the bone microenvironment. Effective to the Bone Bone is the most common site of metastatic spread for prostate cancer, and tumors that have spread to the bone are usually very difficult to treat. Dovitinib is a recently developed drug that inhibits the fibroblast growth factor receptor. Now, Wan et al. have demonstrated that dovitinib is effective for some patients with prostate cancer that has spread to bone. The authors also identified an explanation for some of the observed antitumor effects by showing that the drug interferes with interactions between the prostate cancer cells and surrounding stromal cells in the bone microenvironment. Bone is the most common site of prostate cancer (PCa) progression to a therapy-resistant, lethal phenotype. We found that blockade of fibroblast growth factor receptors (FGFRs) with the receptor tyrosine kinase inhibitor dovitinib has clinical activity in a subset of men with castration-resistant PCa and bone metastases. Our integrated analyses suggest that FGF signaling mediates a positive feedback loop between PCa cells and bone cells and that blockade of FGFR1 in osteoblasts partially mediates the antitumor activity of dovitinib by improving bone quality and by blocking PCa cell–bone cell interaction. These findings account for clinical observations such as reductions in lesion size and intensity on bone scans, lymph node size, and tumor-specific symptoms without proportional declines in serum prostate-specific antigen concentration. Our findings suggest that targeting FGFR has therapeutic activity in advanced PCa and provide direction for the development of therapies with FGFR inhibitors.


Bone | 2012

Effect of transforming growth factor beta (TGF-β) receptor I kinase inhibitor on prostate cancer bone growth

Xinhai Wan; Zhi Gang Li; Jonathan M. Yingling; Jun Yang; Michael W. Starbuck; Murali Ravoori; Vikas Kundra; Elba Vazquez; Nora M. Navone

Transforming growth factor beta 1 (TGF-β1) has been implicated in the pathogenesis of prostate cancer (PCa) bone metastasis. In this study, we tested the antitumor efficacy of a selective TGF-β receptor I kinase inhibitor, LY2109761, in preclinical models. The effect of LY2109761 on the growth of MDA PCa 2b and PC-3 human PCa cells and primary mouse osteoblasts (PMOs) was assessed in vitro by measuring radiolabeled thymidine incorporation into DNA. In vivo, the right femurs of male SCID mice were injected with PCa cells. We monitored the tumor burden in control- and LY2109761-treated mice with MRI analysis and the PCa-induced bone response with X-ray and micro-CT analyses. Histologic changes in bone were studied by performing bone histomorphometric evaluations. PCa cells and PMOs expressed TGF-β receptor I. TGF-β1 induced pathway activation (as assessed by induced expression of p-Smad2) and inhibited cell growth in PC-3 cells and PMOs but not in MDA PCa 2b cells. LY2109761 had no effect on PCa cells but induced PMO proliferation in vitro. As expected, LY2109761 reversed the TGF-β1-induced pathway activation and growth inhibition in PC-3 cells and PMOs. In vivo, LY2109761 treatment for 6weeks resulted in increased volume in normal bone and increased osteoblast and osteoclast parameters. In addition, LY2109761 treatment significantly inhibited the growth of MDA PCa 2b and PC-3 in the bone of SCID mice (p<0.05); moreover, it resulted in significantly less bone loss and change in osteoclast-associated parameters in the PC-3 tumor-bearing bones than in the untreated mice. In summary, we report for the first time that targeting TGF-β receptors with LY2109761 can control PCa bone growth while increasing the mass of normal bone. This increased bone mass in nontumorous bone may be a desirable side effect of LY2109761 treatment for men with osteopenia or osteoporosis secondary to androgen-ablation therapy, reinforcing the benefit of effectively controlling PCa growth in bone. Thus, targeting TGF-β receptor I is a valuable intervention in men with advanced PCa.

Collaboration


Dive into the Murali Ravoori's collaboration.

Top Co-Authors

Avatar

Vikas Kundra

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lin Han

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sheela P. Singh

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

James A. Bankson

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Chunhua Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anil K. Sood

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Lin Ji

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nora M. Navone

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jun Yang

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge