Network


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

Hotspot


Dive into the research topics where Manish Shanker is active.

Publication


Featured researches published by Manish Shanker.


PLOS ONE | 2011

EGFR-Targeted Hybrid Plasmonic Magnetic Nanoparticles Synergistically Induce Autophagy and Apoptosis in Non-Small Cell Lung Cancer Cells

Tomohisa Yokoyama; Justina O. Tam; Shinji Kuroda; Ailing W. Scott; Jesse Aaron; Timothy Larson; Manish Shanker; Arlene M. Correa; Seiji Kondo; Jack A. Roth; Konstantin Sokolov; Rajagopal Ramesh

Background The epidermal growth factor receptor (EGFR) is overexpressed in 80% of non-small cell lung cancer (NSCLC) and is associated with poor survival. In recent years, EGFR-targeted inhibitors have been tested in the clinic for NSCLC. Despite the emergence of novel therapeutics and their application in cancer therapy, the overall survival rate of lung cancer patients remains 15%. To develop more effective therapies for lung cancer we have combined the anti-EGFR antibody (Clone 225) as a molecular therapeutic with hybrid plasmonic magnetic nanoparticles (NP) and tested on non-small cell lung cancer (NSCLC) cells. Methodology/Principal Findings Cell viability was determined by trypan-blue assay. Cellular protein expression was determined by Western blotting. C225-NPs were detected by electron microscopy and confocal microscopy, and EGFR expression using immunocytochemistry. C225-NP exhibited a strong and selective antitumor effect on EGFR-expressing NSCLC cells by inhibiting EGFR-mediated signal transduction and induced autophagy and apoptosis in tumor cells. Optical images showed specificity of interactions between C225-NP and EGFR-expressing NSCLC cells. No binding of C225-NP was observed for EGFR-null NSCLC cells. C225-NP exhibited higher efficiency in induction of cell killing in comparison with the same amount of free C225 antibody in tumor cells with different levels of EGFR expression. Furthermore, in contrast to C225-NP, free C225 antibody did not induce autophagy in cells. However, the therapeutic efficacy of C225-NP gradually approached the level of free antibodies as the amount of C225 antibody conjugated per nanoparticle was decreased. Finally, attaching C225 to NP was important for producing the enhanced tumor cell killing as addition of mixture of free C225 and NP did not demonstrate the same degree of cell killing activity. Conclusions/Significance We demonstrated for the first time the molecular mechanism of C225-NP induced cytotoxic effects in lung cancer cells that are not characteristic for free molecular therapeutics thus increasing efficacy of therapy against NSCLC.


Molecular Cancer | 2007

MDA-7/IL-24 suppresses human ovarian carcinoma growth in vitro and in vivo

Began Gopalan; Manish Shanker; Sunil Chada; Rajagopal Ramesh

BackgroundPrevious studies showed that the human melanoma differentiation-associated gene-7 (mda-7), also known as interleukin-24 (IL-24), has potent antitumor activity against human and murine cancer cells. However, the majority of these studies were limited to in vitro testing. In the present study, we investigated the antitumor activity of mda-7/IL-24 against human ovarian cancer cells both in vitro and in vivo.ResultsIn vitro, treatment of ovarian cancer cells with an adenoviral vector carrying the mda-7 gene (Ad-mda7) resulted in inhibition of cell proliferation and induction of cell cycle arrest, leading to apoptosis. We did not observe inhibitory activity in Ad-mda7-treated normal cells. In vivo, treatment of subcutaneous tumor xenografts with Ad-mda7 resulted in significant tumor growth inhibition when compared with that in control groups (p < 0.001). Molecular analysis of ovarian tumor tissue lysates treated with Ad-mda7 showed that MDA-7 protein expression was associated with activation of the caspase cascade.ConclusionOur results show that treatment of ovarian cancer cells with mda-7/IL-24 results in growth suppression both in vitro and in vivo.


Cancer Gene Therapy | 2005

Selective induction of cell cycle arrest and apoptosis in human prostate cancer cells through adenoviral transfer of the melanoma differentiation-associated -7 (mda-7)/interleukin-24 (IL-24) gene.

Yuji Saito; Ryo Miyahara; Began Gopalan; Anya Litvak; Satoshi Inoue; Manish Shanker; Cynthia D. Branch; Abner M. Mhashilkar; Jack A. Roth; Sunil Chada; Rajagopal Ramesh

We have previously reported that overexpression of the melanoma differentiation-associated gene -7 (mda-7) using a replication-defective adenovirus (Ad-mda7), results in tumor-specific growth suppression and induction of apoptosis in wide variety of cancer cells. In the present study, we investigated the antitumor activity of Ad-mda7 and the underlying mechanism in human prostate cancer cells and normal prostate epithelial cells. Overexpression of MDA-7 induced significant (P=.001) suppression of cell growth and apoptosis in prostate cancer cells (DU 145, LNCaP, and PC-3). In normal prostate epithelial cells (PrEC) some degree of growth inhibition but not apoptosis was observed. However, the inhibitory effects in normal cells were less compared to tumor cells. Growth inhibitory effects were mediated by the intracellular and not by extracellular MDA-7 protein. Molecular effectors that are involved in Ad-mda7-mediated tumor killing included activation of the caspase cascade, and the induction of G2 phase cell cycle arrest through the inhibition of Cdc25C pathway. These results demonstrate the mechanisms by which Ad-mda7 exerts its antitumor activity in human prostate cancer cells. The antitumor activity combined with previously reported antiangiogenic and proimmune properties of Ad-mda7 can serve as a potential therapeutic agent for treatment of primary and disseminated prostate cancer.We have previously reported that overexpression of the melanoma differentiation-associated gene -7 (mda-7) using a replication-defective adenovirus (Ad-mda7), results in tumor-specific growth suppression and induction of apoptosis in wide variety of cancer cells. In the present study, we investigated the antitumor activity of Ad-mda7 and the underlying mechanism in human prostate cancer cells and normal prostate epithelial cells. Overexpression of MDA-7 induced significant (P=.001) suppression of cell growth and apoptosis in prostate cancer cells (DU 145, LNCaP, and PC-3). In normal prostate epithelial cells (PrEC) some degree of growth inhibition but not apoptosis was observed. However, the inhibitory effects in normal cells were less compared to tumor cells. Growth inhibitory effects were mediated by the intracellular and not by extracellular MDA-7 protein. Molecular effectors that are involved in Ad-mda7-mediated tumor killing included activation of the caspase cascade, and the induction of G2 phase cell cycle arrest through the inhibition of Cdc25C pathway. These results demonstrate the mechanisms by which Ad-mda7 exerts its antitumor activity in human prostate cancer cells. The antitumor activity combined with previously reported antiangiogenic and proimmune properties of Ad-mda7 can serve as a potential therapeutic agent for treatment of primary and disseminated prostate cancer.


Lung Cancer: Targets and Therapy | 2010

Drug resistance in lung cancer

Manish Shanker; David Willcutts; Jack A. Roth; Rajagopal Ramesh

Resistance to chemotherapy drugs is a major problem in cancer treatment. Scientific advances made in the last two decades have resulted in the identification of genes and molecular signaling mechanisms that contribute to drug resistance. This has resulted in a better understanding of the biology of cancer cells and the way these cells adapt or undergo subtle molecular changes thereby protecting themselves from the cytotoxic effects of the anticancer drugs. Based on the knowledge gained to-date new molecularly targeted drugs are being developed and tested in clinical studies, in an attempt to overcome drug resistance and improve drug efficacy. Despite these attempts the overall 5-year survival of patients diagnosed with cancer, such as lung cancer, remains dismal and is less than 15%. It is evident that additional mechanisms contributing to drug resistance exist which are yet to be discovered. It is hoped that identification of new targets and understanding their contribution to drug resistance will provide opportunities for innovative therapies in overcoming drug resistance. In an attempt to broaden our knowledge and understanding on drug resistance we have, in this review article, summarized the most common mechanisms associated with drug resistance in lung cancer.


PLOS ONE | 2015

IL-24 Inhibits Lung Cancer Cell Migration and Invasion by Disrupting The SDF-1/CXCR4 Signaling Axis

Janani Panneerselvam; Jiankang Jin; Manish Shanker; Jason Lauderdale; Jonathan Bates; Qi Wang; Yan D. Zhao; Stephen J. Archibald; Timothy J. Hubin; Rajagopal Ramesh

Background The stromal cell derived factor (SDF)-1/chemokine receptor (CXCR)-4 signaling pathway plays a key role in lung cancer metastasis and is molecular target for therapy. In the present study we investigated whether interleukin (IL)-24 can inhibit the SDF-1/CXCR4 axis and suppress lung cancer cell migration and invasion in vitro. Further, the efficacy of IL-24 in combination with CXCR4 antagonists was investigated. Methods Human H1299, A549, H460 and HCC827 lung cancer cell lines were used in the present study. The H1299 lung cancer cell line was stably transfected with doxycycline-inducible plasmid expression vector carrying the human IL-24 cDNA and used in the present study to determine the inhibitory effects of IL-24 on SDF-1/CXCR4 axis. H1299 and A549 cell lines were used in transient transfection studies. The inhibitory effects of IL-24 on SDF1/CXCR4 and its downstream targets were analyzed by quantitative RT-PCR, western blot, luciferase reporter assay, flow cytometry and immunocytochemistry. Functional studies included cell migration and invasion assays. Principal Findings Endogenous CXCR4 protein expression levels varied among the four human lung cancer cell lines. Doxycycline-induced IL-24 expression in the H1299-IL24 cell line resulted in reduced CXCR4 mRNA and protein expression. IL-24 post-transcriptionally regulated CXCR4 mRNA expression by decreasing the half-life of CXCR4 mRNA (>40%). Functional studies showed IL-24 inhibited tumor cell migration and invasion concomitant with reduction in CXCR4 and its downstream targets (pAKTS473, pmTORS2448, pPRAS40T246 and HIF-1α). Additionally, IL-24 inhibited tumor cell migration both in the presence and absence of the CXCR4 agonist, SDF-1. Finally, IL-24 when combined with CXCR4 inhibitors (AMD3100, SJA5) or with CXCR4 siRNA demonstrated enhanced inhibitory activity on tumor cell migration. Conclusions IL-24 disrupts the SDF-1/CXCR4 signaling pathway and inhibits lung tumor cell migration and invasion. Additionally, IL-24, when combined with CXCR4 inhibitors exhibited enhanced anti-metastatic activity and is an attractive therapeutic strategy for lung metastasis.


Cancer Gene Therapy | 2008

MDA-7/IL-24, a novel tumor suppressor/cytokine is ubiquitinated and regulated by the ubiquitin-proteasome system, and inhibition of MDA-7/IL-24 degradation enhances the antitumor activity

B Gopalan; Manish Shanker; A Scott; Cynthia D. Branch; Sunil Chada; Rajagopal Ramesh

Steady-state protein levels are determined by the balance between protein synthesis and degradation. Protein half-lives are determined primarily by degradation, and the major degradation pathways involve either lysosomal destruction or an ATP-dependent process involving ubiquitination to target proteins to the proteosome. Studies have shown that multiple tumor-suppressor proteins are ubiquitinated and degraded by the 26S proteasome. In the present study, we investigated whether the tumor suppressor/cytokine melanoma differentiation-associated gene-7/interleukin-24 gene (MDA-7/IL-24) protein is ubiquitinated and its degradation controlled by the proteasome. Treatment of ovarian (2008) and lung (H1299) tumor cells with adenoviral delivery of mda-7 (Ad-mda7) or Ad-mda7 plus the proteosome inhibitor MG132 showed that MDA-7 protein expression was dependent upon proteosome activity. Western blot and immunoprecipitation analyses verified that the MDA-7 protein was ubiquitinated and that ubiquitinated-MDA-7 levels were increased in MG132-treated cells. These results were confirmed using small interfering RNA (siRNA)-mediated knockdown of ubiquitin. Furthermore, ubiquitinated MDA-7 protein was degraded by the 26S proteasome, as MDA-7 accumulation was observed only when cells were treated with MG132 but not with lysosome or protease inhibitors. Inhibition of the catalytic β-5 subunit of the 20S proteasome using siRNA resulted in MDA-7 protein accumulation. Finally, treatment of tumor cells with Ad-mda7 plus the proteasome inhibitor bortezomib resulted in increased tumor cell killing. Our results show that MDA-7/IL-24 is ubiquitinated and degraded by the 26S proteasome. Furthermore, inhibition of MDA-7 degradation results in enhanced tumor killing, identifying a novel anticancer strategy.


Cancer Research | 2015

Abstract 1713: IL-24 inhibits lung cancer cell migration and invasion by disrupting the SDF-1/CXCR4 signaling axis

Janani Panneerselvam; Jiankang Jin; Manish Shanker; Jason Lauderdale; Jonathan Bates; Qi Wang; Daniel Zhao; Stephen J. Archibald; Timothy J. Hubin; Rajagopal Ramesh

Introduction The stromal cell derived factor (SDF)-1/chemokine receptor (CXCR)-4 signaling pathway plays a key role in lung cancer metastasis. Therefore, disrupting the SDF-1/CXCR4 signaling axis will reduce the incidence of lung metastasis. In the present study we investigated whether interleukin (IL)-24 can inhibit the SDF-1/CXCR4 axis and suppress lung cancer cell migration and invasion in vitro. Further, the efficacy of IL-24 in combination with CXCR4 antagonists was investigated. Methods Human H1299 lung tumor cell line was stably transfected with a tetracycline-inducible plasmid vector carrying the IL-24. Upon addition of doxycycline (Dox; 1μg/ml), cells were induced to express IL-24 protein. The expression levels of CXCR4 and its downstream molecular mechanisms in H1299 cells were analyzed. The inhibitory effect of IL-24 on SDF-1/CXCR4 axis is determined by RT-qPCR, western blot, luciferase reporter assay, flow-cytometry and immuno-cytochemistry and the consequence of its inhibition on cell migration, and invasion. Results Endogenous CXCR4 protein expression levels varied among four human lung cancer cell lines with H1299 cells showing the highest expression. Doxycycline-induced IL-24 expression in the H1299-IL-24 cell line resulted in reduced CXCR4 mRNA and protein expression. IL-24 post-transcriptionally regulated CXCR4 mRNA expression by decreasing the half-life of CXCR4 mRNA (>40%). Associated with CXCR4 inhibition was the reduced protein expression of pAKTS473, pmTORS2448, pPRAS40T246 and HIF-1α. IL-24 inhibited tumor cell migration and invasion both in the presence and absence of the CXCR4 agonist, SDF-1. However, the combinatorial effect of either IL-24 combined with CXCR4 inhibitors (AMD3100, SJA5) or with CXCR4 siRNA demonstrated enhanced inhibitory activity on tumor cell migration. Conclusions Our study results demonstrate that IL-24 inhibits lung tumor cell migration and invasion by disrupting the SDF-1/CXCR4 signaling pathway and exhibits enhanced anti-metastatic activity when combined with CXCR4 inhibitors. Citation Format: Janani Panneerselvam, Jiankang Jin, Manish Shanker, Jason Lauderdale, Jonathan Bates, Qi Wang, Daniel Zhao, Stephen Archibald, Timothy Hubin, Rajagopal Ramesh. IL-24 inhibits lung cancer cell migration and invasion by disrupting the SDF-1/CXCR4 signaling axis. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1713. doi:10.1158/1538-7445.AM2015-1713


Cancer Research | 2010

Abstract 3505: EGFR-targeted hybrid plasmonic magnetic nanoparticles induce autophagy and apoptosis through DNA damage in non-small cell lung cancer cells

Shinji Kuroda; Tomohisa Yokoyama; Justina O. Tam; Ailing W. Scott; Manish Shanker; Arlene M. Correa; Jack A. Roth; Konstantin Sokolov; Rajagopal Ramesh

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DC Nanoparticles represent a novel class of agents for cancer treatment. In the present study, we examined the effects of epidermal growth factor receptor (EGFR)-targeted hybrid plasmonic magnetic nanoparticles (NP) on non-small cell lung cancer (NSCLC) cells. The nanoparticles (NP) consisted of a paramagnetic iron core that is surrounded by a gold layer and is functionalized with monoclonal anti-EGFR antibody (C225) or control rabbit IgG antibody (IgG). The nanoparticles were tested against a panel of human lung cancer cells that were mutant (HCC827, H1819), wild-type (H1299) or null (H520) for EGFR and compared to EGFR-positive normal fibroblasts (MRC9, WI38) and normal human bronchial epithelial (NHBE) cells. Treatment of tumor cells with C225-NP resulted in a strong and significant inhibitory effect of cell growth compared to normal cells. C225-NP-mediated inhibitory effect was observed only in EGFR-positive tumor cells (HCC827, H1819, H1299) but not in EGFR-null H520 cells. EGFR-mutant cells (HCC827) were the most sensitive to C225-NP. No significant growth inhibitory effects were observed in IgG-NP-treated cells when compared to untreated cells. Molecular analysis showed C225-NP selectively inhibited phosphorylated p-EGFR protein expression and EGFR-mediated signal transduction resulting in induction of autophagy followed by apoptosis. Autophagy occurred as early as twenty-four hours after treatment with an increase in LC3-II content and autophagic vacuoles observed by Western blotting and electron microscopy respectively. Apoptosis as determined by flow cytometry and Western blotting showed an increase in the number of sub-G1 population cells and cleavage of caspase-3 and PARP in C225-NP-treated cells. Optical images determined by dark-field microscopy showed an increase in the concentration of C225-NP bound to EGFR-expressing NSCLC cells but not EGFR-null NSCLC cells. C225-NP exhibited 2-7 fold higher efficiency in induction of cell killing in comparison with the same amount of free C225 antibody. Free C225 antibody did not induce autophagy in cells. Investigation into how C225-NP but not C225 antibody or NP-alone triggers autophagy and initiates apoptosis showed induction of a DNA damage response as evidenced by the formation of γH2AX foci in C225-NP-treated cells. Minimal to no γH2AX foci were observed in cells that were untreated, treated with C225 antibody or NP-alone or treated with IgG-NP. Our data show that EGFR specific NP selectively induced cytotoxic effects in EGFR-expressing lung cancer cells via a novel cell-death mechanism that is not characteristic of the free antibody thus increasing efficacy of therapy. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3505.


Journal of Photopolymer Science and Technology | 2006

MDA-7/IL-24-based cancer gene therapy: Translation from the laboratory to the clinic

Satoshi Inoue; Manish Shanker; Ryo Miyahara; Began Gopalan; Suraag Patel; Yasuhisa Oida; Cynthia D. Branch; Anupama Munshi; Raymond E. Meyn; Michael Andreeff; Fumihiro Tanaka; Abner M. Mhashilkar; Sunil Chada; Rajagopal Ramesh


Cancer Letters | 2007

Vitamin E succinate in combination with mda-7 results in enhanced human ovarian tumor cell killing through modulation of extrinsic and intrinsic apoptotic pathways

Manish Shanker; Began Gopalan; Suraag Patel; Dora Bocangel; Sunil Chada; Rajagopal Ramesh

Collaboration


Dive into the Manish Shanker's collaboration.

Top Co-Authors

Avatar

Rajagopal Ramesh

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sunil Chada

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jack A. Roth

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Began Gopalan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Cynthia D. Branch

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jiankang Jin

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ailing W. Scott

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Justina O. Tam

University of Texas at Austin

View shared research outputs
Researchain Logo
Decentralizing Knowledge