Network


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

Hotspot


Dive into the research topics where Rajesh Vij is active.

Publication


Featured researches published by Rajesh Vij.


Nature | 2011

COP1 is a tumour suppressor that causes degradation of ETS transcription factors

Alberto C. Vitari; Kevin G. Leong; Kim Newton; Cindy Yee; Karen O’Rourke; Jinfeng Liu; Lilian Phu; Rajesh Vij; Ronald E. Ferrando; Suzana S. Couto; Sankar Mohan; Ajay Pandita; Jo-Anne Hongo; David Arnott; Ingrid E. Wertz; Wei-Qiang Gao; Dorothy French; Vishva M. Dixit

The proto-oncogenes ETV1, ETV4 and ETV5 encode transcription factors in the E26 transformation-specific (ETS) family, which includes the most frequently rearranged and overexpressed genes in prostate cancer. Despite being critical regulators of development, little is known about their post-translational regulation. Here we identify the ubiquitin ligase COP1 (also known as RFWD2) as a tumour suppressor that negatively regulates ETV1, ETV4 and ETV5. ETV1, which is mutated in prostate cancer more often, was degraded after being ubiquitinated by COP1. Truncated ETV1 encoded by prostate cancer translocation TMPRSS2:ETV1 lacks the critical COP1 binding motifs and was 50-fold more stable than wild-type ETV1. Almost all patient translocations render ETV1 insensitive to COP1, implying that this confers a selective advantage to prostate epithelial cells. Indeed, COP1 deficiency in mouse prostate elevated ETV1 and produced increased cell proliferation, hyperplasia, and early prostate intraepithelial neoplasia. Combined loss of COP1 and PTEN enhanced the invasiveness of mouse prostate adenocarcinomas. Finally, rare human prostate cancer samples showed hemizygous loss of the COP1 gene, loss of COP1 protein, and elevated ETV1 protein while lacking a translocation event. These findings identify COP1 as a tumour suppressor whose downregulation promotes prostatic epithelial cell proliferation and tumorigenesis.


Journal of Biological Chemistry | 2006

Pro-urokinase-type plasminogen activator is a substrate for hepsin.

Paul Moran; Wei Li; Bin Fan; Rajesh Vij; Charles Eigenbrot; Daniel Kirchhofer

Hepsin, a type II transmembrane serine protease, is strongly up-regulated in prostate cancer. Hepsin overexpression in a mouse prostate cancer model resulted in tumor progression and metastasis, associated with basement membrane disorganization. We investigated whether hepsin enzymatic activity was linked to the basement membrane defects by examining its ability to initiate the plasminogen/plasmin proteolytic pathway. Because plasminogen is not processed by hepsin, we investigated the upstream activators, urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator. Enzymatic assays with a recombinant soluble form of hepsin demonstrated that hepsin did not cleave pro-tissue-type plasminogen activator but efficiently converted pro-uPA into high molecular weight uPA by cleavage at the Lys158-Ile159 (P1-P1′) peptide bond. uPA generated by hepsin displayed enzymatic activity toward small synthetic and macromolecular substrates indistinguishable from uPA produced by plasmin. The catalytic efficiency of pro-uPA activation by hepsin (kcat/Km 4.8 × 105 m–1 s–1) was similar to that of plasmin, which is considered the most potent pro-uPA activator and was about 6-fold higher than that of matriptase. Conversion of pro-uPA was also demonstrated with cell surface-expressed full-length hepsin. A stable hepsinoverexpressing LnCaP cell line converted pro-uPA into high molecular weight uPA at a rate of 6.6 ± 1.9 nm uPA h–1, which was about 3-fold higher than LnCaP cells expressing lower hepsin levels on their surface. In conclusion, the ability of hepsin to efficiently activate pro-uPA suggests that it may initiate plasmin-mediated proteolytic pathways at the tumor/stroma interface that lead to basement membrane disruption and tumor progression.


Science Translational Medicine | 2015

Targeting LGR5 + cells with an antibody-drug conjugate for the treatment of colon cancer

Melissa R. Junttila; Weiguang Mao; Xi Wang; Bu-Er Wang; Thinh Pham; John A. Flygare; Shang-Fan Yu; Sharon Yee; David M. Goldenberg; Carter Fields; Jeffrey Eastham-Anderson; Mallika Singh; Rajesh Vij; Jo-Anne Hongo; Ron Firestein; Melissa Schutten; Kelly Flagella; Paul Polakis; Andrew G. Polson

An antibody-drug conjugate targeting LGR5 effectively treats intestinal cancer in preclinical models. Stemming the progression of cancer LGR5 is a well-known marker of intestinal cancer stem cells, which makes it an attractive target for anticancer treatments. Unfortunately, it is also found in healthy intestinal stem cells, giving rise to concerns about the potential toxicity of such treatments. Now, Junttila et al. used preclinical models of intestinal cancer to demonstrate that targeting LGR5 with an antibody-drug conjugate is effective for shrinking tumors without damaging the surrounding normal tissues. These observations of preclinical effectiveness as well as safety suggest that targeting LGR5-expressing cells may be a viable therapeutic strategy and a candidate for evaluation in human studies. Cancer stem cells (CSCs) are hypothesized to actively maintain tumors similarly to how their normal counterparts replenish differentiated cell types within tissues, making them an attractive therapeutic target for the treatment of cancer. Because most CSC markers also label normal tissue stem cells, it is unclear how to selectively target them without compromising normal tissue homeostasis. We evaluated a strategy that targets the cell surface leucine-rich repeat–containing G protein–coupled receptor 5 (LGR5), a well-characterized tissue stem cell and CSC marker, with an antibody conjugated to distinct cytotoxic drugs. One antibody-drug conjugate (ADC) demonstrated potent tumor efficacy and safety in vivo. Furthermore, the ADC decreased tumor size and proliferation, translating to improved survival in a genetically engineered model of intestinal tumorigenesis. These data demonstrate that ADCs can be leveraged to exploit differences between normal and cancer stem cells to successfully target gastrointestinal cancers.


PLOS Pathogens | 2014

A Neutralizing Anti-gH/gL Monoclonal Antibody Is Protective in the Guinea Pig Model of Congenital CMV Infection

Marcy R. Auerbach; Donghong Yan; Rajesh Vij; Jo-Anne Hongo; Gerald R. Nakamura; Jean-Michel Vernes; Y. Gloria Meng; Samantha Lein; Pamela Chan; Jed Ross; Richard A. D. Carano; Rong Deng; Nicholas Lewin-Koh; Min Xu; Becket Feierbach

Human cytomegalovirus (HCMV) is the most common cause of congenital virus infection. Congenital HCMV infection occurs in 0.2–1% of all births, and causes birth defects and developmental abnormalities, including sensorineural hearing loss and developmental delay. Several key studies have established the guinea pig as a tractable model for the study of congenital HCMV infection and have shown that polyclonal antibodies can be protective [1]–[3]. In this study, we demonstrate that an anti-guinea pig CMV (GPCMV) glycoprotein H/glycoprotein L neutralizing monoclonal antibody protects against fetal infection and loss in the guinea pig. Furthermore, we have delineated the kinetics of GPCMV congenital infection, from maternal infection (salivary glands, seroconversion, placenta) to fetal infection (fetus and amniotic fluid). Our studies support the hypothesis that a neutralizing monoclonal antibody targeting an envelope GPCMV glycoprotein can protect the fetus from infection and may shed light on the therapeutic intervention of HCMV congenital infection in humans.


mAbs | 2014

An improved and robust DNA immunization method to develop antibodies against extra-cellular loops of multi-transmembrane proteins

Meredith Hazen; Sunil Bhakta; Rajesh Vij; Steven Randle; Dara Y. Kallop; Vicki Chiang; Isidro Hotzel; Bijay S. Jaiswal; Karen E. Ervin; Bing Li; Robby M. Weimer; Paul Polakis; Richard H. Scheller; Jagath R. Junutula; Jo-Anne Hongo

Multi-transmembrane proteins are especially difficult targets for antibody generation largely due to the challenge of producing a protein that maintains its native conformation in the absence of a stabilizing membrane. Here, we describe an immunization strategy that successfully resulted in the identification of monoclonal antibodies that bind specifically to extracellular epitopes of a 12 transmembrane protein, multi-drug resistant protein 4 (MRP4). These monoclonal antibodies were developed following hydrodynamic tail vein immunization with a cytomegalovirus (CMV) promoter-based plasmid expressing MRP4 cDNA and were characterized by flow cytometry. As expected, the use of the immune modulators fetal liver tyrosine kinase 3 ligand (Flt3L) and granulocyte-macrophage colony-stimulating factor positively enhanced the immune response against MRP4. Imaging studies using CMV-based plasmids expressing luciferase showed that the in vivo half-life of the target antigen was less than 48 h using CMV-based plasmids, thus necessitating frequent boosting with DNA to achieve an adequate immune response. We also describe a comparison of plasmids, which contained MRP4 cDNA with either the CMV or CAG promoters, used for immunizations. The observed luciferase activity in this comparison demonstrated that the CAG promoter-containing plasmid pCAGGS induced prolonged constitutive expression of MRP4 and an increased anti-MRP4 specific immune response even when the plasmid was injected less frequently. The method described here is one that can be broadly applicable as a general immunization strategy to develop antibodies against multi-transmembrane proteins, as well as target antigens that are difficult to express or purify in native and functionally active conformation.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Monoclonal antibody targeting the β-barrel assembly machine of Escherichia coli is bactericidal

Kelly M. Storek; Marcy R. Auerbach; Handuo Shi; Natalie K. Garcia; Dawei Sun; Nicholas N. Nickerson; Rajesh Vij; Zhonghua Lin; Nan Chiang; Kellen Schneider; Aaron T. Wecksler; Elizabeth Skippington; Gerald R. Nakamura; Dhaya Seshasayee; James T. Koerber; Jian Payandeh; Peter A. Smith; Steven T. Rutherford

Significance The outer membrane of Gram-negative bacteria presents a formidable barrier to the discovery of new antibiotics needed to combat infections by multidrug-resistant bacteria. Targeting essential proteins or processes directly exposed to the environment could bypass this obstacle. Here, we describe a monoclonal antibody that selectively and potently antagonizes BamA, which folds and inserts integral outer membrane β-barrel proteins, by binding to a surface-exposed BamA epitope and, as a result, inhibits bacterial cell growth. Mechanisms of resistance to the antibody reveal that membrane fluidity affects BamA activity. This antibody validates the potential therapeutic strategy of targeting essential, exposed functions and provides a powerful tool for dissecting the fundamental process of folding integral membrane β-barrel proteins in vivo. The folding and insertion of integral β-barrel membrane proteins into the outer membrane of Gram-negative bacteria is required for viability and bacterial pathogenesis. Unfortunately, the lack of selective and potent modulators to dissect β-barrel folding in vivo has hampered our understanding of this fundamental biological process. Here, we characterize a monoclonal antibody that selectively inhibits an essential component of the Escherichia coli β-barrel assembly machine, BamA. In the absence of complement or other immune factors, the unmodified antibody MAB1 demonstrates bactericidal activity against an E. coli strain with truncated LPS. Direct binding of MAB1 to an extracellular BamA epitope inhibits its β-barrel folding activity, induces periplasmic stress, disrupts outer membrane integrity, and kills bacteria. Notably, resistance to MAB1-mediated killing reveals a link between outer membrane fluidity and protein folding by BamA in vivo, underscoring the utility of this antibody for studying β-barrel membrane protein folding within a living cell. Identification of this BamA antagonist highlights the potential for new mechanisms of antibiotics to inhibit Gram-negative bacterial growth by targeting extracellular epitopes.


Scientific Reports | 2018

A targeted boost-and-sort immunization strategy using Escherichia coli BamA identifies rare growth inhibitory antibodies

Rajesh Vij; Zhonghua Lin; Nan Chiang; Jean-Michel Vernes; Kelly M. Storek; Summer Park; Joyce Chan; Y. Gloria Meng; Laetitia Comps-Agrar; Peng Luan; Sophia Lee; Kellen Schneider; Jack Bevers; Inna Zilberleyb; Christine Tam; Christopher M. Koth; Min Xu; Avinash Gill; Marcy R. Auerbach; Peter A. Smith; Steven T. Rutherford; Gerald R. Nakamura; Dhaya Seshasayee; Jian Payandeh; James T. Koerber

Outer membrane proteins (OMPs) in Gram-negative bacteria are essential for a number of cellular functions including nutrient transport and drug efflux. Escherichia coli BamA is an essential component of the OMP β-barrel assembly machinery and a potential novel antibacterial target that has been proposed to undergo large (~15 Å) conformational changes. Here, we explored methods to isolate anti-BamA monoclonal antibodies (mAbs) that might alter the function of this OMP and ultimately lead to bacterial growth inhibition. We first optimized traditional immunization approaches but failed to identify mAbs that altered cell growth after screening >3000 hybridomas. We then developed a “targeted boost-and-sort” strategy that combines bacterial cell immunizations, purified BamA protein boosts, and single hybridoma cell sorting using amphipol-reconstituted BamA antigen. This unique workflow improves the discovery efficiency of FACS + mAbs by >600-fold and enabled the identification of rare anti-BamA mAbs with bacterial growth inhibitory activity in the presence of a truncated lipopolysaccharide layer. These mAbs represent novel tools for dissecting the BamA-mediated mechanism of β-barrel folding and our workflow establishes a new template for the efficient discovery of novel mAbs against other highly dynamic membrane proteins.


Science Translational Medicine | 2018

Avidity-based binding to HER2 results in selective killing of HER2-overexpressing cells by anti-HER2/CD3

Dionysos Slaga; Diego Ellerman; T. Noelle Lombana; Rajesh Vij; Ji Li; Maria Hristopoulos; Robyn Clark; Jennifer Johnston; Amy Shelton; Elaine Mai; Kapil Gadkar; Amy A. Lo; James T. Koerber; Klara Totpal; Rodney A. Prell; Genee Lee; Christoph Spiess; Teemu T. Junttila

T cell–dependent bispecific antibodies with bivalent low affinity binding to HER2 are more selective for tumor cells that overexpress the target. Less can be more for tumor targeting HER2 is a receptor tyrosine kinase that is often overexpressed in breast cancer. Unfortunately, many normal tissues also express HER2, resulting in toxicity from HER2-targeted treatments. Slaga et al. have developed a T cell–dependent bispecific antibody that binds to both HER2 and the CD3 protein on T cells, helping redirect the T cells to recognize tumor cells. To improve treatment safety, the authors selected an antibody that binds two HER2 molecules at a time, but with low affinity for each one, making it selective for tumors that have a high density of surface HER2 relative to healthy tissues. A primary barrier to the success of T cell–recruiting bispecific antibodies in the treatment of solid tumors is the lack of tumor-specific targets, resulting in on-target off-tumor adverse effects from T cell autoreactivity to target-expressing organs. To overcome this, we developed an anti-HER2/CD3 T cell–dependent bispecific (TDB) antibody that selectively targets HER2-overexpressing tumor cells with high potency, while sparing cells that express low amounts of HER2 found in normal human tissues. Selectivity is based on the avidity of two low-affinity anti-HER2 Fab arms to high target density on HER2-overexpressing cells. The increased selectivity to HER2-overexpressing cells is expected to mitigate the risk of adverse effects and increase the therapeutic index. Results included in this manuscript not only support the clinical development of anti-HER2/CD3 1Fab–immunoglobulin G TDB but also introduce a potentially widely applicable strategy for other T cell–directed therapies. The potential of this discovery has broad applications to further enable consideration of solid tumor targets that were previously limited by on-target, but off-tumor, autoimmunity.


PLOS ONE | 2018

Analysis of the effect of promoter type and skin pretreatment on antigen expression and antibody response after gene gun-based immunization

Rajesh Vij; Zhonghua Lin; Kellen Schneider; Dhaya Seshasayee; James T. Koerber

Monoclonal antibodies (mAbs) have enabled numerous basic research discoveries and therapeutic approaches for many protein classes. However, there still exist a number of target classes, such as multi-pass membrane proteins, for which antibody discovery is difficult, due in part to lack of high quality, recombinant protein. Here we describe the impact of several parameters on antigen expression and the development of mAbs against human claudin 4 (CLDN4), a potential multi-indication cancer target. Using gene gun-based DNA delivery and bioluminescence imaging, we optimize promoter type by comparing expression profiles of four robust in vivo promoters. In addition, we observe that most vectors rapidly lose expression, ultimately reaching almost background levels by three days post-delivery. Recognizing this limitation, we next explored skin pretreatment strategies as an orthogonal method to further boost the efficiency of mAb generation. We show that SDS pretreatment can boost antigen expression, but fails to significantly increase mAb discovery efficiency. In contrast, we find that sandpaper pretreatment yields 5-fold more FACS+ anti-CLDN4 hybridomas, without impacting antigen expression. Our findings coupled with other strategies to improve DNA immunizations should improve the success of mAb discovery against other challenging targets and enable the generation of critical research tools and therapeutic candidates.


Cancer Research | 2010

Abstract 4289: Early study on LGR5/GPR49 molecule as a potential colon cancer stem cell target for the antibody conjugated drug treatment

Weiguang Mao; Janet Tien; David M. Goldenberg; Scott Stawicki; Rajesh Vij; Xiaoling Xia; Jo-Anne Hongo; Sarajane Ross; Kathy Kozak; Ron Firestein; Yan Wu; Susan D. Spencer; Paul Polakis

LGR5 (GPR49) is a Wnt pathway downstream target gene. It has already been confirmed that LGR5 gene is up regulated by APC or β-catenin mutation. LGR5 has recently been identified as a biomarker on the human and murine intestinal or colon stem cells. Our gene expression data shows that LGR5 is highly expressed in colon cancer, with minimal expression in normal tissue. Our strategy in this project is to eliminate tumor stem cells by drug conjugated antibody. Our goal is to find a tumor stem cell marker and develop antibody to target the tumor stem cells. Here we developed and characterized both anti-LGR5 phage and monoclonal antibodies by multi molecular techniques. We finally focus on one phage antibody (YW353) and three monoclonal antibodies (2H6, 3G12 and 8E11) as therapeutic drugs for further study. We evaluated antibody drug conjugate (ADC) in vitro and in vivo with different animal tumor models derived from cell lines and human tumor explants. Good efficacy was observed in these animal models following a single dose of 5mg/kg. Conclusion: LGR5 is an attractive colon cancer target for the administration of anti-LGR5 ADC in human cancer. More in vivo models will be tested to confirm the efficacy and evaluate safety. 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 4289.

Collaboration


Dive into the Rajesh Vij's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge