Network


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

Hotspot


Dive into the research topics where Lalita Wadhwa is active.

Publication


Featured researches published by Lalita Wadhwa.


Neuro-oncology | 2011

A single intravenous injection of oncolytic picornavirus SVV-001 eliminates medulloblastomas in primary tumor-based orthotopic xenograft mouse models

Litian Yu; Patricia Baxter; Xiumei Zhao; Zhigang Liu; Lalita Wadhwa; Yujing Zhang; Jack Su; Xiaojie Tan; Jianhua Yang; Adekunle M. Adesina; Lazlo Perlaky; Mary Y. Hurwitz; Neeraja Idamakanti; Paul L. Hallenbeck; Susan M. Blaney; Murali Chintagumpala; Richard L. Hurwitz; Xiao-Nan Li

Difficulties of drug delivery across the blood-brain barrier (BBB) and failure to eliminate cancer stem cells (CSCs) are believed to be the major causes of tumor recurrences in children with medulloblastoma (MB). Seneca Valley virus-001 (SVV-001) is a naturally occurring oncolytic picornavirus that can be systemically administered. Here, we report its antitumor activities against MB cells in a panel of 10 primary tumor-based orthotopic xenograft mouse models. We found that SVV-001 killed the primary cultured xenograft cells, infected and replicated in tumor cells expressing CSC surface marker CD133, and eliminated tumor cells capable of forming neurospheres in vitro in 5 of the 10 xenograft models. We confirmed that SVV-001 could pass through BBB in vivo. A single i.v. injection of SVV-001 in 2 anaplastic MB models led to widespread infection of the preformed intracerebellar (ICb) xenografts, resulting in significant increase in survival (2.2-5.9-fold) in both models and complete elimination of ICb xenografts in 8 of the 10 long-term survivors. Mechanistically, we showed that the intracellular replication of SVV-001 is mediated through a subverted autophagy that is different from the bona fide autophagic process induced by rapamycin. Our data suggest that SVV-001 is well suited for MB treatment. This work expands the current views in the oncolytic therapy field regarding the utility of oncolytic viruses in simultaneous targeting of stem and nonstem tumor cells.


Cancer Research | 2007

Treatment of invasive retinoblastoma in a murine model using an oncolytic picornavirus

Lalita Wadhwa; Mary Y. Hurwitz; Patricia Chévez-Barrios; Richard L. Hurwitz

Retinoblastoma, the most common intraocular malignancy of childhood, metastasizes by initial invasion of the choroid and the optic nerve. There is no effective treatment for metastatic retinoblastoma, especially when the central nervous system (CNS) is involved, and prevention of this complication is a treatment priority. Seneca Valley Virus (SVV-001) is a conditionally replication-competent picornavirus that is not pathogenic to normal human cells but can kill human retinoblastoma cells in vitro with an IC(50) of <1 viral particle (vp) per cell. A xenograft murine model of metastatic retinoblastoma was used to examine the therapeutic potential of SVV-001. Histopathologic analysis of ocular and brain tissues after a single tail vein injection of SVV-001 (1 x 10(13) vp/kg) showed effective treatment of choroid and ocular nerve tumor invasion (1 of 20 animals with invasive disease in the treated group versus 7 of 20 animals with invasive disease in the control group; P = 0.017) and prevention of CNS metastasis (0 of 20 animals with CNS metastatic disease in the treated group versus 4 of 20 animals with CNS disease in the control group; P = 0.036). There were no observed adverse events due to the virus in any of the treated animals. SVV-001 may be effective as a treatment of locally invasive and metastatic retinoblastoma.


Neuro-oncology | 2013

Intravenous injection of oncolytic picornavirus SVV-001 prolongs animal survival in a panel of primary tumor–based orthotopic xenograft mouse models of pediatric glioma

Zhigang Liu; Xiumei Zhao; Patricia Baxter; Yulun Huang; Litian Yu; Lalita Wadhwa; J. Su; Adekunle M. Adesina; Lazlo Perlaky; Mary Y. Hurwitz; Neeraja Idamakanti; Paul L. Hallenbeck; Richard L. Hurwitz; Ching C. Lau; Murali Chintagumpala; Susan M. Blaney; Xiao-nan Li

BACKGROUND Seneca Valley virus (SVV-001) is a nonpathogenic oncolytic virus that can be systemically administered and can pass through the blood-brain barrier. We examined its therapeutic efficacy and the mechanism of tumor cell infection in pediatric malignant gliomas. METHODS In vitro antitumor activities were examined in primary cultures, preformed neurospheres, and self-renewing glioma cells derived from 6 patient tumor orthotopic xenograft mouse models (1 anaplastic astrocytoma and 5 GBM). In vivo therapeutic efficacy was examined by systemic treatment of preformed xenografts in 3 permissive and 2 resistant models. The functional role of sialic acid in mediating SVV-001 infection was investigated using neuraminidase and lectins that cleave or competitively bind to linkage-specific sialic acids. RESULTS SVV-001 at a multiplicity of infection of 0.5 to 25 replicated in and effectively killed primary cultures, preformed neurospheres, and self-renewing stemlike single glioma cells derived from 4 of the 6 glioma models in vitro. A single i.v. injection of SVV-001 (5 × 10(12) viral particles/kg) led to the infection of orthotopic xenografts without harming normal mouse brain cells, resulting in significantly prolonged survival in all 3 permissive and 1 resistant mouse models (P < .05). Treatment with neuraminidase and competitive binding using lectins specific for α2,3-linked and/or α2,6-linked sialic acid significantly suppressed SVV-001 infectivity (P < .01). CONCLUSION SVV-001 possesses strong antitumor activity against pediatric malignant gliomas and utilizes α2,3-linked and α2,6-linked sialic acids as mediators of tumor cell infection. Our findings support the consideration of SVV-001 for clinical trials in children with malignant glioma.


Biopreservation and Biobanking | 2016

Assays for Qualification and Quality Stratification of Clinical Biospecimens Used in Research: A Technical Report from the ISBER Biospecimen Science Working Group

Fay Betsou; Alexandre Bulla; Sang Yun Cho; Judith A. Clements; Rodrigo F. Chuaqui; Domenico Coppola; Yvonne De Souza; Annemieke De Wilde; William E. Grizzle; Fiorella Guadagni; Elaine Gunter; Stacey Heil; Verity S. Hodgkinson; Joseph Kessler; Michael Kiehntopf; Hee Sung Kim; Iren Koppandi; Katheryn Shea; Rajeev Singh; Marc Sobel; Stella Somiari; Demetri D. Spyropoulos; Mars Stone; Gunnel Tybring; Klara Valyi-Nagy; Gert Van den Eynden; Lalita Wadhwa

This technical report presents quality control (QC) assays that can be performed in order to qualify clinical biospecimens that have been biobanked for use in research. Some QC assays are specific to a disease area. Some QC assays are specific to a particular downstream analytical platform. When such a qualification is not possible, QC assays are presented that can be performed to stratify clinical biospecimens according to their biomolecular quality.


Journal of Chromatography B | 2003

Determination of 3-keto-4-ene steroids and their hydroxylated metabolites catalyzed by recombinant human cytochrome P450 1B1 enzyme using gas chromatography–mass spectrometry with trimethylsilyl derivatization

Junghan Song; Lalita Wadhwa; Bassem A. Bejjani; William E. O’Brien

A protocol utilizing gas chromatography with selected ion monitoring mass spectrometric detection (GC-SIM-MS) using a simplified trimethylsilyl (TMS) derivatization protocol was developed and validated for the determination of hydroxylated metabolites of 3-keto-4-ene steroids such as testosterone, progesterone and androstenedione. Hydroxylated metabolites catalyzed by human CYP1B1 were extracted with methylene chloride and derivatized with BSTFA-10% TMCS. To get an optimal derivatizing condition, the effect of various incubation times and temperatures was evaluated. When the incubation temperature and time in the presence of the TMS derivatizing agent were increased, the 3-keto group became derivatized with TMS to form a 3-TMS derivative. To minimize the formation of the TMS ether on the 3-keto group, a reaction condition of 56 degrees C for 10 min was used for the routine measurement of the steroids and their hydroxylated metabolite. Performance studies including linearity of calibration curves, extraction efficiency and precision were performed. Linearity of the calibration curves was satisfactory from 0.125 to 5 microM for most compounds except 21-hydroxyprogesterone and 16alpha-hydroxyandrostenedione which deviated from linearity at the lower concentrations. Mean percentage extraction recoveries were greater than 80% for all compounds. Most compounds showed good precisions with C.V.s of within-day precision of less than 5% and C.V.s of between-day precision of less than 10%. The selected ion chromatograms from the recombinant human CYP1B1 incubations with testosterone, progesterone and androstenedione showed evidence of 6beta-, 16alpha-, 2alpha-, and 15alpha-hydroxytestosterone, 6alpha- and 16alpha-hydroxyprogesterone and 6alpha- and 16alpha-hydroxyandrostenedione, respectively. There was no significant interference associated with Escherichia coli membrane extracts in detecting hydroxylated metabolites. This procedure provides a rapid and sensitive method for the evaluation of steroid hydroxylation by CYP isoenzymes.


International Journal of Gynecology & Obstetrics | 2006

Conventional and color Doppler sonography in preoperative assessment of ovarian tumors.

Anjali Tempe; Sompal Singh; Lalita Wadhwa; A. Garg

Objective: To study the vascular patterns of ovarian tumors by color Doppler imaging (CDI) and compare the findings of conventional sonographic studies and CDI with histopathologic findings for the same tumors. Methods: Fifty nonpregnant women scheduled for elective surgery for ovarian tumors were examined by sonographic scanning and CDI by the same physician. Sonographic morphology scores [SMSs] were used, and the pulsatility index (PI) and resistance index (RI) were calculated after locating vessels in and around the tumors by CDI. Results: It was possible to obtain CDI results in 26 of the 50 women. The sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were 100%, 71.43%, 73.33%, and 100%, respectively, for SMS. With CDI, the sensitivity, specificity, PPV, and NPV were 100%, 85.72%, 95%, and 100% for PI and 100%, 71.43%, 90.48%, and 100% for RI. RI sensitivity and specificity were identical to those of SMS, i.e., 100% and 71.43%, respectively, but PI specificity was better (85.72%). Conclusion: In this study, CDI was definitely better than SMS when color flow could be obtained through the tumors. The overall efficiency in diagnosing the nature of tumors is very high if SMS and CDI are both used in patients having ovarian tumors.


Scientific Reports | 2016

Rbfox2 function in RNA metabolism is impaired in hypoplastic left heart syndrome patient hearts.

Sunil Kumar Verma; Vaibhav Deshmukh; Curtis A. Nutter; Elizabeth Jaworski; Wenhao Jin; Lalita Wadhwa; Joshua Abata; Marco Ricci; Joy Lincoln; James F. Martin; Gene W. Yeo; Muge N. Kuyumcu-Martinez

Hypoplastic left heart syndrome (HLHS) is a fatal congenital heart disease in which the left side of the heart is underdeveloped, impairing the systemic circulation. Underdeveloped left ventricle exerts biomechanical stress on the right ventricle that can progress into heart failure. Genome-wide transcriptome changes have been identified at early stages in the right ventricle (RV) of infants with HLHS, although the molecular mechanisms remain unknown. Here, we demonstrate that the RNA binding protein Rbfox2, which is mutated in HLHS patients, is a contributor to transcriptome changes in HLHS patient RVs. Our results indicate that majority of transcripts differentially expressed in HLHS patient hearts have validated Rbfox2 binding sites. We show that Rbfox2 regulates mRNA levels of targets with 3’UTR binding sites contributing to aberrant gene expression in HLHS patients. Strikingly, the Rbfox2 nonsense mutation identified in HLHS patients truncates the protein, impairs its subcellular distribution and adversely affects its function in RNA metabolism. Overall, our findings uncover a novel role for Rbfox2 in controlling transcriptome in HLHS.


Investigative Ophthalmology & Visual Science | 2012

Embryonic Retinal Tumors in SV40 T-Ag Transgenic Mice Contain CD133+ Tumor-Initiating Cells

Lalita Wadhwa; Wesley S. Bond; Laszlo Perlaky; Paul A. Overbeek; Mary Y. Hurwitz; Patricia Chévez-Barrios; Richard L. Hurwitz

PURPOSE Human retinoblastomas form during the proliferative phase of retina development and are caused by mutations that result in absent or functionally defective Rb protein. Similar tumors occur in mice only when multiple Rb gene family members are absent. We asked if retinal tumors can arise from an undifferentiated retinal cell. The tumor-initiating cells isolated from these tumors that formed in early embryonic murine retinas were characterized. METHODS Transgenic mice were created using a Pax6 promoter to target expression of SV40 large T-antigen (T-Ag) in the undifferentiated murine embryonic retina. T-Ag, which sequesters all Rb family proteins and p53, is expressed in the retina and lens by murine embryonic day 10 (E10) and tumors are observed by E12.5. A cell line that is adherent in serum-containing media and forms neurospheres in supplemented serum-free media was developed from retinal tumors isolated on postnatal day 7. RESULTS In all, 1.5% of attached cells form neurospheres when transferred to serum-free medium. All cultured cells express T-Ag, confirming that they derive from the original tumors; 0.5% of adherent cells express detectable levels of CD133. CD133+ FACS-sorted cells cultured in serum-free medium form 3-fold more neurospheres than do CD133- cells. Six of seven mice injected with CD133+ cells and one of seven mice injected with CD133- cells formed tumors during a 6-month period. Unlike primary adherent cells, primary and secondary tumors heterogeneously express markers of stem cells and differentiation similar to human retinoblastoma. CONCLUSIONS CD133+ tumor-initiating cells can originate from proliferating undifferentiated precursor cells.


Journal of Visualized Experiments | 2011

Establishment and propagation of human retinoblastoma tumors in immune deficient mice.

Wesley S. Bond; Lalita Wadhwa; Laszlo Perlaky; Rebecca L. Penland; Mary Y. Hurwitz; Richard L. Hurwitz; Patricia Chévez-Barrios

Culturing retinoblastoma tumor cells in defined stem cell media gives rise to primary tumorspheres that can be grown and maintained for only a limited time. These cultured tumorspheres may exhibit markedly different cellular phenotypes when compared to the original tumors. Demonstration that cultured cells have the capability of forming new tumors is important to ensure that cultured cells model the biology of the original tumor. Here we present a protocol for propagating human retinoblastoma tumors in vivo using Rag2-/- immune deficient mice. Cultured human retinoblastoma tumorspheres of low passage or cells obtained from freshly harvested human retinoblastoma tumors injected directly into the vitreous cavity of murine eyes form tumors within 2-4 weeks. These tumors can be harvested and either further passaged into murine eyes in vivo or grown as tumorspheres in vitro. Propagation has been successfully carried out for at least three passages thus establishing a continuing source of human retinoblastoma tissue for further experimentation. Wesley S. Bond and Lalita Wadhwa are co-first authors.


Fems Microbiology Letters | 2000

Progesterone side-chain cleavage by Bacillus sphaericus

Lalita Wadhwa; Kelvin E. Smith

Collaboration


Dive into the Lalita Wadhwa's collaboration.

Top Co-Authors

Avatar

Mary Y. Hurwitz

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lazlo Perlaky

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wesley S. Bond

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sompal Singh

Maulana Azad Medical College

View shared research outputs
Top Co-Authors

Avatar

J. B. Sharma

Maulana Azad Medical College

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charles D. Fraser

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge