Network


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

Hotspot


Dive into the research topics where Roma Patel is active.

Publication


Featured researches published by Roma Patel.


Cell Death and Disease | 2013

USP7 inhibitor P22077 inhibits neuroblastoma growth via inducing p53-mediated apoptosis

Yihui Fan; Jin Cheng; Sanjeev A. Vasudevan; Jun Dou; H. Zhang; Roma Patel; I. T. Ma; Yesenia Rojas; Yanling Zhao; Y. Yu; Jason M. Shohet; Jed G. Nuchtern; Eugene S. Kim; Jianhua Yang

Neuroblastoma (NB) is a common pediatric cancer and contributes to more than 15% of all pediatric cancer-related deaths. Unlike adult tumors, recurrent somatic mutations in NB, such as tumor protein 53 (p53) mutations, occur with relative paucity. In addition, p53 downstream function is intact in NB cells with wild-type p53, suggesting that reactivation of p53 may be a viable therapeutic strategy for NB treatment. Herein, we report that the ubiquitin-specific protease 7 (USP7) inhibitor, P22077, potently induces apoptosis in NB cells with an intact USP7-HDM2-p53 axis but not in NB cells with mutant p53 or without human homolog of MDM2 (HDM2) expression. In this study, we found that P22077 stabilized p53 by inducing HDM2 protein degradation in NB cells. P22077 also significantly augmented the cytotoxic effects of doxorubicin (Dox) and etoposide (VP-16) in NB cells with an intact USP7-HDM2-p53 axis. Moreover, P22077 was found to be able to sensitize chemoresistant LA-N-6 NB cells to chemotherapy. In an in vivo orthotopic NB mouse model, P22077 significantly inhibited the xenograft growth of three NB cell lines. Database analysis of NB patients shows that high expression of USP7 significantly predicts poor outcomes. Together, our data strongly suggest that targeting USP7 is a novel concept in the treatment of NB. USP7-specific inhibitors like P22077 may serve not only as a stand-alone therapy but also as an effective adjunct to current chemotherapeutic regimens for treating NB with an intact USP7-HDM2-p53 axis.


Apoptosis | 2013

TAK1 inhibitor 5Z-7-oxozeaenol sensitizes neuroblastoma to chemotherapy.

Yihui Fan; Jin Cheng; Sanjeev A. Vasudevan; Roma Patel; Li Liang; Xin Xu; Yanling Zhao; Wei Jia; Fengmin Lu; Hong Zhang; Jed G. Nuchtern; Eugene S. Kim; Jianhua Yang

Treatment failure in high risk neuroblastoma is largely due to development of chemoresistance. NF-κB activation is one of the resistance mechanisms for cancer cells to escape from chemotherapy-induced cell-death. TAK1 is an essential component in genotoxic stresses-induced NF-κB activation; however, the role of TAK1 in the development of chemoresistance in neuroblastoma remains unknown. Using a panel of neuroblastoma cell lines, we found that TAK1 inhibitor 5Z-7-oxozeaenol significantly augmented the cytotoxic effects of doxorubicin (Dox) and etoposide (VP-16) on neuroblastoma cell lines. TAK1 inhibition also enhanced the inhibitory effect of Dox and VP-16 on anchorage-independent growth. Treatment of neuroblastoma cells with 5Z-7-oxozeaenol blocked Dox- and VP16-induced NF-κB activation and enhanced Dox- and VP16-induced apoptosis. Moreover, 5Z-7-oxozeaenol was able to overcome the established chemoresistance in LA-N-6 neuroblastoma cells. Using an orthotopic neuroblastoma mouse model, we found that 5Z-7-oxozeaenol significantly enhanced chemotherapeutic efficacy in vivo. Together, our results provide a proof-of-concept that TAK1 inhibition significantly increases the sensitivity of neuroblastoma cells to chemotherapy-induced cell-death and can serve as an effective adjunct to current chemotherapeutic regimens for high risk diseases.


Apoptosis | 2015

MTOR ATP-competitive inhibitor INK128 inhibits neuroblastoma growth via blocking mTORC signaling

Huiyuan Zhang; Jun Dou; Yang Yu; Yanling Zhao; Yihui Fan; Jin Cheng; Xin Xu; Wei Liu; Shan Guan; Zhenghu Chen; Yan Shi; Roma Patel; Sanjeev A. Vasudevan; Peter E. Zage; Hong Zhang; Jed G. Nuchtern; Eugene S. Kim; Songbin Fu; Jianhua Yang

High-risk neuroblastoma often develops resistance to high-dose chemotherapy. The mTOR signaling cascade is frequently deregulated in human cancers and targeting mTOR signaling sensitizes many cancer types to chemotherapy. Here, using a panel of neuroblastoma cell lines, we found that the mTOR inhibitor INK128 showed inhibitory effects on both anchorage-dependent and independent growth of neuroblastoma cells and significantly enhanced the cytotoxic effects of doxorubicin (Dox) on these cell lines. Treatment of neuroblastoma cells with INK128 blocked the activation of downstream mTOR signaling and enhanced Dox-induced apoptosis. Moreover, INK128 was able to overcome the established chemoresistance in the LA-N-6 cell line. Using an orthotopic neuroblastoma mouse model, we found that INK128 significantly inhibited tumor growth in vivo. In conclusion, we have shown that INK128-mediated mTOR inhibition possessed substantial antitumor activity and could significantly increase the sensitivity of neuroblastoma cells to Dox therapy. Taken together, our results indicate that using INK128 can provide additional efficacy to current chemotherapeutic regimens and represent a new paradigm in restoring drug sensitivity in neuroblastoma.


Cell Death and Disease | 2015

NSC-87877 inhibits DUSP26 function in neuroblastoma resulting in p53-mediated apoptosis

Yan Shi; I. T. Ma; Roma Patel; X Shang; Zhenghu Chen; Yanling Zhao; Jin Cheng; Yihui Fan; Yesenia Rojas; E Barbieri; Yang Yu; Jingling Jin; Eugene S. Kim; Jason M. Shohet; Sanjeev A. Vasudevan; Jianhua Yang

Dual specificity protein phosphatase 26 (DUSP26) is overexpressed in high-risk neuroblastoma (NB) and contributes to chemoresistance by inhibiting p53 function. In vitro, DUSP26 has also been shown to effectively inhibit p38 MAP kinase. We hypothesize that inhibiting DUSP26 will result in decreased NB cell growth in a p53 and/or p38-mediated manner. NSC-87877 (8-hydroxy-7-[(6-sulfo-2-naphthyl)azo]-5-quinolinesulfonic acid), a novel DUSP26 small molecule inhibitor, shows effective growth inhibition and induction of apoptosis in NB cell lines. NB cell lines treated with small hairpin RNA (shRNA) targeting DUSP26 also exhibit a proliferation defect both in vitro and in vivo. Treatment of NB cell lines with NSC-87877 results in increased p53 phosphorylation (Ser37 and Ser46) and activation, increased activation of downstream p38 effector proteins (heat shock protein 27 (HSP27) and MAP kinase-activated protein kinase 2 (MAPKAPK2)) and poly ADP ribose polymerase/caspase-3 cleavage. The cytotoxicity resulting from DUSP26 inhibition is partially reversed by knocking down p53 expression with shRNA and also by inhibiting p38 activity with SB203580 (4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine). In an intrarenal mouse model of NB, NSC-87877 treatment results in decreased tumor growth and increased p53 and p38 activity. Together, these results suggest that DUSP26 inhibition with NSC-87877 is an effective strategy to induce NB cell cytotoxicity in vitro and in vivo through activation of the p53 and p38 mitogen-activated protein kinase (MAPK) tumor-suppressor pathways.


Scientific Reports | 2016

Wip1 inhibitor GSK2830371 inhibits neuroblastoma growth by inducing Chk2/p53-mediated apoptosis

Zhenghu Chen; Long Wang; Dayong Yao; Tianshu Yang; Wen Ming Cao; Jun Dou; Jonathan C. Pang; Shan Guan; Huiyuan Zhang; Yang Yu; Yanling Zhao; Yongfeng Wang; Xin Xu; Yan Shi; Roma Patel; Hong Zhang; Sanjeev A. Vasudevan; Shangfeng Liu; Jianhua Yang; Jed G. Nuchtern

Neuroblastoma (NB) is the most common extracranial tumor in children. Unlike in most adult tumors, tumor suppressor protein 53 (p53) mutations occur with a relatively low frequency in NB and the downstream function of p53 is intact in NB cell lines. Wip1 is a negative regulator of p53 and hindrance of Wip1 activity by novel inhibitor GSK2830371 is a potential strategy to activate p53’s tumor suppressing function in NB. Yet, the in vivo efficacy and the possible mechanisms of GSK2830371 in NB have not yet been elucidated. Here we report that novel Wip1 inhibitor GSK2830371 induced Chk2/p53-mediated apoptosis in NB cells in a p53-dependent manner. In addition, GSK2830371 suppressed the colony-formation potential of p53 wild-type NB cell lines. Furthermore, GSK2830371 enhanced doxorubicin- (Dox) and etoposide- (VP-16) induced cytotoxicity in a subset of NB cell lines, including the chemoresistant LA-N-6 cell line. More importantly, GSK2830371 significantly inhibited tumor growth in an orthotopic xenograft NB mouse model by inducing Chk2/p53-mediated apoptosis in vivo. Taken together, this study suggests that GSK2830371 induces Chk2/p53-mediated apoptosis both in vitro and in vivo in a p53 dependent manner.


Oncotarget | 2017

EWS-FLI1 and RNA helicase A interaction inhibitor YK-4-279 inhibits growth of neuroblastoma

Wenjing Sun; Yesenia Rojas; Hao Wang; Yang Yu; Yongfeng Wang; Zhenghu Chen; Kimal Rajapakshe; Xin Xu; Wei Huang; Saurabh Agarwal; Roma Patel; Sarah E. Woodfield; Yanling Zhao; Jingling Jin; Hong Zhang; Angela Major; M. John Hicks; Jason M. Shohet; Sanjeev A. Vasudevan; Cristian Coarfa; Jianhua Yang; Jed G. Nuchtern

Treatment failure in high risk neuroblastoma (NB) is largely due to the development of chemotherapy resistance. We analyzed the gene expression changes associated with exposure to chemotherapy in six high risk NB tumors with the aid of the Connectivity Map bioinformatics platform. Ten therapeutic agents were predicted to have a high probability of reversing the transcriptome changes associated with neoadjuvant chemotherapy treatment. Among these agents, initial screening showed the EWS-FLI1 and RNA helicase A interaction inhibitor YK-4-279, had obvious cytotoxic effects on NB cell lines. Using a panel of NB cell lines, including MYCN nonamplified (SK-N-AS, SH-SY5Y, and CHLA-255), and MYCN amplified (NB-19, NGP, and IMR-32) cell lines, we found that YK-4-279 had cytotoxic effects on all lines tested. In addition, YK-4-279 also inhibited cell proliferation and anchorage-independent growth and induced cell apoptosis of these cells. YK-4-279 enhanced the cytotoxic effect of doxorubicin (Dox). Moreover, YK-4-279 was able to overcome the established chemoresistance of LA-N-6 NB cells. In an orthotopic xenograft NB mouse model, YK-4-279 inhibited NB tumor growth and induced apoptosis in tumor cells through PARP and Caspase 3 cleavage in vivo. While EWS-FLI1 fusion protein is not frequently found in NB, using the R2 public database of neuroblastoma outcome and gene expression, we found that high expression of EWSR1 was associated with poor patient outcome. Knockdown of EWSR1 inhibited the oncogenic potential of neuroblastoma cell lines. Taken together, our results indicate that YK-4-279 might be a promising agent for treatment of NB that merits further exploration.


Oncology Reports | 2018

Targeting LRH‑1 in hepatoblastoma cell lines causes decreased proliferation

Jingling Jin; Junliang Jin; Sarah E. Woodfield; Roma Patel; Nan Ge Jin; Yan Shi; Bin Liu; Wenjing Sun; Xiangmei Chen; Yang Yu; Sanjeev A. Vasudevan

Hepatoblastoma is the most common malignant liver tumor in children. Since it is often unresectable and exhibits drug resistance, the treatment of advanced hepatoblastoma is challenging. The orphan nuclear receptor liver receptor homolog-1 (LRH-1) serves prominent roles in malignancy; however, to the best of our knowledge, the role of LRH-1 in hepatoblastoma remains unknown. In the present study, human hepatoblastoma cell lines were analyzed; the mRNA and protein expression levels of LRH-1 were significantly higher in HepG2 and HuH6 cells compared with those in HepT1 cells and control THLE-2 cells. Knockdown of LRH-1 resulted in decreased HepG2 and HuH6 cell proliferation via downregulation of cyclin D1 (CCND1) and c-Myc. Furthermore, treatment with an LRH-1 antagonist (LRA) inhibited the proliferation and colony formation of cell lines in a dose-dependent manner, and induced cell cycle arrest at G1 phase through inhibition of CCND1 expression. Finally, LRA treatment enhanced the cytotoxic effects of doxorubicin on hepatoblastoma cells. Collectively, these findings suggested that LRH-1 may have an important role in the progression of hepatoblastoma and implicated LRA as a novel, potential therapeutic agent for the treatment of hepatoblastoma.


Cancer Research | 2018

Abstract B35: Reactivation of p53 signaling in hepatoblastoma with a stapled peptide dual inhibitor of MDM2 and MDM4

Sarah E. Woodfield; Roma Patel; Aryana M. Ibarra; Zhenghu Chen; Sanjeev A. Vasudevan

Introduction: Hepatoblastoma is the most common pediatric liver malignancy. Despite aggressive therapeutic regimens, long-term survival of patients with high-risk, metastatic disease is less than 50%. Most cases of hepatoblastoma do not carry mutations in the TP53 tumor suppressor gene; therefore, we hypothesized that targeting the two major regulators of p53, MDM2 and MDM4, with the stapled peptide dual inhibitor, ATSP-7041, to reactivate p53 signaling would be an effective therapeutic strategy for hepatoblastoma. Methods: Levels of MDM2 and MDM4 gene expression were examined in HB cell lines and in a cohort of 18 primary HB patient samples from stage III (n=12) and IV (n=6) patients in comparison to expression in four adjacent uninvolved liver samples with qPCR experiments. The effects of the dual MDM2/MDM4 inhibitor ATSP-7041 (Aileron Therapeutics) on the same cell lines were analyzed with MTT assays for cytotoxicity and qPCR experiments to look at changes in mRNA expression of p53 targets Bax, Puma, CDKN1A, and MDM2. Results: In 16 of 18 samples, MDM4 expression was higher than average in the uninvolved liver samples; in five samples expression was at least 5-fold higher and in four samples expression was at least 10-fold higher. Of note, all four samples with at least 10-fold higher expression were from stage IV patients. In comparison, MDM2 expression was only elevated in one of the 18 patient samples. All three hepatoblastoma cell lines showed expression of both MDM2 and MDM4. Treatment with ATSP-7041 showed cytotoxic (5.7-8.6 μM) effects in all cell lines tested. All cell lines also showed upregulation of p53 targets Bax (1.4- to 2.4-fold), Puma (2.4- to 3.2-fold), CDKN1A (2.4- to 7.2-fold), and MDM2 (6.1- to 8.3-fold) with treatment. Conclusions: Dual inhibition of MDM2 and MDM4 shows efficacy in preclinical models of hepatoblastoma by upregulating p53 tumor-suppressor signaling in TP53 wild-type cells and is a promising therapeutic strategy for high-risk patients with wild-type TP53. Citation Format: Sarah E. Woodfield, Roma H. Patel, Aryana M. Ibarra, Zhenghu Chen, Sanjeev A. Vasudevan. Reactivation of p53 signaling in hepatoblastoma with a stapled peptide dual inhibitor of MDM2 and MDM4 [abstract]. In: Proceedings of the AACR Special Conference: Pediatric Cancer Research: From Basic Science to the Clinic; 2017 Dec 3-6; Atlanta, Georgia. Philadelphia (PA): AACR; Cancer Res 2018;78(19 Suppl):Abstract nr B35.


Scientific Reports | 2017

A Novel Cell Line Based Orthotopic Xenograft Mouse Model That Recapitulates Human Hepatoblastoma

Sarah E. Woodfield; Yan Shi; Roma Patel; Jingling Jin; Angela Major; Stephen F. Sarabia; Zbigniew Starosolski; Barry Zorman; Siddharth S. Gupta; Zhenghu Chen; Aryana M. Ibarra; Karl-Dimiter Bissig; Ketan B. Ghaghada; Pavel Sumazin; Dolores Lopez-Terrada; Sanjeev A. Vasudevan

Currently, preclinical testing of therapies for hepatoblastoma (HB) is limited to subcutaneous and intrasplenic xenograft models that do not recapitulate the hepatic tumors seen in patients. We hypothesized that injection of HB cell lines into the livers of mice would result in liver tumors that resemble their clinical counterparts. HepG2 and Huh-6 HB cell lines were injected, and tumor growth was monitored with bioluminescence imaging (BLI) and magnetic resonance imaging (MRI). Levels of human α-fetoprotein (AFP) were monitored in the serum of animals. Immunohistochemical and gene expression analyses were also completed on xenograft tumor samples. BLI signal indicative of tumor growth was seen in 55% of HepG2- and Huh-6-injected animals after a period of four to seven weeks. Increased AFP levels correlated with tumor growth. MRI showed large intrahepatic tumors with active neovascularization. HepG2 and Huh-6 xenografts showed expression of β-catenin, AFP, and Glypican-3 (GPC3). HepG2 samples displayed a consistent gene expression profile most similar to human HB tumors. Intrahepatic injection of HB cell lines leads to liver tumors in mice with growth patterns and biologic, histologic, and genetic features similar to human HB tumors. This orthotopic xenograft mouse model will enable clinically relevant testing of novel agents for HB.


Oncotarget | 2017

Small molecule inhibitor regorafenib inhibits RET signaling in neuroblastoma cells and effectively suppresses tumor growth in vivo

Zhenghu Chen; Yanling Zhao; Yang Yu; Jonathan C. Pang; Sarah E. Woodfield; Ling Tao; Shan Guan; Huiyuan Zhang; Shayahati Bieerkehazhi; Yan Shi; Roma Patel; Sanjeev A. Vasudevan; Joanna S. Yi; Jodi A. Muscal; Guo-Tong Xu; Jianhua Yang

Neuroblastoma (NB), the most common extracranial pediatric solid tumor, continues to cause significant cancer-related morbidity and mortality in children. Dysregulation of oncogenic receptor tyrosine kinases (RTKs) has been shown to contribute to tumorigenesis in various human cancers and targeting these RTKs has had therapeutic benefit. RET is an RTK which is commonly expressed in NB, and high expression of RET correlates with poor outcomes in patients with NB. Herein we report that RET is required for NB cell proliferation and that the small molecule inhibitor regorafenib (BAY 73-4506) blocks glial cell derived neurotrophic factor (GDNF)-induced RET signaling in NB cells and inhibits NB growth both in vitro and in vivo. We found that regorafenib significantly inhibited cell proliferation and colony formation ability of NB cells. Moreover, regorafenib suppressed tumor growth in both an orthotopic xenograft NB mouse model and a TH-MYCN transgenic NB mouse model. Finally, regorafenib markedly improved the overall survival of TH-MYCN transgenic tumor-bearing mice. In summary, our study suggests that RET is a potential therapeutic target in NB, and that using a novel RET inhibitor, like regorafenib, should be investigated as a therapeutic treatment option for children with NB.

Collaboration


Dive into the Roma Patel's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianhua Yang

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yanling Zhao

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jed G. Nuchtern

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jingling Jin

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yang Yu

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yihui Fan

Baylor College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge