Network


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

Hotspot


Dive into the research topics where A. Parekh is active.

Publication


Featured researches published by A. Parekh.


Cancer Letters | 2013

IGFBP5 domains exert distinct inhibitory effects on the tumorigenicity and metastasis of human osteosarcoma

Gaurav Luther; Joseph D. Lamplot; Xiang Chen; Richard Rames; Eric R. Wagner; Xing Liu; A. Parekh; Enyi Huang; Stephanie H. Kim; Jikun Shen; Rex C. Haydon; Tong-Chuan He; Hue H. Luu

Osteosarcoma (OS) is the most common primary malignancy of bone. We investigated the roles of insulin-like growth factor binding protein 5 (IGFBP5) domains in modulating OS tumorigenicity and metastasis. The N-terminal (to a lesser extent the C-terminal) domain inhibited cell proliferation and induced apoptosis while the C-terminal domain inhibited cell migration and invasion. The Linker domain had no independent effects. In vivo, the N-terminal domain decreased tumor growth without affecting pulmonary metastases while the C-terminal domain inhibited tumor growth and metastases. In summary, the N- and C-terminal domains modulated OS tumorigenic phenotypes while the C-terminal domain inhibited OS metastatic phenotypes.


Oncotarget | 2016

Cancer therapies activate RIG-I-like receptor pathway through endogenous non-coding RNAs

Diana Rose E. Ranoa; A. Parekh; Sean P. Pitroda; Xiaona Huang; Thomas E. Darga; Antony C. Wong; Lei Huang; Jorge Andrade; Jonathan P. Staley; Takashi Satoh; Shizuo Akira; Ralph R. Weichselbaum; Nikolai N. Khodarev

Emerging evidence indicates that ionizing radiation (IR) and chemotherapy activate Type I interferon (IFN) signaling in tumor and host cells. However, the mechanism of induction is poorly understood. We identified a novel radioprotective role for the DEXH box RNA helicase LGP2 (DHX58) through its suppression of IR-induced cytotoxic IFN-beta [1]. LGP2 inhibits activation of the RIG-I-like receptor (RLR) pathway upon binding of viral RNA to the cytoplasmic sensors RIG-I (DDX58) and MDA5 (IFIH1) and subsequent IFN signaling via the mitochondrial adaptor protein MAVS (IPS1). Here we show that MAVS is necessary for IFN-beta induction and interferon-stimulated gene expression in the response to IR. Suppression of MAVS conferred radioresistance in normal and cancer cells. Germline deletion of RIG-I, but not MDA5, protected mice from death following total body irradiation, while deletion of LGP2 accelerated the death of irradiated animals. In human tumors depletion of RIG-I conferred resistance to IR and different classes of chemotherapy drugs. Mechanistically, IR stimulated the binding of cytoplasmic RIG-I with small endogenous non-coding RNAs (sncRNAs), which triggered IFN-beta activity. We demonstrate that the small nuclear RNAs U1 and U2 translocate to the cytoplasm after IR treatment, thus stimulating the formation of RIG-I: RNA complexes and initiating downstream signaling events. Taken together, these findings suggest that the physiologic responses to radio-/chemo-therapy converge on an antiviral program in recruitment of the RLR pathway by a sncRNA-dependent activation of RIG-I which commences cytotoxic IFN signaling. Importantly, activation of interferon genes by radiation or chemotherapy is associated with a favorable outcome in patients undergoing treatment for cancer. To our knowledge, this is the first demonstration of a cell-intrinsic response to clinically relevant genotoxic treatments mediated by an RNA-dependent mechanism.


Proceedings of the National Academy of Sciences of the United States of America | 2014

RIG-I–like receptor LGP2 protects tumor cells from ionizing radiation

Ryan C. Widau; A. Parekh; M.C. Ranck; Daniel W. Golden; Kiran A. Kumar; Ravi F. Sood; Sean P. Pitroda; Zhengkai Liao; Xiaona Huang; Thomas E. Darga; David Xu; Lei Huang; Jorge Andrade; Bernard Roizman; Ralph R. Weichselbaum; Nikolai N. Khodarev

Significance An undesirable outcome of radiotherapy (ionizing radiation, IR) of cancer is the emergence of radioresistant cells. We report that Laboratory of Genetics and Physiology 2 (LGP2), a resident RIG-I (retinoic acid inducible gene I)–like receptor protein, can induce radioresistance. IR induces interferon and stimulates accumulation of LGP2. In turn, LGP2 shuts off the synthesis of interferon and blocks its cytotoxic effects. Ectopic expression of LGP2 enhances resistance to IR, whereas depletion enhances cytotoxic effects of IR. Here we show that LGP2 is associated with radioresistance in numerous diverse cancer cell lines. Examination of available databases links expression of LGP2 with poor prognosis in cancer patients. An siRNA screen targeting 89 IFN stimulated genes in 14 different cancer cell lines pointed to the RIG-I (retinoic acid inducible gene I)–like receptor Laboratory of Genetics and Physiology 2 (LGP2) as playing a key role in conferring tumor cell survival following cytotoxic stress induced by ionizing radiation (IR). Studies on the role of LGP2 revealed the following: (i) Depletion of LGP2 in three cancer cell lines resulted in a significant increase in cell death following IR, (ii) ectopic expression of LGP2 in cells increased resistance to IR, and (iii) IR enhanced LGP2 expression in three cell lines tested. Studies designed to define the mechanism by which LGP2 acts point to its role in regulation of IFNβ. Specifically (i) suppression of LGP2 leads to enhanced IFNβ, (ii) cytotoxic effects following IR correlated with expression of IFNβ inasmuch as inhibition of IFNβ by neutralizing antibody conferred resistance to cell death, and (iii) mouse embryonic fibroblasts from IFN receptor 1 knockout mice are radioresistant compared with wild-type mouse embryonic fibroblasts. The role of LGP2 in cancer may be inferred from cumulative data showing elevated levels of LGP2 in cancer cells are associated with more adverse clinical outcomes. Our results indicate that cytotoxic stress exemplified by IR induces IFNβ and enhances the expression of LGP2. Enhanced expression of LGP2 suppresses the IFN stimulated genes associated with cytotoxic stress by turning off the expression of IFNβ.


Molecular Therapy | 2017

In Vivo Delivery and Therapeutic Effects of a MicroRNA on Colorectal Liver Metastases

Go Oshima; Nining Guo; Chunbai He; Melinda E. Stack; Christopher Poon; Abhineet Uppal; Sean C. Wightman; A. Parekh; Kinga B. Skowron; Mitchell C. Posner; Wenbin Lin; Nikolai N. Khodarev; Ralph R. Weichselbaum

Multiple therapeutic agents are typically used in concert to effectively control metastatic tumors. Recently, we described microRNAs that are associated with the oligometastatic state, in which a limited number of metastatic tumors progress to more favorable outcomes. Here, we report the effective delivery of an oligometastatic microRNA (miR-655-3p) to colorectal liver metastases using nanoscale coordination polymers (NCPs). The NCPs demonstrated a targeted and prolonged distribution of microRNAs to metastatic liver tumors. Tumor-targeted microRNA miR-655-3p suppressed tumor growth when co-delivered with oxaliplatin, suggesting additive or synergistic interactions between microRNAs and platinum drugs. This is the first known example of systemically administered nanoparticles delivering an oligometastatic microRNA to advanced metastatic liver tumors and demonstrating tumor-suppressive effects. Our results suggest a potential therapeutic strategy for metastatic liver disease by the co-delivery of microRNAs and conventional cytotoxic agents using tumor-specific NCPs.


Molecular Cancer Therapeutics | 2018

JAK2 Inhibitor SAR302503 Abrogates PD-L1 Expression and Targets Therapy-Resistant Non–small Cell Lung Cancers

Sean P. Pitroda; Melinda E. Stack; Gene-Fu Liu; Sui-Sui Song; Lucy F. Chen; Hua Liang; A. Parekh; Xiaona Huang; Paul Roach; Mitchell C. Posner; Ralph R. Weichselbaum; Nikolai N. Khodarev

Lung cancer is the leading cause of cancer-related deaths worldwide. Approximately 85% of all lung cancers are non–small cell histology [non–small cell lung cancer (NSCLC)]. Modern treatment strategies for NSCLC target driver oncogenes and immune checkpoints. However, less than 15% of patients survive beyond 5 years. Here, we investigated the effects of SAR302503 (SAR), a selective JAK2 inhibitor, on NSCLC cell lines and tumors. We show that SAR is cytotoxic to NSCLC cells, which exhibit resistance to genotoxic therapies, such as ionizing radiation, cisplatin, and etoposide. We demonstrate that constitutive IFN-stimulated gene expression, including an IFN-related DNA damage resistance signature, predicts for sensitivity to SAR. Importantly, tumor cell–intrinsic expression of PD-L1 is IFN-inducible and abrogated by SAR. Taken together, these findings suggest potential dual roles for JAK2 inhibitors, both as a novel monotherapy in NSCLCs resistant to genotoxic therapies, and in tandem with immune checkpoint inhibition. Mol Cancer Ther; 17(4); 732–9. ©2018 AACR.


Advances in radiation oncology | 2018

Late toxicity after post-prostatectomy intensity modulated radiation therapy: Evaluating normal-tissue sparing guidelines

Adil S. Akthar; Anthony C. Wong; A. Parekh; Greg Hubert; Christina H. Son; Charles A. Pelizzari; Stanley L. Liauw

Purpose Dose-volume histogram (DVH) toxicity relationships are poorly defined in men who receive radiation after radical prostatectomy (RP). We evaluated Radiation Therapy Oncology Group (RTOG) study 0534 and institutional intact normal-tissue sparing guidelines, as well as dose to bladder trigone, for ability to minimize late toxicity. Methods and materials 164 men received intensity modulated radiation therapy (RT) to a median prostate bed dose of 66.6 Gy at a median of 22 months after RP. 46% of men were prescribed androgen deprivation therapy and pelvic lymph node irradiation to a median dose of 50.4 Gy. DVH relationships for the rectum, bladder, trigone, and bladder excluding the clinical target volume (bladder-CTV) were analyzed against the Common Terminology Criteria for Adverse Events late grade 2 + (G2+) gastrointestinal (GI) and genitourinary (GU) toxicity by log-rank test. RTOG 0534 (rectum V65, 40 Gy ≤35, 55%, and bladder-CTV V65, 40 ≤50, 70%) and intact prostate RT institutional guidelines (rectum V70, 65, 40 ≤20, 40, 80% and bladder V70, 65, 40 ≤30, 60, 80%, respectively) guidelines were evaluated. Results With a median follow-up time of of 33 months, the 4-year freedom from G2 + GI and GU toxicity were both 91%. G2 + GI (n = 12) and GU (n = 15) toxicity included 4% diarrhea (n = 6), 4% hemorrhage (n = 6), 1% proctitis (n = 1), and 4% urinary frequency (n = 7), 1% obstructive (n = 2), 2% cystitis (n = 3), and 3% incontinence (n = 5), respectively. RTOG 0534 rectum and bladder goals were not achieved in 65% and 41% of cases, while the institutional intact prostate goals were not achieved in 21% and 25% of cases, respectively. Neither dose to the bladder trigone nor any of the proposed normal tissue goals were associated with late toxicity (P > .1). In the univariate analysis, age, pelvic RT, RT dose, anticoagulation use, androgen deprivation therapy, time from RP to RT, and tobacco history were not associated with toxicity. Conclusions More than 90% of men were free from late G2 + toxicity 4 years after post-RP intensity modulated RT. No tested parameters were associated with late toxicity. In the absence of established normal-tissue DVH guidelines in the postoperative setting, the use of intact guidelines is reasonable.


International Journal of Radiation Oncology Biology Physics | 2015

Late Toxicity After Post Prostatectomy Radiation Therapy: Evaluating RTOG 0534 Tissue-Sparing Guidelines

A. Parekh; Christina H. Son; J.M. Melotek; C Pelizzari; Greg Hubert; Stanley L. Liauw


International Journal of Radiation Oncology Biology Physics | 2016

Does Regional Radiation Therapy Impact Outcome for High-Risk, Node-Positive Cutaneous Melanoma?

A. Parekh; T. Strom; A.O. Naghavi; Jimmy J. Caudell; Jonathan S. Zager; Jane L. Messina; Javier F. Torres-Roca; Vernon K. Sondak; Andy Trotti; Louis B. Harrison


International Journal of Radiation Oncology Biology Physics | 2014

RIG-1-Like Receptor LGP2 Protects Tumor Cells From Ionizing Radiation

Sean P. Pitroda; Ryan C. Widau; A. Parekh; Daniel W. Golden; M.C. Ranck; Ravi F. Sood; Xiaona Huang; Thomas E. Darga; Bernard Roizman; Nikolai N. Khodarev; Ralph R. Weichselbaum


International Journal of Radiation Oncology Biology Physics | 2013

No Difference in Late Toxicity With Postoperative Radiation Therapy Following Open or Robotic Prostatectomy

A. Parekh; M.C. Ranck; S.K. Chennupati; Stanley L. Liauw

Collaboration


Dive into the A. Parekh's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge