Network


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

Hotspot


Dive into the research topics where Esther H. Chang is active.

Publication


Featured researches published by Esther H. Chang.


Human Gene Therapy | 1999

Transferrin- Liposome-Mediated Systemic p53 Gene Therapy in Combination with Radiation Results in Regression of Human Head and Neck Cancer Xenografts

Liang Xu; Kathleen F. Pirollo; Wenhua Tang; Antonina Rait; Esther H. Chang

The use of cationic liposomes as nonviral vehicles for the delivery of therapeutic molecules is becoming increasingly prevalent in the field of gene therapy. We have previously demonstrated that the use of the transferrin ligand (Tf) to target a cationic liposome delivery system resulted in a significant increase in the transfection efficiency of the complex [Xu, L., Pirollo, K.F., and Chang, E.H. (1997). Hum. Gene Ther. 8, 467-475]. Delivery of wild-type (wt) p53 to a radiation-resistant squamous cell carcinoma of the head and neck (SCCHN) cell line via this ligand-targeted, liposome complex was also able to revert the radiation resistant phenotype of these cells in vitro. Here we optimized the Tf/liposome/DNA ratio of the complex (LipT) for maximum tumor cell targeting, even in the presence of serum. The efficient reestablishment of wtp53 function in these SCCHN tumor cells in vitro, via the LipT complex, restored the apoptotic pathway, resulting in a significant increase in radiation-induced apoptosis that was directly proportional to the level of exogenous wtp53 in the tumor cells. More significantly, intravenous administration of LipT-p53 markedly sensitized established SCCHN nude mouse xenograft tumors to radiotherapy. The combination of systemic LipT-p53 gene therapy and radiation resulted in complete tumor regression and inhibition of their recurrence even 6 months after the end of all treatment. These results indicate that this tumor-specific, ligand-liposome delivery system for p53 gene therapy, when used in concert with conventional radiotherapy, can provide a new and more effective means of cancer treatment.


Cancer Research | 2008

Targeted Delivery of Small Interfering RNA : Approaching Effective Cancer Therapies

Kathleen F. Pirollo; Esther H. Chang

Three of the primary requirements for the development of effective dual-targeting therapeutic modalities for the treatment of cancer are the tumor-targeted delivery of the therapeutic molecules of interest to the tumor site(s) in the body (both primary and metastatic), passage of the molecular therapeutic through the cell membrane, and targeting specifically a growth or apoptotic pathway. However, lack of efficient targeted delivery, low transfection efficiency, instability to nucleases, poor tissue penetration, and nonspecific immune stimulation have hindered the translation of small interfering RNA (siRNA) into clinical applications. The development of a systemically administered, tumor-specific immunoliposome nanocomplex with high transfection efficiency could overcome these limitations and thus realize the potential of siRNAs to become effective anticancer clinical modalities.


Cancer Research | 2007

Materializing the Potential of Small Interfering RNA via a Tumor-Targeting Nanodelivery System

Kathleen F. Pirollo; Antonina Rait; Qi Zhou; Sung Hee Hwang; John A. Dagata; Gerald Zon; Richard I. Hogrefe; Guillermo Palchik; Esther H. Chang

The field of small interfering RNA (siRNA) as potent sequence-selective inhibitors of transcription is rapidly developing. However, until now, low transfection efficiency, poor tissue penetration, and nonspecific immune stimulation by in vivo administered siRNAs have delayed their therapeutic application. Their potential as anticancer therapeutics hinges on the availability of a vehicle that can be systemically administered, safely and repeatedly, and will deliver the siRNA specifically and efficiently to the tumor, both primary tumors and metastases. We have developed a nanosized immunoliposome-based delivery complex (scL) that, when systemically administered, will preferentially target and deliver molecules useful in gene medicine, including plasmid DNA and antisense oligonucleotides, to tumor cells wherever they occur in the body. This tumor-targeting nanoparticle delivery vehicle can also deliver siRNA to both primary and metastatic disease. We have also enhanced the efficiency of this complex by the inclusion of a pH-sensitive histidine-lysine peptide in the complex (scL-HoKC) and by delivery of a modified hybrid (DNA-RNA) anti-HER-2 siRNA molecule. Scanning probe microscopy confirms that this modified complex maintains its nanoscale size. More importantly, we show that this nanoimmunoliposome anti-HER-2 siRNA complex can sensitize human tumor cells to chemotherapeutics, silence the target gene and affect its downstream pathway components in vivo, and significantly inhibit tumor growth in a pancreatic cancer model. Thus, this complex has the potential to help translate the potent effects of siRNA into a clinically viable anticancer therapeutic.


ACS Nano | 2014

A nanoparticle carrying the p53 gene targets tumors including cancer stem cells, sensitizes glioblastoma to chemotherapy and improves survival.

Sang-Soo Kim; Antonina Rait; Eric P. Kim; Kathleen F. Pirollo; Maki Nishida; Natalia Farkas; John A. Dagata; Esther H. Chang

Temozolomide (TMZ)-resistance in glioblastoma multiforme (GBM) has been linked to upregulation of O6-methylguanine-DNA methyltransferase (MGMT). Wild-type (wt) p53 was previously shown to down-modulate MGMT. However, p53 therapy for GBM is limited by lack of efficient delivery across the blood brain barrier (BBB). We have developed a systemic nanodelivery platform (scL) for tumor-specific targeting (primary and metastatic), which is currently in multiple clinical trials. This self-assembling nanocomplex is formed by simple mixing of the components in a defined order and a specific ratio. Here, we demonstrate that scL crosses the BBB and efficiently targets GBM, as well as cancer stem cells (CSCs), which have been implicated in recurrence and treatment resistance in many human cancers. Moreover, systemic delivery of scL-p53 down-modulates MGMT and induces apoptosis in intracranial GBM xenografts. The combination of scL-p53 and TMZ increased the antitumor efficacy of TMZ with enhanced survival benefit in a mouse model of highly TMZ-resistant GBM. scL-p53 also sensitized both CSCs and bulk tumor cells to TMZ, increasing apoptosis. These results suggest that combining scL-p53 with standard TMZ treatment could be a more effective therapy for GBM.


The Journal of Urology | 1993

Expression of the c-erbB-2 (Her-2/neu) Oncoprotein in Human Prostatic Carcinoma

Eric J. Kuhn; Richard A. Kurnot; Isabell A. Sesterhenn; Esther H. Chang; Judd W. Moul

The objective of this study was to determine the expression of the c-erB-2 oncoprotein via immunohistochemistry of archival clinically localized human prostate cancers and to compare these results to known clinical prognostic factors. In addition, positive staining cases were subjected to differential polymerase chain reaction to assess for c-erbB-2 gene amplification. Immunohistochemical staining with a polyclonal antibody (pAb 1) was performed on archival radical prostatectomy specimens. To standardize the staining, positive and negative control material was generated using c-erbB-2 transfected NIH3T3 cells grown on agar plugs, formalin fixed, paraffin embedded and processed on glass slides for immunohistochemistry. Definite positive membranous staining was detected in 18 of 53 neoplastic cases (34%). In addition, 9 cases of benign prostatic hyperplasia were stained without evidence of c-erbB-2 expression detected. Either focal or diffuse membranous staining was identified in 6 of 27 (22%) well, 8 of 20 (40%) moderately and 4 of 6 (66%) poorly differentiated tumors (p = 0.03, chi-square test for trend). Positive staining occurred in 6 of 18 patients (33%) with pathological stage B and 12 of 33 (36%) with pathological stage C disease. At a mean of 36 months, complete followup was available for 16 of the 18 positive cases and 30 of the 35 negative cases. For stage B 1 of 6 positive (16.7%) versus 1 of 12 negative (8%) staining cases showed progression (p = 1.0). For stage C 7 of 12 positive (58.3%) versus 9 of 21 negative (42.9%) cases showed progression (p = 0.48). Deoxyribonucleic acid was extracted from the exact same archival paraffin blocks for the c-erbB-2 protein positive cases and subjected to differential polymerase chain reaction analysis, which revealed no c-erbB-2 gene amplification. This study demonstrates that approximately a third of all clinically localized prostate cancers express the c-erbB-2 oncoprotein via immunohistochemistry using pAb-1 on archival material, c-erbB-2 oncoprotein expression does not appear to be a prognostic marker for prostate cancer although our results are preliminary and, although oncoprotein expression was detected, no positive case demonstrated deoxyribonucleic acid amplification.


Oncogene | 1997

p53 mediated sensitization of squamous cell carcinoma of the head and neck to radiotherapy

Kathleen F. Pirollo; Zhengmei Hao; Antonina Rait; Yng-Ju Jang; Willard E. Fee; Patricia Ryan; Yawen Chiang; Esther H. Chang

Radiation resistant squamous cell carcinoma of the head and neck cell line JSQ-3 carries a mutant form of tumor suppressor gene p53. Treatment of these cells with an adenoviral vector containing wild-type p53 (Av1p53) was able to inhibit their growth in vitro and in vivo while having no effect on normal cells. More significantly, introduction of wtp53 also reduced the radiation-resistance level of this cell line in vitro, in a viral dose-dependent manner. Furthermore, this radiosensitization also carried over to the in vivo situation where the response of JSQ-3 cell-induced mouse xenografts to radiotherapy was markedly enhanced after treatment with Av1p53. Complete, long-term regression of the tumors for up to 162 days was observed when a single dose of Av1p53 was administered in combination with ionizing radiation, demonstrating the effectiveness of this combination of gene therapy and conventional radiotherapy. This sensitization of tumors to radiation therapy by replacement of wtp53 could significantly decrease the rate of recurrence after radiation treatment. Since radiation is one of the most prevalent forms of adjunctive therapy for a variety of cancers, these results have great relevance in moving toward an improved cancer therapy.


Human Gene Therapy | 2002

Self-Assembly of a Virus-Mimicking Nanostructure System for Efficient Tumor-Targeted Gene Delivery

Liang Xu; Peter Frederik; Kathleen F. Pirollo; Wenhua Tang; Antonina Rait; Lai-Man Xiang; Weiqun Huang; Idalia Cruz; Yuzhi Yin; Esther H. Chang

Molecular therapy, including gene therapy, is a promising strategy for the treatment of human disease. However, delivery of molecular therapeutics efficiently and specifically to the target tissue remains a significant challenge. A human transferrin (Tf)-targeted cationic liposome-DNA complex, Tf-lipoplex, has shown high gene transfer efficiency and efficacy with human head and neck cancer in vitro and in vivo (Xu, L., Pirollo, K.F., Tang, W.H., Rait, A., and Chang, E.H. Hum. Gene Ther. 1999;10:2941-2952). Here we explore the structure, size, formation process, and structure-function relationships of Tf-lipoplex. We have observed Tf-lipoplex to have a highly compact structure, with a relatively uniform size of 50-90 nm. This nanostructure is novel in that it resembles a virus particle with a dense core enveloped by a membrane coated with Tf molecules spiking the surface. More importantly, compared with unliganded lipoplex, Tf-lipoplex shows enhanced stability, improved in vivo gene transfer efficiency, and long-term efficacy for systemic p53 gene therapy of human prostate cancer when used in combination with conventional radiotherapy. On the basis of our observations, we propose a multistep self-assembly process and Tf-facilitated DNA cocondensation model that may provide an explanation for the resultant small size and effectiveness of our nanostructural Tf-lipoplex system.


Anti-Cancer Drugs | 2000

Does p53 status influence tumor response to anticancer therapies

Kathleen F. Pirollo; Kerrie B. Bouker; Esther H. Chang

Abnormalities in the tumor suppressor gene p53 have been identified in over 60% of human cancers. Since it plays such a pivotal role in cell growth regulation and apoptosis, the status of the p53 gene has been proposed as one of the major determinants of a tumors response to anticancer therapies. In this review we examine the relationship between functional p53 and sensitivity/resistance to both chemotherapy and radiotherapy, and discuss the potential use of some of the current gene therapy approaches to restore functional p53 to tumors as a means of modulating the effects of radiation and chemotherapy.


Radiation Research | 1993

Oncogene-Transformed NIH 3T3 Cells Display Radiation Resistance Levels Indicative of a Signal Transduction Pathway Leading to the Radiation-Resistant Phenotype

Kathleen F. Pirollo; Yue Ao Tong; Zulema Villegas; Yawen Chen; Esther H. Chang

Oncogenes and their normal counterparts, proto-oncogenes, are functionally important cellular genes which interact with one another as components of signal transduction pathways leading to cell growth and differentiation. Numerous reports in the literature have also begun to link these genes to the phenomenon of cellular radiation resistance. In this report we examine the radiation resistance level of NIH 3T3 cells transformed by various oncogenes in an attempt to define the intracellular pathway to the radiation-resistant phenotype. The results demonstrate that an analogous signaling pathway is apparently involved in acquisition of radiation resistance. Serine/threonine protein kinase oncogenes such as raf, mos, and PKC play a central role in the pathway. Moreover, specific oncogenes upstream (sis, HER-2, met, trk, and ras) and downstream (ets and myc) of these important signaling mediators can also influence the radiation resistance level of the cells.


Journal of General Virology | 1977

Interferon inhibits mouse leukaemia virus release: an electron microscope study.

Esther H. Chang; Sharon Mims; Timothy J. Triche; Robert M. Friedman

Scanning electron microscopy of AKR cells chronically infected with AKR mouse leukaemia virus revealed that the number of budding virions was greatly increased in interferon-treated cells. These results, together with previous biochemical findings, suggest that in this system, interferon inhibits a late stage of virus assembly or release.

Collaboration


Dive into the Esther H. Chang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Liang Xu

University of Kansas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shiv Srivastava

Uniformed Services University of the Health Sciences

View shared research outputs
Top Co-Authors

Avatar

Wenhua Tang

Georgetown University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Qi Zhou

Georgetown University

View shared research outputs
Top Co-Authors

Avatar

Cheng-Cheng Huang

Georgetown University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joe B. Harford

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John A. Dagata

National Institute of Standards and Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge