Network


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

Hotspot


Dive into the research topics where Dhong Hyun Lee is active.

Publication


Featured researches published by Dhong Hyun Lee.


International Journal of Cancer | 2015

Synergistic effect of JQ1 and rapamycin for treatment of human osteosarcoma

Dhong Hyun Lee; Jun Qi; James E. Bradner; Jonathan W. Said; Ngan Doan; Charles Forscher; Henry Yang; H. Phillip Koeffler

Bromodomain and extra terminal domain (BET) proteins are important epigenetic regulators facilitating the transcription of genes in chromatin areas linked to acetylated histones. JQ1, a BET protein inhibitor, has antiproliferative activity against many cancers, mainly through inhibition of c‐MYC and upregulation of p21. In this research, we investigated the use of JQ1 for human osteosarcoma (OS) treatment. JQ1 significantly inhibited the proliferation and survival of OS cells inducing G1 cell cycle arrest, premature senescence, but little effect on apoptosis. Interestingly, c‐MYC protein levels in JQ1‐treated cells remained unchanged, whereas the upregulation of p21 protein was still observable. Although effective in vitro, JQ1 alone failed to reduce the size of the MNNG/HOS xenografts in immunocompromised mice. To overcome the resistance of OS cells to JQ1 treatment, we combined JQ1 with rapamycin, an mammalian target of rapamycin (mTOR) inhibitor. JQ1 and rapamycin synergistically inhibited the growth and survival of OS cells in vitro and in vivo. We also identified that RUNX2 is a direct target of bromodomain‐containing protein 4 (BRD4) inhibition by JQ1 in OS cells. Chromatin immunoprecipitation (ChIP) showed that enrichment of BRD4 protein around RUNX2 transcription start sites diminished with JQ1 treatment in MNNG/HOS cells. Overexpression of RUNX2 protected JQ1‐sensitive OS cells from the effect of JQ1, and siRNA‐mediated inhibition of RUNX2 sensitized the same cells to JQ1. In conclusion, our findings suggest that JQ1, in combination with rapamycin, is an effective chemotherapeutic option for OS treatment. We also show that inhibition of RUNX2 expression by JQ1 partly explains the antiproliferative activity of JQ1 in OS cells.


British Journal of Pharmacology | 2010

Cucurbitacin B, a novel in vivo potentiator of gemcitabine with low toxicity in the treatment of pancreatic cancer

Gabriela B. Iwanski; Dhong Hyun Lee; Shlomit En-Gal; Ngan Bao Doan; Brandon Castor; Marco Vogt; Melvin Toh; Carsten Bokemeyer; Jonathan W. Said; Nils H. Thoennissen; H. Phillip Koeffler

Background and purpose:  Pancreatic cancer is a highly aggressive malignancy, and improvement in systemic therapy is necessary to treat this frequently encountered metastatic disease. The current targeted agents used in combination with gemcitabine improved objective response rates, but with little or no improvements in survival and also increased toxicities in pancreatic cancer patients. Recently, we showed that the triterpenoid cucurbitacin B inhibited tumour growth in pancreatic cancer cells by inhibition of the JAK/STAT pathway, and synergistically increased antiproliferative effects of gemcitabine in vitro.


Molecular Carcinogenesis | 2014

Growth inhibition of pancreatic cancer cells by histone deacetylase inhibitor belinostat through suppression of multiple pathways including HIF, NFkB, and mTOR signaling in vitro and in vivo

Wenwen Chien; Dhong Hyun Lee; Yun Zheng; Peer Wuensche; Rosie Alvarez; Ding Ling Wen; Ahmed M. Aribi; Su Ming Thean; Ngan Doan; Jonathan W. Said; H. Phillip Koeffler

Pancreatic ductal adenocarcinoma is a devastating disease with few therapeutic options. Histone deacetylase inhibitors are a novel therapeutic approach to cancer treatment; and two new pan‐histone deacetylase inhibitors (HDACi), belinostat and panobinostat, are undergoing clinical trials for advanced hematologic malignancies, non‐small cell lung cancers and advanced ovarian epithelial cancers. We found that belinostat and panobinostat potently inhibited, in a dose‐dependent manner, the growth of six (AsPc1, BxPc3, Panc0327, Panc0403, Panc1005, MiaPaCa2) of 14 human pancreatic cancer cell lines. Belinostat increased the percentage of apoptotic pancreatic cancer cells and caused prominent G2/M growth arrest of most pancreatic cancer cells. Belinostat prominently inhibited PI3K‐mTOR‐4EBP1 signaling with a 50% suppression of phorphorylated 4EBP1 (AsPc1, BxPc3, Panc0327, Panc1005 cells). Surprisingly, belinostat profoundly blocked hypoxia signaling including the suppression of hypoxia response element reporter activity; as well as an approximately 10‐fold decreased transcriptional expression of VEGF, adrenomedullin, and HIF1α at 1% compared to 20% O2. Treatment with this HDACi decreased levels of thioredoxin mRNA associated with increased levels of its endogenous inhibitor thioredoxin binding protein‐2. Also, belinostat alone and synergistically with gemcitabine significantly (P = 0.0044) decreased the size of human pancreatic tumors grown in immunodeficiency mice. Taken together, HDACi decreases growth, increases apoptosis, and is associated with blocking the AKT/mTOR pathway. Surprisingly, it blocked hypoxic growth related signals. Our studies of belinostat suggest it may be an effective drug for the treatment of pancreatic cancers when used in combination with other drugs such as gemcitabine.


International Journal of Cancer | 2013

The triterpenoid cucurbitacin B augments the antiproliferative activity of chemotherapy in human breast cancer

Ahmed M. Aribi; Sigal Gery; Dhong Hyun Lee; Nils H. Thoennissen; Gabriela B. Thoennissen; Rocio Alvarez; Quoc Ho; Kunik Lee; Ngan Doan; Kin Tak Chan; Melvin Toh; Jonathan W. Said; H. Phillip Koeffler

Despite recent advances in therapy, breast cancer remains the second most common cause of death from malignancy in women. Chemotherapy plays a major role in breast cancer management, and combining chemotherapeutic agents with nonchemotherapeutic agents is of considerable clinical interest. Cucurbitacins are triterpenes compounds found in plants of the Cucurbitaceae family, reported to have anticancer and anti‐inflammatory activities. Previously, we have shown antiproliferative activity of cucurbitacin B (CuB) in breast cancer, and we hypothesized that combining CuB with chemotherapeutic agents can augment their antitumor effect. Here, we show that a combination of CuB with either docetaxel (DOC) or gemcitabine (GEM) synergistically inhibited the proliferation of MDA‐MB‐231 breast cancer cells in vitro. This antiproliferative effect was accompanied by an increase in apoptosis rates. Furthermore, in vivo treatment of human breast cancer orthotopic xenografts in immunodeficient mice with CuB at either low (0.5 mg/kg) or high (1 mg/kg) doses in combination with either DOC (20 mg/kg) or GEM (12.5mg/kg) significantly reduced tumor volume as compared with monotherapy of each drug. Importantly, no significant toxicity was noted with low‐dose CuB in combination with either DOC or GEM. In conclusion, combination of CuB at a relatively low concentration with either of the chemotherapeutic agents, DOC or GEM, shows prominent antiproliferative activity against breast cancer cells without increased toxicity. This promising combination should be examined in therapeutic trials of breast cancer.


RNA Biology | 2017

High-throughput sequencing of two populations of extracellular vesicles provides an mRNA signature that can be detected in the circulation of breast cancer patients

Andrew Conley; Valentina R. Minciacchi; Dhong Hyun Lee; Beatrice Knudsen; Beth Y. Karlan; Luigi Citrigno; Giuseppe Viglietto; Muneesh Tewari; Michael R. Freeman; Francesca Demichelis; Dolores Di Vizio

ABSTRACT Extracellular vesicles (EVs) contain a wide range of RNA types with a reported prevalence of non-coding RNA. To date a comprehensive characterization of the protein coding transcripts in EVs is still lacking. We performed RNA-Sequencing (RNA-Seq) of 2 EV populations and identified a small fraction of transcripts that were expressed at significantly different levels in large oncosomes and exosomes, suggesting they may mediate specialized functions. However, these 2 EV populations exhibited a common mRNA signature that, in comparison to their donor cells, was significantly enriched in mRNAs encoding E2F transcriptional targets and histone proteins. These mRNAs are primarily expressed in the S-phase of the cell cycle, suggesting that they may be packaged into EVs during S-phase. In silico analysis using subcellular compartment transcriptome data from the ENCODE cell line compendium revealed that EV mRNAs originate from a cytoplasmic RNA pool. The EV signature was independently identified in plasma of patients with breast cancer by RNA-Seq. Furthermore, several transcripts differentially expressed in EVs from patients versus controls mirrored differential expression between normal and breast cancer tissues. Altogether, this largest high-throughput profiling of EV mRNA demonstrates that EVs carry tumor-specific alterations and can be interrogated as a source of cancer-derived cargo.


Journal of Virology | 2017

Roles of M1 and M2 Macrophages in Herpes Simplex Virus 1 Infectivity

Dhong Hyun Lee; Homayon Ghiasi

ABSTRACT Macrophages are the predominant infiltrate in the corneas of mice that have been ocularly infected with herpes simplex virus 1 (HSV-1). However, very little is known about the relative roles of M1 (classically activated or polarized) and M2 (alternatively activated or polarized) macrophages in ocular HSV-1 infection. To better understand these relationships, we assessed the impact of directed M1 or M2 activation of RAW264.7 macrophages and peritoneal macrophages (PM) on subsequent HSV-1 infection. In both the RAW264.7 macrophage and PM in vitro models, HSV-1 replication in M1 macrophages was markedly lower than in M2 macrophages and unstimulated controls. The M1 macrophages expressed significantly higher levels of 28 of the 32 tested cytokines and chemokines than M2 macrophages, with HSV-1 infection significantly increasing the levels of proinflammatory cytokines and chemokines in the M1 versus the M2 macrophages. To examine the effects of shifting the immune response toward either M1 or M2 macrophages in vivo, wild-type mice were injected with gamma interferon (IFN-γ) DNA or colony-stimulating factor 1 (CSF-1) DNA prior to ocular infection with HSV-1. Virus replication in the eye, latency in trigeminal ganglia (TG), and markers of T cell exhaustion in the TG were determined. We found that injection of mice with IFN-γ DNA, which enhances the development of M1 macrophages, increased virus replication in the eye; increased latency; and also increased CD4, CD8, IFN-γ, and PD-1 transcripts in the TG of latently infected mice. Conversely, injection of mice with CSF-1 DNA, which enhances the development of M2 macrophages, was associated with reduced virus replication in the eye and reduced latency and reduced the levels of CD4, CD8, IFN-γ,and PD-1 transcripts in the TG. Collectively, these results suggest that M2 macrophages directly reduce the levels of HSV-1 latency and, thus, T-cell exhaustion in the TG of ocularly infected mice. IMPORTANCE Our findings demonstrate a novel approach to further reducing HSV-1 replication in the eye and latency in the TG by modulating immune components, specifically, by altering the phenotype of macrophages. We suggest that inclusion of CSF-1 as part of any vaccination regimen against HSV infection to coax responses of macrophages toward an M2, rather than an M1, response may further improve vaccine efficacy against ocular HSV-1 replication and latency.


PLOS Pathogens | 2017

Suppression of IL-12p70 formation by IL-2 or following macrophage depletion causes T-cell autoreactivity leading to CNS demyelination in HSV-1-infected mice

Dhong Hyun Lee; Mandana Zandian; Jane Kuo; Kevin R. Mott; Shuang Chen; Moshe Arditi; Homayon Ghiasi

We have established two mouse models of central nervous system (CNS) demyelination that differ from most other available models of multiple sclerosis (MS) in that they represent a mixture of viral and immune triggers. In the first model, ocular infection of different strains of mice with a recombinant HSV-1 that expresses murine IL-2 constitutively (HSV-IL-2) causes CNS demyelination. In the second model, depletion of macrophages causes CNS demyelination in mice that are ocularly infected with wild-type (WT) HSV-1. In the present study, we found that the demyelination in macrophage-intact mice infected with HSV-IL-2 was blocked by depletion of FoxP3-expressing cells, while concurrent depletion of macrophages restored demyelination. In contrast, demyelination was blocked in the macrophage-depleted mice infected with wild-type HSV-1 following depletion of FoxP3-expressing cells. In macrophage-depleted HSV-IL-2-infected mice, demyelination was associated with the activity of both CD4+ and CD8+ T cells, whereas in macrophage-depleted mice infected with WT HSV-1, demyelination was associated with CD4+ T cells. Macrophage depletion or infection with HSV-IL-2 caused an imbalance of T cells and TH1 responses as well as alterations in IL-12p35 and IL-12p40 but not other members of the IL-12 family or their receptors. Demyelination was blocked by adoptive transfer of macrophages that were infected with HSV-IL-12p70 or HSV-IL-12p40 but not by HSV-IL-12p35. These results indicate that suppression of IL-12p70 formation by IL-2 or following macrophage depletion causes T-cell autoreactivity leading to CNS demyelination in HSV-1-infected mice.


Biomedicine & Pharmacotherapy | 2018

Co-targeting poly(ADP-ribose) polymerase (PARP) and histone deacetylase (HDAC) in triple-negative breast cancer: Higher synergism in BRCA mutated cells

Hélène Marijon; Dhong Hyun Lee; Ling-Wen Ding; Haibo Sun; Sigal Gery; Aimery de Gramont; H. Phillip Koeffler

PURPOSE Despite similarities with BRCA-mutated breast cancers, triple-negative breast cancers (TNBC) remain resistant to poly(ADP-ribose) polymerase (PARP) inhibitors as single agents. Histone deacetylase inhibitors (HDACi) can decrease expression of proteins involved in DNA repair. We thus hypothesized that a HDACi (suberoylanilide hydroxamic acid (SAHA) or belinostat) could sensitize TNBC to the PARP inhibitor olaparib. METHODS Human TNBC cells were co-treated with olaparib and either SAHA or belinostat, and their effects on survival, proliferation, cell cycle, apoptosis and DNA repair pathways were evaluated. Subcutaneous xenografts were used to determine the effect of the combination treatment in vivo. RESULTS HDACi and olaparib synergistically inhibited proliferation of a panel of 8 TNBC cell lines in vitro and in nude mice harboring TNBC xenografts in vivo. We noted a weaker synergism in PTEN-deficient TNBC cells and a stronger synergism in BRCA1-mutated TNBC cells. In the BRCA1-mutated cell line HCC-1937, we observed a drastic decrease in the expression of proteins involved in homologous recombination (HR), leading to a large imbalance of the ratio P-H2AX/RAD51. In BRCA1 wild type (wt) cell lines, effect of the combination treatment relied on DNA damage-induced cell cycle arrest followed by induction of apoptosis. CONCLUSION In summary, these results provide a preclinical rationale to combine a HDACi with a PARP inhibitor to reduce HR efficiency in TNBC and sensitize these aggressive tumors to PARP inhibition.


Journal of Virology | 2017

Reply to “Herpes Simplex Virus 1, Macrophages, and the Cornea”

Dhong Hyun Lee; Homayon Ghiasi

In a letter to the editor regarding our report in the Journal of Virology (JVI) ([1][1]), several issues were raised by Daniel Carr, and our responses to his points are as follows. (i) Dr. Carr, in support of his comments, cites papers by Tumpey et al. ([2][2]) and Thomas et al. ([3][3]). However,


Cancer Research | 2014

Abstract 1689: Synergistic effect of JQ1 and rapamycin for treatment of human osteosarcoma

Dhong Hyun Lee; Jun Qi; James E. Bradner; Jonathan W. Said; Ngan Doan; Charles Forscher; H. Phillip Koeffler

Bromodomain and extra terminal domain (BET) proteins are important epigenetic regulators facillitating the transcription of genes in chromatin areas linked to acetylated histones. JQ1, a BET protein inhibitor, has antiproliferative activity againts many hematological and solid cancers, mainly through inhibition of c-myc and upregulation of p21. In this research, we investigated the use of JQ1 for human osteosarcoma (OS) treatment, due to the frequent copy number gain and overexpression of c-myc in OS cells. JQ1 significantly inhibited the proliferation and survival of seven OS cells (U2OS, G292, MG-63, HT-161, MNNG/HOS, SAOS-2, SJSA), at a relatively higher dose than other cancers (median IC50 = 7.35 μM). JQ1 induced G1 cell cycle arrest and premature senescence of OS cells, but produced little apoptosis. Interestingly, c-myc protein levels in JQ1-treated cells remained unchanged, whereas the upregulation of p21 protein was still observable, as noted by Western blots. Although effective in vitro, JQ1 failed to reduce the size of the MNNG/HOS xenografts in immunocompromised mice in vivo. To overcome the resistance of OS cells to JQ1 treatment, we tested JQ1 in combination with rapamycin. Rapamycin inhibits the assembly of mTOR complexes which is one of the important growth signaling pathways in many cancers including OS. JQ1 and rapamycin synergistically inhibited the growth and survival of OS cells in vitro. The drug combination nearly abrogated expression of c-myc protein, enhanced levels of p21, and induced apoptosis in OS cells in vitro. After 24 days of treatment in vivo, JQ1 (50mg/kg body weight) or rapamycin (1.5 mg/kg body weight) alone showed little effect on the size of human OS xenografts in nude mice. However, combination of both drugs significantly inhibited tumor growth by 85% and 73% (p In conclusion, JQ1 inhibited proliferation and survival of OS cells in vitro but failed to reduce growth of OS xenografts in immunodeficient mice. When combined with rapamycin, JQ1 synergistically inhibited proliferation of OS cells both in vitro and in vivo. Citation Format: Dhong Hyun Tony Lee, Jun Qi, James Bradner, Jonathan Said, Ngan Doan, Charles Forscher, H Phillip Koeffler. Synergistic effect of JQ1 and rapamycin for treatment of human osteosarcoma. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1689. doi:10.1158/1538-7445.AM2014-1689

Collaboration


Dive into the Dhong Hyun Lee's collaboration.

Top Co-Authors

Avatar

H. Phillip Koeffler

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ngan Doan

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Catherine Goff

University of California

View shared research outputs
Top Co-Authors

Avatar

Charles Forscher

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Homayon Ghiasi

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ahmed M. Aribi

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge