Network


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

Hotspot


Dive into the research topics where Charles Ng is active.

Publication


Featured researches published by Charles Ng.


Nature | 2011

RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E)

Poulikos I. Poulikakos; Yogindra Persaud; Manickam Janakiraman; Xiangju Kong; Charles Ng; Gatien Moriceau; Hubing Shi; Mohammad Atefi; Bjoern Titz; May Tal Gabay; Maayan Salton; Kimberly B. Dahlman; Madhavi Tadi; Jennifer A. Wargo; Keith T. Flaherty; Mark C. Kelley; Tom Misteli; Paul B. Chapman; Jeffrey A. Sosman; Thomas G. Graeber; Antoni Ribas; Roger S. Lo; Neal Rosen; David B. Solit

Activated RAS promotes dimerization of members of the RAF kinase family. ATP-competitive RAF inhibitors activate ERK signalling by transactivating RAF dimers. In melanomas with mutant BRAF(V600E), levels of RAS activation are low and these drugs bind to BRAF(V600E) monomers and inhibit their activity. This tumour-specific inhibition of ERK signalling results in a broad therapeutic index and RAF inhibitors have remarkable clinical activity in patients with melanomas that harbour mutant BRAF(V600E). However, resistance invariably develops. Here, we identify a new resistance mechanism. We find that a subset of cells resistant to vemurafenib (PLX4032, RG7204) express a 61-kDa variant form of BRAF(V600E), p61BRAF(V600E), which lacks exons 4–8, a region that encompasses the RAS-binding domain. p61BRAF(V600E) shows enhanced dimerization in cells with low levels of RAS activation, as compared to full-length BRAF(V600E). In cells in which p61BRAF(V600E) is expressed endogenously or ectopically, ERK signalling is resistant to the RAF inhibitor. Moreover, a mutation that abolishes the dimerization of p61BRAF(V600E) restores its sensitivity to vemurafenib. Finally, we identified BRAF(V600E) splicing variants lacking the RAS-binding domain in the tumours of six of nineteen patients with acquired resistance to vemurafenib. These data support the model that inhibition of ERK signalling by RAF inhibitors is dependent on levels of RAS–GTP too low to support RAF dimerization and identify a novel mechanism of acquired resistance in patients: expression of splicing isoforms of BRAF(V600E) that dimerize in a RAS-independent manner.


Nature Communications | 2012

Melanoma whole-exome sequencing identifies V600E B-RAF amplification-mediated acquired B-RAF inhibitor resistance

Hubing Shi; Gatien Moriceau; Xiangju Kong; Mi Kyung Lee; Hane Lee; Richard C. Koya; Charles Ng; Thinle Chodon; Richard A. Scolyer; Kimberly B. Dahlman; Jeffrey A. Sosman; Richard F. Kefford; Stanley F. Nelson; Antoni Ribas; Roger S. Lo

The development of acquired drug resistance hampers the long-term success of B-RAF inhibitor (B-RAFi) therapy for melanoma patients. Here we show V600EB-RAF copy number gain as a mechanism of acquired B-RAFi resistance in four out of twenty (20%) patients treated with B-RAFi. In cell lines, V600EB-RAF over-expression and knockdown conferred B-RAFi resistance and sensitivity, respectively. In V600EB-RAF amplification-driven (vs. mutant N-RAS-driven) B-RAFi resistance, ERK reactivation is saturable, with higher doses of vemurafenib down-regulating pERK and re-sensitizing melanoma cells to B-RAFi. These two mechanisms of ERK reactivation are sensitive to the MEK1/2 inhibitor AZD6244/selumetinib or its combination with the B-RAFi vemurafenib. In contrast to mutant N-RAS-mediated V600EB-RAF bypass, which is sensitive to C-RAF knockdown, V600EB-RAF amplification-mediated resistance functions largely independently of C-RAF. Thus, alternative clinical strategies may potentially overcome distinct modes of ERK reactivation underlying acquired B-RAFi resistance in melanoma.


Nature | 2014

Focused specificity of intestinal TH17 cells towards commensal bacterial antigens.

Yi Yang; Miriam B. Torchinsky; Michael Gobert; Huizhong Xiong; Mo Xu; Jonathan L. Linehan; Francis Alonzo; Charles Ng; Alessandra Chen; Xiyao Lin; Andrew Sczesnak; Jia Jun Liao; Victor J. Torres; Marc K. Jenkins; Juan J. Lafaille; Dan R. Littman

T-helper-17 (TH17) cells have critical roles in mucosal defence and in autoimmune disease pathogenesis. They are most abundant in the small intestine lamina propria, where their presence requires colonization of mice with microbiota. Segmented filamentous bacteria (SFB) are sufficient to induce TH17 cells and to promote TH17-dependent autoimmune disease in animal models. However, the specificity of TH17 cells, the mechanism of their induction by distinct bacteria, and the means by which they foster tissue-specific inflammation remain unknown. Here we show that the T-cell antigen receptor (TCR) repertoire of intestinal TH17 cells in SFB-colonized mice has minimal overlap with that of other intestinal CD4+ T cells and that most TH17 cells, but not other T cells, recognize antigens encoded by SFB. T cells with antigen receptors specific for SFB-encoded peptides differentiated into RORγt-expressing TH17 cells, even if SFB-colonized mice also harboured a strong TH1 cell inducer, Listeria monocytogenes, in their intestine. The match of T-cell effector function with antigen specificity is thus determined by the type of bacteria that produce the antigen. These findings have significant implications for understanding how commensal microbiota contribute to organ-specific autoimmunity and for developing novel mucosal vaccines.


PLOS ONE | 2011

Reversing Melanoma Cross-Resistance to BRAF and MEK Inhibitors by Co-Targeting the AKT/mTOR Pathway

Mohammad Atefi; Erika von Euw; Narsis Attar; Charles Ng; Connie Chu; Deliang Guo; Ramin Nazarian; Bartosz Chmielowski; John A. Glaspy; Begonya Comin-Anduix; Paul S. Mischel; Roger S. Lo; Antoni Ribas

Background The sustained clinical activity of the BRAF inhibitor vemurafenib (PLX4032/RG7204) in patients with BRAFV600 mutant melanoma is limited primarily by the development of acquired resistance leading to tumor progression. Clinical trials are in progress using MEK inhibitors following disease progression in patients receiving BRAF inhibitors. However, the PI3K/AKT pathway can also induce resistance to the inhibitors of MAPK pathway. Methodology/Principal Findings The sensitivity to vemurafenib or the MEK inhibitor AZD6244 was tested in sensitive and resistant human melanoma cell lines exploring differences in activation-associated phosphorylation levels of major signaling molecules, leading to the testing of co-inhibition of the AKT/mTOR pathway genetically and pharmacologically. There was a high degree of cross-resistance to vemurafenib and AZD6244, except in two vemurafenib-resistant cell lines that acquired a secondary mutation in NRAS. In other cell lines, acquired resistance to both drugs was associated with persistence or increase in activity of AKT pathway. siRNA-mediated gene silencing and combination therapy with an AKT inhibitor or rapamycin partially or completely reversed the resistance. Conclusions/Significance Primary and acquired resistance to vemurafenib in these in vitro models results in frequent cross resistance to MEK inhibitors, except when the resistance is the result of a secondary NRAS mutation. Resistance to BRAF or MEK inhibitors is associated with the induction or persistence of activity within the AKT pathway in the presence of these drugs. This resistance can be potentially reversed by the combination of a RAF or MEK inhibitor with an AKT or mTOR inhibitor. These combinations should be available for clinical testing in patients progressing on BRAF inhibitors.


Cancer Discovery | 2012

BRAF L597 mutations in melanoma are associated with sensitivity to MEK inhibitors

Kimberly B. Dahlman; Junfeng Xia; Katherine E. Hutchinson; Charles Ng; Donald Hucks; Peilin Jia; Mohammad Atefi; Zengliu Su; Suzanne Branch; Pamela L. Lyle; Donna Hicks; Viviana Bózon; John A. Glaspy; Neal Rosen; David B. Solit; James L. Netterville; Cindy L. Vnencak-Jones; Jeffrey A. Sosman; Antoni Ribas; Zhongming Zhao; William Pao

UNLABELLED Kinase inhibitors are accepted treatment for metastatic melanomas that harbor specific driver mutations in BRAF or KIT, but only 40% to 50% of cases are positive. To uncover other potential targetable mutations, we conducted whole-genome sequencing of a highly aggressive BRAF (V600) and KIT (W557, V559, L576, K642, and D816) wild-type melanoma. Surprisingly, we found a somatic BRAF(L597R) mutation in exon 15. Analysis of BRAF exon 15 in 49 tumors negative for BRAF(V600) mutations as well as driver mutations in KIT, NRAS, GNAQ, and GNA11, showed that two (4%) harbored L597 mutations and another two involved BRAF D594 and K601 mutations. In vitro signaling induced by L597R/S/Q mutants was suppressed by mitogen-activated protein (MAP)/extracellular signal-regulated kinase (ERK) kinase (MEK) inhibition. A patient with BRAF(L597S) mutant metastatic melanoma responded significantly to treatment with the MEK inhibitor, TAK-733. Collectively, these data show clinical significance to BRAF(L597) mutations in melanoma. SIGNIFICANCE This study shows that cells harboring BRAF(L597R) mutants are sensitive to MEK inhibitor treatment, providing a rationale for routine screening and therapy of BRAF(L597R)-mutant melanoma.


Angewandte Chemie | 2013

Polymer Nanofiber‐Embedded Microchips for Detection, Isolation, and Molecular Analysis of Single Circulating Melanoma Cells

Shuang Hou; Libo Zhao; Qinglin Shen; Juehua Yu; Charles Ng; Xiangju Kong; Dongxia Wu; Min Song; Xiaohong Shi; Xiaochun Xu; Wei-Han OuYang; Rongxian He; Xingzhong Zhao; Thomas H. Lee; F. Charles Brunicardi; Mitch A. Garcia; Antoni Ribas; Roger S. Lo; Hsian-Rong Tseng

Circulating tumor cells (CTCs)[1] are cancer cells shed from either the primary tumors or metastatic sites. The presence and number of CTCs in peripheral blood can provide clinically significant data on prognosis and therapeutic response patterns, respectively[2]. Thus, as with traditional invasive tumor biopsies that enable gold-standard pathological analysis, CTCs can be regarded as “liquid biopsies” of the tumor, which enable repeated and relatively non-invasive characterization of tumor evolution, especially important during therapeutic interventions. Currently, FDA-cleared CellSearch™ Assay is costly and inefficient in capturing CTCs, and the enriched CTCs are typically contaminated with a large number of white blood cells (WBCs). As a result, the diagnostic value of CTCs has been underutilized. Over the past decade, a diversity of CTC detection technologies[2d, 3] have been developed to overcome the challenges encountered by the immunomagnetic separation-based CellSearch™ Assay.


Cancer Discovery | 2014

A Novel AKT1 Mutant Amplifies an Adaptive Melanoma Response to BRAF Inhibition

Hubing Shi; Aayoung Hong; Xiangju Kong; Richard C. Koya; Chunying Song; Gatien Moriceau; Willy Hugo; Clarissa C. Yu; Charles Ng; Thinle Chodon; Richard A. Scolyer; Richard F. Kefford; Antoni Ribas; Roger S. Lo

BRAF inhibitor (BRAFi) therapy leads to remarkable anti melanoma responses, but the initial tumor shrinkage is commonly incomplete, providing a nidus for subsequent disease progression. Adaptive signaling may underlie early BRAFi resistance and influence the selection pattern for genetic variants, causing late, acquired resistance. We show here that BRAFi (or BRAFi + MEKi) therapy in patients frequently led to rebound phosphorylated AKT (p-AKT) levels in their melanomas early on-treatment. In cell lines, BRAFi treatment led to rebound levels of receptor tyrosine kinases (RTK; including PDGFRβ), phosphatidyl (3,4,5)-triphosphate (PIP3), pleckstrin homology domain recruitment, and p-AKT. PTEN expression limited this BRAFi-elicited PI3K-AKT signaling, which could be rescued by the introduction of a mutant AKT1 (Q79K) known to confer acquired BRAFi resistance. Functionally, AKT1(Q79K) conferred BRAFi resistance via amplification of BRAFi-elicited PI3K-AKT signaling. In addition, mitogen-activated protein kinase pathway inhibition enhanced clonogenic growth dependency on PI3K or AKT. Thus, adaptive or genetic upregulation of AKT critically participates in melanoma survival during BRAFi therapy.


Nature | 2015

DDX5 and its associated lncRNA Rmrp modulate TH17 cell effector functions

Wendy Huang; Benjamin Thomas; Ryan A. Flynn; Samuel J. Gavzy; Lin Wu; Sangwon V. Kim; Jason A. Hall; Emily R. Miraldi; Charles Ng; Frank Rigo; Sarah Meadows; Nina R. Montoya; Natalia G. Herrera; Ana I. Domingos; Fraydoon Rastinejad; Richard M. Myers; Frances V. Fuller-Pace; Richard Bonneau; Howard Y. Chang; Oreste Acuto; Dan R. Littman

T helper 17 (TH17) lymphocytes protect mucosal barriers from infections, but also contribute to multiple chronic inflammatory diseases. Their differentiation is controlled by RORγt, a ligand-regulated nuclear receptor. Here we identify the RNA helicase DEAD-box protein 5 (DDX5) as a RORγt partner that coordinates transcription of selective TH17 genes, and is required for TH17-mediated inflammatory pathologies. Surprisingly, the ability of DDX5 to interact with RORγt and coactivate its targets depends on intrinsic RNA helicase activity and binding of a conserved nuclear long noncoding RNA (lncRNA), Rmrp, which is mutated in patients with cartilage-hair hypoplasia. A targeted Rmrp gene mutation in mice, corresponding to a gene mutation in cartilage-hair hypoplasia patients, altered lncRNA chromatin occupancy, and reduced the DDX5–RORγt interaction and RORγt target gene transcription. Elucidation of the link between Rmrp and the DDX5–RORγt complex reveals a role for RNA helicases and lncRNAs in tissue-specific transcriptional regulation, and provides new opportunities for therapeutic intervention in TH17-dependent diseases.


Clinical Cancer Research | 2014

Adoptive Transfer of MART-1 T-Cell Receptor Transgenic Lymphocytes and Dendritic Cell Vaccination in Patients with Metastatic Melanoma

Thinle Chodon; Begonya Comin-Anduix; Bartosz Chmielowski; Richard C. Koya; Zhongqi Wu; Martin Auerbach; Charles Ng; Earl Avramis; Elizabeth Seja; Arturo Villanueva; Tara A. McCannel; Akira Ishiyama; Johannes Czernin; Caius G. Radu; Xiaoyan Wang; David W. Gjertson; Alistair J. Cochran; Kenneth Cornetta; Deborah J.L. Wong; Paula Kaplan-Lefko; Omid Hamid; Wolfram E. Samlowski; Peter A. Cohen; Gregory A. Daniels; Bijay Mukherji; Lili Yang; Jerome A. Zack; Donald B. Kohn; James R. Heath; John A. Glaspy

Purpose: It has been demonstrated that large numbers of tumor-specific T cells for adoptive cell transfer (ACT) can be manufactured by retroviral genetic engineering of autologous peripheral blood lymphocytes and expanding them over several weeks. In mouse models, this therapy is optimized when administered with dendritic cell (DC) vaccination. We developed a short 1-week manufacture protocol to determine the feasibility, safety, and antitumor efficacy of this double cell therapy. Experimental Design: A clinical trial (NCT00910650) adoptively transferring MART-1 T-cell receptor (TCR) transgenic lymphocytes together with MART-1 peptide-pulsed DC vaccination in HLA-A2.1 patients with metastatic melanoma. Autologous TCR transgenic cells were manufactured in 6 to 7 days using retroviral vector gene transfer, and reinfused with (n = 10) or without (n = 3) prior cryopreservation. Results: A total of 14 patients with metastatic melanoma were enrolled and 9 of 13 treated patients (69%) showed evidence of tumor regression. Peripheral blood reconstitution with MART-1–specific T cells peaked within 2 weeks of ACT, indicating rapid in vivo expansion. Administration of freshly manufactured TCR transgenic T cells resulted in a higher persistence of MART-1–specific T cells in the blood as compared with cryopreserved. Evidence that DC vaccination could cause further in vivo expansion was only observed with ACT using noncryopreserved T cells. Conclusion: Double cell therapy with ACT of TCR-engineered T cells with a very short ex vivo manipulation and DC vaccines is feasible and results in antitumor activity, but improvements are needed to maintain tumor responses. Clin Cancer Res; 20(9); 2457–65. ©2014 AACR.


Journal of Translational Medicine | 2011

Combination therapy with vemurafenib (PLX4032/RG7204) and metformin in melanoma cell lines with distinct driver mutations

Franziska Niehr; Erika von Euw; Narsis Attar; Deliang Guo; Doug Matsunaga; Hooman Sazegar; Charles Ng; John A. Glaspy; Juan A Recio; Roger S. Lo; Paul S. Mischel; Begonya Comin-Anduix; Antoni Ribas

BackgroundA molecular linkage between the MAPK and the LKB1-AMPK energy sensor pathways suggests that combined MAPK oncogene inhibition and metabolic modulation of AMPK would be more effective than either manipulation alone in melanoma cell lines.Materials and methodsThe combination of the BRAF inhibitor vemurafenib (formerly PLX4032) and metformin were tested against a panel of human melanoma cell lines with defined BRAF and NRAS mutations for effects on viability, cell cycle and apoptosis. Signaling molecules in the MAPK, PI3K-AKT and LKB1-AMPK pathways were studied by Western blot.ResultsSingle agent metformin inhibited proliferation in 12 out of 19 cell lines irrespective of the BRAF mutation status, but in one NRASQ61K mutant cell line it powerfully stimulated cell growth. Synergistic anti-proliferative effects of the combination of metformin with vemurafenib were observed in 6 out of 11 BRAFV600E mutants, including highly synergistic effects in two BRAFV600E mutant melanoma cell lines. Antagonistic effects were noted in some cell lines, in particular in BRAFV600E mutant cell lines resistant to single agent vemurafenib. Seven out of 8 BRAF wild type cell lines showed marginally synergistic anti-proliferative effects with the combination, and one cell line had highly antagonistic effects with the combination. The differential effects were not dependent on the sensitivity to each drug alone, effects on cell cycle or signaling pathways.ConclusionsThe combination of vemurafenib and metformin tended to have stronger anti-proliferative effects on BRAFV600E mutant cell lines. However, determinants of vemurafenib and metformin synergism or antagonism need to be understood with greater detail before any potential clinical utility of this combination.

Collaboration


Dive into the Charles Ng's collaboration.

Top Co-Authors

Avatar

Antoni Ribas

University of California

View shared research outputs
Top Co-Authors

Avatar

Mohammad Atefi

University of California

View shared research outputs
Top Co-Authors

Avatar

Roger S. Lo

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiangju Kong

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Earl Avramis

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard C. Koya

Roswell Park Cancer Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge