Network


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

Hotspot


Dive into the research topics where Kwok Peng Ng is active.

Publication


Featured researches published by Kwok Peng Ng.


Blood | 2012

STAT3 mutations unify the pathogenesis of chronic lymphoproliferative disorders of NK cells and T-cell large granular lymphocyte leukemia

Andres Jerez; Michael J. Clemente; Hideki Makishima; Hanna L M Koskela; Francis LeBlanc; Kwok Peng Ng; Thomas L. Olson; Bartlomiej Przychodzen; Manuel Afable; Inés Gómez-Seguí; Kathryn M Guinta; Lisa Durkin; Eric D. Hsi; Kathy L. McGraw; Dan Zhang; Marcin W. Wlodarski; Kimmo Porkka; Mikkael A. Sekeres; Alan F. List; Satu Mustjoki; Thomas P. Loughran; Jaroslaw P. Maciejewski

Chronic lymphoproliferative disorders of natural killer cells (CLPD-NKs) and T-cell large granular lymphocytic leukemias (T-LGLs) are clonal lymphoproliferations arising from either natural killer cells or cytotoxic T lymphocytes (CTLs). We have investigated for distribution and functional significance of mutations in 50 CLPD-NKs and 120 T-LGL patients by direct sequencing, allele-specific PCR, and microarray analysis. STAT3 gene mutations are present in both T and NK diseases: approximately one-third of patients with each type of disorder convey these mutations. Mutations were found in exons 21 and 20, encoding the Src homology 2 domain. Patients with mutations are characterized by symptomatic disease (75%), history of multiple treatments, and a specific pattern of STAT3 activation and gene deregulation, including increased expression of genes activated by STAT3. Many of these features are also found in patients with wild-type STAT3, indicating that other mechanisms of STAT3 activation can be operative in these chronic lymphoproliferative disorders. Treatment with STAT3 inhibitors, both in wild-type and mutant cases, resulted in accelerated apoptosis. STAT3 mutations are frequent in large granular lymphocytes suggesting a similar molecular dysregulation in malignant chronic expansions of NK and CTL origin. STAT3 mutations may distinguish truly malignant lymphoproliferations involving T and NK cells from reactive expansions.


Nature Genetics | 2013

Somatic SETBP1 mutations in myeloid malignancies

Hideki Makishima; Kenichi Yoshida; Nhu Ngoc Thi Nguyen; Bartlomiej Przychodzen; Masashi Sanada; Yusuke Okuno; Kwok Peng Ng; Kristbjorn Orri Gudmundsson; Bandana A. Vishwakarma; Andres Jerez; Inés Gómez-Seguí; Mariko Takahashi; Yuichi Shiraishi; Yasunobu Nagata; Kathryn M Guinta; Hiraku Mori; Mikkael A. Sekeres; Kenichi Chiba; Hiroko Tanaka; Hideki Muramatsu; Hirotoshi Sakaguchi; Ronald Paquette; Michael A. McDevitt; Seiji Kojima; Yogen Saunthararajah; Satoru Miyano; Lee-Yung Shih; Yang Du; Seishi Ogawa; Jaroslaw P. Maciejewski

Here we report whole-exome sequencing of individuals with various myeloid malignancies and identify recurrent somatic mutations in SETBP1, consistent with a recent report on atypical chronic myeloid leukemia (aCML). Closely positioned somatic SETBP1 mutations encoding changes in Asp868, Ser869, Gly870, Ile871 and Asp880, which match germline mutations in Schinzel-Giedion syndrome (SGS), were detected in 17% of secondary acute myeloid leukemias (sAML) and 15% of chronic myelomonocytic leukemia (CMML) cases. These results from deep sequencing demonstrate a higher mutational detection rate than reported with conventional sequencing methodology. Mutant cases were associated with advanced age and monosomy 7/deletion 7q (–7/del(7q)) constituting poor prognostic factors. Analysis of serially collected samples indicated that SETBP1 mutations were acquired during leukemic evolution. Transduction with mutant Setbp1 led to the immortalization of mouse myeloid progenitors that showed enhanced proliferative capacity compared to cells transduced with wild-type Setbp1. Somatic mutations of SETBP1 seem to cause gain of function, are associated with myeloid leukemic transformation and convey poor prognosis in myelodysplastic syndromes (MDS) and CMML.


Cancer Research | 2007

Podocalyxin Increases the Aggressive Phenotype of Breast and Prostate Cancer Cells In vitro through Its Interaction with Ezrin

Steven T. Sizemore; Muzaffer Cicek; Nywana Sizemore; Kwok Peng Ng; Graham Casey

Podocalyxin is an anti-adhesive transmembrane sialomucin that has been implicated in the development of more aggressive forms of breast and prostate cancer. The mechanism through which podocalyxin increases cancer aggressiveness remains poorly understood but may involve the interaction of podocalyxin with ezrin, an established mediator of metastasis. Here, we show that overexpression of podocalyxin in MCF7 breast cancer and PC3 prostate cancer cell lines increased their in vitro invasive and migratory potential and led to increased expression of matrix metalloproteases 1 and 9 (MMP1 and MMP9). Podocalyxin expression also led to an increase in mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) activity. To determine the role of ezrin in these podocalyxin-dependent phenotypic events, we first confirmed that podocalyxin formed a complex with ezrin in MCF7 and PC3 cells. Furthermore, expression of podocalyxin was associated with a changed ezrin subcellular localization and increased ezrin phosphorylation. Transient knockdown of ezrin protein abrogated MAPK and PI3K signaling as well as MMP expression and invasiveness in cancer cells overexpressing podocalyxin. These findings suggest that podocalyxin leads to increased in vitro migration and invasion, increased MMP expression, and increased activation of MAPK and PI3K activity in MCF7 and PC3 cells through its ability to form a complex with ezrin.


Leukemia | 2012

CpG methylation patterns and decitabine treatment response in acute myeloid leukemia cells and normal hematopoietic precursors

Soledad Negrotto; Kwok Peng Ng; Ania Jankowska; Juraj Bodo; Banu Gopalan; Kathryn M Guinta; James C. Mulloy; Eric D. Hsi; Jaroslaw P. Maciejewski; Yogen Saunthararajah

The DNA hypomethylating drug decitabine maintains normal hematopoietic stem cell (HSC) self-renewal but induces terminal differentiation in acute myeloid leukemia (AML) cells. The basis for these contrasting cell fates, and for selective CpG hypomethylation by decitabine, is poorly understood. Promoter CpGs, with methylation measured by microarray, were classified by the direction of methylation change with normal myeloid maturation. In AML cells, the methylation pattern at maturation-responsive CpGs suggested at least partial maturation. Consistent with partial maturation, in gene expression analyses, AML cells expressed high levels of the key lineage-specifying factor CEBPA, but relatively low levels of the key late-differentiation driver CEBPE. In methylation analysis by mass spectrometry, CEBPE promoter CpGs that are usually hypomethylated during granulocyte maturation were significantly hypermethylated in AML cells. Decitabine-induced hypomethylation was greatest at these and other promoter CpGs that are usually hypomethylated with myeloid maturation, accompanied by cellular differentiation of AML cells. In contrast, decitabine-treated normal HSCs retained immature morphology, and methylation significantly decreased at CpGs that are less methylated in immature cells. High expression of lineage-specifying factor and aberrant epigenetic repression of some key late-differentiation driver genes distinguishes AML cells from normal HSCs, and could explain the contrasting differentiation and methylation responses to decitabine.


Oncogene | 2008

Mutated in Colorectal Cancer, a Putative Tumor Suppressor for Serrated Colorectal Cancer, Selectively Represses β-catenin-Dependent Transcription

Ryuichi Fukuyama; Roxana Niculaita; Kwok Peng Ng; Emmanuel Obusez; Julian A. Sanchez; Matthew F. Kalady; Phyu Phyu Aung; Graham Casey; Nywana Sizemore

Mutated in colorectal cancer (MCC) was originally identified as a candidate gene for familial adenomatous polyposis (FAP) but further study identified adenomatous polyposis coli (APC) as responsible for FAP and the physiologic/pathologic roles of MCC remained poorly understood. Recently, MCC promoter methylation was discovered as a frequent early event in a distinct subset of precursor lesions and colorectal cancer (CRC) associated with the serrated CRC pathway. Here we provide the first evidence of the biological significance of MCC loss in CRC and the molecular pathways involved. We show MCC expression is dramatically decreased in many CRC cell lines and the distinct subset of sporadic CRC characterized by the CpG island methylator phenotype and BRAFV600E mutation due to promoter methylation as reported previously. Importantly, we find MCC interacts with β-catenin and that reexpression of MCC in CRC cells specifically inhibits Wnt signaling, β-catenin/T-cell factor/lymphoid-enhancer factor-dependent transcription and cellular proliferation even in the presence of oncogenic mutant APC. We also show that MCC is localized in the nucleus and identify two functional nuclear localization signals. Taken together, MCC is a nuclear, β-catenin-interacting protein that can act as a potential tumor suppressor in the serrated CRC pathway by inhibiting Wnt/β-catenin signal transduction.


Leukemia | 2011

p53 Independent epigenetic-differentiation treatment in xenotransplant models of acute myeloid leukemia

Kwok Peng Ng; Quteba Ebrahem; Soledad Negrotto; R. Mahfouz; Kevin A. Link; Zhenbo Hu; Xiaorong Gu; Anjali S. Advani; M Kalaycio; Ronald Sobecks; Mikkael A. Sekeres; Edward A. Copelan; Tomas Radivoyevitch; Jaroslaw P. Maciejewski; James C. Mulloy; Yogen Saunthararajah

Suppression of apoptosis by TP53 mutation contributes to resistance of acute myeloid leukemia (AML) to conventional cytotoxic treatment. Using differentiation to induce irreversible cell cycle exit in AML cells could be a p53-independent treatment alternative, however, this possibility requires evaluation. In vitro and in vivo regimens of the deoxycytidine analogue decitabine that deplete the chromatin-modifying enzyme DNA methyl-transferase 1 without phosphorylating p53 or inducing early apoptosis were determined. These decitabine regimens but not equimolar DNA-damaging cytarabine upregulated the key late differentiation factors CCAAT enhancer-binding protein ɛ and p27/cyclin dependent kinase inhibitor 1B (CDKN1B), induced cellular differentiation and terminated AML cell cycle, even in cytarabine-resistant p53- and p16/CDKN2A-null AML cells. Leukemia initiation by xenotransplanted AML cells was abrogated but normal hematopoietic stem cell engraftment was preserved. In vivo, the low toxicity allowed frequent drug administration to increase exposure, an important consideration for S phase specific decitabine therapy. In xenotransplant models of p53-null and relapsed/refractory AML, the non-cytotoxic regimen significantly extended survival compared with conventional cytotoxic cytarabine. Modifying in vivo dose and schedule to emphasize this pathway of decitabine action can bypass a mechanism of resistance to standard therapy.


Blood | 2012

Effects of tetrahydrouridine on pharmacokinetics and pharmacodynamics of oral decitabine

Donald Lavelle; Kestis Vaitkus; Yonghua Ling; Maria Armila Ruiz; R. Mahfouz; Kwok Peng Ng; Soledad Negrotto; Nicola Smith; Pramod Terse; Kory Engelke; Joseph M. Covey; Kenneth K. Chan; Joseph M. DeSimone; Yogen Saunthararajah

The deoxycytidine analog decitabine (DAC) can deplete DNA methyl-transferase 1 (DNMT1) and thereby modify cellular epigenetics, gene expression, and differentiation. However, a barrier to efficacious and accessible DNMT1-targeted therapy is cytidine deaminase, an enzyme highly expressed in the intestine and liver that rapidly metabolizes DAC into inactive uridine counterparts, severely limiting exposure time and oral bioavailability. In the present study, the effects of tetrahydrouridine (THU), a competitive inhibitor of cytidine deaminase, on the pharmacokinetics and pharmacodynamics of oral DAC were evaluated in mice and nonhuman primates. Oral administration of THU before oral DAC extended DAC absorption time and widened the concentration-time profile, increasing the exposure time for S-phase-specific depletion of DNMT1 without the high peak DAC levels that can cause DNA damage and cytotoxicity. THU also decreased interindividual variability in pharmacokinetics seen with DAC alone. One potential clinical application of DNMT1-targeted therapy is to increase fetal hemoglobin and treat hemoglobinopathy. Oral THU-DAC at a dose that would produce peak DAC concentrations of less than 0.2μM administered 2×/wk for 8 weeks to nonhuman primates was not myelotoxic, hypomethylated DNA in the γ-globin gene promoter, and produced large cumulative increases in fetal hemoglobin. Combining oral THU with oral DAC changes DAC pharmacology in a manner that may facilitate accessible noncytotoxic DNMT1-targeted therapy.


Molecular Carcinogenesis | 2007

Role of IKK and oscillatory NFκB kinetics in MMP‐9 gene expression and chemoresistance to 5‐fluorouracil in RKO colorectal cancer cells

Ryuichi Fukuyama; Kwok Peng Ng; Muzaffer Cicek; Clare Kelleher; Roxana Niculaita; Graham Casey; Nywana Sizemore

Nuclear factor kappa B (NFκB) is a central participant in the metastasis and chemoresistance of colorectal cancer (CRC). However, it is not fully understood to what extent NFκB contributes to induction of the metastasis‐associated matrix metalloprotease‐9 (MMP‐9) gene and sensitivity to the commonly used chemotherapeutic 5‐fluorouracil (5‐Fu) in CRC. Using the RKO human CRC cell line and two NFκB signaling deficient RKO mutants, we investigated NFκBs role in the induction of MMP‐9 and 5‐Fu sensitivity in RKO CRC cells. NFκB plays a predominant role in MMP‐9 gene induction in RKO cells, as evidenced by the failure of tumor necrosis factor alpha (TNFα) to induce MMP‐9 in either of the NFκB signaling mutants. RKO cells exhibit a robust, oscillatory NFκB activity in response to TNFα not seen in either of the NFκB mutant cell lines, which instead demonstrate diminished, nonoscillatory NFκB activation. Analysis of TNFα‐induced phosphorylation and MMP‐9 promoter recruitment of the p65 NFκB subunit revealed a significant reduction in p65 phosphorylation as well as reduced and altered recruitment of p65 to the MMP‐9 gene promoter in the mutants compared to the parental RKO cell line. 5‐Fu only activated NFκB in the parental RKO cells through induction of IκB‐kinase (IKK) activity and increased sensitivity to 5‐Fu is observed in both NFκB mutant lines. Our results suggest that TNFα‐dependent induction of MMP‐9 gene expression is tightly regulated by oscillatory/cumulative activation of NFκB and that 5‐Fu stimulates NFκB and RKO CRC cell survival through induction of IKK activity.


Molecular Cancer Therapeutics | 2010

Decitabine Maintains Hematopoietic Precursor Self-Renewal by Preventing Repression of Stem Cell Genes by a Differentiation-Inducing Stimulus

Zhenbo Hu; Soledad Negrotto; Xiaorong Gu; R. Mahfouz; Kwok Peng Ng; Quteba Ebrahem; Edward A. Copelan; Harinder Singh; Jaroslaw P. Maciejewski; Yogen Saunthararajah

The cytosine analogue decitabine alters hematopoietic differentiation. For example, decitabine treatment increases self-renewal of normal hematopoietic stem cells. The mechanisms underlying decitabine-induced shifts in differentiation are poorly understood, but likely relate to the ability of decitabine to deplete the chromatin-modifying enzyme DNA methyltransferase 1 (DNMT1), which plays a central role in transcription repression. HOXB4 is a transcription factor that promotes hematopoietic stem cell self-renewal. In hematopoietic precursors induced to differentiate by the lineage-specifying transcription factor Pu.1 or by the cytokine granulocyte-colony stimulating factor, there is rapid repression of HOXB4 and other stem cell genes. Depletion of DNMT1 using shRNA or decitabine prevents HOXB4 repression by Pu.1 or granulocyte-colony stimulating factor and maintains hematopoietic precursor self-renewal. In contrast, depletion of DNMT1 by decitabine 6 hours after the differentiation stimulus, that is, after repression of HOXB4 has occurred, augments differentiation. Therefore, DNMT1 is required for the early repression of stem cell genes, which occurs in response to a differentiation stimulus, providing a mechanistic explanation for the observation that decitabine can maintain or increase hematopoietic stem cell self-renewal in the presence of a differentiation stimulus. Using decitabine to deplete DNMT1 after this early repression phase does not impair progressive differentiation. Mol Cancer Ther; 9(6); 1536–43. ©2010 AACR.


Cancer Research | 2011

Noncytotoxic Differentiation Treatment of Renal Cell Cancer

Soledad Negrotto; Zhenbo Hu; Oscar Alcazar; Kwok Peng Ng; Pierre L. Triozzi; Daniel J. Lindner; Brian I. Rini; Yogen Saunthararajah

Current drug therapy for metastatic renal cell cancer (RCC) results in temporary disease control but not cure, necessitating continued investigation into alternative mechanistic approaches. Drugs that inhibit chromatin-modifying enzymes involved in transcription repression (chromatin-relaxing drugs) could have a role, by inducing apoptosis and/or through differentiation pathways. At low doses, the cytosine analogue decitabine (DAC) can be used to deplete DNA methyl-transferase 1 (DNMT1), modify chromatin, and alter differentiation without causing apoptosis (cytotoxicity). Noncytotoxic regimens of DAC were evaluated for in vitro and in vivo efficacy against RCC cell lines, including a p53-mutated RCC cell line developed from a patient with treatment-refractory metastatic RCC. The cell division-permissive mechanism of action-absence of early apoptosis or DNA damage, increase in expression of HNF4α (hepatocyte nuclear factor 4α), a key driver associated with the mesenchymal to epithelial transition, decrease in mesenchymal marker expression, increase in epithelial marker expression, and late increase in cyclin-dependent kinase inhibitor CDKN1B (p27) protein-was consistent with differentiation-mediated cell-cycle exit. In vivo blood counts and animal weights were consistent with minimal toxicity of therapy. The distinctive mechanism of action of a dose and schedule of DAC designed for noncytotoxic depletion of DNMT1 suggests a potential role in treating RCC.

Collaboration


Dive into the Kwok Peng Ng'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