Network


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

Hotspot


Dive into the research topics where Yung-Mae Yao is active.

Publication


Featured researches published by Yung-Mae Yao.


Cancer Research | 2009

PI3K Pathway Activation Mediates Resistance to MEK Inhibitors in KRAS Mutant Cancers

Susan Wee; Zainab Jagani; Kay Xiaoqin Xiang; Alice Loo; Marion Dorsch; Yung-Mae Yao; William R. Sellers; Christoph Lengauer; Frank Stegmeier

The RAS pathway is one of the most frequently deregulated pathways in cancer. RAS signals through multiple effector pathways, including the RAF/mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK MAPK and phosphatidylinositol 3-kinase (PI3K)-AKT signaling cascades. The oncogenic potential of these effector pathways is illustrated by the frequent occurrence of activating mutations in BRAF and PIK3CA as well as loss-of-function mutations in the tumor suppressor PTEN, a negative regulator of PI3K. Previous studies have found that whereas BRAF mutant cancers are highly sensitive to MEK inhibition, RAS mutant cancers exhibit a more variable response. The molecular mechanisms responsible for this heterogeneous response remain unclear. In this study, we show that PI3K pathway activation strongly influences the sensitivity of RAS mutant cells to MEK inhibitors. Activating mutations in PIK3CA reduce the sensitivity to MEK inhibition, whereas PTEN mutations seem to cause complete resistance. We further show that down-regulation of PIK3CA resensitizes cells with co-occurring KRAS and PIK3CA mutations to MEK inhibition. At the molecular level, the dual inhibition of both pathways seems to be required for complete inhibition of the downstream mammalian target of rapamycin effector pathway and results in the induction of cell death. Finally, we show that whereas inactivation of either the MEK or PI3K pathway leads to partial tumor growth inhibition, targeted inhibition of both pathways is required to achieve tumor stasis. Our study provides molecular insights that help explain the heterogeneous response of KRAS mutant cancers to MEK pathway inhibition and presents a strong rationale for the clinical testing of combination MEK and PI3K targeted therapies.


Haematologica | 2010

In vitro and in vivo rationale for the triple combination of panobinostat (LBH589) and dexamethasone with either bortezomib or lenalidomide in multiple myeloma

Enrique M. Ocio; David Vilanova; Peter Atadja; Patricia Maiso; Edvan Crusoe; Diego Fernández-Lázaro; Mercedes Garayoa; Laura San-Segundo; Teresa Hernández-Iglesias; Enrique de Alava; Wenlin Shao; Yung-Mae Yao; Atanasio Pandiella; Jesús F. San-Miguel

Background Combinations of drug treatments based on bortezomib or lenalidomide plus steroids have resulted in very high response rates in multiple myeloma. However, most patients still relapse, indicating the need for novel combination partners to increase duration of response or to treat relapsed disease. We explored the antimyeloma activity of triple combinations of these well-established schemes with panobinostat, a novel deacetylase inhibitor with a multi-targeted profile. Design and Methods The activity of these combinations was explored in vitro in cell lines by using MTT and annex-in V, ex vivo by flow cytometry, and in vivo using two different murine models of human myeloma: one bearing a subcutaneous plasmacytoma and another with a disseminated myeloma. Moreover, gene expression profiling and immunohistochemical studies were performed. Results The addition of panobinostat (LBH589) to dexamethasone and either bortezomib or lenalidomide resulted in clear potentiation in multiple myeloma cell lines, freshly isolated plasma cells, and murine models of multiple myeloma. The quantification of the potency of these combinations by using the Chou-Talalay method showed synergistic combination indices for all of them. This effect derived from the deregulation of a cluster of genes that was completely different from the sum of genes affected by the single agents (895 and 1323 genes exclusively deregulated by panobinostat and dexamethasone plus bortezomib or lenalidomide, respectively). Functional experiments, such as annexin V staining, cell cycle analysis, and immunohistochemical studies also supported this potentiation. Anti-myeloma efficacy was confirmed in an extramedullary plasmacytoma model and a disseminated luciferized model, in which panobinostat also provided a marked benefit in bone disease. Conclusions The potent activity, together with the exclusive mechanistic profile, provides the rationale for the clinical evaluation of these drug combinations in multiple myeloma.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Inhibition of tumorigenesis driven by different Wnt proteins requires blockade of distinct ligand-binding regions by LRP6 antibodies

Seth Ettenberg; Olga Charlat; Michael P. Daley; Shanming Liu; Karen Vincent; Darrin Stuart; Alwin Schuller; Jing Yuan; Beatriz Ospina; John Green; Qunyan Yu; Renee Walsh; Sharon Li; Rita Schmitz; Holger Heine; Sanela Bilic; Lance Ostrom; Rebecca A. Mosher; K. Felix Hartlepp; Zhenping Zhu; Stephen E. Fawell; Yung-Mae Yao; David Stover; Peter Finan; Jeffery A. Porter; William R. Sellers; Ingo Klagge; Feng Cong

Disregulated Wnt/β-catenin signaling has been linked to various human diseases, including cancers. Inhibitors of oncogenic Wnt signaling are likely to have a therapeutic effect in cancers. LRP5 and LRP6 are closely related membrane coreceptors for Wnt proteins. Using a phage-display library, we identified anti-LRP6 antibodies that either inhibit or enhance Wnt signaling. Two classes of LRP6 antagonistic antibodies were discovered: one class specifically inhibits Wnt proteins represented by Wnt1, whereas the second class specifically inhibits Wnt proteins represented by Wnt3a. Epitope-mapping experiments indicated that Wnt1 class-specific antibodies bind to the first propeller and Wnt3a class-specific antibodies bind to the third propeller of LRP6, suggesting that Wnt1- and Wnt3a-class proteins interact with distinct LRP6 propeller domains. This conclusion is further supported by the structural functional analysis of LRP5/6 and the finding that the Wnt antagonist Sclerostin interacts with the first propeller of LRP5/6 and preferentially inhibits the Wnt1-class proteins. We also show that Wnt1 or Wnt3a class-specific anti-LRP6 antibodies specifically block growth of MMTV-Wnt1 or MMTV-Wnt3 xenografts in vivo. Therapeutic application of these antibodies could be limited without knowing the type of Wnt proteins expressed in cancers. This is further complicated by our finding that bivalent LRP6 antibodies sensitize cells to the nonblocked class of Wnt proteins. The generation of a biparatopic LRP6 antibody blocks both Wnt1- and Wnt3a-mediated signaling without showing agonistic activity. Our studies provide insights into Wnt-induced LRP5/6 activation and show the potential utility of LRP6 antibodies in Wnt-driven cancer.


Cancer Research | 2011

Protein Kinase C Inhibitor Sotrastaurin Selectively Inhibits the Growth of CD79 Mutant Diffuse Large B-Cell Lymphomas

Tara L. Naylor; Huaping Tang; Boris A. Ratsch; Andreas Enns; Alice Loo; Liqing Chen; Peter Lenz; Nigel J. Waters; Walter Schuler; Bernd Dörken; Yung-Mae Yao; Markus Warmuth; Georg Lenz; Frank Stegmeier

The activated B-cell-like (ABC) subtype of diffuse large B-cell lymphoma (DLBCL) correlates with poor prognosis. The ABC subtype of DLBCL is associated with constitutive activation of the NF-κB pathway, and oncogenic lesions have been identified in its regulators, including CARD11/CARMA1 (caspase recruitment domain-containing protein 11), A20/TNFAIP3, and CD79A/B. In this study, we offer evidence of therapeutic potential for the selective PKC (protein kinase C) inhibitor sotrastaurin (STN) in preclinical models of DLBCL. A significant fraction of ABC DLBCL cell lines exhibited strong sensitivity to STN, and we found that the molecular nature of NF-κB pathway lesions predicted responsiveness. CD79A/B mutations correlated with STN sensitivity, whereas CARD11 mutations rendered ABC DLBCL cell lines insensitive. Growth inhibitory effects of PKC inhibition correlated with NF-κB pathway inhibition and were mediated by induction of G₁-phase cell-cycle arrest and/or cell death. We found that STN produced significant antitumor effects in a mouse xenograft model of CD79A/B-mutated DLBCL. Collectively, our findings offer a strong rationale for the clinical evaluation of STN in ABC DLBCL patients who harbor CD79 mutations also illustrating the necessity to stratify DLBCL patients according to their genetic abnormalities.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Maintenance of adenomatous polyposis coli (APC)-mutant colorectal cancer is dependent on Wnt/β-catenin signaling

Alix Scholer-Dahirel; Michael R. Schlabach; Alice Loo; Linda Bagdasarian; Ronald Meyer; Ribo Guo; Steve Woolfenden; Kristine Yu; Judit Markovits; Karen Killary; Dmitry Sonkin; Yung-Mae Yao; Markus Warmuth; William R. Sellers; Robert A. Schlegel; Frank Stegmeier; Rebecca Mosher; Margaret McLaughlin

Persistent expression of certain oncogenes is required for tumor maintenance. This phenotype is referred to as oncogene addiction and has been clinically validated by anticancer therapies that specifically inhibit oncoproteins such as BCR-ABL, c-Kit, HER2, PDGFR, and EGFR. Identifying additional genes that are required for tumor maintenance may lead to new targets for anticancer drugs. Although the role of aberrant Wnt pathway activation in the initiation of colorectal cancer has been clearly established, it remains unclear whether sustained Wnt pathway activation is required for colorectal tumor maintenance. To address this question, we used inducible β-catenin shRNAs to temporally control Wnt pathway activation in vivo. Here, we show that active Wnt/β-catenin signaling is required for maintenance of colorectal tumor xenografts harboring APC mutations. Reduced tumor growth upon β-catenin inhibition was due to cell cycle arrest and differentiation. Upon reactivation of the Wnt/β-catenin pathway colorectal cancer cells resumed proliferation and reacquired a crypt progenitor phenotype. In human colonic adenocarcinomas, high levels of nuclear β-catenin correlated with crypt progenitor but not differentiation markers, suggesting that the Wnt/β-catenin pathway may also control colorectal tumor cell fate during the maintenance phase of tumors in patients. These results support efforts to treat human colorectal cancer by pharmacological inhibition of the Wnt/β-catenin pathway.


International Journal of Cancer | 2010

Activity of deacetylase inhibitor panobinostat (LBH589) in cutaneous T-cell lymphoma models: Defining molecular mechanisms of resistance.

Wenlin Shao; Joseph D. Growney; Yun Feng; Gregory O'Connor; Minying Pu; Wenjing Zhu; Yung-Mae Yao; Paul Kwon; Stephen Fawell; Peter Atadja

Panobinostat (LBH589) is a highly potent deacetylase inhibitor that has demonstrated clinical efficacy in patients with advanced cutaneous T‐cell lymphoma (CTCL). To gain a better understanding of the compound activity in this tumor type, we investigated the cellular and molecular effects of panobinostat using both in vitro and in vivo models of CTCL. All 4 tested CTCL cell lines exhibited very high sensitivity to panobinostat‐induced growth inhibition. However, only 2 of 4 lines exhibited significant response to the cytotoxic activity of panobinostat. In a CTCL xenograft mouse tumor model, panobinostat treatment resulted in complete tumor regression. The difference in cell sensitivity to panobinostat‐induced death enabled us to further investigate potential mechanisms responsible for tumor sensitivity or resistance. In CTCL cell lines that were insensitive to panobinostat‐induced apoptosis, constitutively activated NF‐κB and high levels of Bcl‐2 were observed. Inhibition of Bcl‐2 sensitized cells to the cytotoxic activity of panobinostat. Conversely, knockdown of Bax diminished the CTCL cell sensitivity. Interestingly, panobinostat could induce cytotoxicity in vorinostat‐resistant CTCL cells by downregulating phosphorylated STAT3 and STAT5 proteins. These studies suggest distinct mechanisms responsible for resistance to different deacetylase inhibitors. We show that the intrinsic apoptotic signaling plays an essential role in mediating panobinostat anticancer activity. Moreover, cancer cell sensitivity to panobinostat treatment may be further improved by combination with inhibition of anti‐apoptotic factors. These data provide preclinical support that panobinostat, as a single agent or in combination with other anticancer agents, is a promising therapy for CTCL.


Journal of Medicinal Chemistry | 2011

Optimization of the in Vitro Cardiac Safety of Hydroxamate-Based Histone Deacetylase Inhibitors

Michael Shultz; Xueying Cao; Christine Hiu-Tung Chen; Young Shin Cho; Nicole R. Davis; Joe Eckman; Jianmei Fan; Alex Fekete; Brant Firestone; Julie Flynn; Jack Green; Joseph D. Growney; Mats Holmqvist; Meier Hsu; Daniel Jansson; Lei Jiang; Paul Kwon; Gang Liu; Franco Lombardo; Qiang Lu; Dyuti Majumdar; Christopher Meta; Lawrence Blas Perez; Minying Pu; Tim Ramsey; Stacy W. Remiszewski; Suzanne Skolnik; Martin Traebert; Laszlo Urban; Vinita Uttamsingh

Histone deacetylase (HDAC) inhibitors have shown promise in treating various forms of cancer. However, many HDAC inhibitors from diverse structural classes have been associated with QT prolongation in humans. Inhibition of the human ether a-go-go related gene (hERG) channel has been associated with QT prolongation and fatal arrhythmias. To determine if the observed cardiac effects of HDAC inhibitors in humans is due to hERG blockade, a highly potent HDAC inhibitor devoid of hERG activity was required. Starting with dacinostat (LAQ824), a highly potent HDAC inhibitor, we explored the SAR to determine the pharmacophores required for HDAC and hERG inhibition. We disclose here the results of these efforts where a high degree of pharmacophore homology between these two targets was discovered. This similarity prevented traditional strategies for mitigating hERG binding/modulation from being successful and novel approaches for reducing hERG inhibition were required. Using a hERG homology model, two compounds, 11r and 25i, were discovered to be highly efficacious with weak affinity for the hERG and other ion channels.


Journal of Medicinal Chemistry | 2012

VX-322: A Novel Dual Receptor Tyrosine Kinase Inhibitor for the Treatment of Acute Myelogenous Leukemia

David K. Heidary; George Huang; Diane M. Boucher; Jianguo Ma; Cornelia Forster; Ron Grey; Jinwang Xu; Michael J. Arnost; Deborah Choquette; Guanjing Chen; Jiehua Zhou; Yung-Mae Yao; Edward D. Ball; Mark Namchuk; Robert J. Davies; Greg Henkel

In acute myelogenous leukemia (AML), the FLT3 receptor tyrosine kinase (RTK) is highly expressed with 30% of patients expressing a mutated, constitutively active form of this protein. To inhibit this receptor, VX-322 was developed and found to be very potent against both the FLT3 and c-KIT RTKs with enzyme K(i) values of <1 nM and a cellular IC(50) between 1 and 5 nM. It was efficacious in a FLT3-ITD dependent myeloproliferative mouse model, doubling survival compared to other FLT3 inhibitors, with 25% of the mice cured. Upon treatment of primary AML patient blast cells, the dual inhibition of FLT3 and c-KIT was superior to inhibitors targeting a single RTK. Thus, this compound may represent an improved pharmacologic and selectivity profile that could be effective in the treatment of AML.


Molecular Cancer Therapeutics | 2009

Abstract B27: Correlation between TNFα and LCL161 anti‐tumor activity in patient derived xenograft models of human cancer

Brant Firestone; Colleen Conway; Guizhi Yang; Hui Gao; Dale Porter; Joanna Slisz; Dan He; Rebecca Mosher; John E. Monahan; Christopher Sean Straub; Michael Morrissey; Yung-Mae Yao; Leigh Zawel

LCL161, a small molecule antagonist of inhibitor of apoptosis proteins (IAPs), induces TNF ‐mediated apoptosis in a subset of tumor cell lines including the MDA‐MB‐231 breast cancer line. To investigate the in vivo activity of LCL161, MDA‐MB‐231 tumor bearing mice were treated with a once weekly oral dose. We observed anti‐tumor activity that was associated with pharmacodynamic responses including degradation of CIAP1 and induction of cleaved caspase 3. The loss of CIAP1, specifically the E3 ligase activity of this protein, has been shown to directly impact the stability of a number of client proteins including NIK (NF‐κB inducing kinase), Mad1 (MAX‐binding protein), and others. Gene expression analysis on LCL161 treated MDA‐MB‐231 tumors revealed a striking upregulation of NF‐κB regulated target genes. These data were further supported by the observation that TNFα, a direct target of NF‐κB, was induced in LCL161 treated MDAMB‐231 tumor lysates. To explore whether TNFα expression levels could be used to predict single agent efficacy in a more clinically relevant context, a series of human primary tumor xenografts were profiled for baseline TNFα mRNA levels and evaluated for response to LCL161 in vivo. We observed that tumors with the highest TNFα expression levels showed the greatest sensitivity to LCL161. These studies demonstrate an essential role of the NF‐κB pathway in IAP antagonist anti‐tumor activity. Ongoing efforts focus on delineating the contribution of the canonical and non‐canonical NF‐κB pathways to efficacy and on leveraging this mechanism for LCL161 patient selection. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):B27.


Cancer Research | 2010

Abstract 4290: The smoothened antagonist NVP-LDE225 targets stroma and cancer stem cells in primary human pancreatic tumor xenografts

Silvia Buonamici; Beatriz Ospina; Julia Xing Zhu; Jing Yuan; Kathy Hsiao; Anthony Vattay; Fang Li; James Deeds; Lance Ostrom; John E. Monahan; Juliet Williams; Joseph F. Kelleher; Stefan Peukert; Shifeng Pan; Xu Wu; Markus Warmuth; Rebecca Mosher; Yung-Mae Yao; William R. Sellers; Marion Dorsch

Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer mortality. Despite surgical intervention and treatment with Gemcitabine, the standard of care, the 5-year survival rate remains less than 5%. Because the aberrant activation of the Hedgehog (Hh) pathway in stroma cells and cancer stem cells in PDAC has become evident during the last few years, therapeutic agents able to inhibit Smoothened (Smo), the positive regulator of the pathway, are under vigorous investigation. We have developed a selective Smo antagonist, NVP-LDE225, which is orally bioavailable and potently inhibits Hh signaling in vitro and in vivo. Using a combination of in vitro and in vivo assays, we evaluated the effects of NVP-LDE225 alone or in combination with Gemcitabine in primary human tumor xenograft models. In these models, NVP-LDE225 dosed together with Gemcitibine resulted in a trend towards combination activity. Tumor samples collected at the end of the studies showed that the Hh pathway was completely inhibited in the stroma of the NVP-LDE225 treated animals. Moreover, analysis of the ratio of tumor to stroma area demonstrated no differences between the drug treated as compared to vehicle treated tumors. After subcutaneous implantation of the human tumors in mice, the mouse stroma replaces the human stroma. Using mouse and human Affymetrix analysis of tumors following in vivo treatment, we were able to differentiate the changes occurring in murine stromal cells versus human epithelial cells. This analysis revealed changes in the regulation of several key developmental and growth factor pathways in the mouse stroma. To study cancer stem cells, we isolated Aldehyde Dehydrogenase (ALDH) positive cells from untreated xenograft tumors and confirmed up regulation of the Hh pathway genes, and that ALDH positive cells were more tumorigenic than ALDH negative cells when implanted in animals. Furthermore, we used 2 different stem cell functional assays, in vitro sphere formation and in vivo serial transplantation to evaluate the effect of NVP-LDE225 on cancer stem cells. We observed that upon NVP-LDE225 treatment the cells had impaired ability to form spheres in vitro and similarly, tumor growth was delayed following in vivo serial transplantation. These experiments suggest that Smo inhibitors can have a high impact on the treatment of this very aggressive cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 4290.

Collaboration


Dive into the Yung-Mae Yao's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shifeng Pan

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Xu Wu

Harvard University

View shared research outputs
Researchain Logo
Decentralizing Knowledge