Network


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

Hotspot


Dive into the research topics where Phuong N. Le is active.

Publication


Featured researches published by Phuong N. Le.


Pharmacology & Therapeutics | 2015

Targeting the Wnt pathway in human cancers: therapeutic targeting with a focus on OMP-54F28.

Phuong N. Le; Jessica D. McDermott; Antonio Jimeno

The Wnt signaling pathways are a group of signal transduction pathways that play an important role in cell fate specification, cell proliferation and cell migration. Aberrant signaling in these pathways has been implicated in the development and progression of multiple cancers by allowing increased proliferation, angiogenesis, survival and metastasis. Activation of the Wnt pathway also contributes to the tumorigenicity of cancer stem cells (CSCs). Therefore, inhibiting this pathway has been a recent focus of cancer research with multiple targetable candidates in development. OMP-54F28 is a fusion protein that combines the cysteine-rich domain of frizzled family receptor 8 (Fzd8) with the immunoglobulin Fc domain that competes with the native Fzd8 receptor for its ligands and antagonizes Wnt signaling. Preclinical models with OMP-54F28 have shown reduced tumor growth and decreased CSC frequency as a single agent and in combination with other chemotherapeutic agents. Due to these findings, a phase 1a study is nearing completion with OMP-54F28 in advanced solid tumors and 3 phase 1b studies have been opened with OMP-54F28 in combination with standard-of-care chemotherapy backbones in ovarian, pancreatic and hepatocellular cancers. This article will review the Wnt signaling pathway, preclinical data on OMP-54F28 and other Wnt pathway inhibitors and ongoing clinical trials.


Molecular Oncology | 2013

A patient tumor transplant model of squamous cell cancer identifies PI3K inhibitors as candidate therapeutics in defined molecular bins

Stephen B. Keysar; David P. Astling; Ryan T. Anderson; Brian W. Vogler; Daniel W. Bowles; J. Jason Morton; Jeramiah J. Paylor; Magdalena J. Glogowska; Phuong N. Le; Justin R. Eagles-Soukup; Severine Kako; Sarah M. Takimoto; Daniel Sehrt; Adrian Umpierrez; Morgan A. Pittman; Sarah M. Macfadden; Ryan M. Helber; Scott Peterson; Diana F. Hausman; Sherif Said; Ted H. Leem; Julie A. Goddard; John J. Arcaroli; Wells A. Messersmith; William A. Robinson; Fred R. Hirsch; Marileila Varella-Garcia; David Raben; Xiao-Jing Wang; John I. Song

Targeted therapy development in head and neck squamous cell carcinoma (HNSCC) is challenging given the rarity of activating mutations. Additionally, HNSCC incidence is increasing related to human papillomavirus (HPV). We sought to develop an in vivo model derived from patients reflecting the evolving HNSCC epidemiologic landscape, and use it to identify new therapies. Primary and relapsed tumors from HNSCC patients, both HPV+ and HPV−, were implanted on mice, giving rise to 25 strains. Resulting xenografts were characterized by detecting key mutations, measuring protein expression by IHC and gene expression/pathway analysis by mRNA‐sequencing. Drug efficacy studies were run with representative xenografts using the approved drug cetuximab as well as the new PI3K inhibitor PX‐866. Tumors maintained their original morphology, genetic profiles and drug susceptibilities through serial passaging. The genetic makeup of these tumors was consistent with known frequencies of TP53, PI3KCA, NOTCH1 and NOTCH2 mutations. Because the EGFR inhibitor cetuximab is a standard HNSCC therapy, we tested its efficacy and observed a wide spectrum of efficacy. Cetuximab‐resistant strains had higher PI3K/Akt pathway gene expression and protein activation than cetuximab‐sensitive strains. The PI3K inhibitor PX‐866 had anti‐tumor efficacy in HNSCC models with PIK3CA alterations. Finally, PI3K inhibition was effective in two cases with NOTCH1 inactivating mutations. In summary, we have developed an HNSCC model covering its clinical spectrum whose major genetic alterations and susceptibility to anticancer agents represent contemporary HNSCC. This model enables to prospectively test therapeutic‐oriented hypotheses leading to personalized medicine.


Cancer Research | 2013

Hedgehog Signaling Alters Reliance on EGF Receptor Signaling and Mediates Anti-EGFR Therapeutic Resistance in Head and Neck Cancer

Stephen B. Keysar; Phuong N. Le; Ryan T. Anderson; J. Jason Morton; Daniel W. Bowles; Jeramiah J. Paylor; Brian W. Vogler; Jackie Thorburn; Pamela Fernandez; Magdalena J. Glogowska; Sarah M. Takimoto; Daniel Sehrt; Gregory Gan; Justin R. Eagles-Soukup; Hilary S. Serracino; Fred R. Hirsch; M. Scott Lucia; Andrew Thorburn; John I. Song; Xiao-Jing Wang; Antonio Jimeno

The EGF receptor (EGFR)-directed monoclonal antibody cetuximab is the only targeted therapy approved for the treatment of squamous cell carcinoma of the head and neck (HNSCC) but is only effective in a minority of patients. Epithelial-to-mesenchymal transition (EMT) has been implicated as a drug resistance mechanism in multiple cancers, and the EGFR and Hedgehog pathways (HhP) are relevant to this process, but the interplay between the two pathways has not been defined in HNSCC. Here, we show that HNSCC cells that were naturally sensitive to EGFR inhibition over time developed increased expression of the HhP transcription factor GLI1 as they became resistant after long-term EGFR inhibitor exposure. This robustly correlated with an increase in vimentin expression. Conversely, the HhP negatively regulated an EGFR-dependent, EMT-like state in HNSCC cells, and pharmacologic or genetic inhibition of HhP signaling pushed cells further into an EGFR-dependent phenotype, increasing expression of ZEB1 and VIM. In vivo treatment with cetuximab resulted in tumor shrinkage in four of six HNSCC patient-derived xenografts; however, they eventually regrew. Cetuximab in combination with the HhP inhibitor IPI-926 eliminated tumors in two cases and significantly delayed regrowth in the other two cases. Expression of EMT genes TWIST and ZEB2 was increased in sensitive xenografts, suggesting a possible resistant mesenchymal population. In summary, we report that EGFR-dependent HNSCC cells can undergo both EGFR-dependent and -independent EMT and HhP signaling is a regulator in both processes. Cetuximab plus IPI-926 forces tumor cells into an EGFR-dependent state, delaying or completely blocking tumor recurrence.


Oncogene | 2016

XactMice: humanizing mouse bone marrow enables microenvironment reconstitution in a patient-derived xenograft model of head and neck cancer

J. Jason Morton; Gregory H. Bird; Stephen B. Keysar; David P. Astling; Traci R. Lyons; Ryan T. Anderson; Magdalena J. Glogowska; Patricia A. Estes; Justin R. Eagles; Phuong N. Le; Gregory Gan; Brett McGettigan; Pamela Fernandez; Nuria Padilla-Just; Marileila Varella-Garcia; John I. Song; Daniel W. Bowles; Pepper Schedin; Aik Choon Tan; Dennis R. Roop; Xiao-Jing Wang; Yosef Refaeli; Antonio Jimeno

The limitations of cancer cell lines have led to the development of direct patient-derived xenograft models. However, the interplay between the implanted human cancer cells and recruited mouse stromal and immune cells alters the tumor microenvironment and limits the value of these models. To overcome these constraints, we have developed a technique to expand human hematopoietic stem and progenitor cells (HSPCs) and use them to reconstitute the radiation-depleted bone marrow of a NOD/SCID/IL2rg−/− (NSG) mouse on which a patient’s tumor is then transplanted (XactMice). The human HSPCs produce immune cells that home into the tumor and help replicate its natural microenvironment. Despite previous passage on nude mice, the expression of epithelial, stromal and immune genes in XactMice tumors aligns more closely to that of the patient tumor than to those grown in non-humanized mice—an effect partially facilitated by human cytokines expressed by both the HSPC progeny and the tumor cells. The human immune and stromal cells produced in the XactMice can help recapitulate the microenvironment of an implanted xenograft, reverse the initial genetic drift seen after passage on non-humanized mice and provide a more accurate tumor model to guide patient treatment.


Cancer Research | 2014

Hedgehog signaling drives radioresistance and stroma-driven tumor repopulation in head and neck squamous cancers

Gregory Gan; Justin R. Eagles; Stephen B. Keysar; Guoliang Wang; Magdalena J. Glogowska; Cem Altunbas; Ryan T. Anderson; Phuong N. Le; J. Jason Morton; Barbara Frederick; David Raben; Xiao-Jing Wang; Antonio Jimeno

Local control and overall survival in patients with advanced head and neck squamous cell cancer (HNSCC) remains dismal. Signaling through the Hedgehog (Hh) pathway is associated with epithelial-to-mesenchymal transition, and activation of the Hh effector transcription factor Gli1 is a poor prognostic factor in this disease setting. Here, we report that increased GLI1 expression in the leading edge of HNSCC tumors is further increased by irradiation, where it contributes to therapeutic inhibition. Hh pathway blockade with cyclopamine suppressed GLI1 activation and enhanced tumor sensitivity to radiotherapy. Furthermore, radiotherapy-induced GLI1 expression was mediated in part by the mTOR/S6K1 pathway. Stroma exposed to radiotherapy promoted rapid tumor repopulation, and this effect was suppressed by Hh inhibition. Our results demonstrate that Gli1 that is upregulated at the tumor-stroma intersection in HNSCC is elevated by radiotherapy, where it contributes to stromal-mediated resistance, and that Hh inhibitors offer a rational strategy to reverse this process to sensitize HNSCC to radiotherapy.


Journal of the National Cancer Institute | 2017

Regulation of Head and Neck Squamous Cancer Stem Cells by PI3K and SOX2.

Stephen B. Keysar; Phuong N. Le; Bettina Miller; Brian C. Jackson; Justin R. Eagles; Cera Nieto; Jihye Kim; Binwu Tang; Magdalena J. Glogowska; J. Jason Morton; Nuria Padilla-Just; Karina Gomez; Emily Warnock; Julie Reisinger; John J. Arcaroli; Wells A. Messersmith; Lalage M. Wakefield; Dexiang Gao; Aik Choon Tan; Hilary S. Serracino; Vasilis Vasiliou; Dennis R. Roop; Xiao-Jing Wang; Antonio Jimeno

Background: We have an incomplete understanding of the differences between cancer stem cells (CSCs) in human papillomavirus–positive (HPV-positive) and –negative (HPV-negative) head and neck squamous cell cancer (HNSCC). The PI3K pathway has the most frequent activating genetic events in HNSCC (especially HPV-positive driven), but the differential signaling between CSCs and non-CSCs is also unknown. Methods: We addressed these unresolved questions using CSCs identified from 10 HNSCC patient-derived xenografts (PDXs). Sorted populations were serially passaged in nude mice to evaluate tumorigenicity and tumor recapitulation. The transcription profile of HNSCC CSCs was characterized by mRNA sequencing, and the susceptibility of CSCs to therapy was investigated using an in vivo model. SOX2 transcriptional activity was used to follow the asymmetric division of PDX-derived CSCs. All statistical tests were two-sided. Results: CSCs were enriched by high aldehyde dehydrogenase (ALDH) activity and CD44 expression and were similar between HPV-positive and HPV-negative cases (percent tumor formation injecting ⩽ 1x103 cells: ALDH+CD44high = 65.8%, ALDH-CD44high = 33.1%, ALDH+CD44high = 20.0%; and injecting 1x105 cells: ALDH-CD44low = 4.4%). CSCs were resistant to conventional therapy and had PI3K/mTOR pathway overexpression (GSEA pathway enrichment, P < .001), and PI3K inhibition in vivo decreased their tumorigenicity (40.0%–100.0% across cases). PI3K/mTOR directly regulated SOX2 protein levels, and SOX2 in turn activated ALDH1A1 (P < .001 013C and 067C) expression and ALDH activity (ALDH+ [%] empty-control vs SOX2, 0.4% ± 0.4% vs 14.5% ± 9.8%, P = .03 for 013C and 1.7% ± 1.3% vs 3.6% ± 3.4%, P = .04 for 067C) in 013C and 067 cells. SOX2 enhanced sphere and tumor growth (spheres/well, 013C P < .001 and 067C P = .04) and therapy resistance. SOX2 expression prompted mesenchymal-to-epithelial transition (MET) by inducing CDH1 (013C P = .002, 067C P = .01), followed by asymmetric division and proliferation, which contributed to tumor formation. Conclusions: The molecular link between PI3K activation and CSC properties found in this study provides insights into therapeutic strategies for HNSCC. Constitutive expression of SOX2 in HNSCC cells generates a CSC-like population that enables CSC studies.


Molecular Cancer Therapeutics | 2013

The Dual Pathway Inhibitor Rigosertib Is Effective in Direct Patient Tumor Xenografts of Head and Neck Squamous Cell Carcinomas

Ryan T. Anderson; Stephen B. Keysar; Daniel W. Bowles; Magdalena J. Glogowska; David P. Astling; J. Jason Morton; Phuong N. Le; Adrian Umpierrez; Justin R. Eagles-Soukup; Gregory Gan; Brian W. Vogler; Daniel Sehrt; Sarah M. Takimoto; Dara L. Aisner; Francois Wilhelm; Barbara Frederick; Marileila Varella-Garcia; Aik Choon Tan; Antonio Jimeno

The dual pathway inhibitor rigosertib inhibits phosphoinositide 3-kinase (PI3K) pathway activation as well as polo-like kinase 1 (PLK1) activity across a broad spectrum of cancer cell lines. The importance of PIK3CA alterations in squamous cell carcinoma of the head and neck (HNSCC) has raised interest in exploring agents targeting PI3K, the product of PIK3CA. The genetic and molecular basis of rigosertib treatment response was investigated in a panel of 16 HNSCC cell lines, and direct patient tumor xenografts from eight patients with HNSCC [four HPV-serotype16 (HPV16)–positive]. HNSCC cell lines and xenografts were characterized by pathway enrichment gene expression analysis, exon sequencing, gene copy number, Western blotting, and immunohistochemistry (IHC). Rigosertib had potent antiproliferative effects on 11 of 16 HPV− HNSCC cell lines. Treatment sensitivity was confirmed in two cell lines using an orthotopic in vivo xenograft model. Growth reduction after rigosertib treatment was observed in three of eight HNSCC direct patient tumor lines. The responsive tumor lines carried a combination of a PI3KCA-activating event (amplification or mutation) and a p53-inactivating event (either HPV16- or mutation-mediated TP53 inactivation). In this study, we evaluated the in vitro and in vivo efficacy of rigosertib in both HPV+ and HPV− HNSCCs, focusing on inhibition of the PI3K pathway. Although consistent inhibition of the PI3K pathway was not evident in HNSCC, we identified a combination of PI3K/TP53 events necessary, but not sufficient, for rigosertib sensitivity. Mol Cancer Ther; 12(10); 1994–2005. ©2013 AACR.


Clinical Cancer Research | 2018

Salivary Gland Cancer Patient-Derived Xenografts Enable Characterization of Cancer Stem Cells and New Gene Events Associated with Tumor Progression

Stephen B. Keysar; Justin R. Eagles; Bettina Miller; Brian C. Jackson; Farshad N. Chowdhury; Julie Reisinger; Tugs-Saikhan Chimed; Phuong N. Le; John J. Morton; Hilary Somerset; Marileila Varella-Garcia; Aik Choon Tan; John I. Song; Daniel W. Bowles; Mary E. Reyland; Antonio Jimeno

Purpose: Salivary gland cancers (SGC) frequently present with distant metastases many years after diagnosis, suggesting a cancer stem cell (CSC) subpopulation that initiates late recurrences; however, current models are limited both in their availability and suitability to characterize these rare cells. Experimental Design: Patient-derived xenografts (PDX) were generated by engrafting patient tissue onto nude mice from one acinic cell carcinoma (AciCC), four adenoid cystic carcinoma (ACC), and three mucoepidermoid carcinoma (MEC) cases, which were derived from successive relapses from the same MEC patient. Patient and PDX samples were analyzed by RNA-seq and Exome-seq. Sphere formation potential and in vivo tumorigenicity was assessed by sorting for Aldefluor (ALDH) activity and CD44-expressing subpopulations. Results: For successive MEC relapses we found a time-dependent increase in CSCs (ALDH+CD44high), increasing from 0.2% to 4.5% (P=0.033), but more importantly we observed an increase in individual CSC sphere formation and tumorigenic potential. A 50% increase in mutational burden was documented in subsequent MEC tumors, and this was associated with increased expression of tumor-promoting genes (MT1E, LGR5, and LEF1), decreased expression of tumor-suppressor genes (CDKN2B, SIK1, and TP53), and higher expression of CSC-related proteins such as SOX2, MYC, and ALDH1A1. Finally, genomic analyses identified a novel NFIB–MTFR2 fusion in an ACC tumor and confirmed previously reported fusions (NTRK3–ETV6 and MYB–NFIB). Conclusions: Sequential MEC PDX models preserved key patient features and enabled the identification of genetic events putatively contributing to increases in both CSC proportion and intrinsic tumorigenicity, which mirrored the patients clinical course. Clin Cancer Res; 24(12); 2935–43. ©2018 AACR.


Cancer Research | 2003

A Selective Small Molecule Inhibitor of c-Met Kinase Inhibits c-Met-Dependent Phenotypes in Vitro and Exhibits Cytoreductive Antitumor Activity in Vivo

James G. Christensen; Randall Schreck; Jon Burrows; Poonam Kuruganti; Emily Chan; Phuong N. Le; Jeffrey H. Chen; Xueyan Wang; Lany Ruslim; Robert A. Blake; Kenneth E. Lipson; John Ramphal; Steven Do; Jingrong J. Cui; Julie M. Cherrington; Dirk B. Mendel


Molecular Cancer Therapeutics | 2003

Potent and selective inhibitors of the Met (hepatocyte growth factor/scatter factor (HGF/SF) receptor) tyrosine kinase block HGF/SF-induced tumor cell growth and invasion

Xueyan Wang; Phuong N. Le; Congxin Liang; Julie Chan; David Kiewlich; Todd W. Miller; Dave Harris; Li Sun; Audie Rice; Stefan Vasile; Robert A. Blake; Anthony R. Howlett; Neela Patel; Gerald McMahon; Kenneth E. Lipson

Collaboration


Dive into the Phuong N. Le's collaboration.

Top Co-Authors

Avatar

Antonio Jimeno

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Stephen B. Keysar

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

J. Jason Morton

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Daniel W. Bowles

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Ryan T. Anderson

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gregory Gan

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar

Justin R. Eagles

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge