Network


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

Hotspot


Dive into the research topics where Jason H. Cheng is active.

Publication


Featured researches published by Jason H. Cheng.


Nature Biotechnology | 2010

Assessing therapeutic responses in Kras mutant cancers using genetically engineered mouse models

Mallika Singh; Anthony Lima; Rafael Molina; Patricia Hamilton; Anne C Clermont; Vidusha Devasthali; Jennifer D. Thompson; Jason H. Cheng; Hani Bou Reslan; Calvin C K Ho; Timothy C Cao; Chingwei V. Lee; Michelle Nannini; Germaine Fuh; Richard A. D. Carano; Hartmut Koeppen; Ron Yu; William F. Forrest; Gregory D. Plowman; Leisa Johnson

The low rate of approval of novel anti-cancer agents underscores the need for better preclinical models of therapeutic response as neither xenografts nor early-generation genetically engineered mouse models (GEMMs) reliably predict human clinical outcomes. Whereas recent, sporadic GEMMs emulate many aspects of their human disease counterpart more closely, their ability to predict clinical therapeutic responses has never been tested systematically. We evaluated the utility of two state-of-the-art, mutant Kras-driven GEMMs—one of non-small-cell lung carcinoma and another of pancreatic adenocarcinoma—by assessing responses to existing standard-of-care chemotherapeutics, and subsequently in combination with EGFR and VEGF inhibitors. Standard clinical endpoints were modeled to evaluate efficacy, including overall survival and progression-free survival using noninvasive imaging modalities. Comparisons with corresponding clinical trials indicate that these GEMMs model human responses well, and lay the foundation for the use of validated GEMMs in predicting outcome and interrogating mechanisms of therapeutic response and resistance.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Oncogenic RAS pathway activation promotes resistance to anti-VEGF therapy through G-CSF–induced neutrophil recruitment

Vernon Phan; Xiumin Wu; Jason H. Cheng; Sheng Rx; Alicia S. Chung; Guanglei Zhuang; Christopher Tran; Qinghua Song; Marcin Kowanetz; Sambrone A; Martha Tan; Yu-Ju Gloria Meng; Jackson El; Franklin Peale; Melissa R. Junttila; Napoleone Ferrara

Granulocyte-colony stimulating factor (G-CSF) promotes mobilization of CD11b+Gr1+ myeloid cells and has been implicated in resistance to anti-VEGF therapy in mouse models. High G-CSF production has been associated with a poor prognosis in cancer patients. Here we show that activation of the RAS/MEK/ERK pathway regulates G-CSF expression through the Ets transcription factor. Several growth factors induced G-CSF expression by a MEK-dependent mechanism. Inhibition of G-CSF release with a MEK inhibitor markedly reduced G-CSF production in vitro and synergized with anti-VEGF antibodies to reduce CD11b+Ly6G+ neutrophil mobilization and tumor growth and led to increased survival in animal models of cancer, including a genetically engineered mouse model of pancreatic adenocarcinoma. Analysis of biopsies from pancreatic cancer patients revealed increased phospho-MEK, G-CSF, and Ets expression and enhanced neutrophil recruitment compared with normal pancreata. These results provide insights into G-CSF regulation and on the mechanism of action of MEK inhibitors and point to unique anticancer strategies.


The Journal of Pathology | 2012

Anti-VEGF antibody therapy does not promote metastasis in genetically engineered mouse tumour models†

Mallika Singh; Suzana S. Couto; William F. Forrest; Anthony Lima; Jason H. Cheng; Rafael Molina; Jason E. Long; Patricia Hamilton; Angela McNutt; Ian Kasman; Michelle Nannini; Hani Bou Reslan; Tim C. Cao; Calvin C K Ho; Kai H. Barck; Richard A. D. Carano; Oded Foreman; Jeffrey Eastham-Anderson; Adrian M. Jubb; Napoleone Ferrara; Leisa Johnson

Resistance to anti‐angiogenic therapy can occur via several potential mechanisms. Unexpectedly, recent studies showed that short‐term inhibition of either VEGF or VEGFR enhanced tumour invasiveness and metastatic spread in preclinical models. In an effort to evaluate the translational relevance of these findings, we examined the consequences of long‐term anti‐VEGF monoclonal antibody therapy in several well‐validated genetically engineered mouse tumour models of either neuroendocrine or epithelial origin. Anti‐VEGF therapy decreased tumour burden and increased overall survival, either as a single agent or in combination with chemotherapy, in all four models examined. Importantly, neither short‐ nor long‐term exposure to anti‐VEGF therapy altered the incidence of metastasis in any of these autochthonous models, consistent with retrospective analyses of clinical trials. In contrast, we observed that sunitinib treatment recapitulated previously reported effects on tumour invasiveness and metastasis in a pancreatic neuroendocrine tumour (PNET) model. Consistent with these results, sunitinib treatment resulted in an up‐regulation of the hypoxia marker GLUT1 in PNETs, whereas anti‐VEGF did not. These results indicate that anti‐VEGF mediates anti‐tumour effects and therapeutic benefits without a paradoxical increase in metastasis. Moreover, these data underscore the concept that drugs targeting VEGF ligands and receptors may affect tumour metastasis in a context‐dependent manner and are mechanistically distinct from one another. Copyright


Clinical Cancer Research | 2013

Identification and Analysis of In Vivo VEGF Downstream Markers Link VEGF Pathway Activity with Efficacy of Anti- VEGF Therapies

Matthew J. Brauer; Guanglei Zhuang; Maike Schmidt; Jenny Yao; Xiumin Wu; Joshua S. Kaminker; Stefanie S. Jurinka; Ganesh Kolumam; Alicia S. Chung; Adrian M. Jubb; Zora Modrusan; Tomoko Ozawa; C. David James; Heidi S. Phillips; Benjamin Haley; Rachel Tam; Anne C Clermont; Jason H. Cheng; Sherry X. Yang; Sandra M. Swain; Daniel Chen; Stefan J. Scherer; Hartmut Koeppen; Ru Fang Yeh; Peng Yue; Jean Philippe Stephan; Priti Hegde; Napoleone Ferrara; Mallika Singh; Carlos Bais

Purpose: The aim of this study was to identify conserved pharmacodynamic and potential predictive biomarkers of response to anti-VEGF therapy using gene expression profiling in preclinical tumor models and in patients. Experimental Design: Surrogate markers of VEGF inhibition [VEGF-dependent genes or VEGF-dependent vasculature (VDV)] were identified by profiling gene expression changes induced in response to VEGF blockade in preclinical tumor models and in human biopsies from patients treated with anti-VEGF monoclonal antibodies. The potential value of VDV genes as candidate predictive biomarkers was tested by correlating high or low VDV gene expression levels in pretreatment clinical samples with the subsequent clinical efficacy of bevacizumab (anti-VEGF)-containing therapy. Results: We show that VDV genes, including direct and more distal VEGF downstream endothelial targets, enable detection of VEGF signaling inhibition in mouse tumor models and human tumor biopsies. Retrospective analyses of clinical trial data indicate that patients with higher VDV expression in pretreatment tumor samples exhibited improved clinical outcome when treated with bevacizumab-containing therapies. Conclusions: In this work, we identified surrogate markers (VDV genes) for in vivo VEGF signaling in tumors and showed clinical data supporting a correlation between pretreatment VEGF bioactivity and the subsequent efficacy of anti-VEGF therapy. We propose that VDV genes are candidate biomarkers with the potential to aid the selection of novel indications as well as patients likely to respond to anti-VEGF therapy. The data presented here define a diagnostic biomarker hypothesis based on translational research that warrants further evaluation in additional retrospective and prospective trials. Clin Cancer Res; 19(13); 3681–92. ©2013 AACR.


Molecular Cancer Therapeutics | 2015

Modeling Targeted Inhibition of MEK and PI3 Kinase in Human Pancreatic Cancer

Melissa R. Junttila; Vidusha Devasthali; Jason H. Cheng; Joseph Castillo; Ciara Metcalfe; Anne C Clermont; Douglas Den Otter; Emily Chan; Hani Bou-Reslan; Tim C. Cao; William F. Forrest; Michelle Nannini; Dorothy French; Richard A. D. Carano; Mark Merchant; Klaus P. Hoeflich; Mallika Singh

Activating mutations in the KRAS oncogene occur in approximately 90% of pancreatic cancers, resulting in aberrant activation of the MAPK and the PI3K pathways, driving malignant progression. Significant efforts to develop targeted inhibitors of nodes within these pathways are underway and several are currently in clinical trials for patients with KRAS-mutant tumors, including patients with pancreatic cancer. To model MEK and PI3K inhibition in late-stage pancreatic cancer, we conducted preclinical trials with a mutant Kras-driven genetically engineered mouse model that faithfully recapitulates human pancreatic ductal adenocarcinoma development. Treatment of advanced disease with either a MEK (GDC-0973) or PI3K inhibitor (GDC-0941) alone showed modest tumor growth inhibition and did not significantly enhance overall survival. However, combination of the two agents resulted in a significant survival advantage as compared with control tumor-bearing mice. To model the clinical scenario, we also evaluated the combination of these targeted agents with gemcitabine, the current standard-of-care chemotherapy for pancreatic cancer. The addition of MEK or PI3K inhibition to gemcitabine, or the triple combination regimen, incrementally enhanced overall survival as compared with gemcitabine alone. These results are reminiscent of the survival advantage conferred in this model and in patients by the combination of gemcitabine and erlotinib, an approved therapeutic regimen for advanced nonresectable pancreatic cancer. Taken together, these data indicate that inhibition of MEK and PI3K alone or in combination with chemotherapy do not confer a dramatic improvement as compared with currently available therapies for patients with pancreatic cancer. Mol Cancer Ther; 14(1); 40–47. ©2014 AACR.


Journal of Clinical Investigation | 2013

Anti-EGFL7 antibodies enhance stress-induced endothelial cell death and anti-VEGF efficacy

Leisa Johnson; Mahrukh Huseni; Tanya Smyczek; Anthony Lima; Stacey Yeung; Jason H. Cheng; Rafael Molina; David Kan; Ann De Mazière; Judith Klumperman; Ian Kasman; Yin Zhang; Mark S. Dennis; Jeffrey Eastham-Anderson; Adrian M. Jubb; Olivia Hwang; Rupal Desai; Maike Schmidt; Michelle Nannini; Kai H. Barck; Richard A. D. Carano; William F. Forrest; Qinghua Song; Daniel S. Chen; Louie Naumovski; Mallika Singh; Weilan Ye; Priti Hegde

Many oncology drugs are administered at their maximally tolerated dose without the knowledge of their optimal efficacious dose range. In this study, we describe a multifaceted approach that integrated preclinical and clinical data to identify the optimal dose for an antiangiogenesis agent, anti-EGFL7. EGFL7 is an extracellular matrix-associated protein expressed in activated endothelium. Recombinant EGFL7 protein supported EC adhesion and protected ECs from stress-induced apoptosis. Anti-EGFL7 antibodies inhibited both of these key processes and augmented anti-VEGF-mediated vascular damage in various murine tumor models. In a genetically engineered mouse model of advanced non-small cell lung cancer, we found that anti-EGFL7 enhanced both the progression-free and overall survival benefits derived from anti-VEGF therapy in a dose-dependent manner. In addition, we identified a circulating progenitor cell type that was regulated by EGFL7 and evaluated the response of these cells to anti-EGFL7 treatment in both tumor-bearing mice and cancer patients from a phase I clinical trial. Importantly, these preclinical efficacy and clinical biomarker results enabled rational selection of the anti-EGFL7 dose currently being tested in phase II clinical trials.


PLOS ONE | 2013

Early Detection of Lewis Lung Carcinoma Tumor Control by Irradiation Using Diffusion-Weighted and Dynamic Contrast-Enhanced MRI

Jason H. Cheng; Ang Yuan; Jyh-Horng Chen; Yi Chien Lu; Kuan Hung Cho; Jian Kuen Wu; Chien Jang Wu; Yeun-Chung Chang; Pan-Chyr Yang

Purpose To investigate the correlation between diffusion-weighted (DW) and dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) derived parameters and radioresponsiveness of Lewis lung carcinoma (LLC) tumor. Materials and Methods LLC tumor growth in C57BL/6 mouse limb was used for the experiment. The tumors were irradiated with 10 Gy×5, or 30 Gy×2 vs. sham irradiation. Fourteen tumors were subjected to DW-MRI and DCE-MRI pre-radiotherapy and weekly imaging after radiotherapy. The temporal changes in apparent diffusion coefficient (ADC) and DCE-MRI derived parameters (Ktrans, kep, ve, and vp) were correlated with tumor size, and were histologically compared with CD31 staining of resected tumors. Results The 10 Gy×5 dose inhibited tumor growth for a week, while 30 Gy×2 controlled tumor growth for a 3-week observation period. One week after radiotherapy (week 2), irradiated tumors showed significantly higher values of ADC than untreated ones (10 Gy×5, p = 0.004; 30 Gy×2, p = 0.01). Significantly higher values of ve were shown earlier by 30 Gy×2 vs. sham (p = 0.01) and 10 Gy×5 vs. sham irradiation (p = 0.05). Sustained higher ve from 10 Gy×5 compared to sham irradiated tumors was evident at week 3 (p = 0.016) and week 4 (p = 0.046). A 13.8% early increase in ADC for 30 Gy×2 tumor group (p = 0.002) and a 16.5% increase for 10 Gy×5 group were noted (p = 0.01) vs. sham irradiation (which showed a 2.2% decrease). No differences were found for Ktrans, kep, or vp. Both radiotherapy groups demonstrated significant reduction in microvessel counts. Conclusion Early increase in ADC and ve correlated with tumor control by irradiation.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Kras mutant genetically engineered mouse models of human cancers are genomically heterogeneous

Wei-Jen Chung; Anneleen Daemen; Jason H. Cheng; Jason E. Long; Jonathan E. Cooper; Bu-Er Wang; Christopher Tran; Mallika Singh; Florian Gnad; Zora Modrusan; Oded Foreman; Melissa R. Junttila

Significance RAS mutant cancers represent a large unmet clinical need. Kras mutant genetically engineered mouse models (GEMMs) of cancer recapitulate disease characteristics and are relied upon preclinically to validate targets and test therapies. Our integrative analysis of GEMM tumors revealed significantly evolved genetic heterogeneity, a common feature of human tumors that undermines therapeutic responses. Moreover, interspecies comparative analyses showed the extent of gene-level fidelity between altered oncogenes and tumor suppressors. The genomic diversity represents an unrecognized opportunity to identify therapeutically susceptible genomic subsets preclinically. Moreover, this more-thorough understanding of the unappreciated complexity in these model systems ultimately allows for better interpretation and translatability of preclinical GEMM data for the benefit of cancer patients. KRAS mutant tumors are largely recalcitrant to targeted therapies. Genetically engineered mouse models (GEMMs) of Kras mutant cancer recapitulate critical aspects of this disease and are widely used for preclinical validation of targets and therapies. Through comprehensive profiling of exomes and matched transcriptomes of >200 KrasG12D-initiated GEMM tumors from one lung and two pancreatic cancer models, we discover that significant intratumoral and intertumoral genomic heterogeneity evolves during tumorigenesis. Known oncogenes and tumor suppressor genes, beyond those engineered, are mutated, amplified, and deleted. Unlike human tumors, the GEMM genomic landscapes are dominated by copy number alterations, while protein-altering mutations are rare. However, interspecies comparative analyses of the genomic landscapes demonstrate fidelity between genes altered in KRAS mutant human and murine tumors. Genes that are spontaneously altered during murine tumorigenesis are also among the most prevalent found in human indications. Using targeted therapies, we also demonstrate that this inherent tumor heterogeneity can be exploited preclinically to discover cancer-specific and genotype-specific therapeutic vulnerabilities. Focusing on Kras allelic imbalance, a feature shared by all three models, we discover that MAPK pathway inhibition impinges uniquely on this event, indicating distinct susceptibility and fitness advantage of Kras-mutant cells. These data reveal previously unknown genomic diversity among KrasG12D-initiated GEMM tumors, places them in context of human patients, and demonstrates how to exploit this inherent tumor heterogeneity to discover therapeutic vulnerabilities.


PLOS ONE | 2017

Combined MEK and ERK inhibition overcomes therapy-mediated pathway reactivation in RAS mutant tumors

Mark Merchant; John Moffat; Gabriele Schaefer; Jocelyn Chan; Xi Wang; Christine Orr; Jason H. Cheng; Thomas Hunsaker; Lily Shao; Stephanie J. Wang; Marie-Claire Wagle; Eva Lin; Peter M. Haverty; Sheerin Shahidi-Latham; Hai Ngu; Margaret Solon; Jeffrey Eastham-Anderson; Hartmut Koeppen; Shih-Min A. Huang; Jacob B. Schwarz; Marcia Belvin; Daniel C. Kirouac; Melissa R. Junttila

Mitogen-activated protein kinase (MAPK) pathway dysregulation is implicated in >30% of all cancers, rationalizing the development of RAF, MEK and ERK inhibitors. While BRAF and MEK inhibitors improve BRAF mutant melanoma patient outcomes, these inhibitors had limited success in other MAPK dysregulated tumors, with insufficient pathway suppression and likely pathway reactivation. In this study we show that inhibition of either MEK or ERK alone only transiently inhibits the MAPK pathway due to feedback reactivation. Simultaneous targeting of both MEK and ERK nodes results in deeper and more durable suppression of MAPK signaling that is not achievable with any dose of single agent, in tumors where feedback reactivation occurs. Strikingly, combined MEK and ERK inhibition is synergistic in RAS mutant models but only additive in BRAF mutant models where the RAF complex is dissociated from RAS and thus feedback productivity is disabled. We discovered that pathway reactivation in RAS mutant models occurs at the level of CRAF with combination treatment resulting in a markedly more active pool of CRAF. However, distinct from single node targeting, combining MEK and ERK inhibitor treatment effectively blocks the downstream signaling as assessed by transcriptional signatures and phospho-p90RSK. Importantly, these data reveal that MAPK pathway inhibitors whose activity is attenuated due to feedback reactivation can be rescued with sufficient inhibition by using a combination of MEK and ERK inhibitors. The MEK and ERK combination significantly suppresses MAPK pathway output and tumor growth in vivo to a greater extent than the maximum tolerated doses of single agents, and results in improved anti-tumor activity in multiple xenografts as well as in two Kras mutant genetically engineered mouse (GEM) models. Collectively, these data demonstrate that combined MEK and ERK inhibition is functionally unique, yielding greater than additive anti-tumor effects and elucidates a highly effective combination strategy in MAPK-dependent cancer, such as KRAS mutant tumors.


Nature Communications | 2018

Manic Fringe deficiency imposes Jagged1 addiction to intestinal tumor cells

Erika López-Arribillaga; Verónica Rodilla; Carlota Colomer; Anna Vert; Amy Shelton; Jason H. Cheng; Bing Yan; Abel Gonzalez-Perez; Melissa R. Junttila; Mar Iglesias; Ferran Torres; Joan Albanell; Alberto Villanueva; Anna Bigas; Christian W. Siebel; Lluis Espinosa

Delta ligands regulate Notch signaling in normal intestinal stem cells, while Jagged1 activates Notch in intestinal adenomas carrying active β-catenin. We used the ApcMin/+ mouse model, tumor spheroid cultures, and patient-derived orthoxenografts to address this divergent ligand-dependent Notch function and its implication in disease. We found that intestinal-specific Jag1 deletion or antibody targeting Jag1 prevents tumor initiation in mice. Addiction to Jag1 is concomitant with the absence of Manic Fringe (MFNG) in adenoma cells, and its ectopic expression reverts Jag1 dependence. In 239 human colorectal cancer patient samples, MFNG imposes a negative correlation between Jag1 and Notch, being high Jag1 in the absence of MFNG predictive of poor prognosis. Jag1 antibody treatment reduces patient-derived tumor orthoxenograft growth without affecting normal intestinal mucosa. Our data provide an explanation to Jag1 dependence in cancer, and reveal that Jag1–Notch1 interference provides therapeutic benefit in a subset of colorectal cancer and FAP syndrome patients.In intestinal adenomas carrying active β-catenin, Jagged1 (Jag1) ligand is responsible for Notch1 activation. Here, the authors show the reliance of Notch on Jag1 in cancer, and investigate how Jag1–Notch1 signaling interference may provide therapeutic benefits in some colorectal cancer patients

Collaboration


Dive into the Jason H. Cheng's collaboration.

Researchain Logo
Decentralizing Knowledge