Network


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

Hotspot


Dive into the research topics where Joshua Korn is active.

Publication


Featured researches published by Joshua Korn.


Nature Medicine | 2015

High-throughput screening using patient-derived tumor xenografts to predict clinical trial drug response

Hui Gao; Joshua Korn; Stephane Ferretti; John E. Monahan; Youzhen Wang; Mallika Singh; Chao Zhang; Christian Schnell; Guizhi Yang; Yun Zhang; O Alejandro Balbin; Stéphanie Barbe; Hongbo Cai; Fergal Casey; Susmita Chatterjee; Derek Y. Chiang; Shannon Chuai; Shawn M Cogan; Scott D Collins; Ernesta Dammassa; Nicolas Ebel; Millicent Embry; John Green; Audrey Kauffmann; Colleen Kowal; Rebecca J. Leary; Joseph Lehar; Ying Liang; Alice Loo; Edward Lorenzana

Profiling candidate therapeutics with limited cancer models during preclinical development hinders predictions of clinical efficacy and identifying factors that underlie heterogeneous patient responses for patient-selection strategies. We established ∼1,000 patient-derived tumor xenograft models (PDXs) with a diverse set of driver mutations. With these PDXs, we performed in vivo compound screens using a 1 × 1 × 1 experimental design (PDX clinical trial or PCT) to assess the population responses to 62 treatments across six indications. We demonstrate both the reproducibility and the clinical translatability of this approach by identifying associations between a genotype and drug response, and established mechanisms of resistance. In addition, our results suggest that PCTs may represent a more accurate approach than cell line models for assessing the clinical potential of some therapeutic modalities. We therefore propose that this experimental paradigm could potentially improve preclinical evaluation of treatment modalities and enhance our ability to predict clinical trial responses.


Nature Medicine | 2014

Pharmacological and genomic profiling identifies NF-κB–targeted treatment strategies for mantle cell lymphoma

Rami Rahal; Mareike Frick; Rodrigo Romero; Joshua Korn; Robert Kridel; Fong Chun Chan; Barbara Meissner; Hyo-eun C. Bhang; Dave Ruddy; Audrey Kauffmann; Ali Farsidjani; Adnan Derti; Daniel Rakiec; Tara L. Naylor; Estelle Pfister; Steve Kovats; Sunkyu Kim; Kerstin Dietze; Bernd Dörken; Christian Steidl; Alexandar Tzankov; Michael Hummel; John E. Monahan; Michael Morrissey; Christine Fritsch; William R. Sellers; Vesselina G. Cooke; Randy D. Gascoyne; Georg Lenz; Frank Stegmeier

Mantle cell lymphoma (MCL) is an aggressive malignancy that is characterized by poor prognosis. Large-scale pharmacological profiling across more than 100 hematological cell line models identified a subset of MCL cell lines that are highly sensitive to the B cell receptor (BCR) signaling inhibitors ibrutinib and sotrastaurin. Sensitive MCL models exhibited chronic activation of the BCR-driven classical nuclear factor-κB (NF-κB) pathway, whereas insensitive cell lines displayed activation of the alternative NF-κB pathway. Transcriptome sequencing revealed genetic lesions in alternative NF-κB pathway signaling components in ibrutinib-insensitive cell lines, and sequencing of 165 samples from patients with MCL identified recurrent mutations in TRAF2 or BIRC3 in 15% of these individuals. Although they are associated with insensitivity to ibrutinib, lesions in the alternative NF-κB pathway conferred dependence on the protein kinase NIK (also called mitogen-activated protein 3 kinase 14 or MAP3K14) both in vitro and in vivo. Thus, NIK is a new therapeutic target for MCL treatment, particularly for lymphomas that are refractory to BCR pathway inhibitors. Our findings reveal a pattern of mutually exclusive activation of the BCR–NF-κB or NIK–NF-κB pathways in MCL and provide critical insights into patient stratification strategies for NF-κB pathway–targeted agents.


Nature Medicine | 2015

Studying clonal dynamics in response to cancer therapy using high-complexity barcoding

Hyo-eun C. Bhang; David A. Ruddy; Viveksagar Krishnamurthy Radhakrishna; Justina X. Caushi; Rui Zhao; Matthew M Hims; Angad P. Singh; Iris Kao; Daniel Rakiec; Pamela Shaw; Marissa Balak; Alina Raza; Elizabeth Ackley; Nicholas Keen; Michael R. Schlabach; Michael Palmer; Rebecca J. Leary; Derek Y. Chiang; William R. Sellers; Franziska Michor; Vesselina G. Cooke; Joshua Korn; Frank Stegmeier

Resistance to cancer therapies presents a significant clinical challenge. Recent studies have revealed intratumoral heterogeneity as a source of therapeutic resistance. However, it is unclear whether resistance is driven predominantly by pre-existing or de novo alterations, in part because of the resolution limits of next-generation sequencing. To address this, we developed a high-complexity barcode library, ClonTracer, which enables the high-resolution tracking of more than 1 million cancer cells under drug treatment. In two clinically relevant models, ClonTracer studies showed that the majority of resistant clones were part of small, pre-existing subpopulations that selectively escaped under therapeutic challenge. Moreover, the ClonTracer approach enabled quantitative assessment of the ability of combination treatments to suppress resistant clones. These findings suggest that resistant clones are present before treatment, which would make up-front therapeutic combinations that target non-overlapping resistance a preferred approach. Thus, ClonTracer barcoding may be a valuable tool for optimizing therapeutic regimens with the goal of curative combination therapies for cancer.


Cancer Discovery | 2016

CRISPR Screens Provide a Comprehensive Assessment of Cancer Vulnerabilities but Generate False-Positive Hits for Highly Amplified Genomic Regions

Diana M Munoz; Pamela J. Cassiani; Li Li; Eric Billy; Joshua Korn; Michael D. Jones; Javad Golji; David A. Ruddy; Kristine Yu; Gregory McAllister; Antoine deWeck; Dorothee Abramowski; Jessica Wan; Matthew D. Shirley; Sarah Y. Neshat; Daniel Rakiec; Rosalie de Beaumont; Odile Weber; Audrey Kauffmann; E. Robert McDonald; Nicholas Keen; Francesco Hofmann; William R. Sellers; Tobias Schmelzle; Frank Stegmeier; Michael R. Schlabach

UNLABELLED CRISPR/Cas9 has emerged as a powerful new tool to systematically probe gene function. We compared the performance of CRISPR to RNAi-based loss-of-function screens for the identification of cancer dependencies across multiple cancer cell lines. CRISPR dropout screens consistently identified more lethal genes than RNAi, implying that the identification of many cellular dependencies may require full gene inactivation. However, in two aneuploid cancer models, we found that all genes within highly amplified regions, including nonexpressed genes, scored as lethal by CRISPR, revealing an unanticipated class of false-positive hits. In addition, using a CRISPR tiling screen, we found that sgRNAs targeting essential domains generate the strongest lethality phenotypes and thus provide a strategy to rapidly define the protein domains required for cancer dependence. Collectively, these findings not only demonstrate the utility of CRISPR screens in the identification of cancer-essential genes, but also reveal the need to carefully control for false-positive results in chromosomally unstable cancer lines. SIGNIFICANCE We show in this study that CRISPR-based screens have a significantly lower false-negative rate compared with RNAi-based screens, but have specific liabilities particularly in the interrogation of regions of genome amplification. Therefore, this study provides critical insights for applying CRISPR-based screens toward the systematic identification of new cancer targets. Cancer Discov; 6(8); 900-13. ©2016 AACR.See related commentary by Sheel and Xue, p. 824See related article by Aguirre et al., p. 914This article is highlighted in the In This Issue feature, p. 803.


Journal of Biological Chemistry | 2012

Isocitrate Dehydrogenase (IDH) Mutations Promote a Reversible ZEB1/MicroRNA (miR)-200-dependent Epithelial-Mesenchymal Transition (EMT)

Alexandra R. Grassian; Fallon Lin; Rosemary Barrett; Yue Liu; Wei Jiang; Manav Korpal; Holly Astley; Daniel Gitterman; Thomas Henley; Rob Howes; Julian Levell; Joshua Korn; Raymond Pagliarini

Background: Isocitrate dehydrogenase (IDH) mutations occur in diverse tumor types, leading to production of the oncometabolite 2-hydroxyglutarate (2-HG). Results: High 2-HG levels lead to a reversible epithelial-mesenchymal transition (EMT) phenotype, which is dependent on ZEB1/miR-200. Conclusion: Mutant IDH reversibly disrupts normal epithelial morphology through EMT induction, a possible tumorigenic mechanism. Significance: This is the first report of a reversible mutant IDH-dependent signaling phenotype. Mutations in the genes encoding isocitrate dehydrogenase 1 and 2 (IDH1/2) occur in a variety of tumor types, resulting in production of the proposed oncometabolite, 2-hydroxyglutarate (2-HG). How mutant IDH and 2-HG alter signaling pathways to promote cancer, however, remains unclear. Additionally, there exist relatively few cell lines with IDH mutations. To examine the effect of endogenous IDH mutations and 2-HG, we created a panel of isogenic epithelial cell lines with either wild-type IDH1/2 or clinically relevant IDH1/2 mutations. Differences were noted in the ability of IDH mutations to cause robust 2-HG accumulation. IDH1/2 mutants that produce high levels of 2-HG cause an epithelial-mesenchymal transition (EMT)-like phenotype, characterized by changes in EMT-related gene expression and cellular morphology. 2-HG is sufficient to recapitulate aspects of this phenotype in the absence of an IDH mutation. In the cells types examined, mutant IDH-induced EMT is dependent on up-regulation of the transcription factor ZEB1 and down-regulation of the miR-200 family of microRNAs. Furthermore, sustained knockdown of IDH1 in IDH1 R132H mutant cells is sufficient to reverse many characteristics of EMT, demonstrating that continued expression of mutant IDH is required to maintain this phenotype. These results suggest mutant IDH proteins can reversibly deregulate discrete signaling pathways that contribute to tumorigenesis.


Oncogene | 2015

TMPRSS2:ERG blocks neuroendocrine and luminal cell differentiation to maintain prostate cancer proliferation

Zineb Mounir; Fallon Lin; V G Lin; Joshua Korn; Y Yu; R Valdez; O H Aina; Gilles Buchwalter; A B Jaffe; M Korpal; P Zhu; Myles Brown; R D Cardiff; J L Rocnik; Y Yang; Raymond Pagliarini

The biological outcome of TMPRSS2:ERG chromosomal translocations in prostate cancer (PC) remains poorly understood. To address this, we compared the transcriptional effects of TMPRSS2:ERG expression in a transgenic mouse model with those of ERG knockdown in a TMPRSS2:ERG-positive PC cell line. This reveals that ERG represses the expression of a previously unreported set of androgen receptor (AR)—independent neuronal genes that are indicative of neuroendocrine (NE) cell differentiation—in addition to previously reported AR-regulated luminal genes. Cell sorting and proliferation assays performed after sustained ERG knockdown indicate that ERG drives proliferation and blocks the differentiation of prostate cells to both NE and luminal cell types. Inhibition of ERG expression in TMPRSS2:ERG-positive PC cells through blockade of AR signaling is tracked with increased NE gene expression. We also provide evidence that these NE cells are resistant to pharmacological AR inhibition and can revert to the phenotype of parental cells upon restoration of AR/ERG signaling. Our findings highlight an ERG-regulated mechanism capable of repopulating the parent tumor through the transient generation of an anti-androgen therapy-resistant cell population, suggesting that ERG may have a direct role in preventing resistance to anti-androgen therapy.


Cancer Research | 2014

Inhibiting Tankyrases sensitizes KRAS mutant cancer cells to MEK inhibitors via FGFR2 feedback signaling

Marie Schoumacher; Kristen E. Hurov; Joseph Lehar; Yan Yan-Neale; Yuji Mishina; Dmitriy Sonkin; Joshua Korn; Daisy Flemming; Michael D. Jones; Brandon Antonakos; Vesselina G. Cooke; Janine Steiger; Jebediah Ledell; Mark Stump; William R. Sellers; Nika N. Danial; Wenlin Shao

Tankyrases (TNKS) play roles in Wnt signaling, telomere homeostasis, and mitosis, offering attractive targets for anticancer treatment. Using unbiased combination screening in a large panel of cancer cell lines, we have identified a strong synergy between TNKS and MEK inhibitors (MEKi) in KRAS-mutant cancer cells. Our study uncovers a novel function of TNKS in the relief of a feedback loop induced by MEK inhibition on FGFR2 signaling pathway. Moreover, dual inhibition of TNKS and MEK leads to more robust apoptosis and antitumor activity both in vitro and in vivo than effects observed by previously reported MEKi combinations. Altogether, our results show how a novel combination of TNKS and MEK inhibitors can be highly effective in targeting KRAS-mutant cancers by suppressing a newly discovered resistance mechanism.


Cancer Research | 2015

Inhibition of casein kinase 1 alpha prevents acquired drug resistance to erlotinib in EGFR-mutant non-small cell lung cancer

Alexandra B. Lantermann; Dongshu Chen; Kaitlin J McCutcheon; Gregory R. Hoffman; Elizabeth Frias; David A. Ruddy; Daniel Rakiec; Joshua Korn; Gregory McAllister; Frank Stegmeier; Matthew John Meyer; Sreenath V. Sharma

Patients with lung tumors harboring activating mutations in the EGF receptor (EGFR) show good initial treatment responses to the EGFR tyrosine kinase inhibitors (TKI) erlotinib or gefitinib. However, acquired resistance invariably develops. Applying a focused shRNA screening approach to identify genes whose knockdown can prevent and/or overcome acquired resistance to erlotinib in several EGFR-mutant non-small cell lung cancer (NSCLC) cell lines, we identified casein kinase 1 α (CSNK1A1, CK1α). We found that CK1α suppression inhibits the NF-κB prosurvival signaling pathway. Furthermore, downregulation of NF-κB signaling by approaches independent of CK1α knockdown can also attenuate acquired erlotinib resistance, supporting a role for activated NF-κB signaling in conferring acquired drug resistance. Importantly, CK1α suppression prevented erlotinib resistance in an HCC827 xenograft model in vivo. Our findings suggest that patients with EGFR-mutant NSCLC might benefit from a combination of EGFR TKIs and CK1α inhibition to prevent acquired drug resistance and to prolong disease-free survival.


eLife | 2016

ERG signaling in prostate cancer is driven through PRMT5-dependent methylation of the Androgen Receptor

Zineb Mounir; Joshua Korn; Thomas Westerling; Fallon Lin; Christina A. Kirby; Markus Schirle; Gregg McAllister; Greg Hoffman; Nadire Ramadan; Anke Hartung; Yan Feng; David Randal Kipp; Christopher Quinn; Michelle Fodor; Jason Baird; Marie Schoumacher; Ronald Meyer; James Deeds; Gilles Buchwalter; Travis Stams; Nicholas Keen; William R. Sellers; Myles Brown; Raymond Pagliarini

The TMPRSS2:ERG gene fusion is common in androgen receptor (AR) positive prostate cancers, yet its function remains poorly understood. From a screen for functionally relevant ERG interactors, we identify the arginine methyltransferase PRMT5. ERG recruits PRMT5 to AR-target genes, where PRMT5 methylates AR on arginine 761. This attenuates AR recruitment and transcription of genes expressed in differentiated prostate epithelium. The AR-inhibitory function of PRMT5 is restricted to TMPRSS2:ERG-positive prostate cancer cells. Mutation of this methylation site on AR results in a transcriptionally hyperactive AR, suggesting that the proliferative effects of ERG and PRMT5 are mediated through attenuating AR’s ability to induce genes normally involved in lineage differentiation. This provides a rationale for targeting PRMT5 in TMPRSS2:ERG positive prostate cancers. Moreover, methylation of AR at arginine 761 highlights a mechanism for how the ERG oncogene may coax AR towards inducing proliferation versus differentiation. DOI: http://dx.doi.org/10.7554/eLife.13964.001


Molecular Cancer Research | 2017

BRAF-inhibitor associated MEK Mutations Increase RAF-dependent and-independent Enzymatic Activity

Caroline Emery; Kelli-Ann Monaco; Ping Wang; Marissa Balak; Alyson K Freeman; Jodi Meltzer; Scott Delach; Daniel Rakiec; David A. Ruddy; Joshua Korn; Jacob R. Haling; Michael G. Acker; Giordano Caponigro

Alterations in MEK1/2 occur in cancers, both in the treatment-naïve state and following targeted therapies, most notably BRAF and MEK inhibitors in BRAF-V600E–mutant melanoma and colorectal cancer. Efforts were undertaken to understand the effects of these mutations, based upon protein structural location, and MEK1/2 activity. Two categories of MEK1/2 alterations were evaluated, those associated with either the allosteric pocket or helix-A. Clinically, MEK1/2 alterations of the allosteric pocket are rare and we demonstrate that they confer resistance to MEK inhibitors, while retaining sensitivity to BRAF inhibition. Most mutations described in patients fall within, or are associated with, helix-A. Mutations in this region reduce sensitivity to both BRAF and MEK inhibition and display elevated phospho-ERK1/2 levels, independent from increases in phospho-MEK1/2. Biochemical experiments with a representative helix-A variant, MEK1-Q56P, reveal both increased catalytic efficiency of the activated enzyme, and phosphorylation-independent activity relative to wild-type MEK1. Consistent with these findings, MEK1/2 alterations in helix A retain sensitivity to downstream antagonism via pharmacologic inhibition of ERK1/2. This work highlights the importance of classifying mutations based on structural and phenotypic consequences, both in terms of pathway signaling output and response to pharmacologic inhibition. Implications: This study suggests that alternate modes of target inhibition, such as ERK inhibition, will be required to effectively treat tumors harboring these MEK1/2-resistant alleles. Mol Cancer Res; 15(10); 1431–44. ©2017 AACR.

Collaboration


Dive into the Joshua Korn's collaboration.

Researchain Logo
Decentralizing Knowledge