Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2008
    In:  Cancer Research Vol. 68, No. 19 ( 2008-10-01), p. 7947-7955
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 68, No. 19 ( 2008-10-01), p. 7947-7955
    Abstract: The Mre11-Rad50-Nbs1 complex and autophosphorylated Ser1981-ATM are involved in recognizing and repairing DNA damage, such as double-strand breaks (DSB). However, the role of these factors in response to stalled replication forks is not clear. Nucleoside analogues are agents that are incorporated into DNA during replication, which cause stalling of replication forks. The molecular mechanisms that sense these events may signal for DNA repair and contribute to survival but are poorly understood. Cellular responses to both DSBs and stalled replication forks are marked by H2AX phosphorylation on Ser139 (γ-H2AX), which forms nuclear foci at sites of DNA damage. Here, concentrations of the nucleoside analogues 1-β-d-arabinofuranosylcytosine (cytarabine; ara-C), gemcitabine, and troxacitabine, which inhibited DNA synthesis by 90% within 2 hours, were determined for each agent. Using γ-H2AX as a marker for changes in chromatin structure, we show that Mre11, Rad50, Nbs1, and phosphorylated ATM respond to nucleoside analogue–induced stalled replication forks by forming nuclear foci that colocalize with γ-H2AX within 2 hours. Because neither DSBs nor single-strand breaks were detectable after nucleoside analogue exposure, we conclude that this molecular response is not due to the presence of DNA breaks. Deficiencies in ATM, Mre11, or Rad50 led to a 2- to 5-fold increase in clonogenic sensitization to gemcitabine, whereas Nbs1 and H2AX deficiency did not affect reproductive growth. Taken together, these results suggest that ATM, Mre11, and Rad50 are required for survival after replication fork stalling, whereas Nbs1 and H2AX are inconsequential. [Cancer Res 2008;68(19):7947–55]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 2 ( 2020-01-15), p. 204-218
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 2 ( 2020-01-15), p. 204-218
    Abstract: Dissemination is an essential early step in metastasis but its molecular basis remains incompletely understood. To define the essential targetable effectors of this process, we developed a 3D mammary epithelial culture model, in which dissemination is induced by overexpression of the transcription factor Twist1. Transcriptomic analysis and ChIP-PCR together demonstrated that protein kinase D1 (Prkd1) is a direct transcriptional target of Twist1 and is not expressed in the normal mammary epithelium. Pharmacologic and genetic inhibition of Prkd1 in the Twist1-induced dissemination model demonstrated that Prkd1 was required for cells to initiate extracellular matrix (ECM)–directed protrusions, release from the epithelium, and migrate through the ECM. Antibody-based protein profiling revealed that Prkd1 induced broad phosphorylation changes, including an inactivating phosphorylation of β-catenin and two microtubule depolymerizing phosphorylations of Tau, potentially explaining the release of cell–cell contacts and persistent activation of Prkd1. In patients with breast cancer, TWIST1 and PRKD1 expression correlated with metastatic recurrence, particularly in basal breast cancer. Prkd1 knockdown was sufficient to block dissemination of both murine and human mammary tumor organoids. Finally, Prkd1 knockdown in vivo blocked primary tumor invasion and distant metastasis in a mouse model of basal breast cancer. Collectively, these data identify Prkd1 as a novel and targetable signaling node downstream of Twist1 that is required for epithelial invasion and dissemination. Significance: Twist1 is a known regulator of metastatic cell behaviors but not directly targetable. This study provides a molecular explanation for how Twist1-induced dissemination works and demonstrates that it can be targeted.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 3926-3926
    Abstract: Small cell lung cancer (SCLC) is the most aggressive form of lung malignancies and accounts for 15-20% of all lung cancers. It has the tendency to metastasize early, thus limited-stage SCLC patients receive systemic chemo-radiotherapy (XRT) treatments. SCLC is exceptionally sensitive to XRT and exhibits high response rates; however, the recurrence rate is almost 100% and patients relapse with tumors that resist further chemotherapy. Clearly, elucidating the mechanisms of chemo-radiation resistance in SCLC will contribute to understanding how SCLC resists further treatments, to develop improved therapies and positively impact patient outcomes. Significant limitations for SCLC therapeutic development have been the lack of germane reliable and tractable model systems. Recent advances in establishing 3D organotypic culture have shown that this model can preserve the majority of pathways, key genes, histology and behavior of in situ tumors. Furthermore, patient-derived organoids (PDO) represent a powerful preclinical model that enable real-time cellular and molecular analysis of patient-derived xenograft (PDX) behavior ex vivo. Here, we present a novel patient-derived cancer organoid model to study the molecular underpinnings of XRT resistance in SCLC. Classic and variant SCLC PDX tumor tissues were isolated from mice and mechanically dissociated. Derived organoids were cultured in basal organoid medium. PDOs have been characterized using the SCLC molecular subtype classification reported in literature. RNA for transcriptomic analyses has been obtained to further characterize gene expression profiles of primary PDXs and PDOs, and to reconstruct gene regulatory network associated with XRT resistance. A SCLC PDX served as in vivo system to characterize the response to chemo-radiation resistance. Briefly, PDX tumor bearing mice were treated with: 1) vehicle control; 2) Cisplatin 5mg/kg on d1 plus Etoposide 8mg/kg on d1-2 (EP); 3) Radiotherapy 3Gy x1 on d3 (RT); and 4) both EP/RT. Whole transcriptome profiling among all treatments arms reveals molecular pathways and biological processes associated with XRT resistance. Also, by comparing our data with two previous SCLC patient cohort studies, we identified ideal candidates for functional analyses. SCLC XRT resistance candidate genes will be tested by either treating PDOs with small molecule inhibitors or by cDNA/shRNA lentiviral infection. To assess changes in chemo-radiation sensitivity, chemo-radiation protocols have been established and immunofluorescence staining for Ki67, γH2AX and cleaved caspase 3 served as markers for proliferation, DNA damage and apoptosis, respectively. Although further in-depth characterization is required, we aim to utilize our novel SCLC PDO model as a tool to identify candidate biomarkers to be used for developing therapy responses and translational research. Citation Format: Francesca A. Carrieri, Nick Connis, Eloise M. Grasset, Isaac S. Chan, Eddie Luidy-Imada, Christine Lam, Hailun Wang, Andrew J. Ewald, Luigi Marchionni, Christine L. Hann, Phuoc T. Tran. Establishment of patient-derived organoids as ex vivo tool to characterize the molecular mechanisms of SCLC chemo-radiation resistance [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3926.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 24_Supplement ( 2016-12-15), p. A48-A48
    Abstract: Genome-wide molecular profiles have been proven to be beneficial for the identification of clinically relevant tumor subtypes in many neoplastic diseases. While pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related death, very few genomic and transcriptomic studies have been conducted. This is mainly due to the difficulty to obtain a suitable cohort of PDAC tumor samples. The major obstacle is the usually high proportion of non-transformed stromal cells, which can greatly hinder the analysis of carcinogenic-specific processes. Moreover, tumor cohorts requiring resection samples can be biased by the exclusion of inoperable patients, representing 85% of all patients presenting PDAC. In this study, we generated Patient Derived Xenografts (PDX) with samples collected from 29 patients using either Endoscopic Ultrasound-Guided Fine-Needle Aspirates or, for operable patients, resections. The transcriptomic profiles, of both mRNA and miRNA, and the epigenetic landscape of early PDX passages consistently identified two tumor-specific molecular subtypes: a well differentiated group often referred to as classical, and an undifferentiated group previously recognized as Basal, Quasi-Mesenchymal or Squamous. Of all the genetic alterations determined by Copy Number analysis and exome sequencing, none were specific to any of the two subtypes, which therefore could not be discriminated solely on genetic basis. These two PDAC subtypes are characterized by distinct epigenetic and transcriptomic profiles of which the analysis revealed the deregulation of several pathways previously imputed in PDAC development and carcinogenesis in general. In particular, we showed that the Wnt pathway as well as several metabolic pathways, including cytochrome P450 genes recently implicated in drug resistance, are both epigenetically and transcriptionally deregulated. Altogether, our results provide new insights on pancreatic carcinogenesis and suggest that PDAC phenotypical heterogeneity is mainly driven by epigenetic rather than genetic events. Citation Format: Remy Nicolle, Yuna Blum, Laetitia Marisa, Jonathan Garnier, Benjamin Bian, Celine Loncle, Martin Bigonnet, Odile Gayet, Vincent Moutardier, Pauline Duconseil, Mohamed Gasmi, Mehdi Ouaissi, Olivier Turrini, Marc Giovannini, Aurélie Maignan, Jean-Marie Boher, Jacques Ewald, Erwan Bories, Marc Barthet, Anthony Goncalves, Flora Poizat, Jean-Luc Raoul, Veronique Secq, Stephane Garcia, Philippe Grandval, Marine Barraud-Blanc, Emmanuelle Norguet, Marine Gilabert, Jean-Robert Delpero, Ezequiel Calvo, Aurélien de Reyniès, Nelson Dusetti, Juan Iovanna.{Authors}. Multi-omics characterization of PDAC subtypes using PDX reveals that epigenetic but not genetic analysis permit a clinically relevant classification. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2016 May 12-15; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2016;76(24 Suppl):Abstract nr A48.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 19_Supplement ( 2018-10-01), p. B45-B45
    Abstract: Introduction: Recurrent somatic alterations associated with pediatric, childhood, and young adult cancers have not been as intensively studied as those associated with adult cancers. Consequently, whole-exome and transcriptome approaches are still being used to support discovery efforts. However, due to several initiatives aimed at profiling genomic alterations associated with childhood cancers, a set of recurrent somatic alterations has been defined. To accelerate research in this area, we have developed a novel targeted next-generation sequencing (NGS) assay to detect relevant somatic alterations previously reported in these cancer types. Methods: The assay was developed using Ion AmpliSeq targeted sequencing technology to cover the major gene variants associated with childhood cancers, including both solid tumor and hematologic cancer types. Over 200 gene targets were included on the basis of consultation with expert pediatric oncologists, literature review of the recent pediatric cancer genomic publications, as well as inclusion of relevant markers from adult cancers that are also observed in childhood cancers. Variant classes include mutations, copy number variations, gene fusions, and gene expression. Mutations in 130 genes, copy number variants in 28 genes, and over 1,400 distinct fusion isoforms in 88 fusion driver genes are analyzed. Variant calling algorithms for both DNA and RNA were optimized and combined into a single Ion Reporter workflow. Results: The assay generated an average read depth of & gt;3,000 reads per DNA amplicon with high uniformity ( & gt;95%), when up to 7 sample DNA-RNA pairs were analyzed with the 540 chip of the Ion S5 sequencing instrument. Minimal allele frequency detected for key hotspots was 5%. Sensitive and reproducible detection of CNV and fusion variants associated with pediatric solid tumors (EWSR1-FL1 and KIAA1549-BRAF fusions, MYC and EGFR amplification) and hematologic cancers (ETV6-RUNX1 and PML-RARA fusions) was demonstrated in orthogonally profiled FFPE, blood, and bone marrow samples. Performance was robust across sample types. Similar results were observed with manual and automated library preparation. Conclusions: A novel NGS assay, designed specifically for pediatric, childhood, and young adult cancers, and capable of detecting relevant DNA and RNA alterations from the same sample, was developed and validated. The assay is useful for characterizing relevant alterations in a wide range of cancers, including childhood leukemias and lymphomas as well as solid tumors including neuroblastoma, rhabdomyosarcoma, retinoblastoma, osteosarcoma, Ewing sarcoma, Wilms tumor, and brain and spinal cord tumors. A review of the analytical studies will be presented. Citation Format: Nickolay A. Khazanov, Chaitali Parikh, Habib Hamidi, Scott P. Myrand, Efren Ballesteros-Villagrana, Jingwei Ni, Paul D. Williams, Karen L. Clyde, Dinesh Cyanam, Armand Bankhead, III, Manimozhi Manivannan, Mark Tomilo, Susan Ewald, Jon K. Sherlock, Janice K. Au-Young, Jaclyn Biegel, Jonathan Buckley, Matthew Hiemenz, Dejerianne Ostrow, Alex Judkins, Xiaowu Gai, Tracy Busse, Alan Wayne, Deepa Bhojwani, Raca Gordana, Matthew Oberley, David Parham, Seth Sadis, Timothy Triche. Development of a next-generation sequencing (NGS) assay for pediatric, childhood, and young adult cancer research with comprehensive DNA and RNA variant detection [abstract]. In: Proceedings of the AACR Special Conference: Pediatric Cancer Research: From Basic Science to the Clinic; 2017 Dec 3-6; Atlanta, Georgia. Philadelphia (PA): AACR; Cancer Res 2018;78(19 Suppl):Abstract nr B45.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1526-1526
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1526-1526
    Abstract: Members of the Retinoic acid receptor (RAR) and HOX gene families are known to be associated with cellular differentiation. Nonetheless, how HOX genes drive differentiation in mammalian cells is poorly understood. HOX genes are transcription factors that orchestrate body segmentation during embryogenesis. But the continued expression of several members in adulthood indicates the necessity of their biological functions in specifying cell fate in adulthood. The mammary gland is an organ that continues to go through multiple cycles of development during life. Here, loss of specific HOX genes could be detrimental to normal development. We have previously shown that expression of HOXA5, a downstream target of the retinoic acid signaling pathway, is often lost in breast cancer. Loss of HOXA5 expression correlates with increasing pathological grades of breast cancer. Expression array analysis of HOXA5-depleted MCF10A (immortalized line of human breast epithelium) revealed its potential role in regulating several traits of the epithelial lineage, including expression of E-cadherin and CD24. Depleting HOXA5 in MCF10A enhanced cell motility, invasiveness and increased stem-cell population (CD24lo/CD44hi). Using an inducible HOXA5-depletion system, differentiation of the stem cells in MCF10A was shown to be mediated by HOXA5. Conversely, ectopic expression of HOXA5 in SUM149 breast cancer cells significantly reduced plasticity and stem cell population, confirmed by in vivo limiting dilution assays in mice. MCF10A-KRAS-HOXA5 depleted cells formed invasive structures in 3D matrigel cultures, and formed larger tumors, implanted orthotopically in mice. MCF10A-KRAS-tumors were histologically dedifferentiated and lost expression of luminal cell markers such as E-cadherin and cytokeratin 18. Collectively, our findings demonstrate the role of HOXA5 in retinoid-induced cell differentiation by regulating cell fate specific surface adhesion molecules. Citation Format: Wei Wen Teo, Vanessa Merino, Sean Cho, Preethi Korangath, Neil M. Neumann, Andrew J. Ewald, Saraswati Sukumar. HOXA5 inhibits cell plasticity and stemness in breast cancer cells by reinforcing epithelial traits. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1526. doi:10.1158/1538-7445.AM2015-1526
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 4396-4396
    Abstract: Patient-derived xenografts (PDX) are appearing as a prime approach for preclinical studies despite being insufficiently characterized as a model of the human disease and its diversity. In this work, 29 PDX were obtained from either surgery or endoscopic ultrasound-guided fine needle aspirate of pancreatic adenocarcinoma. The extensive genomic profiling of these pancreatic PDX, revealed two clinically-relevant subtypes having broad similarities with human primary tumors. These subtypes are defined by highly specific DNA methylation and transcriptomic profiles (mRNA, miRNA or lncRNA) but are not distinguishable by exonic mutations or copy number aberrations. Moreover, by specifically analyzing the stroma transcriptome, as defined by the expression of murine transcripts, we found that it is able to stratify the patients with the same efficiency than the analysis of grafted human tumor cells. This finding suggest that transformed pancreatic cells drive the composition of their own stroma. Finally, the multiomics analysis pinpoints novel therapeutic targets, one of which we demonstrate to be an efficient method for treating pancreatic cancer. Overall, we show that PDX are trustworthy pre-clinical models of pancreatic adenocarcinoma including of unresectable tumors. Their multiomics profiling allow the independent analysis of the uncontaminated cancer or stromal compartments and discloses several original therapeutics targets. Citation Format: Remy Nicolle, Yuna Blum, Laetitia Marisa, Celine Loncle, Odile Gayet, Vincent Moutardier, Olivier Turrini, Marc Giovannini, Benjamin Bian, Martin Bigonnet, Marion Rubis, Nabila Elarouci, Lucile Armenoult, Mira Ayadi, Pauline Duconseil, Mohamed Gasmi, Mehdi Ouaissi, Aurélie Maignan, Gwen Lomberk, Jean-Marie Boher, Jacques Ewald, Erwan Bories, Jonathan Garnier, Anthony Goncalves, Flora Poizat, Jean-Luc Raoul, Veronique Secq, Stephane Garcia, Philippe Grandval, Marine Barraud-Blanc, Emmanuelle Norguet, Marine Gilabert, Jean-Robert Delpero, Julie Roques, Ezequiel Calvo, Fabienne Guillaumond, Sophie Vasseur, Raul Urrutia, Aurélien de Reyniès, Nelson Dusetti, Juan Iovanna. Multiomics assessment of the cancer and stromal compartments of patient-derived pancreatic xenografts reveals clinically-relevant subtypes and novel targeted therapies [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4396. doi:10.1158/1538-7445.AM2017-4396
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 9_Supplement ( 2015-05-01), p. BS2-1-BS2-1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 9_Supplement ( 2015-05-01), p. BS2-1-BS2-1
    Abstract: Metastasis is the major driver of mortality and morbidity from breast cancer. Unfortunately, there are very few therapies that are effective in the metastatic setting. The overall hypothesis guiding our work is that a deeper biological understanding of the cellular and molecular mechanisms driving metastatic progression would facilitate the development of novel therapeutic concepts. A major barrier to achieving this goal is that the essential cellular steps occur deep within the body over a period of months to years. To overcome this barrier, we developed a novel organoid invasion assay that enables us to prospectively identify the most invasive cells within primary breast tumors. We initially explanted organoids from advanced tumors derived from the MMTV-PyMT transgenic mouse model of luminal B breast cancer. Surprisingly, we discovered that the cells that lead invasion into the extracellular matrix express basal epithelial genes, such as keratin-14, p63, and P-cadherin, both in 3D culture and in vivo. We next demonstrated that the cells leading invasion in mouse models of Her2 overexpressing (MMTV-Neu) and basal breast cancer (C3(1)-Tag) also expressed basal epithelial markers. We then adapted our organoid invasion assay to study primary human breast tumors in real-time and found that keratin-14+ cells lead collective invasion across diverse molecular subtypes of breast cancer, both in 3D culture and in vivo. With these correlations firmly established, we returned to the MMTV-PyMT mouse model to dissect the mechanisms underlying collective invasion. Using a keratin-14 reporter line we revealed that the basal invasion program can be induced in luminal phenotype cancer cells and that these newly keratin-14+ cells can lead collective invasion. We then identified shRNA constructs to knockdown keratin-14 and demonstrated that it is required for collective invasion in 3D culture and in vivo. Our current work focuses on using next generation sequencing to identify targetable genes required for the invasive process and using in vivo lineage analysis to study the relationship between collective invasion and distant metastasis. Citation Format: Andrew J Ewald. Collective invasion in breast cancer is led by specialized cancer cells with basal gene expression [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr BS2-1.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 7_Supplement ( 2016-04-01), p. IA17-IA17
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 7_Supplement ( 2016-04-01), p. IA17-IA17
    Abstract: The spread of cancer to distant organs is the major driver of mortality in cancer and, unfortunately, few therapies are effective in patients with extensive metastatic disease. Our hypothesis is that deeper mechanistic insight into the biological basis of metastasis will lead to novel therapeutic concepts. We have focused our laboratory investigations on understanding how epithelial cells acquire the ability to release their connections with their neighbors, migrate through the stroma, and enter the systemic circulation. The prevailing model when we began was the epithelial to mesenchymal transition (EMT), which posited a loss of epithelial features in the cancer cells during metastasis. The EMT model was controversial, in part because metastatic tumors typically retain many epithelial features. However, it remained possible that epithelial features were lost during stages of transit that are difficult to impossible to observe in vivo. To distinguish these possibilities, we developed novel 3D organoid assays to model normal epithelial growth, to determine the molecular requirements for Twist1 induced dissemination and to identify molecular programs driving the colonization of distant organs. The conclusions drawn across these studies are (1) that there are normal epithelial migratory programs that can be rapidly co-opted through activation of RTK signaling, (2) that the epithelial cell adhesion machinery can facilitate dissemination, and (3) that clusters of breast cancer cells utilize epithelial-specific gene expression (e.g. keratin-14) as they invade, disseminate, and colonize distant organs. These insights lead us to view metastasis as a process that can be accomplished without loss of epithelial character and that these epithelial routes to metastasis may have unique vulnerabilities compared to EMT models. We will discuss the latest progress in these studies and also technical advances in 3D culture that will accelerate metastasis research across organ sites. Citation Format: Andrew J. Ewald. Collective epithelial routes to metastatic spread. [abstract]. In: Proceedings of the AACR Special Conference on Tumor Metastasis; 2015 Nov 30-Dec 3; Austin, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(7 Suppl):Abstract nr IA17.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 368-368
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 368-368
    Abstract: Pancreatic cancer mortality is rising due to late detection and established metastasis. Despite the recent advances in cancer research, metastasis research has been hampered by the lack of models that recapitulate this complex process. Traditional cell culture and animal models provided insights on cancer cell migration and dissemination. But local invasion, which precedes all the events in the metastatic cascade, remains poorly understood. Patient-derived organoids (PDO) present an innovative platform in which the earliest step of metastasis can be observed and manipulated ex vivo. We developed a pancreatic cancer organoid model using surgically resected tumors from The Johns Hopkins Hospital. We previously reported that there are two invasive patterns in pancreatic cancer PDOs - Mesenchymal and Collective invasion, with one dominating pattern in every PDO culture. Here, we describe the gene expression signature associated with local invasion in pancreatic cancer by comparing the whole transcriptome of matched invasive and non-invasive organoids from the same tumor. For this study, PDOs were cultured for 7 days ex vivo and we manually collected 50 invasive and non-invasive organoids from each culture for RNA-seq analysis. To date, the invasive and non-invasive organoids from 6 PDO lines have been analyzed. The list of differentially expressed genes between invasive and non-invasive organoids includes 1,106 genes with FDR 0.001, with DACT1, DKK3 and KIF26B being most differentially expressed. Pathway analysis identifies that the invasive phenotype is associated with extracellular matrix receptor interaction, focal adhesion and PI3K-Akt signaling pathways. In conclusion, our findings revealed a distinct gene expression signature between invasive and non-invasive cells in PDOs. Interestingly, we did not find significant up or down-regulation in the classical EMT markers in our results, which suggests possible pancreatic cancer-specific metastasis programs exist. We believe our results can inform new diagnostic and prognostic tools and help identify anti-metastasis targets in pancreatic cancer. Citation Format: Yea Ji Jeong, Michael G. Lerner, Yuchen Ge, Hildur Knutsdottir, Bernat Navarro-Serer, Peter E. Chianchiano, Andrew J. Ewald, Joel S. Bader, Laura D. Wood. Defining the invasive gene signature using patient derived pancreatic cancer organoids [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 368.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages