GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (11)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 22 ( 2018-11-15), p. 6497-6508
    Abstract: Dasatinib is a multi-tyrosine kinase inhibitor approved for treatment of Ph+ acute lymphoblastic leukemia (ALL), but its efficacy is limited by resistance. Recent preclinical studies suggest that dasatinib may be a candidate therapy in additional ALL subtypes including pre-BCR+ ALL. Here we utilized shRNA library screening and global transcriptomic analysis to identify several novel genes and pathways that may enhance dasatinib efficacy or mitigate potential resistance in human pre-BCR+ ALL. Depletion of the transcriptional coactivator CBP increased dasatinib sensitivity by downregulating transcription of the pre-BCR signaling pathway previously associated with dasatinib sensitivity. Acquired resistance was due, in part, to upregulation of alternative pathways including WNT through a mechanism, suggesting transcriptional plasticity. Small molecules that disrupt CBP interactions with the CREB KID domain or β-catenin showed promising preclinical efficacy in combination with dasatinib. These findings highlight novel modulators of sensitivity to targeted therapies in human pre-BCR+ ALL, which can be reversed by small-molecule inhibitors. They also identify promising therapeutic approaches to ameliorate dasatinib sensitivity and prevent resistance in ALL. Significance: These findings reveal mechanisms that modulate sensitivity to dasatinib and suggest therapeutic strategies to improve the outcome of patients with acute lymphoblastic leukemia. Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/22/6497/F1.large.jpg. Cancer Res; 78(22); 6497–508. ©2018 AACR.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 23 ( 2016-12-01), p. 6937-6949
    Abstract: There is limited understanding of how signaling pathways are altered by oncogenic fusion transcription factors that drive leukemogenesis. To address this, we interrogated activated signaling pathways in a comparative analysis of mouse and human leukemias expressing the fusion protein E2A-PBX1, which is present in 5%–7% of pediatric and 50% of pre-B-cell receptor (preBCR+) acute lymphocytic leukemia (ALL). In this study, we describe remodeling of signaling networks by E2A-PBX1 in pre-B-ALL, which results in hyperactivation of the key oncogenic effector enzyme PLCγ2. Depletion of PLCγ2 reduced proliferation of mouse and human ALLs, including E2A-PBX1 leukemias, and increased disease-free survival after secondary transplantation. Mechanistically, E2A-PBX1 bound promoter regulatory regions and activated the transcription of its key target genes ZAP70, SYK, and LCK, which encode kinases upstream of PLCγ2. Depletion of the respective upstream kinases decreased cell proliferation and phosphorylated levels of PLCγ2 (pPLCγ2). Pairwise silencing of ZAP70, SYK, or LCK showed additive effects on cell growth inhibition, providing a rationale for combination therapy with inhibitors of these kinases. Accordingly, inhibitors such as the SRC family kinase (SFK) inhibitor dasatinib reduced pPLCγ2 and inhibited proliferation of human and mouse preBCR+/E2A-PBX1+ leukemias in vitro and in vivo. Furthermore, combining small-molecule inhibition of SYK, LCK, and SFK showed synergistic interactions and preclinical efficacy in the same setting. Our results show how the oncogenic fusion protein E2A-PBX1 perturbs signaling pathways upstream of PLCγ2 and renders leukemias amenable to targeted therapeutic inhibition. Cancer Res; 76(23); 6937–49. ©2016 AACR.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 96-96
    Abstract: Gene fusions frequently result from rearrangements in cancer genomes. In many instances, they play an important role in oncogenesis; in other instances, they are thought to be passenger events. Regulatory element rearrangements and copy number alterations resulting from these structural variants (SVs) lead to transcriptional dysregulation across cancers, though the extent to which these events result in functional dependencies with an impact on cancer cell survival is variable. We demonstrated enrichment of fusion partner genes and “collateral genes”, or genes in the same topologically associating domains (TADs) as fusion partners, among CRISPR-Cas9 dependencies across 209 cell lines with whole-genome sequencing data. We subsequently used CRISPR-Cas9 dependency screens to evaluate the fitness impact of 3,277 fusions across 645 cell lines with RNAseq data from the Cancer Dependency Map (DepMap). We found that 35% of cell lines harbored either a fusion partner dependency or a collateral dependency. CRISPR-Cas9 loss-of-function screening identified fusion-associated dependencies in 20 of 35 (57%) COSMIC fusions from our dataset. Fusion-associated dependencies revealed numerous novel oncogenic drivers and clinically translatable alterations. We observed several instances of intrachromosomal FOXR1 fusions associated with FOXR1 as a strong dependency, occurring independently in osteosarcoma, lung adenocarcinoma, and bladder carcinoma cell lines; these dependencies were validated through in vitro CRISPR-Cas9 knockout. FOXR1 fusions leading to FOXR1 overexpression were seen in an independent cohort of clinical tumor samples, speaking to the clinical relevance of this finding. FGFR3, a targetable kinase, was the key dependency in t(4;14) multiple myeloma cells that harbored an IgH-NSD2 fusion. We also showed that the implications of fusion-associated dependencies extended beyond two-dimensional cell line space, exemplified by dependency on EML4 in the context of a THADA-MTA3 fusion persisting in spheroid models. Broadly, fusions can result in partner and collateral dependencies that have biological and clinical relevance across cancer types. Citation Format: Riaz Gillani, Bo Kyung A. Seong, Jett Crowdis, Jake R. Conway, Neekesh V. Dharia, Saif Alimohamed, Brian J. Haas, Kyuho Han, Jihye Park, David Liu, Felix Dietlein, Meng Xiao He, Alma Imamovic, Clement Ma, Michael C. Bassik, Jesse S. Boehm, Francisca Vazquez, Alexander Gusev, Katherine A. Janeway, James M. McFarland, Kimberly Stegmaier, Eliezer M. Van Allen. Gene fusions create partner and collateral dependencies that are essential to cancer cell survival [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 96.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4362-4362
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4362-4362
    Abstract: Lung cancer is the number one cause of cancer-related deaths worldwide. The most prevalent type of lung cancer is Non-Small Cell Lung Cancer (NSCLC). A significant number of patients with NSCLC carry oncogenic KRAS mutations. However, the efforts to target KRAS directly have thus far proven unsuccessful and tumors harboring mutations in this gene remain the most difficult to treat, highlighting the need for alternative approaches. One promising strategy is to target KRAS-dependent cancers through synthetic lethality. However, KRAS activates multiple effector pathways, suggesting that targeting one gene may not be sufficient to fully inhibit KRAS oncogenesis. Therefore, we propose that targeting combinations of genes that together are synthetic lethal with KRAS may constitute a better therapeutic strategy. Furthermore, we hypothesize that a targeted approach focused on the protein-protein interaction network proximal to KRAS may be more effective than the current emphasis on genome-wide screens. To discover novel, combinatorial KRAS synthetic lethal genes, we used affinity purification/mass spectrometry (AP/MS), to systematically identify KRAS interacting proteins and construct a detailed map of protein-protein interactions centered on KRAS. Based on this network we designed a CRISPR/Cas9 library targeting pairwise combinations of KRAS-interacting genes. Using this library we simultaneously knocked-out pairs of 119 genes in two KRAS-driven non-small cell lung cancer (NSCLC) cell lines (A549 and H23). Knock-out of many gene pairs synergistically impaired growth of these cells, while the knock-out of each of the genes alone had no or little effect. We chose 20 most promising targets for further screening in vitro and in vivo in a panel of 9 KRAS-mutant and KRAS wild type Cas9-expressing NSCLC cell lines. We also selected six gene pairs that had the most synergistic effect on growth in A549 and H23 cells for individual validation in Cas9-expressing NSCLC cell lines and normal human bronchial epithelial cells (HBECs). We found that the simultaneous knock-out of one pair of genes, Rap1GDS1 and RhoA, significantly decreased growth of KRAS-dependent NSCLC cells, while having a limited effect on KRAS-independent cells or HBECs. Moreover the knock-out of either of these genes alone had no effect on growth in any of the cell lines, suggesting that only the combination of these two genes is synthetically lethal with KRAS. We are currently performing further validation in organoid cultures and in vivo. Additional validation and human relevance will be determined using patient-derived xenografts (PDX). Citation Format: Kaja Kostyrko, Marcus R. Kelly, Kyuho Han, Edwin E. Jeng, David W. Morgens, Michael C. Bassik, Peter K. Jackson, Alejandro Sweet-Cordero. Identification of novel combinatorial synthetic lethal vulnerabilities in KRAS-driven lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4362.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 702-702
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 702-702
    Abstract: Successful identification of the genetic determinants of drug sensitivity has been a long-standing goal of cancer research and, more recently, of precision medicine. In cancer cells, drug treatment can induce both growth arrest and death. The distinction between these outcomes is important but can be overlooked in large-scale analyses that rely on traditional metabolism-based methods for quantifying cell viability. We recently demonstrated that Scalable Time-Lapse Analysis of Cell Death Kinetics (STACK) effectively quantifies cell death in response to diverse lethal stimuli. We investigated cases in which this approach captured drug-induced lethality more effectively than traditional assays. We found that inhibitors of mitogen activated protein kinase 1 and 2 (MEK1/2) arrested proliferation in diverse cancer cell lines, but induced cell death in only a subset of these lines. We sought to identify the factors that determine whether a cancer cell will undergo growth arrest or death in response to MEK inhibition. MAP kinase pathway activation has previously been reported as a determinant of MEK inhibitor sensitivity; we determined that the dynamics of Bcl-2 family proteins in response to MEK inhibitor treatment correlate with death sensitivity independent of pathway activation. In a genome-wide CRISPR screen, we identified a lipid metabolic enzyme that is a novel modulator of MEKi sensitivity. Our results suggest new approaches for predicting and enhancing sensitivity to cancer therapeutics in tumors and demonstrate that direct measurement of cell death provides clear advantages over proliferation-based metrics. Citation Format: Zintis Inde, Kyuho Han, Michael C. Bassik, Scott J. Dixon. Kinetic analysis identifies determinants of sensitivity to MEK inhibitor-induced cell death [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 702.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 15 ( 2021-08-01), p. 3971-3984
    Abstract: Gene fusions frequently result from rearrangements in cancer genomes. In many instances, gene fusions play an important role in oncogenesis; in other instances, they are thought to be passenger events. Although regulatory element rearrangements and copy number alterations resulting from these structural variants are known to lead to transcriptional dysregulation across cancers, the extent to which these events result in functional dependencies with an impact on cancer cell survival is variable. Here we used CRISPR-Cas9 dependency screens to evaluate the fitness impact of 3,277 fusions across 645 cell lines from the Cancer Dependency Map. We found that 35% of cell lines harbored either a fusion partner dependency or a collateral dependency on a gene within the same topologically associating domain as a fusion partner. Fusion-associated dependencies revealed numerous novel oncogenic drivers and clinically translatable alterations. Broadly, fusions can result in partner and collateral dependencies that have biological and clinical relevance across cancer types. Significance: This study provides insights into how fusions contribute to fitness in different cancer contexts beyond partner-gene activation events, identifying partner and collateral dependencies that may have direct implications for clinical care.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 10, No. 12 ( 2020-12-01), p. 1950-1967
    Abstract: Activating mutations in RAS GTPases drive many cancers, but limited understanding of less-studied RAS interactors, and of the specific roles of different RAS interactor paralogs, continues to limit target discovery. We developed a multistage discovery and screening process to systematically identify genes conferring RAS-related susceptibilities in lung adenocarcinoma. Using affinity purification mass spectrometry, we generated a protein–protein interaction map of RAS interactors and pathway components containing hundreds of interactions. From this network, we constructed a CRISPR dual knockout library targeting 119 RAS-related genes that we screened for KRAS-dependent genetic interactions (GI). This approach identified new RAS effectors, including the adhesion controller RADIL and the endocytosis regulator RIN1, and & gt;250 synthetic lethal GIs, including a potent KRAS-dependent interaction between RAP1GDS1 and RHOA. Many GIs link specific paralogs within and between gene families. These findings illustrate the power of multiomic approaches to uncover synthetic lethal combinations specific for hitherto untreatable cancer genotypes. Significance: We establish a deep network of protein–protein and genetic interactions in the RAS pathway. Many interactions validated here demonstrate important specificities and redundancies among paralogous RAS regulators and effectors. By comparing synthetic lethal interactions across KRAS-dependent and KRAS-independent cell lines, we identify several new combination therapy targets for RAS-driven cancers. This article is highlighted in the In This Issue feature, p. 1775
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 12, No. 5_Supplement ( 2013-05-01), p. A08-A08
    Abstract: Multiple myeloma (MM) is characterized by the clonal expansion of malignant plasma cells in the bone marrow. MM remains incurable; thus, effective therapies remain an unmet clinical need. MM cells produce large amounts of paraproteins, which impose a sustained demand on the endoplasmic reticulum (ER) resulting in chronic ER stress. Standard-of-care therapies, like the proteasome inhibitor bortezomib, take advantage of the reliance of MM cells on ER function. By blocking the proteasome, client protein load exceeds the capacity of the proteasome, inducing unmitigatable ER stress and apoptosis. However, most MM cells develop resistance to bortezomib, indicating that genetic escape routes are activated upon selective pressure. Adaptive measures to counteract ER stress are controlled by the unfolded protein response (UPR), which is orchestrated by a network of three intertwined signaling pathways, the most conserved of which is activated by the ER stress sensor IRE1 and its effector, the transcription factor XBP1. IRE1/XBP1 are part of a cell-protective response that, in healthy cells, permits adaptation in response to upsurges in ER function, while in MM, they are thought to promote survival. We show that genetic susceptibilities of MM cells can be identified to guide combination therapies using bortezomib or a chemical inhibitor of IRE1. To this end, we systematically interrogated genetic vulnerabilities involving the ER stress response in MM cells using an unbiased genome-wide RNA interference approach that employs screening of ultra-complex shRNA libraries. Conducting pooled shRNA screens in conventional and microcarrier-based cell culture systems, we show that, unlike bortezomib, IRE1 inhibition alone does not lead to tumor cell death, suggesting that IRE1 is not a single fate determinant. Rather, we show that IRE1 represents an Achilles heel that allows identification of combinatorial genetic vulnerabilities resulting in synthetic lethality. The same rationale was applied to proteasome inhibition, where we identified several genes whose diminished function resulted in synthetic lethality. In this way, we identified novel pathways that when modulated in parallel with proteasome inhibition block escape routes leading to drug resistance. By applying our screening method, we validated known genetic susceptibilities of MM cells, identified new ER-associated MM survival factors, and uncovered vulnerabilities that result in synthetic lethality when combined with pharmacological inhibition of IRE1 or the proteasome. Analyses of published gene expression datasets from MM patient samples suggest that the expression levels of some of the identified genes are predictive of poor survival. Taken together, our data validate the screening method as a powerful tool for the discovery of diagnostic biomarkers and novel therapy targets. Citation Format: Diego Acosta-Alvear, Martin Kampmann, Michael C. Bassik, Crystal P. Lee, Marc A. Shuman, Jonathan S. Weissman, Peter Walter. Systematic identification of genetic vulnerabilities synergizing with proteasome and IRE1 inhibition in multiple myeloma cells using ultra-complex shRNA libraries. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Synthetic Lethal Approaches to Cancer Vulnerabilities; May 17-20, 2013; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(5 Suppl):Abstract nr A08.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 19_Supplement ( 2013-10-01), p. C01-C01
    Abstract: Multiple myeloma (MM) remains a devastating incurable disease. Because of their secretory nature, MM cells rely on proper endoplasmic reticulum (ER) function. The unfolded protein response (UPR) oversees the folding capacity of the ER and adjusts it according to need. Paraprotein production by MM cells imposes an ER burden that provides an opportunity for therapeutic intervention. High ER function exceeds the buffering capacity of the cell for protein degradation, rendering MM cells exquisitely sensitive to proteasome inhibitors. While successful, standard-of-care proteasome inhibitors, like bortezomib, remain palliative, as patients ultimately become refractory, suggesting the selection of genetic escape routes. ER and proteasome functions are intimately connected. Proteasomal degradation of unfolded ER client proteins is accomplished after dislocation and ubiquitination of unfolded proteins from the ER. Therefore, genetic escape routes can exist within the ubiquitin-proteasome system and the UPR. The most conserved branch of the UPR, overseen by the sensor kinase IRE1 and its downstream effector, XBP1, has been a primary focus of the MM research community in recent years. In healthy cells IRE1/XBP1 allow adaptation to rises in ER function, while in MM cells IRE1/XBP1 are thought to confer a survival advantage. For these reasons, we hypothesized that modulation of the UPR, alone or in combination with proteasome inhibitors, while therapeutically beneficial, might also provide the driving force for acquired drug resistance. Therefore, a single agent is unlikely to lead to long-term remission. Here we show that the therapeutic benefit of blocking the proteasome or IRE1 in MM cells can be enhanced when combined with additional therapies targeting synthetic-lethal interactions, and that these can be systematically identified using an shRNA screening method developed by our groups. Employing an ultra-complex shRNA library, we performed pooled shRNA screens in RPMI-8226 and MM1-S MM cells and identified genes whose diminished function impact the cell's response to proteasome or IRE1 inhibition. Importantly, we show that unlike proteasome inhibition, blocking IRE1 function does not lead to substantial MM cell death, strongly suggesting that IRE1 alone is not a fate determinant. Instead, we posit IRE1 is a handicap that might be therapeutically exploited through the identification of synthetic-lethal combinations. We applied the same rationale to proteasome inhibition and found several synthetic-lethal pairs. Some of these genes can be targeted pharmacologically, which allowed us to query the susceptibility of a panel of MM cells to combinations of bortezomib and several drug-like molecules, including novel Hsp70 inhibitors. Our results inform which pathways may be selectively blocked to shut down potential escape routes that may lead to drug resistance. Notably, retrospective analyses on publicly available gene expression datasets of MM patients indicate that several of the genes we found on our screens are predictors of poor survival. Taken together, our preliminary results indicate our screening platform is a powerful tool for the discovery of (i) novel therapy targets, (ii) synthetic-lethal pairs that can be exploited in potential combination therapies, and (iii) diagnostics biomarkers. Currently, we continue to validate our results in additional cell lines prior to verification in primary patient samples and in an orthotopic xenograft model developed by our groups (DAA and PW, unpublished). Citation Format: Diego Acosta-Alvear, Martin Kampmann, Michael C. Bassik, Crystal P. Lee, Min Y. Cho, Blake T. Aftab, Xiaokai Li, Jason E. Gestwicki, Marc A. Shuman, Jonathan S. Weissman, Peter Walter. Systematic blueprinting of genetic vulnerabilities using ultra-complex shRNA libraries identifies genes synergizing with proteasome inhibition or blockade of IRE1 in multiple myeloma cells. [abstract]. In: Proceedings of the Third AACR International Conference on Frontiers in Basic Cancer Research; Sep 18-22, 2013; National Harbor, MD. Philadelphia (PA): AACR; Cancer Res 2013;73(19 Suppl):Abstract nr C01.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Molecular Cancer Therapeutics Vol. 12, No. 5_Supplement ( 2013-05-01), p. PR06-PR06
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 12, No. 5_Supplement ( 2013-05-01), p. PR06-PR06
    Abstract: Systematic, high-density mapping of genetic interactions is a powerful approach to elucidate functional pathways and reveal synthetic lethal gene pairs, and has successfully been applied in microorganisms. We have recently developed a functional genomics platform that enables the construction of high-density genetic interaction maps in mammalian cells (Bassik*, Kampmann* et al. Cell 152, Feb. 14, 2013 *Equal contribution). In a first step, we conduct pooled primary screens using an ultracomplex shRNA library that targets each protein-coding gene with 25 independent shRNAs and contains & gt;1,000 negative control shRNAs. This strategy enables us to robustly identify hit genes and shRNAs that target them effectively, while minimizing the identification of false-positive hits which has plagued many genome-wide RNAi screens. In a second step, we construct and screen a double-shRNA library that targets all pairwise combination of hit genes to contruct a high-density genetic interaction map. Our approach enables us to determine 100,000s of genetic interactions in a single experiment. Here, we present the application of our platform to identify synthetic-lethal vulnerabilities in the stress response network of leukemia and multiple myeloma cells. Stress response pathways, including the endoplasmic reticulum stress response (unfolded protein response), heat shock, hypoxia and oxidative stress responses, and the associated induction of autophagy, have been shown to play important roles in cancer cell survival, drug resistance and disease progression. The goal of the research presented here is to systematically characterize vulnerabilities in the stress response network of cancer cells, and in particular, to identify synthetic-lethal vulnerabilities that are potential novel targets for combination drug therapy. We conducted our first experiments in two human cell lines derived from hematologic cancers, K562 (leukemia) and RPMI-8226 (multiple myeloma), for which we determined genetic vulnerabilities and their genetic interactions in the absence and presence of stress-inducing agents. We discovered previously unknown genetic vulnerabilities, several of which are considered “druggable”. We externally validated several of the synthetic-lethal vulnerabilities we identified by demonstrating synergistic effects of drug combinations targeting these gene pairs. Intriguingly, expression levels of several vulnerabilities we identified are prognostic of patient survival in a published multiple myeloma clinical trial. We are in the process of testing the therapeutic potential of targeting the synthetic-lethal vulnerabilities we identified in the multiple myeloma stress response network in primary patient cells and in multiple myeloma mouse models. This abstract is also presented as Poster A15. Citation Format: Martin Kampmann, Diego Acosta-Alvear, Michael C. Bassik, Yuwen Chen, Peter Walter, Jonathan S. Weissman. Systematic genetic interaction maps reveal synthetic-lethal vulnerabilities in leukemia and multiple myeloma. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Synthetic Lethal Approaches to Cancer Vulnerabilities; May 17-20, 2013; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(5 Suppl):Abstract nr PR06.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...