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
  • 1
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 624-624
    Abstract: Achieving durable clinical responses to immune checkpoint inhibitors still remains a challenge. Here we demonstrate in preclinical models that immunotherapy with anti-CTLA-4 and its combination with anti-PD-1 rely on tumor cell-intrinsic activation of the cytosolic RNA receptor RIG-I (Fig. 1A). Mechanistically, tumor cell-intrinsic RIG-I signaling induced caspase-3-mediated tumor cell death, cross-presentation of tumor-associated antigen by CD103+ dendritic cells, subsequent expansion of tumor antigen-specific CD8+ T cells, and their accumulation within tumor tissue. Consistently, therapeutic targeting of RIG-I with 5'-triphosphorylated-RNA in both tumor and non-malignant host cells potently augmented the efficacy of CTLA-4 checkpoint blockade in several tumor models. In humans, transcriptome analysis of primary melanoma samples revealed a strong association between high expression of DDX58 (the gene encoding RIG-I), T cell receptor and antigen presentation pathway activity and prolonged overall survival (Fig. 1B). Moreover, in melanoma patients treated with anti-CTLA-4 checkpoint blockade, high RIG-I transcriptional activity significantly associated with durable clinical responses (Fig. 1C). Our preclinical data further demonstrate that tumor cell-intrinsic RIG-I signaling is also an essential pathway for the efficacy of other immunomodulating anticancer treatments including radiotherapy or hypomethylating agents such as 5-azacytidine. We thus identify aberrant tumor cell-intrinsic RIG-I signaling to be a crucial mechanism underlying cancer resistance to checkpoint inhibitor-based and other immunotherapies. These data have immediate translational potential as a RIG-I agonist for human application has been tested in phase I/II clinical trials with local administration in solid tumors and lymphomas (NCT03065023). Intratumoral RIG-I gene expression may not only serve as a biomarker to select patients that will likely benefit from anti-CTLA-4 therapy, but clinical RIG-I targeting in patients may also increase overall response rates of checkpoint inhibitor-based immunotherapy of malignancy including lymphoma. Figure 1. (A) Wild-type (WT) mice were bilaterally challenged with either WT or RIG-I-deficient (RIG-I-/-) B16.OVA melanoma cells.Recipients were repeatedly treated with anti-CTLA-4. Some mice were additionally injected with the RIG-I ligand 3pRNA into the right-sided tumor. Overall survival of treated mice bearing WT or RIG-I-/- B16.OVA tumors. (B) Overall survival in 456 TCGA melanoma patients by expression of DDX58 (RIG-I) in tumor samples. (C)DDX58 (RIG-I) expression in tumor samples from 18 patients with durable clinical response to anti-CTLA-4 treatment versus non-responders. Date give values from individual patients + geometric mean. Figure 1 Disclosures van den Brink: Seres Therapeutics: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Flagship Ventures: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Evelo: Consultancy, Honoraria; Jazz Pharmaceuticals: Consultancy, Honoraria; Therakos: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Merck & Co, Inc.: Consultancy, Honoraria; Acute Leukemia Forum (ALF): Consultancy, Honoraria; Magenta and DKMS Medical Council: Membership on an entity's Board of Directors or advisory committees; Juno Therapeutics: Other: Licensing.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Life Science Alliance, Life Science Alliance, LLC, Vol. 2, No. 2 ( 2019-04), p. e201900367-
    Abstract: Adoptive transfer of TCR transgenic T cells holds great promise for treating various cancers. So far, mainly semi-randomly integrating vectors have been used to genetically modify T cells. These carry the risk of insertional mutagenesis, and the sole addition of an exogenous TCR potentially results in the mispairing of TCR chains with endogenous ones. Established approaches using nonviral vectors, such as transposons, already reduce the risk of insertional mutagenesis but have not accomplished site-specific integration. Here, we used CRISPR-Cas9 RNPs and adeno-associated virus 6 for gene targeting to deliver an engineered TCR gene specifically to the TCR alpha constant locus, thus placing it under endogenous transcriptional control. Our data demonstrate that this approach replaces the endogenous TCR, functionally redirects the edited T cells’ specificity in vitro , and facilitates potent tumor rejection in an in vivo xenograft model .
    Type of Medium: Online Resource
    ISSN: 2575-1077
    Language: English
    Publisher: Life Science Alliance, LLC
    Publication Date: 2019
    detail.hit.zdb_id: 2948687-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Science Immunology, American Association for the Advancement of Science (AAAS), Vol. 4, No. 39 ( 2019-09-13)
    Abstract: Achieving durable clinical responses to immune checkpoint inhibitors remains a challenge. Here, we demonstrate that immunotherapy with anti–CTLA-4 and its combination with anti–PD-1 rely on tumor cell–intrinsic activation of the cytosolic RNA receptor RIG-I. Mechanistically, tumor cell–intrinsic RIG-I signaling induced caspase-3–mediated tumor cell death, cross-presentation of tumor-associated antigen by CD103 + dendritic cells, subsequent expansion of tumor antigen–specific CD8 + T cells, and their accumulation within the tumor tissue. Consistently, therapeutic targeting of RIG-I with 5′– triphosphorylated RNA in both tumor and nonmalignant host cells potently augmented the efficacy of CTLA-4 checkpoint blockade in several preclinical cancer models. In humans, transcriptome analysis of primary melanoma samples revealed a strong association between high expression of DDX58 (the gene encoding RIG-I), T cell receptor and antigen presentation pathway activity, and prolonged overall survival. Moreover, in patients with melanoma treated with anti–CTLA-4 checkpoint blockade, high DDX58 RIG-I transcriptional activity significantly associated with durable clinical responses. Our data thus identify activation of RIG-I signaling in tumors and their microenvironment as a crucial component for checkpoint inhibitor–mediated immunotherapy of cancer.
    Type of Medium: Online Resource
    ISSN: 2470-9468
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2019
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Science, American Association for the Advancement of Science (AAAS), Vol. 358, No. 6367 ( 2017-12)
    Abstract: Kinase inhibitors are important cancer therapeutics. Polypharmacology is commonly observed, requiring thorough target deconvolution to understand drug mechanism of action. Using chemical proteomics, we analyzed the target spectrum of 243 clinically evaluated kinase drugs. The data revealed previously unknown targets for established drugs, offered a perspective on the “druggable” kinome, highlighted (non)kinase off-targets, and suggested potential therapeutic applications. Integration of phosphoproteomic data refined drug-affected pathways, identified response markers, and strengthened rationale for combination treatments. We exemplify translational value by discovering SIK2 (salt-inducible kinase 2) inhibitors that modulate cytokine production in primary cells, by identifying drugs against the lung cancer survival marker MELK (maternal embryonic leucine zipper kinase), and by repurposing cabozantinib to treat FLT3-ITD–positive acute myeloid leukemia. This resource, available via the ProteomicsDB database, should facilitate basic, clinical, and drug discovery research and aid clinical decision-making.
    Type of Medium: Online Resource
    ISSN: 0036-8075 , 1095-9203
    RVK:
    RVK:
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2017
    detail.hit.zdb_id: 128410-1
    detail.hit.zdb_id: 2066996-3
    detail.hit.zdb_id: 2060783-0
    SSG: 11
    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...