GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
  • 1
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3607-3607
    Kurzfassung: Bst1/CD157 is a GPI-anchored transmembrane protein belonging to the ADP-ribosyl-cyclase family and it is expressed on some blood cells such as monocytes and neutrophils. Importantly, we demonstrated that this antigen is expressed in blast cells from almost 100% of Acute Myeloid Leukemia patients (either at primary diagnosis or at relapse). MEN1112 is a humanized, de-fucosylated antibody targeting the Bst1/CD157 antigen with high affinity and inducing potent in vitro and ex vivo Antibody Dependent Cell-mediated Cytotoxicity (ADCC) responses against AML cell lines and blasts, respectively. MEN1112 was discovered as a mouse Fab antibody from a phage display library raised against recombinant Bst1/CD157 protein. The Fab was selected base upon its ability to recognize recombinant Bst1/CD157 protein by ELISA and for its ability to recognize Bst1/CD157 positive endogenous cancer cell lines by FACS (fluorescence activated cell sorting). The Fab was also chosen based upon its ability to recognize cynomolgus Bst1/CD157 antigen on recombinant and endogenous cells. The selected Fab sequence was humanized and incorporated into the Biowa Potelligent cell line to produce a de-fucosylated full IgG1 version (MEN1112). The binding affinity of MEN1112 for the Bst1/CD157 antigen showed an EC50 of 1nM as assessed by flow cytometry (FACS) analysis using multiple cell lines. No binding was observed on antigen-negative cell lines demonstrating specificity of MEN1112. High affinity of MEN1112 for the recombinant antigen was confirmed in an ELISA assay (EC50 0.4 nM). Beyond the affinity for the antigen, it is of relevance for its ADCC activity that MEN1112 is characterized by an improved affinity for Fc receptors on effector cells over the parental antibody. In particular, MEN1112, lacking fucose residues in its Fc domain, showed an impressive high affinity binding to the Fc receptor CD16A both high affinity and low affinity alleles. Internalization studies demonstrate the slow internalization of the Bst1/CD157 antigen following binding to MEN1112 which favors the ADCC efficacy of the antibody. Overall our results indicate that MEN1112 has the potential to exert anti-leukemia actions through a powerful ADCC in view of: (i) superior affinity for blast cell membrane antigen Bst1/CD157, (ii) enhanced affinity for all variants of CD16A Fc receptor expressed on effector cells and (iii) minimal internalization following antigen binding. Disclosures Aud: Oxford BioTherapeutics: Employment. Dusek:Oxford BioTherapeutics: Employment. Bisht:Oxford BioTherapeutics: Employment. Swaminithan:Oxford BioTherapeutics: Employment. Awdew:Oxford BioTherapeutics: Employment. Trang:Oxford BioTherapeutics: Employment. Lin Lou:Oxford BioTherapeutics: Employment. Manzini:Menarini Ricerche SpA: Employment. Martinez Mogarra:Menarini Biotech srl: Employment. Bressan:Menarini Ricerche SpA: Employment. Binaschi:Menarini Ricerche SpA: Employment.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2014
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 753-753
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 753-753
    Kurzfassung: Solid tumors comprises of two distinct compartments: cancer cells and the stroma that the cancer cells induce and are dispersed in. This stroma contains stromal (Fibroblasts, endothelial and pericyte cells) and infiltrating immune cells (Lymphocytes, macrophages, granulocytes and myeloid derived suppressor cells). Recent oncology research has implicated these stroma cells as promoters of tumor progression and there is thus a strong need to profile the cell membrane proteins present on these cells. For tumors which do not contain infiltrating T cells (non-immunogenic tumors) it has been recognized that it will be necessary to employ therapeutic agents to disrupt the tumor micro-environment (TME); allowing the release and processing of tumor antigens by antigen presenting cells, entry and migration T cells into intrastromal sites and potent activation of T cell responses to tumor antigens. We have conducted in-depth proteomic profiling of tumors from 14 different solid cancer indications with varying degrees of stroma involvement to characterize their immune and stromal cell composition. The data from this work was analyzed in OGAP®; a unique proteomic database that integrates information at the tissue, disease and protein isoform level across diseases, indications, and normal tissues to clarify membrane protein expression levels and profiles. It is one of the world's largest proprietary cell-membrane focused proteomic database, with data on over 5000 cancer membrane proteins. Purified stroma cell populations were also profiled to facilitate the interpretation of whole tumor proteomic data. Cell membrane proteins present in tumors from different cancer indications were analyzed by mass spectrometry. We classified tumors as immunogenic or non-immunogenic using validated T cell markers (CD3, CD4, CD8 and CD69). We then profiled the non-immunogenic tumors for fibroblast, macrophage and B cell type markers to try and identify strong stroma cell expression patterns not detected in normal tissues. Importantly, most current antibody therapeutic stroma cell targets (CSF-1R, FAP and VEGF-A) were re-discovered. Several novel stroma cell therapeutic targets were identified which if targeted by a therapeutic antibody have the potential to enhance the T cell immune response in tumors resistant to current immunotherapy. Citation Format: James Ackroyd, Jason Allen, Jo Berry, Lisa Roesch, Martin Barnes, Arnima Bisht, Christian Rohlff, Keith Wilson, Dee Aud, Robert Boyd. The use of proteomics to analyse whole tumors and identify unique stroma cell targets for antibody-based therapeutics. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 753.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2016
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1321-1321
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1321-1321
    Kurzfassung: The recent clinical success of the mAb therapeutics targeting immune checkpoint inhibitor proteins (PD-1/PD-L1, CTLA-4) has led to an increased appreciation of the potential of utilizing the immune system in oncology. There are two major strategies to elicit either a novel immune anti-tumor response or to reactivate a pre-existing anti-tumor response: by releasing a checkpoint inhibitory pathway via cell surface receptors (such as PD-1/PD-L1, CTLA-4) or by activation of co-stimulatory receptors (such as CD40, OX40, or GITR). Both of these strategies of immune modulation utilize cell surface receptors, and the targeting of antibody therapeutics with the appropriate functional activity to those receptors, to modify immune cell responses and allow for anti-tumor activity. The identification of novel immune-modulatory receptors with the potential to be immune-oncology therapeutic targets could be of high value to this anti-tumor approach. OGAP is a unique proteomic database that integrates information at the tissue, disease and protein isoform level across diseases, indications, and normal tissues to clarify membrane protein expression levels and profiles. Specifically, it currently holds information on ∼16,000 human proteins sequenced, ∼7,000 membrane proteins, ∼35 tissues/organs, and ∼17 cancers. OGAP is fed by a proprietary sample preparation and processing workflow that relies on state-of-the-art high-throughput mass spectrometry and data processing to provide quantitative information on over 4,000 membrane-enriched proteins. OGAP has been used to identify novel oncology therapeutic targets for both ADC and BiTE-like approaches. Utilizing OGAP, membrane proteins present in tumors from five cancer indications (Pancreatic, Lung, Breast, Colorectal and Esophageal cancer) and multiple normal tissues or cells were analyzed. Validated immune cell markers (such as CD8, OX40, CD79B, TLR1, TLR2, TLR4, TLR7, CD56, CD204 and CD207) were profiled across different normal and tumor proteomic data sets. This analysis demonstrated we detect key immune cell markers in tumors and that different immune cell populations are found in tumors from the same or different cancer indications. The proteomic data sets were next analyzed for the presence of validated immuno-oncology targets (TIM3, PDL-1 and B7-H3). The expression patterns of these immune-oncology targets were analyzed to try and identify a unique protein signature. Using this protein expression profiling approach we identified most known immune-oncology targets, validating this as an approach to identify a pool of candidate novel immune-oncology targets. To further develop our approach we also used sequence based homology searching of uncharacterized membrane proteins to improve the quality of the pool of candidate novel immune-oncology targets. Several potential novel immune-oncology targets will be presented. Citation Format: Jim Ackroyd, Arnima Bisht, Jason Allen, Lindsey Hudson, Martin Barnes, Christian Rohlff, Keith Wilson, Robert Boyd, Dee Aud. The use of proteomics to analyze whole tumors and identify unique immuno-oncology targets for antibody-based therapeutics. [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 1321. doi:10.1158/1538-7445.AM2015-1321
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2015
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    Online-Ressource
    Online-Ressource
    Springer Science and Business Media LLC ; 2006
    In:  Nature Clinical Practice Rheumatology Vol. 2, No. 8 ( 2006-8), p. 434-442
    In: Nature Clinical Practice Rheumatology, Springer Science and Business Media LLC, Vol. 2, No. 8 ( 2006-8), p. 434-442
    Materialart: Online-Ressource
    ISSN: 1745-8382 , 1745-8390
    Sprache: Englisch
    Verlag: Springer Science and Business Media LLC
    Publikationsdatum: 2006
    ZDB Id: 2491533-6
    ZDB Id: 2232992-4
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: Immunity, Elsevier BV, Vol. 2, No. 3 ( 1995-03), p. 211-222
    Materialart: Online-Ressource
    ISSN: 1074-7613
    RVK:
    Sprache: Englisch
    Verlag: Elsevier BV
    Publikationsdatum: 1995
    ZDB Id: 2001966-X
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    Online-Ressource
    Online-Ressource
    American Society for Pharmacology & Experimental Therapeutics (ASPET) ; 2008
    In:  Journal of Pharmacology and Experimental Therapeutics Vol. 327, No. 3 ( 2008-12), p. 610-619
    In: Journal of Pharmacology and Experimental Therapeutics, American Society for Pharmacology & Experimental Therapeutics (ASPET), Vol. 327, No. 3 ( 2008-12), p. 610-619
    Materialart: Online-Ressource
    ISSN: 0022-3565 , 1521-0103
    Sprache: Englisch
    Verlag: American Society for Pharmacology & Experimental Therapeutics (ASPET)
    Publikationsdatum: 2008
    ZDB Id: 1475023-5
    SSG: 15,3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    In: Nature Immunology, Springer Science and Business Media LLC, Vol. 1, No. 3 ( 2000-9), p. 221-226
    Materialart: Online-Ressource
    ISSN: 1529-2908 , 1529-2916
    Sprache: Englisch
    Verlag: Springer Science and Business Media LLC
    Publikationsdatum: 2000
    ZDB Id: 2026412-4
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    In: Physiological Genomics, American Physiological Society, Vol. 25, No. 3 ( 2006-05-16), p. 502-513
    Kurzfassung: Complex airway diseases such as asthma and chronic obstructive pulmonary disease exhibit stereotyped traits (especially airway hyperreactivity and mucous cell metaplasia) that are variably expressed in each patient. Here, we used a mouse model for virus-induced long-term expression of these traits to determine whether individual traits can be genetically segregated and thereby linked to separate determinants. We showed that an F2 intercross population derived from susceptible and nonsusceptible mouse strains can manifest individual phenotypic extremes that exhibit one or the other disease trait. Functional genomic analysis of these extremes further indicated that a member of the calcium-activated chloride channel ( CLCA) gene family designated mClca3 was inducible with mucous cell metaplasia but not airway hyperreactivity. In confirmation of this finding, we found that mClca3 gene transfer to mouse airway epithelium was sufficient to induce mucous cell metaplasia but not airway hyperreactivity. However, newly developed mClca3 −/− mice exhibited the same degree of mucous cell metaplasia and airway hyperreactivity as wild-type mice. Bioinformatic analysis of the Clca locus led to the identification of mClca5, and gene transfer indicated that mClca5 also selectively drives mucous cell metaplasia. Thus, in addition to the capacity of CLCA family members to exhibit diverse functional activities, there is also preserved function so that more than one family member mediates mucous cell metaplasia. Nonetheless, Clca expression appears to be a selective determinant of mucous cell metaplasia so that shared homologies between CLCA family members may still represent a useful target for focused therapeutic intervention in hypersecretory airway disease.
    Materialart: Online-Ressource
    ISSN: 1094-8341 , 1531-2267
    Sprache: Englisch
    Verlag: American Physiological Society
    Publikationsdatum: 2006
    ZDB Id: 2031330-5
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    In: Oncotarget, Impact Journals, LLC, Vol. 8, No. 22 ( 2017-05-30), p. 35707-35717
    Materialart: Online-Ressource
    ISSN: 1949-2553
    URL: Issue
    Sprache: Englisch
    Verlag: Impact Journals, LLC
    Publikationsdatum: 2017
    ZDB Id: 2560162-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 3869-3869
    Kurzfassung: Antibody-drug conjugates (ADCs) are a recent and exciting development for targeted therapy of cancer. Their efficacy is governed by ADC-intrinsic characteristics such as avidity, drug load and linker chemistry, and mechanisms of activation and action, which can be controlled or clarified in the early stages of ADC development. In contrast, the properties that define a promising ADC target are still somewhat unclear. OGAP is a unique proteomic database that integrates information at the tissue, disease and protein isoform level across diseases, indications, and normal tissues to clarify protein expression levels and profiles. Specifically, it currently holds information on ∼2,000,000 human protein peptide sequences, ∼16,000 human proteins sequenced, ∼7,000 cancer membrane proteins, ∼50 tissues/organs, and ∼60 diseases. Building on OGAP and a proprietary sample preparation and processing workflow that relies on state-of-the-art high-throughput mass spectrometry and data processing to provide quantitative information on over 4,000 membrane-enriched proteins from ∼ 15,000 unique peptide sequences per analysis, we have established a novel predictive tool to establish each protein's potential to serve as a target for ADC development. The tool considers proteomic and target-specific information on antigenicity, structure, function, expression level, regulation, and tissue distribution in order to highlight the most suitable candidates for ADC development. We will demonstrate the utility of this process for the protein family of G-protein coupled receptors (GPCRs), which according to a recent bioinformatics prediction encompasses 899 distinct members in the human genome. These cell surface receptors are the target of more than one third of conventional drugs, yet their potential for ADCs is largely unexplored. Here we show that proteomics in the context of the OGAP database can highlight which of this large family of receptors have the potential to become true ADC targets. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 3869. doi:1538-7445.AM2012-3869
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2012
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...