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: Bioorganic & Medicinal Chemistry, Elsevier BV, Vol. 16, No. 24 ( 2008-12-15), p. 10326-10331
    Type of Medium: Online Resource
    ISSN: 0968-0896
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2008
    detail.hit.zdb_id: 1501507-5
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 35, No. 15_suppl ( 2017-05-20), p. e14001-e14001
    Abstract: e14001 Background: The epidermal growth factor receptor is a target for the treatment of solid tumors, like CRC, HNSCC and NSCLC. Therapeutic success of EGFR-targeting agents depends on mutations defining resistance to these agents, thus an effective EGFR-targeting therapy overcoming resistance would offer an attractive new treatment option. Natural killer (NK-) cells play a central role in the innate immune system and can destroy cancer cells. NK-cell-engaging bispecific antibodies may offer a safe and effective therapy to target EGFR-expressing tumor cells irrespective of their mutational status. Methods: AFM24 was generated using proprietary human anti-EGFR and anti-CD16A variable domains and characterized for binding, stability, manufacturability, efficacy and safety in biophysical and functional assays in vitro and in vivo. Assays for binding to target/effector cells and cytotoxicity were performed in the presence of physiologically relevant IgG levels using EGFR + tumor cell lines with and without RAS. The safety profile of AFM24 was investigated in vitro for off-target activity and in vivo in cynomolgus monkeys. Results: AFM24 showed excellent biophysical properties and picomolar EC 50 values in cytotoxicity assays. It was more potent than cetuximab, or an antibody with cetuximab-derived anti-EGFR variable domain. Importantly, AFM24 was less prone to IgG interference and eliminated cell lines harboring negative predictive biomarkers in in vitro cytotoxicity assays, even in the presence of relevant serum IgG levels. AFM24 demonstrated in vivo efficacy in a humanized hu-NOG mouse model. Furthermore, AFM24 exhibited a favorable safety profile in cynomolgus monkeys. It was well tolerated up to 94 mg/kg in a single dose, maximum tolerated dose study (MTD) when administered as a 2 hour infusion. Conclusions: Our data suggest that AFM24 is a highly potent and potentially safer drug candidate suitable for the treatment of EGFR-expressing cancers with the potential to overcome resistance to other EGFR-targeting agents and to avoid toxic side effects seen with other anti-EGFR therapeutics.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2017
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Frontiers in Oncology, Frontiers Media SA, Vol. 12 ( 2022-9-14)
    Abstract: The epidermal growth factor receptor (EGFR) is a key player in the normal tissue physiology and the pathology of cancer. Therapeutic approaches have now been developed to target oncogenic genetic aberrations of EGFR, found in a subset of tumors, and to take advantage of overexpression of EGFR in tumors. The development of small-molecule inhibitors and anti-EGFR antibodies targeting EGFR activation have resulted in effective but limited treatment options for patients with mutated or wild-type EGFR-expressing cancers, while therapeutic approaches that deploy effectors of the adaptive or innate immune system are still undergoing development. This review discusses EGFR-targeting therapies acting through distinct molecular mechanisms to destroy EGFR-expressing cancer cells. The focus is on the successes and limitations of therapies targeting the activation of EGFR versus those that exploit the cytotoxic T cells and innate immune cells to target EGFR-expressing cancer cells. Moreover, we discuss alternative approaches that may have the potential to overcome limitations of current therapies; in particular the innate cell engagers are discussed. Furthermore, this review highlights the potential to combine innate cell engagers with immunotherapies, to maximize their effectiveness, or with unspecific cell therapies, to convert them into tumor-specific agents.
    Type of Medium: Online Resource
    ISSN: 2234-943X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2022
    detail.hit.zdb_id: 2649216-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: mAbs, Informa UK Limited, Vol. 11, No. 5 ( 2019-07-04), p. 899-918
    Type of Medium: Online Resource
    ISSN: 1942-0862 , 1942-0870
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2019
    detail.hit.zdb_id: 2537838-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Wiley ; 2007
    In:  PROTEOMICS Vol. 7, No. 16 ( 2007-08), p. 2870-2880
    In: PROTEOMICS, Wiley, Vol. 7, No. 16 ( 2007-08), p. 2870-2880
    Abstract: Glycosylation is the most frequent PTM and contributes significantly to the function of proteins depending on the type of glycosylation. Especially glycan structures like the glycosaminoglycans are considered to constitute themselves the major function of the glycoconjugate which is therefore termed proteoglycan. Here we review recent views on and novel tools for analysing the proteoglycanome, which are directly related to the type of glycanation under investigation. We define the major function of the proteoglycanome to be its interaction with various proteins in many different (patho‐)physiological conditions. This is exemplified by the differential glycosaminoglycan‐interactome of healthy versus arthritic patient sera.
    Type of Medium: Online Resource
    ISSN: 1615-9853 , 1615-9861
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2007
    detail.hit.zdb_id: 2037674-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: mAbs, Informa UK Limited, Vol. 13, No. 1 ( 2021-01-01)
    Type of Medium: Online Resource
    ISSN: 1942-0862 , 1942-0870
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2021
    detail.hit.zdb_id: 2537838-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Leukemia, Springer Science and Business Media LLC, Vol. 36, No. 4 ( 2022-04), p. 1006-1014
    Type of Medium: Online Resource
    ISSN: 0887-6924 , 1476-5551
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2008023-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Immunology Research Vol. 6, No. 5 ( 2018-05-01), p. 517-527
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 6, No. 5 ( 2018-05-01), p. 517-527
    Abstract: CD16A is a potent cytotoxicity receptor on human natural killer (NK) cells, which can be exploited by therapeutic bispecific antibodies. So far, the effects of CD16A-mediated activation on NK cell effector functions beyond classical antibody-dependent cytotoxicity have remained poorly elucidated. Here, we investigated NK cell responses after exposure to therapeutic antibodies such as the tetravalent bispecific antibody AFM13 (CD30/CD16A), designed for the treatment of Hodgkin lymphoma and other CD30+ lymphomas. Our results reveal that CD16A engagement enhanced subsequent IL2- and IL15-driven NK cell proliferation and expansion. This effect involved the upregulation of CD25 (IL2Rα) and CD132 (γc) on NK cells, resulting in increased sensitivity to low-dose IL2 or to IL15. CD16A engagement initially induced NK cell cytotoxicity. The lower NK cell reactivity observed 1 day after CD16A engagement could be recovered by reculture in IL2 or IL15. After reculture in IL2 or IL15, these CD16A-experienced NK cells exerted more vigorous IFNγ production upon restimulation with tumor cells or cytokines. Importantly, after reculture, CD16A-experienced NK cells also exerted increased cytotoxicity toward different tumor targets, mainly through the activating NK cell receptor NKG2D. Our findings uncover a role for CD16A engagement in priming NK cell responses to restimulation by cytokines and tumor cells, indicative of a memory-like functionality. Our study suggests that combination of AFM13 with IL2 or IL15 may boost NK cell antitumor activity in patients by expanding tumor-reactive NK cells and enhancing NK cell reactivity, even upon repeated tumor encounters. Cancer Immunol Res; 6(5); 517–27. ©2018 AACR.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation 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. 14_Supplement ( 2016-07-15), p. 593-593
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 593-593
    Abstract: Constitutive EGFR activation through amplification or dysregulation plays an important role in the pathophysiology of numerous solid cancers, such as colorectal cancer (CRC), non-small cell lung cancer (NSCLC) or squamous cell carcinomas of the head and neck (SCCHN), thus providing a strong rationale for the development of therapeutic strategies targeting EGFR. Different therapeutic approaches have been approved for treatment of such cancers, including tyrosine kinase inhibitors that interfere with signal transduction by blocking the kinase domain or monoclonal antibodies (mAbs) that prevent EGFR ligand binding, dimerization and activation. However, despite demonstrated clinical efficacy, intrinsic or acquired resistance to such treatments has been described for a larger number of patients. Natural killer cells (NK-cells) play a central role in the innate immune system and have the capacity to destroy virally-infected or neoplastic cells. To specifically utilize the cytotoxic potential of NK-cells for the elimination of EGFR+ cancer cells, we developed tetravalent bispecific EGFR/CD16A NK-cell TandAbs, with two binding sites for EGFR being expressed on tumor cells, and two binding sites for CD16A expressed on NK-cells. Using antibody phage display technologies, we identified high affinity scFvs recognizing conformational epitopes in the extracellular domain of EGFR that were different to those targeted by other therapeutic antibodies. We engineered a set of bispecific EGFR/CD16A TandAbs and analysed their binding, thermostability and cytotoxic properties in a panel of in vitro assays. TandAbs containing our EGFR-specific domain 21 were highly potent in cytotoxicity assays towards endogenously EGFR-expressing tumor cell lines or transfected CHO cells with single digit picomolar or subpicomolar EC50 values. In contrast to comparator NK-cell recruiting TandAbs containing the Fv sequences from cetuximab, the TandAbs containing our EGFR-binding domain 21 did not exhibit any signs of temperature-induced instability or aggregation in thermostability studies. Taken together, our data suggest that EGFR/CD16A TandAbs are novel, highly potent drug candidates suitable for the treatment of EGFR-overexpressing malignancies and overcoming resistance to other therapeutic agents. Citation Format: Kristina Ellwanger, Uwe Reusch, Ivica Fucek, Michael Weichel, Erich Rajkovic, Martin Treder. Highly cytotoxic EGFR/CD16A TandAbs specifically recruit NK cells to potently kill various types of solid tumors. [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 593.
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2471-2471
    Abstract: To harness the cytotoxic capacity of immune effector cells for the treatment of several types of solid tumors, we developed tetravalent bifunctional antibodies that recognize EGFRvIII, the deletion variant III of EGFR. Their second functionality binds with high affinity to CD3, thereby directing T-cells to eliminate EGFRvIII+ cancer cells. The expression of EGFRvIII on various solid tumor types, and its absence from healthy tissues, provides an opportunity to develop cytotoxic antibodies that solely target cancer; these would spare normal tissues, and substantially reduce the side effects associated with EGFR therapy. Using phage display, we identified scFvs that selectively bind to EGFRvIII but not to EGFR. The binding affinities of these highly EGFRvIII-specific antibodies were substantially improved, employing affinity maturation techniques, and achieved KDs in the 100pM range and lower. We engineered a panel of bispecific EGFRvIII/CD3 TandAbs with a broad range of binding and cytotoxic properties. Mono- and bivalent binding, specificity for both EGFRvIII and CD3, T cell-mediated cytotoxic activity, and target-mediated T-cell activation were characterized in a panel of in vitro assays. EGFRvIII/CD3 TandAbs exhibit exquisite specificity towards the EGFRvIII antigen in Western Blot, SPR, ELISA, and FACS assays of EGFRvIII+ cells. No specific binding was observed to recombinant EGFR antigen or to EGFR-expressing cells. Apparent affinities of EGFRvIII/CD3 TandAbs to EGFRvIII were up to 25fold improved relative to the monovalently binding scFvs, and achieved a KD of 11pM for the best binding EGFRvIII/CD3 TandAb. Improvement of the binding affinities in the bivalent TandAb format was largely due to slower dissociation. TandAbs with high affinity for EGFRvIII were most potent in killing assays, displaying cytotoxicity towards EGFRvIII-expressing F98 glioma and CHO cells with EC50 in the range of 1pM-10pM. No cytotoxicity was observed on EGFR+ cells or EGFRvIII- cells up to the maximally-evaluated TandAb concentration of 0.5μM (which is 100000 fold higher than the EC50) demonstrating the high selectivity of EGFRvIII/CD3 TandAbs for the tumor-specific EGFRvIII. High affinity binding to CD3 was necessary for efficacious T cell recruitment as shown by the correlation of CD3-binding and cytotoxic potency of EGFRvIII/CD3 TandAbs. Importantly, in the absence of EGFRvIII+ target cells in vitro, TandAbs did not elicit T cell activation, as demonstrated by their lack of proliferation: this specificity contributed to a good preclinical safety profile. Biophysical and pharmacological characterization of several candidates is currently ongoing, whereby the first EGFRvIII/CD3 TandAbs demonstrated a robust dose-dependent growth retardation of EGFRvIII+ subcutaneous xenograft tumors. In summary, EGFRvIII/CD3 TandAbs are specific and highly potent drug candidates for the treatment of EGFRvIII+ malignancies. Citation Format: Kristina Ellwanger, Uwe Reusch, Ivica Fucek, Michael Weichel, Carmen Herbrecht, Stefan Knackmuss, Erich Rajkovic, Vera Molkenthin, Eugene A. Zhukovsky. EGFRvIII T-cell TandAbs are specific and highly potent drug candidates for the treatment of a variety of solid tumors. [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 2471. doi:10.1158/1538-7445.AM2015-2471
    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
    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...