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: Frontiers in Plant Science, Frontiers Media SA, Vol. 11 ( 2020-8-14)
    Type of Medium: Online Resource
    ISSN: 1664-462X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2020
    detail.hit.zdb_id: 2687947-5
    detail.hit.zdb_id: 2613694-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5522-5522
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5522-5522
    Abstract: Background: Siglec-15 is a single-pass type I membrane protein that plays an important role in the immune-suppressive tumor microenvironment (TME). Siglec-15 has low expression levels in most normal human tissues but it is highly expressed in a subset of myeloid cells of the TME and over-expressed in some solid tumors. Siglec-15 on tumor associated macrophages and tumor cells inhibits T cell proliferation and pro-inflammatory cytokine release. Therefore, targeting Siglec-15 may overcome a suppressive TME and enhance the anti-tumor activity of other immune checkpoint inhibitors. Experimental procedures: Humanized mice were used immunized with recombinant Siglec-15-ECD-Fc. The Biosion proprietary H3 (High-throughput, High-content and High-efficiency) antibody screening platform was used to identify a lead anti-Siglec-15 mAb candidate-BSI-060T. Siglec-15 expression in head and neck, lung and other cancer types was assessed by immunohistochemistry (IHC) in conjunction with PD-L1. The ex vivo release of suppression of T cell activity was determined by stimulating human peripheral blood mononuclear cells with a suboptimal dose of immobilized OKT3 in the presence of recombinant human Siglec-15-Fc with and without BSI-060T. A pharmacokinetic study was carried out in cynomolgus monkeys to determine the exposure of BSI-060T over time. Tumor inhibition of BSI-060T was evaluated in Siglec-15 humanized mice that were inoculated with MC38 cells overexpressing human Siglec-15. Summary: BSI-060T is a fully human IgG1κ monoclonal antibody that binds to Siglec-15 protein with high affinity and blocks the interaction between Siglec-15 and its putative receptor LRRC4C. BSI-060T shows cross-reactivity to monkey and mouse Siglec-15. In ex vivo T cell response assays, BSI-060T exhibits strong activity on reverting Siglec-15-mediated inhibition of CD8+ and CD4+ T cell proliferation and interferon-γ release. In a humanized Siglec-15 mouse syngeneic tumor model, BSI-060T shows significant inhibition of tumor growth. BSI-060T also exhibits excellent monkey PK. In addition, an IHC assay has been developed and used to identify tumor types overexpressing Siglec-15. This assay will be used for patient recruitment in early clinical development and has the potential to be a companion diagnostic in the future. Conclusion: BSI-060T exhibits best-in-class biophysical properties and functional characteristics, supporting the initiation of development activities including manufacturing and IND-enabling studies. Citation Format: Zeyu Peng, Xiaodong F. Liu, Shukai Xia, Jinyu Liu, Hongyan Li, Yuxiang Liu, Hugh M. Davis, Mingjiu Chen, Mark Z. Ma. BSI-060T, a high affinity, fully human anti-siglec-15 antibody as an alternative immune checkpoint blocker [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5522.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 2958-2958
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 2958-2958
    Abstract: Introduction: While anti-PD1/PDL1 mAbs have shown success in the market only a small fraction of patients actually benefit from the therapies. Novel approaches to improve outcomes for patients who are resistant or refractory to current checkpoint inhibitors (CPIs) are needed. CD47 expressed on tumor cells can provide a “don’t eat me” signal to antigen-presenting cells (APCs) via the binding to SIRPα. Blocking this interaction, enhancing the phagocytosis of tumor cells, has emerged as an effective alternative to current CPIs. Here we report on the generation and characterization of a novel bispecific fusion molecule BSI-508, composed of an anti-PD1 antibody fused with a SIRPα extracellular domain (ECD). The SIRPα-ECD binds CD47 and blocks the CD47/SIRPα-mediated cell signaling pathway. BSI-508 is designed to target PD1 on T cells and CD47 on tumor cells, providing tumor killing by T cells and macrophages synergistically. Methods: An anti-PD1 mAb was identified from PD1 KO mice immunized with PD1-ECD-Fc and screened by our proprietary H3 (High-throughput, High-content and High-efficiency) platform. Human SIRPα-V2-ECD1 was fused to the N-terminus of the heavy chain of the anti-PD1 antibody via a flexible linker. The binding activities and affinities to PD1 and CD47 were evaluated by ELISA, FACS and SPR. Ligand blocking activity was measured by ELISA and FACS. A cell-based reporter assay was used to evaluate the function of the anti-PD1 portion and a macrophage mediated phagocytosis assay was used to evaluate the function of the SIRPα-V2-ECD1 portion of the bispecific. B-hPD-1/hPD-L1/hSIRPα/hCD47 mice bearing B-hPDL1/hCD47 MC38 tumors were used to evaluate the tumor inhibitory activity of the bispecific fusion molecule. Results: BSI-508 demonstrated comparable activity to the parental anti-PD1 monoclonal antibody regarding PD1 binding and PD1/PDL1 blocking. It also exhibited comparable activity to the SIRPα-V2-ECD1-Fc fusion protein in CD47 binding and CD47/SIRPα blocking. BSI-508 was able to reverse PD1 mediated T-cell suppression and exhibited comparable bioactivity to the parental anti-PD1 antibody. The bispecific molecule showed comparable bioactivity to the SIRPα-V2-ECD1-Fc fusion protein in inducing macrophage mediated phagocytosis of Jurkat cells. In addition, BSI-508 showed superior antitumor efficacy in vivo. Summary: BSI-508 is a novel anti-PD1xanti-CD47 bispecific fusion molecule combining both T-cell activation and macrophage mediated phagocytosis together for superior tumor killing. BSI-508 demonstrates favorable biophysical and functional characteristics, supporting the initiation of development activities including manufacturing and IND-enabling studies. Citation Format: Zeyu Peng, Xiaodong F. Liu, Hongyan Li, Wenwen Dai, Jinyu Liu, Xiaoyao Hao, Shukai Xia, Qun Lv, Hugh M. Davis, Mingjiu Chen, Mark Z. Ma. BSI-508, a novel bispecific fusion molecule targeting PD1 and CD47 for cancer immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2958.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6367-6367
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6367-6367
    Abstract: Introduction: Despite the success of anti-PD1/PDL1 therapies, only a small fraction of patients benefit from these checkpoint inhibitors (CPIs). Novel approaches to improve outcomes for patients who are resistant to current CPIs are needed. PVRIG is expressed on CD4+ and CD8+ T cells, NKT and NK cells. PVRIG binds with high affinity with its ligand PVRL2, which is expressed on tumor cells and some myeloid cells. The PVRIG-PVRL2 axis exerts an inhibitory effect on the cytotoxic activity of lymphocytes. Bispecific antibodies that exhibit dual blockade of PD1 and PVRIG provide a promising strategy to enhance anti-tumor immune response. Methods: An anti-PD1 mAb was identified from PD1 KO mice immunized with PD1-ECD-Fc and screened by our proprietary H3 (High-throughput, High-content and High-efficiency) platform. An anti-PVRIG mAb was identified from rats immunized with recombinant PVRIG-ECD-Fc and screened by the H3 platform. Both anti-PD1 and anti-PVRIG antibodies were humanized, and the anti-PD1 scFv was fused to the N-terminus of the heavy chain of the anti-PD1 antibody via a flexible linker. The binding activities and affinities were evaluated by ELISA, FACS and SPR, and the ligand blocking activities were measured by ELISA and FACS. Cell-based reporter assays were used to evaluate the functions of the anti-PD1 and anti-PVRIG mAbs alone and the bispecific antibody. In addition, the activity of the bispecific antibody to reverse PD1 and PVRIG mediated suppression of CMV pp65495-503 antigen specific CD8+ T-cell cytotoxicity was evaluated. Results: BSI-507, an anti-PD1xanti-PVRIG bispecific antibody demonstrated comparable activity to the parental anti-PD1 antibody regarding PD1 binding and PD1/PDL1 blocking. It also exhibited comparable activity to the parental anti-PVRIG antibody in PVRIG binding and PVRIG/PVRL2 blocking. Based on cell-based reporter assays, BSI-507 was able to reverse either PD1 or PVRIG mediated T-cell suppression and exhibited comparable potency to the parental antibodies. BSI-507 was also able to show enhanced reversal of both PD1 and PVRIG mediated T-cell suppression, much better than anti-PD1 or anti-PVRIG alone. In addition, BSI-507 showed the ability to reverse PD1 and PVRIG mediated suppression of CMV pp65495-503 antigen specific CD8+ T-cell cytotoxicity, stronger than either the anti-PD1 or anti-PVRIG monoclonal antibody. Summary: BSI-507 is a first-in-class anti-PD1xanti-PVRIG bispecific antibody for dual blockade of PD-1 and PVRIG pathways to enhance reversal of T cell inhibition. BSI-507 demonstrates favorable biophysical and functional characteristics, supporting the initiation of development activities including manufacturing and IND-enabling studies. Citation Format: Zeyu Peng, Xiaodong F. Liu, Hongyan Li, Wenwen Dai, Jinyu Liu, Xiaoyao Hao, Shukai Xia, Qun Lv, Hugh M. Davis, Mingjiu Chen, Mark Z. Ma. BSI-507, a first-in-class bispecific antibody targeting PD1 and PVRIG for cancer immunotherapy. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6367.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5290-5290
    Abstract: Background: Trastuzumab and pertuzumab are anti-HER2 antibodies that bind to two different epitopes of HER2. Although pertuzumab itself did not exhibit substantial clinical benefit as monotherapy, it was approved to be used in combination with trastuzumab because of its ability to enhance the clinical efficacy of trastuzumab. Currently, the combination of trastuzumab and pertuzumab is being used as first-line treatment for HER2-positive metastatic breast cancer. However, while the combination of trastuzumab and pertuzumab has been effective, there is still opportunity for greater effectiveness by identifying anti-HER2 mAbs that will be better synergistic partners of trastuzumab, allowing for better efficacy and/or broader indications. Methods: A SynAb࣪ Platform was used to identify BSI-001, an anti-HER2 mAb, through trastuzumab-based synergistic functional screening. A competitive ELISA was used to perform epitope binning of the anti-HER2 antibodies. BSI-001 was further characterized in combination with trastuzumab for its bioactivity in vitro, including inhibition of cell proliferation and antigen-mediated internalization of HER2 positive cell lines. The synergistic effect of BSI-001 with trastuzumab was also evaluated in trastuzumab-resistant cell lines. Multiple animal models were then employed to investigate the synergistic antitumor activity of the combination in vivo. Results: BSI-001 is the best functional partner of trastuzumab identified through the SynAb࣪ platform. BSI-001 binds to a unique epitope, distinct from the binding epitopes of trastuzumab and pertuzumab. It exhibits superior bioactivity and efficacy in vitro and in vivo when combined with trastuzumab. The combination of BSI-001 and trastuzumab inhibits HER2-positive cancer cell growth more potently than that of trastuzumab and pertuzumab. The combination of BSI-001 with trastuzumab also shows inhibition of cell growth of the trastuzumab-resistant cell line BT-474 Clone 5, whereas the combination of pertuzumab and trastuzumab did not. In addition, BSI-001 is able to synergize with trastuzumab to promote antigen-antibody internalization. In animal models, BSI-001 demonstrates superior synergistic antitumor activity when combined with trastuzumab as compared to pertuzumab. Summary: The combination of BSI-001 and trastuzumab showed significantly better in vitro and in vivo activity than the combination of trastuzumab and pertuzumab. In addition, because of the activity of the BSI-001/trastuzumab combination in a trastuzumab-resistant cancer cell line, there is the potential for this combination to be used to treat patients with trastuzumab-resistant cancer or patients who experienced relapse following trastuzumab and pertuzumab treatment. Citation Format: Shukai Xia, Zeyu Peng, Xiaoyu Ding, Wanjian Gu, Yujie Zhong, Jinyu Liu, Hongyan Li, Xiaoyao Hao, Xiaodong F. Liu, Mark Z. Ma, Lan Luo, Chunni Zhang, Mingjiu Chen. BSI-001, a novel anti-HER2 antibody exhibiting potent synergistic efficacy with trastuzumab [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5290.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5312-5312
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5312-5312
    Abstract: Background: SIRPα is an inhibitory immunoreceptor, expressed on macrophages that interacts with CD47 expressed on tumor cells to release a “don't eat me” signal, resulting in immune evasion of tumor cells. Antibodies targeting CD47 or SIRPα are expected to block the CD47/SIRPα signaling pathway, thereby stimulating macrophage-based phagocytosis of tumor cells. Anti-CD47 antibodies usually have side effects related to anemia and thrombocytopenia since CD47 is expressed on normal cells, including red blood cells and platelets. Targeting SIRPα on the macrophage is an alternative approach to blocking CD47/SIRPα signaling without the side effects observed in anti-CD47 antibody treatment. In addition to SIRPα there are two other members of the SIRP family - SIRPβ and SIRPγ. These isoforms share high sequence homology with SIRPα but have very different functionality. Moreover, SIRPα is polymorphic in humans, among which SIRPαV1, SIRPαV2 and SIRPαV8 are the main SIRPα variants in diverse ethnic groups. Together, the key challenge of anti-SIRPα antibody discovery is to identify lead candidates that specifically bind to SIRPα and block the SIRPα/CD47 interaction while also recognizing the majority of SIRPα variants. Experimental procedures: Humanized mice were used for immunization and the proprietary H3 (High-throughput, High-content and High-efficiency) antibody screening platform was used to identify a lead candidate - BSI-082. Affinity was measured by SPR while specificity of antibodies to SIRPα and blocking activity of CD47/SIRPα were evaluated by ELISA and FACS. Functional activity of antibodies was investigated using monocyte-derived macrophages and Raji cells in the presence of Rituximab with and without anti-SIRPα antibody. Antibody internalization was evaluated by using the Biosion SynTracer™ antibody discovery platform. Results: BSI-082 is a fully human IgG1/κ mAb that binds to SIRPα and blocks the SIRPα/CD47-mediated signaling pathway. BSI-082 is a highly selective SIRPα binder with very weak or no binding to SIRPβ and no binding to SIRPγ. In addition, BSI-082 binds three main SIRPα variants (V1, V2 and V8) with high affinity. Through antibody engineering, BSI-082 harbors an engineered Fc domain that prevents FcγR binding and enhances FcRn binding, thus significantly reducing ADCC while also extending half-life. In the presence of rituximab, BSI-082 promotes macrophage-mediated phagocytosis. BSI-082 exhibits effective internalization, which would deplete SIRPα on macrophages and enhance immune response. Conclusion: BSI-082 exhibits best-in-class biophysical properties, SIRPα specificity, broad SIRPα variant binding and superior functional characteristics, supporting the initiation of development activities including manufacturing and IND-enabling studies. Citation Format: Zeyu Peng, Jinyu Liu, Wenwen Dai, Xiaodong F. Liu, Shukai Xia, Hongyan Li, Lei Shi, Hugh M. Davis, Mingjiu Chen, Mark Z. Ma. BSI-082, a fully human anti-SIRPα antibody with both high specificity and broad coverage of SIRPα variant populations [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5312.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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
    Online Resource
    Online Resource
    Science China Press., Co. Ltd. ; 2012
    In:  Chinese Science Bulletin Vol. 57, No. 28-29 ( 2012-10-1), p. 2765-2770
    In: Chinese Science Bulletin, Science China Press., Co. Ltd., Vol. 57, No. 28-29 ( 2012-10-1), p. 2765-2770
    Type of Medium: Online Resource
    ISSN: 0023-074X
    Language: English
    Publisher: Science China Press., Co. Ltd.
    Publication Date: 2012
    SSG: 6,25
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    Wiley ; 2004
    In:  Molecular Reproduction and Development Vol. 69, No. 1 ( 2004-09), p. 66-76
    In: Molecular Reproduction and Development, Wiley, Vol. 69, No. 1 ( 2004-09), p. 66-76
    Type of Medium: Online Resource
    ISSN: 1040-452X , 1098-2795
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2004
    detail.hit.zdb_id: 1493888-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Elsevier BV ; 2014
    In:  Applied Mathematics and Computation Vol. 245 ( 2014-10), p. 520-525
    In: Applied Mathematics and Computation, Elsevier BV, Vol. 245 ( 2014-10), p. 520-525
    Type of Medium: Online Resource
    ISSN: 0096-3003
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2014
    detail.hit.zdb_id: 1465428-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 2018
    In:  Medicine Vol. 97, No. 32 ( 2018-08), p. e11806-
    In: Medicine, Ovid Technologies (Wolters Kluwer Health), Vol. 97, No. 32 ( 2018-08), p. e11806-
    Type of Medium: Online Resource
    ISSN: 0025-7974
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2018
    detail.hit.zdb_id: 2049818-4
    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...