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
Filter
  • American Association for Cancer Research (AACR)  (3)
Materialart
Verlag/Herausgeber
  • American Association for Cancer Research (AACR)  (3)
Sprache
Erscheinungszeitraum
Fachgebiete(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6326-6326
    Kurzfassung: Background: Recent studies on tumor microenvironment (TME) have been conferred considerable attention, especially the transforming growth factor-β (TGF-β) signal axis and the PD-L1/PD-1 signal axis. TGF-β is a crucial enforcer in promoting tumor cells angiogenesis, plasticity, fibrosis, and in regulating immune homeostasis and tolerance. The TGF-β signaling controls innate and adaptive immunity in TME by inhibiting the activity of natural killer (NK) cells and effector T cells, while promoting the expansion of regulatory T cells (Tregs). Many studies have revealed that free TGF-β mainly released from glycoprotein A repetitions predominant (GARP)-TGFβ complex at the surface of activated Tregs, B, NK, and tumor cells. Existing antibodies or fusion proteins targeting TGF-β cannot simultaneously block the TGF-β releasing and naturalized free TGF-β in TME, thus limiting the anti-tumor effect. Herein, we designed a bi-specific antibody named as BPB-101 with triple functions: specifically target GARP-TGFβ complex and/or small latent complex (SLC), free TGF-β, and PD-L1. BPB-101 can stop tumor expansion and recruitment, and may enhance the immune effector cells to eliminate the tumor. Methods: An anti-GARP mAb was discovered using a hybridoma platform and selected for binding with GARP-TGFβ complex/SLC and free TGF-β. An anti-PD-L1 nAb was screened out by VHH library and optimized in tumor-bearing mice. After humanizing, these two antibodies were successfully assembled to form a unique bi-specific antibody, BPB-101. The binding affinity of BPB-101 to GARP-TGFβ complex/SLC, free TGF-β and PD-L1 was determined by ELISA, FACS and SPR/BLI. Report gene assay was utilized to detect the blocking activity of BPB-101. The TGF-β secretion from activated Tregs was also analyzed. Finally, the anti-tumor efficacy of BPB-101 was assessed with a transgenic mouse model (B6-hLRRC32). To address the biosafety risks, the nonclinical safety profile was also evaluated in cynomolgus monkeys. Results and Conclusion: BPB-101 is a high-affinity antibody aims to reverse immunosuppressive TME by triple-targeting functions. The blocking ability of TGF-β in not only latent form but also free form makes our BPB-101 distinct from M7824, any other existing anti-TGF-β mAbs and anti-GARP mAbs. In a MC38-hPD-L1 tumor-bearing B6-hLRRC32 model, BPB-101 showed significant tumor growth inhibition of 93 % at the dose of 5 mg/kg (p & lt;0.001), showing superior efficacy to monoclonal antibody alone or in combination with anti-PD-L1 nAb. In addition, BPB-101 showed durable responsiveness and a splendid safety profile with manageable adverse events. These striking results support the clinical development potential of our trifunctional agent as a monotherapy and in combination with other immunotherapies. Citation Format: Wenxin Xu, Jieying Xu, Deyu Xu, Hongjie Cheng, Li Zhang, Huan Zheng, Siyuan Ye, Mengshi Jiang, Jiabing Wang, Lieming Ding. Discovery & preclinical evaluation of BPB-101: A novel triple functional bi-specific antibody targeting GARP-TGFβ complex/SLC, free TGF-β and PD-L1 [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 6326.
    Materialart: Online-Ressource
    ISSN: 1538-7445
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2023
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 3 ( 2011-02-01), p. 976-987
    Kurzfassung: It is well known that tumor-derived proangiogenic factors induce neovascularization to facilitate tumor growth and malignant progression. However, the concept of “angiocrine” signaling, in which signals produced by endothelial cells elicit tumor cell responses distinct from vessel function, has been proposed, yet remains underinvestigated. Here, we report that angiocrine factors secreted from endothelium regulate tumor growth and motility. We found that Slit2, which is negatively regulated by endothelial EphA2 receptor, is one such tumor suppressive angiocrine factor. Slit2 activity is elevated in EphA2-deficient endothelium. Blocking Slit activity restored angiocrine-induced tumor growth/motility, whereas elevated Slit2 impaired growth/motility. To translate our findings to human cancer, we analyzed EphA2 and Slit2 expression in human cancer. EphA2 expression inversely correlated with Slit2 in the vasculature of invasive human ductal carcinoma samples. Moreover, analysis of large breast tumor data sets revealed that Slit2 correlated positively with overall and recurrence-free survival, providing clinical validation for the tumor suppressor function for Slit2 in human breast cancer. Together, these data support a novel, clinically relevant mechanism through which EphA2 represses Slit2 expression in endothelium to facilitate angiocrine-mediated tumor growth and motility by blocking a tumor suppressive signal. Cancer Res; 71(3); 976–87. ©2010 AACR.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2011
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 17 ( 2016-09-01), p. 5054-5067
    Kurzfassung: Epithelial–mesenchymal transition (EMT) is an essential mechanism of metastasis, including in colorectal cancer. Although EMT processes are often triggered in cancer cells by their surrounding microenvironment, how EMT-relevant genes control these processes is not well understood. In multiple types of cancers, the transcription factor MEF2D has been implicated in cell proliferation, but its contributions to metastasis have not been addressed. Here, we show MEF2D is overexpressed in clinical colorectal cancer tissues where its high expression correlates with metastatic process. Functional investigations showed that MEF2D promoted cancer cell invasion and EMT and that it was essential for certain microenvironment signals to induce EMT and metastasis in vivo. Mechanistically, MEF2D directly regulated transcription of the EMT driver gene ZEB1 and facilitated histone acetylation at the ZEB1 promoter. More importantly, MEF2D responded to various tumor microenvironment signals and acted as a central integrator transducing multiple signals to activate ZEB1 transcription. Overall, our results define a critical function for MEF2D in upregulating EMT and the metastatic capacity of colorectal cancer cells. Further, they offer new insights into how microenvironment signals activate EMT-relevant genes and deepen the pathophysiologic significance of MEF2D, with potential implications for the prevention and treatment of metastatic colorectal cancer. Cancer Res; 76(17); 5054–67. ©2016 AACR.
    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 ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...