Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (2)
  • 1
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 22, No. 12_Supplement ( 2023-12-01), p. B121-B121
    Abstract: Background: Bispecific antibodies rapidly emerge as a powerful therapeutic tool in cancer treatment. Usage of the bispecific antibodies in oncology therapies targeting more than one antigen on cancer cells enhances the specificity of the therapy, increasing its effectiveness and decreasing the resistance rate. This approach may be used to modulate the mechanism of the defense and development of cancer cells, such as high-level expression on the cell surface the ligands of the immune checkpoints like PD-L1, and growth factor receptors like EGFR, VEGFR, or AXL. The latest reports reveal the correlation between the expression level of PD-L1 and AXL/EGFR1 in cancer. Notably, the disruption of the signaling pathway of AXL leads to a reduction of the PD-L1 on the cell surface. Adoption of this mechanism into oncology therapies may enhance the immune response against cancer cells. Moreover, the use of the bispecific antibodies simultaneously targeting PD-L1 and AXL may provoke internalization which maximizes the biological effect of the inhibition of the receptor/ligand complexes (AXL/GAS6, PD-1/PD-L1) formation. Materials and Methods: Bispecific antibody CPL976 was developed as a VHH-Fc-VHH format, with two VHHs raised against PD-L1 and AXL, selected from immune libraries generated after llamas’ immunization. Antibodies' biological activity and selectivity were studied by the Western blot technique on breast cancer cell line (MDA-MB-231) and functional assays constructed on Jurkat E6.1 and Raji cells with stable expression of the PD-1 and PD-L1, respectively. The immunomodulatory functions were also verified in cancer cell lines and PBMC co-cultures. Bispecific binding potential to AXL and PD-L1 and its effect on receptor internalization were evaluated with the use of surface plasmon resonance, Western blot technique, flow cytometry, and pH-dependent dye assays. Results: CPL976 (PD-L1 × AXL biAb), presents excellent binding kinetics with subnanomolar KDs and an innovative mechanism of action based on simultaneous internalization and degradation of both therapeutic targets at 150 nM. The compound also modulates the cell viability and migration in the breast cancer line stronger than the single agents. This data is supported by the observed decrease of the EMT (Epithelial-Mesenchymal Transition) markers. The immunomodulatory effect was confirmed in the co-culture, where the T-cell activity is blocked. The efficacy of the antibody in the tumor was checked in a murine cancer model. Conclusions: We present CPL976, an innovative bispecific antibody, with excellent binding parameters, strong biological effect on receptor presentation on the cell surface, and promising in vitro and in vivo results. These combined properties may establish a new generation of anticancer antibodies, that effectively block cancer development and break the PD-1/PD-L1 axis in patients with primary and secondary resistance to PD-1/PD-L1 targeted therapies. Such unique properties might also be later used as an effective carrier of toxic payloads in antibody-drug conjugate format. Citation Format: Agnieszka Bojko-Matuszek, Damian Kołakowski, Magdalena Bojko, Aleksandra Sowińska, Filip Mituła, Krzysztof Lacek, Delfina Popiel, Sebastian Kwiatkowski, Tomasz Kornatowski, Beata Kliszcz, Anna Jabłońska, Krzysztof Flis, Bartosz Wiernicki, Maciej Wieczorek, Jerzy Pieczykolan, Olga Abramczyk. CPL976, an innovative bispecific antibody targeting AXL and PD-L1 axis as a potential new anticancer therapeutic [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2023 Oct 11-15; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2023;22(12 Suppl):Abstract nr B121.
    Type of Medium: Online Resource
    ISSN: 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2062135-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 22, No. 12_Supplement ( 2023-12-01), p. B146-B146
    Abstract: Background: Antibody-drug conjugates (ADCs) combine the specificity of antibodies with the potency of highly cytotoxic agents, reducing the off-target effects and improve effectiveness with more precising delivery of the payload to the tumor site. Strong correlation has been established between AXL and PD-L1 expression levels in many types of cancer. Therefore, targeting both proteins simultaneously ensure high cancer specificity in delivering the toxic payload. We have developed a bispecific antibody, that strongly induces receptor internalization. Our bispecific ADC combines anti-AXL and anti-PD-L1 construct with toxic MMAE (Monomethyl auristatin E, a potent tubulin inhibitor toxin), for better targeting to solid tumor cells and greatly reducing off-target effects. Materials and Methods: CPL976-MMAE was prepared with site-specific conjugation technology using Fc glycan remodeling and click-chemistry. Briefly, after enzymatic trimming of the N-linked glycans in the Fc pocket of antibody, the terminal GlcNAc was extended with 6-N3-GalNAc using GalNAc-transferase, allowing metal-free click conjugation of the MMAE payload. Bispecific antibodies chosen for conjugation were first characterized in vitro for the interaction with the extracellular domain of human AXL and PD-L1 using surface plasmon resonance and flow cytometry. After the conjugation with MMAE, ADCs cytotoxicity, and selectivity were evaluated in the human breast cancer model (MDA-MB-231) with high expression of both targets, AXL and PD-L1, using SytoX and Hoechst. The human embryonic kidney cell line (HEK-293) with minimal expression of targets, was used as a negative control. The efficacy of conjugates were evaluated in xenograft mouse model. Results: Analytical characterization by RP-HPLC confirmed efficient conjugation and homogeneity of the conjugates. Conjugates showed improved cellular uptake, selectivity, and anticancer activity, compared to unconjugated MMAE in the AXL/PD-L1 overexpressing cancer cell lines. Additionally, HEK-293 cells were not affected by conjugates in concentrations applied in the assay with use of the breast cancer cell lines. This implies enhanced safety by reducing off-target side effects compared to free auristatin. The efficacy of the compound was confirmed in xenograft mouse model. Conclusions: We have designed a new, potentially first-in-class anti-AXL/PD-L1 bispecific antibody conjugated with MMAE, which showed robust and selective antitumor effects in human cancer models. Citation Format: Delfina Popiel, Krzysztof Lacek, Anna Jabłońska, Aleksandra Sowińska, Agnieszka Bojko - Matuszek, Damian Kołakowski, Filip Mituła, Sebastian Kwiatkowski, Magdalena Bojko, Tomasz Kornatowski, Beata Kliszcz, Tomasz Banach, Michał Górka, Jerzy Pieczykolan, Maciej Wieczorek, Olga Abramczyk. Design and preclinical evaluation of CPL976-MMAE - novel, potent AXL-PD-L1 bispecific antibody conjugated with MMAE in targeted anticancer therapy [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2023 Oct 11-15; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2023;22(12 Suppl):Abstract nr B146.
    Type of Medium: Online Resource
    ISSN: 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2062135-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages