Abstract

Abstract Introduction: Multiple myeloma (MM) is the second most prevalent hematologic malignancy. Chimeric antigen receptor (CAR)-T cells and bispecific T-cell engagers targeting BCMA, CS1, and CD38 are promising approaches for MM. However, using these targets individually limits the efficacy of these immunotherapy options due to the variable expression levels of these markers, leading to inefficient eradication of MM. Studies have attempted to develop CAR-T cells and T-cell engagers with multispecific targeting; however, these are still technically very challenging and are highly expensive. In this study, we developed nanoparticles with monovalent or multivalent specificity against MM targets and engage T cells, by chemical conjugation of monoclonal antibodies against each molecule to the surface of the liposome. Bi-specific nanoparticles with anti CD38/CD3, BCMA/CD3 or CS1/CD3 were termed nanoBiTEs, while the multispecific particles with anti BCMA/CS1/CD38/CD3 were termed nanoMuTEs. Methods: Liposomes were prepared using the thin-film hydration method followed by extrusion. nanoBiTEs and nanoMuTEs were developed by chemical conjugation of monoclonal antibodies against BCMA, CS1, and/or CD38; together with anti CD3 onto the liposomes. We have tested the expression of BCMA, CS1, and CD38 antigens in MM cell lines and primary MM patient samples, and we analyzed the binding of the CD38/CD3, BCMA/CD3, and CS1/CD3 nanoBiTEs, as well as the BCMA/CS1/CD38/CD3 nanoMuTEs to these cells by flow cytometry and confocal microscopy. Binding specificity was assessed by blocking the target with corresponding free antibodies. CD25 expression, a T-cell activation marker, was assessed for CD8 and CD4 T cells when incubated with or without nanoBiTEs and nanoMuTEs. Moreover, we tested the efficacy of the nanoBiTEs and nanoMuTEs against MM cells in vitro using the 3D tissue engineered bone marrow (3DTEBM) myeloma model. Finally, we tested the efficacy of the nanoBiTEs and nanoMuTE against MM tumors in vivo in xenograft MM models. Results: The expression of BCMA, CS1 and CD38 was variable in MM cell lines and patient samples, and so was the binding of each nanoBiTEs, while the binding of the nanoMuTE was higher and more universal than each nanoBiTE. Similar patterns were observed in activation of CD8 T cells (no significant activation was observed in CD4 T cells) and in eradication of MM cell lines and primary patient samples in vitro. In vivo, the nanoBiTEs extended the survival and reduced the tumor burden in the MM bearing mice compared to untreated and T cell alone groups, and nanoMuTE experiments are still ongoing. Conclusions: The nanoBiTEs and nanoMuTEs were bound specifically to MM cells and initiated CD8 T-cell activation that resulted in MM eradication in vitro and in vivo, while the effect of the nanoMuTEs was more universal and efficacious than each nanoBiTE. These nanoparticles are an inexpensive method of engaging T cells for the treatment of MM and represent a new class of immunotherapy that can be expanded to other tumors. Citation Format: Kinan Alhallak, Jennifer Sun, Barbara Muz, Christopher Egbulefu, Pilar de la Puente, Cinzia Federico, Justin King, Danny Kohnen, Ravi Vij, Samuel Achilefu, Abdel Kareem Azab. Nanoparticle multispecific T-cell engagers for the treatment of multiple myeloma [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2018 Nov 27-30; Miami Beach, FL. Philadelphia (PA): AACR; Cancer Immunol Res 2020;8(4 Suppl):Abstract nr B01.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call