Background: B cell depletion therapy (BCDT), such as monoclonal antibodies, bispecific T cell engagers and CAR-T, has an extensive history of effectively treating B cell malignancies. Certain BCDT monoclonal antibodies, e.g. rituximab, are also approved to treat several autoimmune diseases [1], however demonstrate poor B cells depletion in deep tissue which may limit the long-term disease control or remission in patients. Recently, anti-CD19 CAR-T therapies, through thorough depletion of pathogenic B cells and “resetting” of normal B cell populations, have achieved compelling preliminary clinical efficacy including multi-year drug-free remission in multiple autoimmune diseases [2, 3], however CAR-T modality continues to pose challenges regarding safety, cost, and logistical complexity. T Cell Engagers (TCEs) represent an attractive alternate option to CAR-T, with approved CD19-targeting bispecific TCE Blincyto demonstrating strong potential in RA [4]. However, existing bispecifics have displayed key limitations such as T cell exhaustion, poor T cell proliferation, and insufficient durability of response. Here, we developed the tri-specific TCE CC312, targeting CD3 and the key co-stimulatory receptor CD28 on T cells and CD19 on B cells. By adding critical CD28 “two signal” co-stimulation, CC312 showed efficacy consistent with CAR-T therapies, achieving greater CD4/CD8 T cell activation, proliferation, T cell survival and B cell depletion versus Blincyto in a prior study in B cell malignancies, indicating strong potential to treat autoimmune disease.
Objectives: In the current study, the potential of CC312 in treating autoimmune disease was explored in translational models, including ex vivo studies with SLE/RA donor PBMCs, an in vivo efficacy study in SLE patient derived-PBMC NCG mouse models, etc.
Methods: B cell killing of CC312 was assessed in ex vivo cytotoxicity assays using human peripheral blood mononuclear cells (PBMCs) isolated from patients with autoimmune disease. T cell activation, proliferation and memory cell differentiation were also evaluated in response to patient-derived PBMCs. Moreover, to evaluate CC312’s durability of response compared with Blincyto, SLE/RA patient derived-T cells were repeat rechallenged with B cells from the same donor. The in vivo efficacy for SLE was investigated in an animal model in which NCG mice were engrafted with SLE patient-derived PBMCs. B cell depletion in blood was analyzed by flow cytometry and in kidney tissues by immunohistochemistry.
Results: Ex vivo , CC312 induced a dose-dependent potent depletion of B cells in PBMCs derived from donors with autoimmune diseases, with EC 50 in pM range across donors and disease types. Compared with Blincyto, CC312 significantly enhanced T cell activation and proliferation due to CD28 co-stimulation. In addition, CC312 also exhibited augmented memory T cell differentiation, with higher proportion of central memory T cells (T cm ) and effector memory T cells (T em ). Moreover, CC312 displayed sustained T cell fitness, proliferation and killing effect in repeat rechallenge assay with SLE donor PBMCs, showing more durable response versus Blincyto. In vivo , CC312 thoroughly depleted blood B cells and plasmablasts as well as eliminated IgG and anti-dsDNA in peripheral blood in an SLE mouse model. At the end of this study, neither renal IgG deposits nor CD20 + B cell infiltrates were observed within kidney tissues.
Conclusion: CC312’s capacity to induce more complete T cell activation and longer T cell durability of response as well as more potently and thoroughly eliminate pathogenic B cells versus leading bispecific TCE Blincyto has been demonstrated in both ex vivo and in vivo studies. This highlights the importance of CD28 co-stimulation and indicates strong potential benefit of CC312 in achieving the depth of response needed to effectively treat B cell-related autoimmune diseases in the clinical setting.
REFERENCES: [1] M J Leandro, et al. Rheumatology (Oxford ). 2005 Dec;44(12):1542-5.
[2] Fabian Müller, et al. N Engl J Med. 2024 Feb 22;390(8):687-700.
[3] Carlo Tur, et al. Ann Rheum Dis. 2024 Sep 11:ard-2024-226142.
[4] Laura Bucci, et al. Nat Med. 2024 Jun;30(6):1593-1601.
Acknowledgements: NIL.
Disclosure of Interests: None declared.
© The Authors 2025. This abstract is an open access article published in Annals of Rheumatic Diseases under the CC BY-NC-ND license (