Bispecific antibodies are on the cusp of coming of age as therapeutics more than half a century after they were first described. Two bispecific antibodies, catumaxomab (Removab(®), anti-EpCAM×anti-CD3) and blinatumomab (Blincyto(®), anti-CD19×anti-CD3) are approved for therapy, and >30 additional bispecific antibodies are currently in clinical development. Many of these investigational bispecific antibody drugs are designed to retarget T cells to kill tumor cells, whereas most others are intended to interact with two different disease mediators such as cell surface receptors, soluble ligands and other proteins. The modular architecture of antibodies has been exploited to create more than 60 different bispecific antibody formats. These formats vary in many ways including their molecular weight, number of antigen-binding sites, spatial relationship between different binding sites, valency for each antigen, ability to support secondary immune functions and pharmacokinetic half-life. These diverse formats provide great opportunity to tailor the design of bispecific antibodies to match the proposed mechanisms of action and the intended clinical application.
Download full-text PDF |
Source |
---|---|
http://dx.doi.org/10.1016/j.molimm.2015.01.003 | DOI Listing |
J Transl Med
January 2025
Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
Background: Multiple myeloma (MM) is an incurable plasma cell malignancy with increasing global incidence. Chimeric antigen receptor (CAR) T-cell therapy targeting BCMA has shown efficacy in relapsed or refractory MM, but it faces resistance due to antigen loss and the tumor microenvironment. Bispecific T-cell engaging (BITE) antibodies also encounter clinical challenges, including short half-lives requiring continuous infusion and potential toxicities.
View Article and Find Full Text PDFJ Transl Med
January 2025
Department of Medical Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, 230031, Anhui, China.
Background: Agonistic monoclonal antibodies targeting 4-1BB/CD137 have shown preclinical promise, but their clinical development has been limited by severe liver toxicity or limited efficacy. Therefore, a safe and efficient immunostimulatory molecule is urgently needed for cancer immunotherapy.
Methods: A novel anti-MSLN×4-1BB bispecific antibody (bsAb) was generated via antibody engineering, and its affinity and activity were detected via enzyme-linked immunosorbent assay (ELISA), flow cytometry, and T-cell activation and luciferase reporter assays.
Rheumatol Immunol Res
December 2024
Department of Toxicology, School of Public Health, Peking University; Peking China.
Autoimmune diseases arise from immune system dysfunction that immune cells mistakenly attack the body's own tissues, resulting in systemic disorders or localized lesions such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Autoreactive B cells play a critical role in the pathogenesis of many autoimmune diseases and B cell depletion using anti-CD20 monoclonal antibody (mAb) has been shown to effectively mitigate disease progression in both preclinical and clinical studies. Recently, bispecific antibody (bsAb) targeting CD20/CD3 have demonstrated substantial clinical benefits in the treatment of various hematologic malignancies.
View Article and Find Full Text PDFJ Adv Pract Oncol
September 2024
St Luke's Health System - Pharmacy, Boise, Idaho.
Bispecific antibodies (BsAbs) have emerged as crucial therapeutic agents for patients with relapsed/refractory diffuse large B-cell lymphoma, multiple myeloma, and most recently, lung cancer. These therapies have demonstrated remarkable efficacy in clinical trials; however, multidisciplinary collaboration is essential to ensure optimal patient outcomes amid the operational complexities associated with BsAb therapy. As BsAbs are being prepared for broader adoption, clinicians and treatment centers must navigate operational challenges, including financial considerations, patient selection, caregiver involvement, and transitions of care.
View Article and Find Full Text PDFJ Immunother Cancer
January 2025
Medical Oncology, Sarah Cannon Research Institute, Nashville, Tennessee, USA.
Background: SL-172154 is a hexameric fusion protein adjoining the extracellular domain of SIRPα to the extracellular domain of CD40L via an inert IgG-derived Fc domain. In preclinical studies, a murine equivalent SIRPα-Fc-CD40L fusion protein provided superior antitumor immunity in comparison to CD47- and CD40-targeted antibodies. A first-in-human phase I trial of SL-172154 was conducted in patients with platinum-resistant ovarian cancer.
View Article and Find Full Text PDFEnter search terms and have AI summaries delivered each week - change queries or unsubscribe any time!