Construction and preclinical evaluation of a I-labelled bispecific antibody targeting PD-L1 and PD-L2.

Eur J Nucl Med Mol Imaging

Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Peking University, No. 52 Fu-Cheng Rd, Beijing, 100142, People's Republic of China.

Published: December 2024

AI Article Synopsis

  • The study focused on developing a bispecific antibody, NB12, that targets PD-L1 and PD-L2, aimed at monitoring cancer patient responses to immune checkpoint inhibitors (ICI) through a radiotracer called [I]I-NB12.
  • In vitro testing showed that [I]I-NB12 had a high affinity for its targets, was effectively taken up by certain cancer cells, and demonstrated favorable pharmacokinetics.
  • Micro-PET/CT imaging indicated that [I]I-NB12 successfully accumulated in tumor regions, confirming its potential for dynamic imaging in cancer diagnostics.

Article Abstract

Purpose: NB12 is a bispecific antibody that consists of two anti-programmed cell death-ligand 1 (PD-L1) nanobodies and two anti-programmed cell death-ligand 2 (PD-L2) nanobodies. The aim of this study was to design a novel tracer, [I]I-NB12, targeting PD-L1/2 and perform preclinical evaluations to dynamically monitor PD-L1/2 expression for determining cancer patient responsiveness to ICI therapy.

Methods: NB12 was labelled with the radionuclide I at room temperature (RT). An in vitro binding assay was performed to assess the affinity of [I]I-NB12 for PD-L1 and PD-L2. Cellular uptake, pharmacokinetic, and biodistribution experiments were performed to evaluate the biological properties. Micro-PET/CT imaging with [I]I-NB12 was conducted at different time points. Immunohistochemical and haematoxylin and eosin (HE) staining experiments were carried out using tumour tissues. Routine blood, biochemical indices and major organ pathology were used to evaluate the biosafety of the tracers.

Results: The radiochemical yield of [I]I-NB12 was 84.62 ± 3.90%, and the radiochemical purity (RCP) was greater than 99%. [I]I-NB12 had a high affinity for the PD-L1 (Kd = 19.82 nM) and PD-L2 (Kd = 2.93 nM). Cellular uptake experiments confirmed that the uptake of [I]I-NB12 by A549-PDL1/2 cells was greater than that by A549 cells. The half-lives of the distribution phase and elimination phase were 0.26 h and 4.08 h, respectively. Micro-PET/CT showed significant [I]I-NB12 uptake in the tumour region of A549-PDL1/2 tumour-bearing mice compared with A549 tumour-bearing mice 24 h postinjection. Immunohistochemical and HE staining experiments confirmed that tumour-bearing mice was successfully constructed.

Conclusion: We constructed a bispecific antibody that targets PD-L1 and PD-L2, namely, [I]I-NB12. Biological evaluation revealed its specificity and affinity for PD-L1/2, and micro-PET/CT confirmed the feasibility of visualizing tumour PD-L1/2 in vivo. Using [I]I-NB12 may be a promising strategy for identifying cancer patients that can potentially benefit from ICI therapy.

Download full-text PDF

Source
http://dx.doi.org/10.1007/s00259-024-06886-5DOI Listing

Publication Analysis

Top Keywords

bispecific antibody
12
pd-l1 pd-l2
12
tumour-bearing mice
12
[i]i-nb12
9
anti-programmed cell
8
cell death-ligand
8
cellular uptake
8
staining experiments
8
experiments confirmed
8
pd-l1
5

Similar Publications

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 PDF

A humanized anti-MSLN×4-1BB bispecific antibody exhibits potent antitumour activity through 4-1BB signaling activation and fc function without systemic toxicity.

J 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.

View Article and Find Full Text PDF

Background And Hypothesis: Teclistamab, a novel bispecific monoclonal antibody targeting CD3 and B-cell maturation antigen (BCMA), and chimeric antigen receptor T-cell (CAR-T) therapy are promising options for treating relapsed/refractory multiple myeloma (MM). However, the rates of acute kidney injury (AKI) associated with teclistamab remain inadequately characterized. This study aims to compare the incidence, severity, and outcomes of AKI between patients receiving teclistamab and CAR-T therapy.

View Article and Find Full Text PDF

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 PDF

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 PDF

Want AI Summaries of new PubMed Abstracts delivered to your In-box?

Enter search terms and have AI summaries delivered each week - change queries or unsubscribe any time!