Development and evaluation of nanobody tracers for noninvasive nuclear imaging of the immune-checkpoint TIGIT

Front Immunol. 2023 Sep 20:14:1268900. doi: 10.3389/fimmu.2023.1268900. eCollection 2023.

Abstract

Introduction: T cell Ig and ITIM domain receptor (TIGIT) is a next-generation immune checkpoint predominantly expressed on activated T cells and NK cells, exhibiting an unfavorable prognostic association with various malignancies. Despite the emergence of multiple TIGIT-blocking agents entering clinical trials, only a fraction of patients responded positively to anti-TIGIT therapy. Consequently, an urgent demand arises for noninvasive techniques to quantify and monitor TIGIT expression, facilitating patient stratification and enhancing therapeutic outcomes. Small antigen binding moieties such as nanobodies, are promising candidates for such tracer development.

Methods: We generated a panel of anti-human or anti-mouse TIGIT nanobodies from immunized llamas. In addition, we designed a single-chain variable fragment derived from the clinically tested monoclonal antibody Vibostolimab targeting TIGIT, and assessed its performance alongside the nanobodies. In vitro characterization studies were performed, including binding ability and affinity to cell expressed or recombinant TIGIT. After Technetium-99m labeling, the nanobodies and the single-chain variable fragment were evaluated in vivo for their ability to detect TIGIT expression using SPECT/CT imaging, followed by ex vivo biodistribution analysis.

Results: Nine nanobodies were selected for binding to recombinant and cell expressed TIGIT with low sub-nanomolar affinities and are thermostable. A six-fold higher uptake in TIGIT-overexpressing tumor was demonstrated one hour post- injection with Technetium-99m labeled nanobodies compared to an irrelevant control nanobody. Though the single-chain variable fragment exhibited superior binding to TIGIT-expressing peripheral blood mononuclear cells in vitro, its in vivo behavior yielded lower tumor-to-background ratios at one hour post- injection, indicating that nanobodies are better suited for in vivo imaging than the single-chain variable fragment. Despite the good affinity, high specificity and on-target uptake in mice in this setting, imaging of TIGIT expression on tumor- infiltrating lymphocytes within MC38 tumors remained elusive. This is likely due to the low expression levels of TIGIT in this model.

Discussion: The excellent affinity, high specificity and rapid on-target uptake in mice bearing TIGIT- overexpressing tumors showed the promising diagnostic potential of nanobodies to noninvasively image high TIGIT expression within the tumor. These findings hold promise for clinical translation to aid patient selection and improve therapy response.

Keywords: TIGIT; immune checkpoint (ICP); nanobodies; noninvasive diagnosis; nuclear imaging; tracer development.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Humans
  • Leukocytes, Mononuclear
  • Mice
  • Neoplasms* / diagnostic imaging
  • Receptors, Immunologic
  • Single-Chain Antibodies*
  • Single-Domain Antibodies* / chemistry
  • Technetium
  • Tissue Distribution
  • Tomography, Emission-Computed, Single-Photon

Substances

  • Technetium
  • Single-Chain Antibodies
  • Single-Domain Antibodies
  • Receptors, Immunologic
  • TIGIT protein, human

Grants and funding

This research was performed with the financial support of the Fonds Wetenschappelijk Onderzoek-Vlaanderen (FWO-V, grant I001618N) and personal grant from the Fonds Wetenschappelijk Onderzoek-Vlaanderen (FWO-SB, grant 1S61021N and 1S61023N). The Wetenschappelijk Fonds Willy Gepts of the UZ Brussel with project code WFWG20-26 and the Strategic Research Programme from the Vrije Universiteit Brussel.