1. Academic Validation
  2. Harnessing ExDNA for precision exatecan delivery in cancer: a novel antibody-drug conjugate approach

Harnessing ExDNA for precision exatecan delivery in cancer: a novel antibody-drug conjugate approach

  • Mol Cancer. 2025 Oct 13;24(1):253. doi: 10.1186/s12943-025-02462-z.
Zaira Ianniello 1 Huimei Lu 2 Elias Quijano 1 3 Daniel A Colón-Ríos 1 3 Madison Rackear 1 3 Venu Bommireddy 4 Dale L Ludwig 4 Zhiyuan Shen 2 Peter M Glazer 5 6
Affiliations

Affiliations

  • 1 Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06520, USA.
  • 2 Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA.
  • 3 Department of Genetics, Yale University School of Medicine, New Haven, CT, 06520, USA.
  • 4 Gennao Bio, Hopewell, NJ, 08525, USA.
  • 5 Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, CT, 06520, USA. peter.glazer@yale.edu.
  • 6 Department of Genetics, Yale University School of Medicine, New Haven, CT, 06520, USA. peter.glazer@yale.edu.
Abstract

Background: Current antibody-drug conjugates (ADCs) face limitations due to a lack of tumor-selective targets, inefficient internalization, and challenges in reaching tumors in challenging sites, ultimately limiting their therapeutic efficacy. We developed and characterized V66-exatecan, a novel ADC composed of V66, a humanized antibody with high affinity for extracellular DNA (exDNA), conjugated to exatecan via a Cleavable Linker. This ADC employs a dual-targeting mechanism based on exDNA and ENT2 transporter expression to enhance nuclear drug delivery and tumor specificity. This study evaluates its anti-tumor activity, mechanism of action, ability to treat challenging tumors, and safety profile.

Methods: To validate tumor selectivity, V66 or a control antibody were conjugated to a fluorescent tag and injected intravenously into tumor-bearing mice; biodistribution analysis demonstrated selective accumulation in tumors and nuclear localization within tumor cells. V66 was then conjugated to exatecan via a Cleavable Linker. In vitro assays across diverse Cancer cell lines assessed cytotoxicity, DNA damage response (DDR) activation, and TOP1 degradation. In vivo efficacy was evaluated in xenograft models of triple-negative breast Cancer (TNBC) and BRCA1/2-deficient tumors, including intracranial medulloblastoma. These models were used to assess tumor growth inhibition, survival benefit, and blood-brain barrier (BBB) permeability. Toxicity was assessed through a dose-escalation study, with analysis of hematologic parameters, histopathology of major organs, and liver and kidney function tests (ALT, AST, BUN, total protein) following short- and long-term treatment.

Results: V66-exatecan demonstrated potent anti-tumor activity in multiple Cancer cell lines but not on healthy mouse primary fibroblasts, with EC50 values in the low nanomolar range. It induced robust DDR signaling, TOP1 degradation, and bystander killing effects. BRCA1/2-deficient models exhibited enhanced penetration and sensitivity, with up to 17-fold lower EC50 compared to BRCA-proficient controls. In vivo, V66-exatecan significantly inhibited tumor growth and extended survival in both TNBC and BRCA-mutant CNS tumors, including complete regressions and prolonged median survival in BRCA2-deficient models. Toxicology studies revealed no significant hematologic, renal, hepatic, or bone marrow toxicity, even at high or repeated doses.

Conclusions: V66-exatecan represents a next-generation of ADCs that overcomes key limitations of traditional platforms by exploiting exDNA-driven tumor selectivity and ENT2-mediated nuclear delivery. It demonstrates broad therapeutic efficacy and a favorable safety profile, supporting its potential for treating DDR-deficient and hard-to-reach tumors.

Supplementary Information: The online version contains supplementary material available at 10.1186/s12943-025-02462-z.

Keywords

Antibody-Drug conjugate; BRCA-deficient tumors; DNA damage; Drug delivery system; ENT2; ExDNA; Nuclear penetration; Targeted cancer therapy; Topoisomerase-I; Tumor microenvironment.

Figures
Products