Journal for ImmunoTherapy of Cancer (Jan 2019)

A novel anti-HER2 anthracycline-based antibody-drug conjugate induces adaptive anti-tumor immunity and potentiates PD-1 blockade in breast cancer

  • Lucia D’Amico,
  • Ulrike Menzel,
  • Michael Prummer,
  • Philipp Müller,
  • Mélanie Buchi,
  • Abhishek Kashyap,
  • Ulrike Haessler,
  • Alexander Yermanos,
  • Rémy Gébleux,
  • Manfred Briendl,
  • Tamara Hell,
  • Fabian I. Wolter,
  • Roger R. Beerli,
  • Iva Truxova,
  • Špíšek Radek,
  • Tatjana Vlajnic,
  • Ulf Grawunder,
  • Sai Reddy,
  • Alfred Zippelius

DOI
https://doi.org/10.1186/s40425-018-0464-1
Journal volume & issue
Vol. 7, no. 1
pp. 1 – 15

Abstract

Read online

Abstract Increasing evidence suggests that antibody-drug conjugates (ADCs) can enhance anti-tumor immunity and improve clinical outcome. Here, we elucidate the therapeutic efficacy and immune-mediated mechanisms of a novel HER2-targeting ADC bearing a potent anthracycline derivate as payload (T-PNU) in a human HER2-expressing syngeneic breast cancer model resistant to trastuzumab and ado-trastuzumab emtansine. Mechanistically, the anthracycline component of the novel ADC induced immunogenic cell death leading to exposure and secretion of danger-associated molecular signals. RNA sequencing derived immunogenomic signatures and TCRβ clonotype analysis of tumor-infiltrating lymphocytes revealed a prominent role of the adaptive immune system in the regulation of T-PNU mediated anti-cancer activity. Depletion of CD8 T cells severely reduced T-PNU efficacy, thus confirming the role of cytotoxic T cells as drivers of the T-PNU mediated anti-tumor immune response. Furthermore, T-PNU therapy promoted immunological memory formation in tumor-bearing animals protecting those from tumor rechallenge. Finally, the combination of T-PNU and checkpoint inhibition, such as α-PD1, significantly enhanced tumor eradication following the treatment. In summary, a novel PNU-armed, HER2-targeting ADC elicited long-lasting immune protection in a murine orthotopic breast cancer model resistant to other HER2-directed therapies. Our findings delineate the therapeutic potential of this novel ADC payload and support its clinical development for breast cancer patients and potentially other HER2 expressing malignancies.

Keywords