PTX3 from vascular endothelial cells contributes to trastuzumab-induced cardiac complications

Cardiovasc Res. 2023 May 22;119(5):1250-1264. doi: 10.1093/cvr/cvad012.

Abstract

Aims: Trastuzumab, the first humanized monoclonal antibody that targets human epidermal growth factor receptor 2 (ERBB2/HER2), is currently used as a first-line treatment for HER2 (+) tumours. However, trastuzumab increases the risk of cardiac complications without affecting myocardial structure, suggesting a distinct mechanism of cardiotoxicity.

Methods and results: We used medium from trastuzumab-treated human umbilical vein endothelial cells (HUVECs) to treat CCC-HEH-2 cells, the human embryonic cardiac tissue-derived cell lines, and human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) to assess the crosstalk between vascular endothelial cells (VECs) and cardiomyocytes. Protein mass spectrometry analysis was used to identify the key factors from VECs that regulate the function of cardiomyocytes. We applied RNA-sequencing to clarify the mechanism, by which PTX3 causes cardiac dysfunction. We used an anti-human/rat HER2 (neu) monoclonal antibody to generate a rat model that was used to evaluate the effects of trastuzumab on cardiac structure and function and the rescue effects of lapatinib on trastuzumab-induced cardiac side effects. Medium from trastuzumab-treated HUVECs apparently impaired the contractility of CCC-HEH-2 cells and iPSC-CMs. PTX3 from VECs caused defective cardiomyocyte contractility and cardiac dysfunction in mice, phenocopying trastuzumab treatment. PTX3 affected calcium homoeostasis in cardiomyocytes, which led to defective contractile properties. EGFR/STAT3 signalling in VECs contributed to the increased expression and release of PTX3. Notably, lapatinib, a dual inhibitor of EGFR/HER2, could rescue the cardiac complications caused by trastuzumab by blocking the release of PTX3.

Conclusion: We identified a distinct mode of cardiotoxicity, wherein the activation of EGFR/STAT3 signalling by trastuzumab in VECs promotes PTX3 excretion, which contributes to the impaired contractility of cardiomyocytes by inhibiting cellular calcium signalling. We confirmed that lapatinib could be a feasible preventive agent against trastuzumab-induced cardiac complications and provided the rationale for the combined application of lapatinib and trastuzumab in cancer therapy.

Keywords: Cardiac complications; Lapatinib; PTX3; Trastuzumab; Vascular endothelial cells.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antibodies, Monoclonal / adverse effects
  • Antineoplastic Agents* / toxicity
  • Breast Neoplasms* / drug therapy
  • Calcium / metabolism
  • Cardiotoxicity / metabolism
  • Cardiotoxicity / pathology
  • Endothelial Cells / metabolism
  • Female
  • Heart Diseases* / chemically induced
  • Heart Diseases* / metabolism
  • Heart Diseases* / prevention & control
  • Humans
  • Induced Pluripotent Stem Cells* / metabolism
  • Lapatinib / adverse effects
  • Lapatinib / metabolism
  • Mice
  • Quinazolines / adverse effects
  • Rats
  • Receptor, ErbB-2 / metabolism
  • Trastuzumab / metabolism
  • Trastuzumab / toxicity

Substances

  • Trastuzumab
  • Lapatinib
  • Calcium
  • Quinazolines
  • Receptor, ErbB-2
  • Antibodies, Monoclonal
  • Antineoplastic Agents