Diagnostics (Aug 2020)

Personalized Treatment Selection and Disease Monitoring Using Circulating Tumor DNA Profiling in Real-World Cancer Patient Management

  • Julius Wehrle,
  • Ulrike Philipp,
  • Martina Jolic,
  • Marie Follo,
  • Saskia Hussung,
  • Silvia Waldeck,
  • Max Deuter,
  • Michael Rassner,
  • Jan Braune,
  • Justyna Rawluk,
  • Christine Greil,
  • Cornelius F. Waller,
  • Heiko Becker,
  • Jesús Duque-Afonso,
  • Anna L. Illert,
  • Ralph M. Fritsch,
  • Frank Meiss,
  • Justus Duyster,
  • Nikolas von Bubnoff,
  • Florian Scherer

DOI
https://doi.org/10.3390/diagnostics10080550
Journal volume & issue
Vol. 10, no. 8
p. 550

Abstract

Read online

Background: Circulating tumor DNA (ctDNA) in the blood plasma of cancer patients is an emerging biomarker used across oncology, facilitating noninvasive disease monitoring and genetic profiling at various disease milestones. Digital droplet PCR (ddPCR) technologies have demonstrated high sensitivity and specificity for robust ctDNA detection at relatively low costs. Yet, their value for ctDNA-based management of a broad population of cancer patients beyond clinical trials remains elusive. Methods: We developed mutation-specific ddPCR assays that were optimized for their use in real-world cancer management, covering 12 genetic aberrations in common cancer genes, such as EGFR, BRAF, KIT, KRAS, and NRAS. We assessed the limit of detection (LOD) and the limit of blank (LOB) for each assay and validated their performance for ctDNA detection using matched tumor sequencing. Results: We applied our custom ddPCR assays to 352 plasma samples from 96 patients with solid tumors. Mutation detection in plasma was highly concordant with tumor sequencing, demonstrating high sensitivity and specificity across all assays. In 20 cases, radiographic cancer progression was mirrored by an increase of ctDNA concentrations or the occurrence of novel mutations in plasma. Moreover, ctDNA profiling at diagnosis and during disease progression reflected personalized treatment selection through the identification of actionable gene targets in 20 cases. Conclusion: Collectively, our work highlights the potential of ctDNA assessment by sensitive ddPCR for accurate disease monitoring, robust identification of resistance mutations, and upfront treatment selection in patients with solid tumors. We envision an increasing future role for ctDNA profiling within personalized cancer management in daily clinical routine.

Keywords