Your browser doesn't support javascript.
loading
Serial monitoring of genomic alterations in circulating tumor cells of ER-positive/HER2-negative advanced breast cancer: feasibility of precision oncology biomarker detection.
Cani, Andi K; Dolce, Emily M; Darga, Elizabeth P; Hu, Kevin; Liu, Chia-Jen; Pierce, Jackie; Bradbury, Kieran; Kilgour, Elaine; Aung, Kimberly; Schiavon, Gaia; Carroll, Danielle; Carr, T Hedley; Klinowska, Teresa; Lindemann, Justin; Marshall, Gayle; Rowlands, Vicky; Harrington, Elizabeth A; Barrett, J Carl; Sathiyayogan, Nitharsan; Morrow, Christopher; Sero, Valeria; Armstrong, Anne C; Baird, Richard; Hamilton, Erika; Im, Seock-Ah; Jhaveri, Komal; Patel, Manish R; Dive, Caroline; Tomlins, Scott A; Udager, Aaron M; Hayes, Daniel F; Paoletti, Costanza.
Afiliação
  • Cani AK; Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
  • Dolce EM; Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
  • Darga EP; Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
  • Hu K; Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
  • Liu CJ; Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
  • Pierce J; Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
  • Bradbury K; Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
  • Kilgour E; Michigan Center for Translational Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
  • Aung K; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
  • Schiavon G; Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
  • Carroll D; Michigan Center for Translational Pathology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
  • Carr TH; Cancer Research UK, Manchester Institute Cancer Biomarker Centre, University of Manchester, UK.
  • Klinowska T; Cancer Research UK, Manchester Institute Cancer Biomarker Centre, University of Manchester, UK.
  • Lindemann J; Cancer Research UK, Manchester Institute Cancer Biomarker Centre, University of Manchester, UK.
  • Marshall G; Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
  • Rowlands V; Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA.
  • Harrington EA; Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
  • Barrett JC; Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
  • Sathiyayogan N; Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
  • Morrow C; Late Development, Oncology R&D, AstraZeneca, Cambridge, UK.
  • Sero V; Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
  • Armstrong AC; Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
  • Baird R; Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
  • Hamilton E; Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
  • Im SA; Research and Early Development, Oncology R&D, AstraZeneca, Waltham, MA, USA.
  • Jhaveri K; Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
  • Patel MR; Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
  • Dive C; Menarini Silicon Biosystems, Inc., San Diego, CA, USA.
  • Tomlins SA; Department of Medical Oncology, The Christie NHS Foundation Trust and the Faculty of Biology, Medicine and Health, The University of Manchester, UK.
  • Udager AM; Breast Cancer and Early Phase Clinical Trials Teams, Cancer Research UK, Cambridge Centre, UK.
  • Hayes DF; Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, TN, USA.
  • Paoletti C; Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
Mol Oncol ; 16(10): 1969-1985, 2022 05.
Article em En | MEDLINE | ID: mdl-34866317
ABSTRACT
Nearly all estrogen receptor (ER)-positive (POS) metastatic breast cancers become refractory to endocrine (ET) and other therapies, leading to lethal disease presumably due to evolving genomic alterations. Timely monitoring of the molecular events associated with response/progression by serial tissue biopsies is logistically difficult. Use of liquid biopsies, including circulating tumor cells (CTC) and circulating tumor DNA (ctDNA), might provide highly informative, yet easily obtainable, evidence for better precision oncology care. Although ctDNA profiling has been well investigated, the CTC precision oncology genomic landscape and the advantages it may offer over ctDNA in ER-POS breast cancer remain largely unexplored. Whole-blood (WB) specimens were collected at serial time points from patients with advanced ER-POS/HER2-negative (NEG) advanced breast cancer in a phase I trial of AZD9496, an oral selective ER degrader (SERD) ET. Individual CTC were isolated from WB using tandem CellSearch® /DEPArray™ technologies and genomically profiled by targeted single-cell DNA next-generation sequencing (scNGS). High-quality CTC (n = 123) from 12 patients profiled by scNGS showed 100% concordance with ctDNA detection of driver estrogen receptor α (ESR1) mutations. We developed a novel CTC-based framework for precision medicine actionability reporting (MI-CTCseq) that incorporates novel features, such as clonal predominance and zygosity of targetable alterations, both unambiguously identifiable in CTC compared to ctDNA. Thus, we nominated opportunities for targeted therapies in 73% of patients, directed at alterations in phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), fibroblast growth factor receptor 2 (FGFR2), and KIT proto-oncogene, receptor tyrosine kinase (KIT). Intrapatient, inter-CTC genomic heterogeneity was observed, at times between time points, in subclonal alterations. Our analysis suggests that serial monitoring of the CTC genome is feasible and should enable real-time tracking of tumor evolution during progression, permitting more combination precision medicine interventions.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias da Mama / DNA Tumoral Circulante / Células Neoplásicas Circulantes Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias da Mama / DNA Tumoral Circulante / Células Neoplásicas Circulantes Idioma: En Ano de publicação: 2022 Tipo de documento: Article