Download PDF

Blood

Publication date: 2021-02-11
Volume: 137 Pages: 801 - 811
Publisher: American Society of Hematology

Author:

Alberti-Servera, Llucia
Demeyer, Sofie ; Govaerts, Inge ; Swings, Toon ; De Bie, Jolien ; Gielen, Olga ; Brociner, Marco ; Michaux, Lucienne ; Maertens, Johan ; Uyttebroeck, Anne ; De Keersmaecker, Kim ; Boeckx, Nancy ; Segers, Heidi ; Cools, Jan

Keywords:

Science & Technology, Life Sciences & Biomedicine, Hematology, MUTATIONS, GENOME, GENETICS, Blood Cells, Bone Marrow Cells, Child, Clonal Evolution, DNA, Neoplasm, Humans, INDEL Mutation, Male, Mutation, Neoplasm Proteins, Neoplasm, Residual, PTEN Phosphohydrolase, Phylogeny, Polymorphism, Single Nucleotide, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma, Receptor, Notch1, Recurrence, Salvage Therapy, Sensitivity and Specificity, Sequence Analysis, DNA, Single-Cell Analysis, C14/18/104#54689614, 1102 Cardiorespiratory Medicine and Haematology, 1103 Clinical Sciences, 1114 Paediatrics and Reproductive Medicine, Immunology, 3101 Biochemistry and cell biology, 3201 Cardiovascular medicine and haematology, 3213 Paediatrics

Abstract:

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive leukemia that is most frequent in children and is characterized by the presence of few chromosomal rearrangements and 10 to 20 somatic mutations in protein-coding regions at diagnosis. The majority of T-ALL cases harbor activating mutations in NOTCH1 together with mutations in genes implicated in kinase signaling, transcriptional regulation, or protein translation. To obtain more insight in the level of clonal heterogeneity at diagnosis and during treatment, we used single-cell targeted DNA sequencing with the Tapestri platform. We designed a custom ALL panel and obtained accurate single-nucleotide variant and small insertion-deletion mutation calling for 305 amplicons covering 110 genes in about 4400 cells per sample and time point. A total of 108 188 cells were analyzed for 25 samples of 8 T-ALL patients. We typically observed a major clone at diagnosis (>35% of the cells) accompanied by several minor clones of which some were less than 1% of the total number of cells. Four patients had >2 NOTCH1 mutations, some of which present in minor clones, indicating a strong pressure to acquire NOTCH1 mutations in developing T-ALL cells. By analyzing longitudinal samples, we detected the presence and clonal nature of residual leukemic cells and clones with a minor presence at diagnosis that evolved to clinically relevant major clones at later disease stages. Therefore, single-cell DNA amplicon sequencing is a sensitive assay to detect clonal architecture and evolution in T-ALL.