Single-cell multiomics reveals increased plasticity, resistant populations, and stem-cell–like blasts in KMT2A-rearranged leukemia

KMT2A-rearranged (KMT2A-r) infant acute lymphoblastic leukemia (ALL) is a devastating malignancy with a dismal outcome, and younger age at diagnosis is associated with increased risk of relapse. To discover age-specific differences and critical drivers that mediate poor outcome in KMT2A-r ALL, we su...

Full description

Saved in:
Bibliographic Details
Published inBlood Vol. 139; no. 14; pp. 2198 - 2211
Main Authors Chen, Changya, Yu, Wenbao, Alikarami, Fatemeh, Qiu, Qi, Chen, Chia-hui, Flournoy, Jennifer, Gao, Peng, Uzun, Yasin, Fang, Li, Davenport, James W., Hu, Yuxuan, Zhu, Qin, Wang, Kai, Libbrecht, Clara, Felmeister, Alex, Rozich, Isaiah, Ding, Yang-yang, Hunger, Stephen P., Felix, Carolyn A., Wu, Hao, Brown, Patrick A., Guest, Erin M., Barrett, David M., Bernt, Kathrin M., Tan, Kai
Format Journal Article
LanguageEnglish
Published United States Elsevier Inc 07.04.2022
American Society of Hematology
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:KMT2A-rearranged (KMT2A-r) infant acute lymphoblastic leukemia (ALL) is a devastating malignancy with a dismal outcome, and younger age at diagnosis is associated with increased risk of relapse. To discover age-specific differences and critical drivers that mediate poor outcome in KMT2A-r ALL, we subjected KMT2A-r leukemias and normal hematopoietic cells from patients of different ages to single-cell multiomics analyses. We uncovered the following critical new insights: leukemia cells from patients <6 months have significantly increased lineage plasticity. Steroid response pathways are downregulated in the most immature blasts from younger patients. We identify a hematopoietic stem and progenitor-like (HSPC-like) population in the blood of younger patients that contains leukemic blasts and form an immunosuppressive signaling circuit with cytotoxic lymphocytes. These observations offer a compelling explanation for the ability of leukemias in young patients to evade chemotherapy and immune-mediated control. Our analysis also revealed preexisting lymphomyeloid primed progenitors and myeloid blasts at initial diagnosis of B-ALL. Tracking of leukemic clones in 2 patients whose leukemia underwent a lineage switch documented the evolution of such clones into frank acute myeloid leukemia (AML). These findings provide critical insights into KMT2A-r ALL and have clinical implications for molecularly targeted and immunotherapy approaches. Beyond infant ALL, our study demonstrates the power of single-cell multiomics to detect tumor intrinsic and extrinsic factors affecting rare but critical subpopulations within a malignant population that ultimately determines patient outcome. •Single-cell multiomics analysis reveals higher plasticity and stem-cell-like blasts in younger KMT2A-r ALL patients with worse prognosis.•The most immature leukemic cells exhibit steroid resistance and stem–cell-like cells contribute to immune evasion in younger patients. [Display omitted]
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
C.C., W.Y., and F.A. contributed equally to this study.
ISSN:0006-4971
1528-0020
DOI:10.1182/blood.2021013442