Abstract

Simple SummaryRelapse after apparent remission remains a major cause of death in patients with acute myeloid leukemia (AML). On the cellular level, leukemia relapse is considered to emerge from subpopulations of therapy-resistant leukemic stem cells (LSC). Identification and targeting of LSC are thus most important goals for AML treatment. However, AML and their LSC are highly heterogeneous. Here, we review the current knowledge on AML LSC identification and targeting via surface antigens with a focus on heterogeneity among different AML subgroups and genetic backgrounds.Patients suffering from acute myeloid leukemia (AML) show highly heterogeneous clinical outcomes. Next to variabilities in patient-specific parameters influencing treatment decisions and outcome, this is due to differences in AML biology. In fact, different genetic drivers may transform variable cells of origin and co-exist with additional genetic lesions (e.g., as observed in clonal hematopoiesis) in a variety of leukemic (sub)clones. Moreover, AML cells are hierarchically organized and contain subpopulations of more immature cells called leukemic stem cells (LSC), which on the cellular level constitute the driver of the disease and may evolve during therapy. This genetic and hierarchical complexity results in a pronounced phenotypic variability, which is observed among AML cells of different patients as well as among the leukemic blasts of individual patients, at diagnosis and during the course of the disease. Here, we review the current knowledge on the heterogeneous landscape of AML surface markers with particular focus on those identifying LSC, and discuss why identification and targeting of this important cellular subpopulation in AML remains challenging.

Highlights

  • Acute myeloid leukemia (AML) is a devastating, rapidly-evolving disease characterized by an abnormal proliferation of poorly-differentiated cells which impairs normal hematopoiesis

  • Since acute myeloid leukemia (AML) is highly heterogeneous with respect to genetic alterations, epigenetics, and leukemia cell of origin, it is not surprising that considerable heterogeneity is observed among surface markers of AML cells and their leukemic stem cells (LSC) [2], making immunological targeting of LSC a constant challenge [7,8,9]

  • Its expression may regulate niche interactions and it was first described as LSC marker in chronic myeloid leukemia [110], but later shown to be expressed on MLL-rearranged (MLLr) AML [70], on the CD34+ CD38− compartment

Read more

Summary

Introduction

Acute myeloid leukemia (AML) is a devastating, rapidly-evolving disease characterized by an abnormal proliferation of poorly-differentiated cells which impairs normal hematopoiesis. Human CD34+ leukemic cells were shown to repopulate the bone marrow (BM) of severe combined immunodeficient (SCID) mice, while CD34− leukemic blasts remained non-leukemogenic [3,4]. These CD34+ cells responsible for leukemia initiation and maintenance were termed LSC. AML is organized hierarchically and contains subpopulations of LSC that share functional and molecular properties with their cells of origin, the healthy hematopoietic stem and progenitor cells (HSPC) [4,13,14,15]. LSC in only some, but not all AML (e.g., CD93, TIM3, CD44, CD123, etc. [9,20,21,22,23,24,25,26])

The Relevance of Immunomodulatory Proteins for LSC Detection
CD34 Expressing
CD34 Non-Expressing AML and Their LSC
Absence of NKG2D Ligands
Phenotypic LSC Evolution and Intra-Patient Heterogeneity
Association between the Genetic Background and the LSC Phenotype in AML
Therapeutic Targeting of LSC
Concluding Remarks
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call