-Author name in bold denotes the presenting author
-Asterisk * with author name denotes a Non-ASH member
Clinically Relevant Abstract denotes an abstract that is clinically relevant.

PhD Trainee denotes that this is a recommended PHD Trainee Session.

Ticketed Session denotes that this is a ticketed session.

1859 The Novel Leukemia Stem Cell Marker GPR56 Discriminates Leukemic Subclones with Divergent Stem Cell Properties in Human Acute Myeloid Leukemia

Malignant Stem and Progenitor Cells
Program: Oral and Poster Abstracts
Session: 661. Malignant Stem and Progenitor Cells: Poster I
Saturday, December 5, 2015, 5:30 PM-7:30 PM
Hall A, Level 2 (Orange County Convention Center)

Caroline Pabst, M.D.1,2*, Anne Bergeron, PhD3*, Vincent-Philippe Lavallée, M.D.4, Jonathan Yeh, PhD5*, Patrick Gendron, M.Sc2*, Jana Krosl, PhD6*, Geneviève Boucher, M.Sc2*, Sébastien Lemieux, Ph.D.2,7*, Josee Hébert, MD8,9,10,11, Guy Sauvageau, M.D., Ph.D.2,9,12 and Frédéric Barabé, MD13,14,15

1Martin-Luther-University of Halle-Wittenberg, Department of Internal Medicine IV- Hematology and Oncology, Halle (Saale), Germany
2The Leucegene project at Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
3Centre de Recherche du CHU de Québec, Centre de Recherche en Infectiologie, Quebec City, Canada
4Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
5University of Montreal, Institute for Research in Immunology and Cancer, Montreal, Canada
6IRIC/Unv. de Montreal Faculty of Medicine, Montreal, QC, Canada
7Department of Computer Science and Operations Research, Université de Montréal, Montréal, QC, Canada
8Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
9Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
10Hematology-Oncology Division, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
11Quebec Leukemia Cell Bank, Montréal, QC, Canada
12Quebec Leukemia Cell Bank, Maisonneuve-Rosemont Hospital, Montréal, QC, Canada
13Centre de Recherche du CHU de Québec, Centre de Recherche en Infectiologie, Québec, Canada
14CHU de Québec, Hopital Enfant-Jésus, Québec, Canada
15Department of Medicine, Université Laval, Québec, Canada

Insights into the complex clonal architecture of acute myeloid leukemia (AML) unravelled by deep sequencing technologies have challenged the concept of AML as a hierarchically organised disease initiated and driven by rare self-renewing leukemic stem cells (LSCs). In contrast to normal human hematopoietic stem cells (HSCs), which are highly enriched in the CD34+CD38- population, LSCs have also been found in the CD34- and the CD38+ fractions questioning the existence of a consistent LSC surface marker profile for AML. Besides, low LSC frequencies in primary samples, rapid onset of differentiation upon ex vivo culture, and genetic inter-specimen heterogeneity hamper the dissection of the molecular machinery that drives LSC self-renewal.

We performed RNA-Sequencing of primary human AML samples and assessed LSC frequencies by limiting dilution analyses for 56 of these in NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice. By comparing gene expression profiles between high vs low LSC frequency leukemias, we identified the G-protein coupled receptor 56 (GPR56) has significantly more expressed in high LSC frequency leukemias. We validated the RNA-seq data with protein expression by FACS and found an excellent correlation. To determine whether GPR56 positive cells overlapped with the known LSC-associated phenotype CD34+CD38-, we stained 45 AML samples with CD34, CD38, GPR56, and antibodies against other described LSC markers. Although CD34+GPR56+ and CD34+CD38- compartments identified the same population in some samples, we found in the majority of samples that GPR56 further subdivided the CD34+CD38- compartment. Accordingly, not only the proportions of total GPR56+ and CD34+GPR56+ cells were significantly higher in LSChigh versus LSClow samples, but also the proportion of GPR56+ cells within the CD34+CD38- compartment was significantly different between the groups indicating that GPR56 might be of additional value to what is currently considered the best described LSC phenotype. The percentage of total CD34 positive cells did not correlate with LSC frequency clearly distinguishing GPR56 from CD34 or CD38, which are only suitable LSC markers when used in combination. We analysed other potential LSC markers (TIM3, CD96, CD44, CD123, CLL1 and CD47) in our RNA-Seq dataset and by FACS analysis in combination with CD34 as we did for GPR56 and none of them correlated with LSC frequency in our sample collection.

To determine whether GPR56 discriminates engrafting LSCs from non-LSCs, we sorted GPR56+ and GPR56- cells within the CD34-positive and -negative compartments from selected specimens with known engraftment potential. We found that GPR56 identified the engrafting fraction in CD34 positive AML samples, with a  >50 fold enrichment in LSC in the CD34+GRP56+ fraction vs the CD34+GPR56- fraction within the same sample, demonstrating that GPR56 is a good LSC marker.

Specimens with high molecular or cytogenetic risk such as chromosome 5 or 7 anomalies and EVI1-rearrangement expressed high levels of both, GPR56 and CD34, while samples with coexistent FLT3-ITD, DNMT3A, and NPM1 mutations displayed a unique CD34lowGPR56high profile. Moreover, we found a divergent distribution of variant allele frequencies in GPR56+ versus GPR56- fractions identifying GPR56 as a discriminator of leukemic sub-clones with high and low NSG engrafting capacity. Analysis of engrafted cells re-sorted based on GPR56 after being harvested from mouse bone marrow revealed reduced complexity of the clonal composition. Most importantly, GPR56 positive cells differentiated to GPR56 negative cells in mice, which did not happen in the human niche, in which GPR56 positive and negative fractions represented two independently evolved subclones.

In summary our work identifies GPR56 as a novel LSC marker in AML and also shows that GPR56 readily identifies a functionally distinct LSC-rich subclone in the majority of human AML patients and reveals hitherto unforeseen complexity in the interaction between human LSCs and the NSG mouse environment.

Disclosures: No relevant conflicts of interest to declare.

Previous Abstract | Next Abstract >>

*signifies non-member of ASH