Sei sulla pagina 1di 9

From www.bloodjournal.org by on December 17, 2010. For personal use only.

2007 110: 2659-2666


Prepublished online Jul 3, 2007;
doi:10.1182/blood-2007-03-083048

The novel AML stem cellassociated antigen CLL-1 aids in


discrimination between normal and leukemic stem cells
Anna van Rhenen, Guus A. M. S. van Dongen, Angèle Kelder, Elwin J. Rombouts, Nicole Feller,
Bijan Moshaver, Marijke Stigter-van Walsum, Sonja Zweegman, Gert J. Ossenkoppele and Gerrit Jan
Schuurhuis

Updated information and services can be found at:


http://bloodjournal.hematologylibrary.org/cgi/content/full/110/7/2659
Articles on similar topics may be found in the following Blood collections:
Hematopoiesis and Stem Cells (2756 articles)
Neoplasia (4217 articles)
Stem Cells in Hematology (166 articles)

Information about reproducing this article in parts or in its entirety may be found online at:
http://bloodjournal.hematologylibrary.org/misc/rights.dtl#repub_requests

Information about ordering reprints may be found online at:


http://bloodjournal.hematologylibrary.org/misc/rights.dtl#reprints

Information about subscriptions and ASH membership may be found online at:
http://bloodjournal.hematologylibrary.org/subscriptions/index.dtl

Blood (print ISSN 0006-4971, online ISSN 1528-0020), is published weekly


by the American Society of Hematology, 2021 L St, NW, Suite 900,
Washington DC 20036.
Copyright 2010 by The American Society of Hematology; all rights reserved.
From www.bloodjournal.org by on December 17, 2010. For personal use only.
NEOPLASIA

The novel AML stem cell–associated antigen CLL-1 aids in discrimination


between normal and leukemic stem cells
Anna van Rhenen,1 Guus A. M. S. van Dongen,2 Angèle Kelder,1 Elwin J. Rombouts,3 Nicole Feller,1 Bijan Moshaver,1
Marijke Stigter-van Walsum,2 Sonja Zweegman,1 Gert J. Ossenkoppele,1 and Gerrit Jan Schuurhuis1
1Department of Hematology and 2Department of Otolaryngology, VU [Vrije Universiteit] University Medical Center, Amsterdam; 3Department of Hematology,

Erasmus University Medical Center, Rotterdam, the Netherlands

In CD34ⴙ acute myeloid leukemia (AML), also present on the CD34ⴙCD38ⴚ stem- cells in normal (n ⴝ 11) and in regenerat-
the malignant stem cells reside in the cell compartment in AML (77/89 patients). ing bone marrow controls (n ⴝ 6). This
CD38ⴚ compartment. We have shown be- The CD34ⴙCLL-1ⴙ population, containing AML stem-cell specificity of the anti-
fore that the frequency of such the CD34ⴙCD38ⴚCLL-1ⴙ cells, does en- CLL-1 antibody under all conditions of
CD34ⴙCD38ⴚ cells at diagnosis corre- graft in nonobese diabetic/severe com- disease and the leukemia-initiating prop-
lates with minimal residual disease (MRD) bined immunodeficiency (NOD/SCID) mice erties of CD34ⴙCLL-1ⴙ cells indicate that
frequency after chemotherapy and with with outgrowth to CLL-1ⴙ blasts. CLL-1 anti–CLL-1 antibody enables both AML-
survival. Specific targeting of expression was not different between di- specific stem-cell detection and possibly
CD34ⴙCD38ⴚ cells might thus offer thera- agnosis and relapse (n ⴝ 9). In remission, antigen-targeting in future. (Blood. 2007;
peutic options. Previously, we found that both CLL-1ⴚ normal and CLL-1ⴙ malig- 110:2659-2666)
C-type lectin-like molecule-1 (CLL-1) has nant CD34ⴙCD38ⴚ cells were present.
high expression on the whole blast com- A high CLL-1ⴙ fraction was associated
partment in the majority of AML cases. with quick relapse. CLL-1 expression is
We now show that CLL-1 expression is completely absent both on CD34ⴙCD38ⴚ © 2007 by The American Society of Hematology

Introduction
Despite high-dose chemotherapy, only 30% to 40% of patients with therapy-) treated patient cannot be considered normal, it is ex-
acute myeloid leukemia (AML) survive, which is due mainly to tremely important to study whether after treatment normal stem
relapse of the disease.1 AML is generally regarded as a stem-cell cells in such regenerating bone marrow remain negative for the
disease. However, there is debate whether normal stem cells antigen of interest. So far, this has not been examined for CD33,
undergoing leukemogenic mutations is the explanation for leukemo- CD44, and CD123.
genesis. Alternatively, leukemogenic mutations occurring at a later In this paper, we focus on the newly discovered antigen CLL-1,
developmental stage, resulting in stem cell–like behavior, might be which we have described to be present on the majority of CD34⫹ as
an alternative or additional option.2-4 For CD34⫹ AML, several well as CD34⫺ AML cases.12 In peripheral blood, both monocytes
authors have shown that leukemic stem cells are present in the and granulocytes show some CLL-1 expression, while it is absent
CD34⫹CD38⫺ compartment.5,6 It has been proven in vitro that in other tissues.12 The intracellular domain of CLL-1 contains both
these stem cells are more resistant to chemotherapy, compared with an immunotyrosine-based inhibition motif as well as a YXXM
the progenitor CD34⫹CD38⫹ cells.7 In vivo, after chemotherapy,
motif, suggesting a role for CLL-1 as a signaling receptor.
the residual malignant CD34⫹CD38⫺ cells are thought to differen-
Phosphorylation of immunotyrosine-based inhibition motif–
tiate to a limited extent, producing leukemic cells with an
containing receptors on a variety of cells leads to inhibition of
immunophenotype, which usually reflects that at diagnosis. Sensi-
activation pathways via recruitment of the protein tyrosine phospha-
tive techniques allow early detection of small numbers of these
differentiated leukemic cells, called minimal residual disease tases SHP-1, SHP-2, and SHIP.13 The YXXM motif, on the other
(MRD), which eventually causes relapse of the disease.8 Since in hand, encompasses a potential SH2 domain–binding site for the
this concept the stem cell is the origin of MRD and relapse, stem p85 subunit of phosphatidylinositol 3⬘-kinase,14 an enzyme impli-
cell–targeted therapy would be of potentially high benefit for AML cated in cellular activation pathways. Whether these properties can
patients. Moreover, early detection of leukemic stem cells after be translated to CLL-1 is still unknown.
chemotherapeutic treatment might offer prognostic value in predict- We now demonstrate that CD34⫹CLL-1⫹ cells of AML patients
ing relapse of the disease. Different options for stem-cell identifica- showed engraftment in nonobese diabetic/severe combined immunode-
tion and/or targeted therapy have been described such as anti- ficient (NOD/SCID) mice, indicating that CLL-1 is expressed on the
CD123, anti-CD44, and anti-CD33, but all have some (potential) leukemic stem-cell population in these patients. Also, we found CLL-1
disadvantages, including expression on normal stem cells and/or to be present on AML and absent on normal CD34⫹CD38⫺ cells at
nonhematologic tissues.9-11 Since the bone marrow of a (chemo- different time points of disease/treatment, which holds potential to serve

Submitted March 30, 2007; accepted June 29, 2007. Prepublished online as payment. Therefore, and solely to indicate this fact, this article is hereby
Blood First Edition paper, July 3, 2007; DOI 10.1182/blood-2007-03-083048. marked ‘‘advertisement’’ in accordance with 18 USC section 1734.

The publication costs of this article were defrayed in part by page charge © 2007 by The American Society of Hematology

BLOOD, 1 OCTOBER 2007 䡠 VOLUME 110, NUMBER 7 2659


From www.bloodjournal.org by on December 17, 2010. For personal use only.
2660 VAN RHENEN et al BLOOD, 1 OCTOBER 2007 䡠 VOLUME 110, NUMBER 7

as a tool to detect residual leukemic CD34⫹CD38⫺ cells after therapy on ice with 10 mL lysis buffer (155 mM NH4Cl, 10 mM KHCO3, 0.1 mM
and as a possible target for therapy. Na2EDTA, pH 7.4) and washed twice with phosphate-buffered saline (PBS)
with 0.1% human serum albumin (HSA) added. Frozen samples were
prepared using a Ficoll gradient (1.077 g/mL; Amersham Biosciences,
Freiburg, Germany) and subsequent red blood cell lysis. Cells were then
Patients, materials, and methods frozen in RPMI (Gibco, Paisley, United Kingdom) with 20% heat-
inactivated fetal bovine serum (FBS; Greiner, Alphen aan den Rijn, the
Patient and control samples Netherlands) and 10% DMSO (Riedel-de Haen, Seelze, Germany) in
The AML clinical protocols and the biologic studies were approved by the isopropanol-filled containers and subsequently stored in liquid nitrogen.
scientific research committee and the medical ethics committee of the VU When needed for analysis, cells were thawed and suspended in prewarmed
University Medical Center (Amsterdam, the Netherlands). Leukemic cells RPMI with 40% FCS at 37°C. Cells were washed and enabled to recover for
of 89 patients presenting with CD34⫹ AML at our institute were obtained 45 minutes in RPMI with 40% FCS at 37°C. Cells were washed again and
after informed consent in accordance with the Declaration of Helsinki, at suspended in PBS with 0.1% HSA.17
diagnosis, and after chemotherapeutic treatment. CD34⫹ AML was defined
as samples with a CD34 percentage more than 1 because we have Fluorescence-activated cell sorting analysis
previously shown that in samples with less than 1% CD34⫹ cells, these of CD34ⴙCD38ⴚ cells
CD34⫹ cells are in general of normal origin.15 In 16 cases, bone marrow
Fluorescence-activated cell sorting (FACS) procedures have been described in
(BM) was not available at diagnosis and peripheral blood (PB) was used
detail before.17 In short, fresh cells were incubated with monoclonal antibodies
instead. After chemotherapeutic treatment, only BM was used. Relapse
for 15 minutes at room temperature, washed once in PBS containing 0.1% HSA,
AML BM samples were obtained from 9 patients. Diagnosis of patients was
and analyzed by flow cytometry. Monoclonal antibody combinations contained
based on morphology using French-American-British (FAB) classification,
fluorescein isothiocyanate (FITC)–, phycoerythrin (PE)–, peridinyl chlorophyllin
immunophenotyping, and cytogenetics.16 Control normal bone marrow
(PerCP)–, or allophycocyanin (APC)–labeled monoclonal antibodies. Anti-
(NBM) was obtained from 11 patients undergoing cardiac surgery after
CD34 FITC, anti-CD34 PerCP, anti-CD45 PerCP, anti-CD45 APC, anti-CD38
informed consent. Control regenerating bone marrow (RBM) was obtained
APC, anti-CD123 PE, anti-CD7 PE, anti-CD19 PE, anti-CD33 FITC, anti-CD33
from 3 patients with acute lymphoblastic leukemia, one patient with
APC, and Via-Probe (7-amino-actinomycin D, 7AAD) were all from BD
non-Hodgkin lymphoma, one patient with completely CD34⫺ AML, and
Biosciences (Basel, Switzerland); anti-CD34 FITC, which was used in some of
one patient with CLL-1⫺ AML. Mobilized peripheral blood (MPB) was
the samples, was from Immunotech (Marseille, France); and annexinV FITC was
obtained from 6 non-AML patients after granulocyte colony-stimulating
from Nexins Research (Kattendijke, the Netherlands). Anti–CLL-1 and isotype
factor (G-CSF) stimulation.
controls (GBS and DNP) were from Crucell (Leiden, the Netherlands).
Patient characteristics are shown in Table 1. AML samples were
When frozen-thawed cells were used, annexinV FITC was included in
analyzed fresh (n ⫽ 52) or after storage in liquid nitrogen (n ⫽ 37). For
the majority of samples to gate out apoptotic/dead cells before stem-cell
expression studies after chemotherapy and in controls fresh material was
assessment. In the remaining samples, this was done by Syto16 (Molecular
used. For relapse studies (n ⫽ 9) in 4 cases frozen-thawed material was
Probes, Eugene, OR) together with 7AAD in one tube, which enabled to
used. NOD/SCID mice repopulation experiments were performed using
gate out apoptotic/dead cells.18 The scatter properties of the viable cells
frozen-thawed material. Procedures used in this study have previously been
were then used in the tube containing CLL-1.
validated for the use of both fresh and frozen-thawed material.17 In fresh
PBS was used as a negative control, since for these specific antibodies
samples, red blood cells were lysed using a 10-minute red blood cell lysis
isotype controls offered the same results.8 Data acquisition was performed using
a FACScalibur (BD Biosciences) equipped with an argon and red diode laser, and
analysis was performed using Cellquest software (BD Biosciences).
Table 1. Patient characteristics
Blasts were identified by CD45dim/low side scatter characteristics according
Characteristic Quantity
to Lacombe, taking into account that the CD34⫹CD38⫺ population is a
Patients, no. 89 minor population.19
Male/female 45/44
Age at diagnosis, y, mean (range) 55 (16-79) CD34ⴙ cell selection
WBC count at diagnosis,109/L, mean (range) 38 (0.7-278)
FAB classification, no. (%) CD34⫹ cells were selected for the colony-forming unit (CFU) assays from NBM
M0 5 (6) and for the fluorescent in situ hybridization (FISH) analysis of a sample from an
M1 12 (13) AML patient in complete remission. After Ficoll separation and red blood cell
M2 14 (16) lysis, mononuclear cells were incubated in PBS containing 5 mM EDTA and
M3 3 (3) 0.1% HSA for 30 minutes at room temperature with CD34 Reagent (Miltenyi
M4 11 (12) Biotec, Bergisch Gladbach, Germany), according to the manufacturer’s proce-
M5 20 (23) dure. Cells were washed and allowed to flow through a positive-selection column
M6 3 (3) in a magnetic field (Automacs; Miltenyi Biotec). After 2 rounds of selection,
Refractory anemia with excess blasts 10 (11) CD34⫹ cells were collected. Purity was checked using flow cytometry and was
Refractory anemia with excess blasts in transformation 5 (6) more than 95% in all cases.
Not classified 6 (7)
Cytogenetic risk group, no. (%) FACS sorting of CD34ⴙ cells
Favorable 11 (12)
FACS-sorted cells were used for CFU assays, FISH analysis, and NOD/
Intermediate 49 (55)
SCID mice experiments. Cells were incubated for 15 minutes at room
Poor 9 (10)
temperature with Via-Probe (7AAD), anti-CD34 FITC, anti-CD38 APC,
No metaphases 20 (23)
and anti–CLL-1 PE and washed in PBS containing 0.1% HSA. Cells were
Flt3 ITD, no. (%)
subsequently sorted using a FACS Vantage cell sorter with Turbo Sort
Present 22 (25)
upgrade (BD Biosciences) equipped with an argon (377G) and a helium-
Absent 54 (61)
neon (127) laser, both from Spectra-Physics (Mountain View, CA) or a
Not analyzed 13 (14)
FACSAria equipped with solid-state lasers (red, blue, and violet; BD
WBC indicates white blood cell; FAB, French-American-British. Biosciences). Cells were sorted based on viability (7AAD negative),
From www.bloodjournal.org by on December 17, 2010. For personal use only.
BLOOD, 1 OCTOBER 2007 䡠 VOLUME 110, NUMBER 7 CLL-1 IS AN AML STEM CELL–ASSOCIATED ANTIGEN 2661

CD34⫹ expression, CD38⫹ expression for the CFU assays, absence of medium containing ␣-methylcellulose (Methocult GF H4434; StemCell
CD38 for the FISH analysis, and CLL-1 expression. Purity of the sorted Technologies, Vancouver, BC). The number of colonies was evaluated after
populations was more than 95%. 14 days culture in semisolid medium.20 CFU–megakaryocyte (CFU-MK)
assays were performed as described before.21
Transplantation of AML blasts in nonobese diabetic/severe
combined immunodeficiency mice Statistical analyses
Engraftment of CD34⫹CLL-1⫹ cells obtained at diagnosis was studied in Statistical analysis of the data was performed using the SPSS 9.0 software
nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice package (SPSS, Chicago, IL).
(Charles River Laboratories, Wilmington, MA). The mice were irradiated Mann-Whitney was used for CLL-1 expression in BM controls and
with 3.5 Gy using a linear accelerator, delivering a 15-MV photon beam; AML samples. Wilcoxon signed-rank and Spearman correlation analysis
tungsten jaws and multileafs collimated and shaped the irradiation field. were used for the analysis of the diagnosis-relapse samples. Spearman
The beam is calibrated, that is, the output for well-defined standard correlation analysis was also used for the correlation between the frequency
irradiation geometry is adjusted to a well-measured signal of the monitor of CD34⫹CD38⫺CLL-1⫹ cells and MRD frequency.
chamber. The monitor chamber is the system in the accelerator controlling P values less than .05 were considered to indicate a statistically
with high accuracy the dose delivery to the mice. significant difference.
Cells used were from 3 patients; samples were selected for CD34 expression,
CLL-1 expression on the CD34⫹CD38⫺ cells, and engraftment in NOD/SCID
mice. Patient 1 had AML with FAB M2 and Flt3 ITD; patient 2, with FAB M1 Results
and Flt3 ITD; and patient 3, with FAB M6, Flt3 ITD, and ⫹i(8)(q10)x2.
CD34⫹CLL-1⫹ cells (3.5-8 ⫻ 106) were injected in the lateral tail vein. The mice CLL-1 expression of CD34ⴙCD38ⴚ cells in AML at diagnosis
were killed after 6 weeks in accordance with the institutional animal research and in normal bone marrow
regulations or at onset of clinical symptoms. BM was isolated from 2 femurs per
mouse. Chimerism was determined using flow cytometric detection of human Previously we have shown that C-type lectin-like molecule-1
CD45 expression. Engrafted cells were analyzed for myeloid origin (CD33⫹ and (CLL-1) is present on leukemic blasts at diagnosis in the majority
CD19⫺), for malignant origin (presence of the leukemia-associated phenotype), of AML cases (68 of 74).12 Now CD34⫹ AML samples at diagnosis
and for CLL-1 expression. were analyzed for CLL-1 expression in the CD34⫹CD38⫺ stem
cell compartment. Representative examples of CLL-1 staining
FISH analysis
on CD34⫹CD38⫺ cells are depicted in Figure 1. In 77 of 89
FISH analysis for t(8;21) was performed using LSI ETO/AML1 from Vysis CD34⫹AML cases the CD34⫹CD38⫺ cells showed a positive shift
(Downers Grove, IL) on FACS-sorted populations of a patient sample after (compared with the isotype control) for CLL-1; because in part of
chemotherapeutic treatment. Procedures have been described before.15 the cases these shifts were small, overall (n ⫽ 89) a median CLL-1
expression of 33% was found for the CD34⫹CD38⫺ compartment
CFU assays (Figure 2). CLL-1 expression in the CD34⫹CD38⫺ compartment
For CFU assays, cells obtained after CD34 isolation and FACS sorting, was found throughout all FAB subtypes studied. CLL-1 expression
were cultured at 37°C in 5% CO2, in a humidified incubator, in semisolid on the CD34⫹CD38⫺ cells correlated neither with any AML

Figure 1. Examples of CLL-1 expression in AML samples at diagnosis and in normal bone marrow. After labeling of the cells with the appropriate antibody combinations,
the CD45dimCD34⫹CD38⫺ cells were identified by a precise gating strategy (as described in “Fluorescence-activated cell sorting analysis of CD34⫹CD38⫺ cells”; not shown)
with subsequent detection of isotype and CLL-1 expression. For the CD34⫺ AML (E-F) only blast gating (CD45dim) was performed. (A-F) Isotype and CLL-1 expression in AML
samples. The percentages in the lower right quadrant indicate CLL-1 expression as a percentage of the CD34⫹CD38⫺ compartment; in the lower left quadrant, the percentage
CLL-1⫺ cells within the CD34⫹CD38⫺ compartment is shown. (B) An example of CLL-1 expression on CD34⫹CD38⫺ cells close to the median. (D) An example of a high CLL-1
expression. (F) A representative example of CLL-1 expression in a CD34⫺ AML sample. (H) The absence of CLL-1 expression on CD34⫹CD38⫺ cells of normal bone marrow.
Note that CLL-1 is expressed on part of the progenitor population.
From www.bloodjournal.org by on December 17, 2010. For personal use only.
2662 VAN RHENEN et al BLOOD, 1 OCTOBER 2007 䡠 VOLUME 110, NUMBER 7

a CD34⫹CD38⫺ compartment that was homogeneously positive


for CLL-1) the CD34⫹CLL-1⫹ cell population was FACS sorted
and injected into sublethally irradiated NOD/SCID mice. After
6 weeks, the mice were killed. The engrafted CD45⫹ cells were of
myeloid origin (ie, CD33⫹ and CD19⫺), had a leukemia-associated
immunophenotype, and showed CLL-1 expression on all blasts
(Figure 3A-I). Engraftment of human cells was found for samples
of all 3 patients and in all mice (Figure 4). Therefore, we can
conclude that the CD34⫹CLL-1⫹ population of these 3 patients
does contain CLL-1–expressing leukemic stem cells.
Figure 2. CLL-1 expression on CD34ⴙCD38ⴚ stem cells in AML, normal bone Expression of CLL-1 in regenerating bone marrow
marrow (NBM), regenerating bone marrow (RBM), and G-CSF–mobilized periph-
eral blood (MPB). After labeling of the cells with the appropriate antibody combina-
and mobilized peripheral blood
tions, the CD34⫹CD38⫺ cells were identified by a precise gating strategy (as
described in “Fluorescence-activated cell sorting analysis of CD34⫹CD38⫺”) and Both for possible future therapeutic use and for specific detection
subsequently CLL-1 expression was determined. The percentage of CLL-1 expres- of malignant stem cells, it is of utmost importance that during and
sion on CD34⫹CD38⫺ cells for every individual patient and control is depicted with a after treatment, the nonmalignant CD34⫹CD38⫺ normal hematopoi-
symbol. In NBM, median CLL-1 expression was 0% (range, 0%-11%; n ⫽ 10); in
RBM, 0% (range, 0%-5%; n ⫽ 6); and in mobilized peripheral blood (MPB), 0.6% etic stem cells should remain CLL-1⫺. To establish that, remission
(range, 0%-3.7%; n ⫽ 6). The CD34⫹ FAB M3 samples of this study showed CLL-1 bone marrow of both non-AML patients and CD34⫺ or CLL-1⫺
expression on the CD34⫹CD38⫺ cells of 17%, 83%, and 89%. AML patients was investigated. No CLL-1 expression on the
CD34⫹CD38⫺ stem-cell compartment was observed (Figure 2).
Furthermore, since G-CSF mobilization is part of the several
subtype nor with prognosis in our patient cohort. Although in NBM current AML treatment protocols, CLL-1 expression was deter-
samples the CD34⫹CD38⫹ progenitor population was partly CLL- mined in G-CSF–mobilized blood. The CD34⫹CD38⫺ population
1⫹,12 the CD34⫹CD38⫺ cells were CLL-1⫺ (Figures 1G,H, 2). To was almost completely CLL-1⫺ in all samples (Figure 2).
conclude, CLL-1 is specifically expressed on AML CD34⫹CD38⫺ To conclude, normal CD34⫹CD38⫺ cells remain CLL-1⫺ in
cells and not on NBM CD34⫹CD38⫺ cells. BM recovering after chemotherapy and in PB after mobilization
with growth factors.
CLL-1–expressing CD34ⴙ cells contain leukemia-initiating
stem cells Identity of CLL-1ⴙ and CLL-1ⴚ normal bone marrow progenitors

NOD/SCID transplantation experiments were performed to show Our previous finding that CLL-1 is expressed on part of the
that the CD34⫹CLL-1⫹ compartment contains leukemic stem cells. CD34⫹CD38⫹ progenitor population in NBM12 was confirmed in
Of the 89 AML patients, we had cells for NOD/SCID experiments 11 additional samples (median expression, 40%; ranging from
from 10 patients. The total blast population from these patients was 26%-60%). Next, the nature of the CD34⫹CD38⫹CLL-1⫹ and
transplanted into NOD/SCID mice, which resulted in engraftment CD34⫹CD38⫹CLL-1⫺ subpopulations in normal BM was deter-
of human cells in 3 of 10 patients. From these 3 AML patients (with mined by FACS sorting and subsequent application of different

Figure 3. Flow cytometry of engrafted cells in NOD/SCID mice. FACS-sorted CD34⫹CLL-1⫹ cells from 3 different AML patients were transplanted into NOD/SCID mice. Six
weeks after transplantation, the mice were killed and engraftment of human cells in these mice was studied using human anti-CD45 labeling. The engrafted cells were analyzed
for myeloid origin (CD19⫺ and CD33⫹, data not shown), leukemia-associated phenotype, and CLL-1 expression. Patient numbers are the same as in Figure 4. (A-E) The
results of engrafted cells in 1 mouse of 3 that received a transplant of cells from patient 2 are shown. (A) CLL-1 expression on CD34⫹ cells at diagnosis (the CD34⫺ cells in this
sample were also CLL-1⫹, not shown). (B) Aberrant expression of CD7 on part of the leukemic CD34⫹ cells at diagnosis. (C) The CD45⫹ human population within the white
blood cell compartment of the mouse. (D) The human cells that grew out of the transplanted CD34⫹CLL-1⫹ cells are all CLL-1⫹, similar to the whole blast compartment at
diagnosis. (E) Similar to diagnosis, part of the cells again show aberrant CD7 expression. (F,H) The human CD45⫹ population within the white blood cell compartment of mice
that received a transplant of cells from patients 3 and 1. CLL-1 expression on engrafted cells is shown in panels G and I.
From www.bloodjournal.org by on December 17, 2010. For personal use only.
BLOOD, 1 OCTOBER 2007 䡠 VOLUME 110, NUMBER 7 CLL-1 IS AN AML STEM CELL–ASSOCIATED ANTIGEN 2663

Figure 4. Engraftment of CD34ⴙCLL-1ⴙ AML blasts in NOD/SCID mice. FACS- Figure 5. Colony-forming capacity of CLL-1–defined CD34 subpopulations in
sorted CD34⫹CLL-1⫹ cells from 3 different AML patients were transplanted into normal bone marrow. CD34⫹CD38⫹CLL-1⫺ and CD34⫹CD38⫹CLL-1⫹ subpopula-
NOD/SCID mice. Six weeks after transplantation, the mice were killed and engraft- tions were purified from NBM cells. Cell input was 2500 cells/mL for BFU-Es/CFU-
ment of human cells in these mice was studied using human anti-CD45. In this figure, GMs. Cell input for CFU-MKs was 5000 cells/mL. The number of colonies was
the overall results are depicted on a logarithmic scale; every symbol represents the determined after 14 days of culture. f shows the output of the CD34⫹CD38⫹CLL-1⫹
percentage engraftment of human CD45⫹ cells in one mouse. In a mouse that population; , the CD34⫹CD38⫹CLL-1⫺ population. The error bars show the
received a transplant of the whole CD34⫹ population from patient 2, which included standard deviation. Two (CFU-MK) and 3 (burst-forming unit–erythroid [BFU-E]/CFU-
the CD34⫹CD38⫺CLL⫹, but also a clear CD34⫹CD38⫺CLL-1⫺ population (15% of GM) independent experiments were performed, each in duplicate.
the CD34⫹CD38⫺ population), outgrowth of CD45dimCD33⫹CD19⫺ AML cells was
likewise accompanied by outgrowth of CD45dimCD33⫺CD19⫹ cells (18% of human
cells), which are presumably normal (data not shown).22 Horizontal lines represent
the mean percentage engraftment of leukemic cells.
CLL-1⫹ (Figure 6D), showing that the majority of BM stem cells
were of malignant origin. Patient 3 (Figure 6E,F) reached CR after
colony assays. Results are depicted in Figure 5. Most CFU– the first cycle of chemotherapy. The MRD frequency at that time
granulocyte macrophages (CFU-GMs) grew out of the point was 0.82%, which decreased to a prognostically favorable
CD34⫹CD38⫹CLL-1⫹ population (Figure 5 second pair of rows). 0.02% after the second cycle of chemotherapy. Despite this, the
All monocytic colonies originated from the CLL-1⫹ fraction, patient relapsed within 6 months. In contrast to MRD, the fraction
whereas the granulocytic colonies originated only in part from the of CLL-1⫹ cells remained high; Figure 6F shows the first cycle
CLL-1⫹ fraction (data not shown). These observations are in with 72% CLL-1 expression on the CD34⫹CD38⫺ cells. Since this
accordance with granulocytes and monocytes being CLL-1⫹.12 In patient was diagnosed with t(8;21), we were able to FACS sort the
contrast, erythroid colonies (BFU-Es) and megakaryocytic colo- CD34⫹CD38⫺ cells after the first cycle and to show by FISH
nies (CFU-MKs) originated from the CD34⫹CD38⫹CLL-1⫺ com- analysis that, in very good agreement with Figure 6F, these were
partment. Therefore, targeting CLL-1⫹ cells using anti–CLL-1– 80% positive for t(8;21). Quantitative polymerase chain reaction
based therapy would most likely affect only granulocyte recovery. (PCR) for t(8;21) confirmed these results (data not shown). The
malignant nature of the CD34⫹CD38⫺CLL-1⫹ population was
CLL-1 expression of AML CD34ⴙCD38ⴚ cells at diagnosis further confirmed by aberrant expression of the natural killer (NK)
versus relapse cell marker CD56 (72%) on CD34⫹CD38⫺ cells. The CD34⫹CD56⫹
immunophenotype has been shown to be a leukemia-associated
For reliable assessment during treatment/disease of the AML
phenotype and is used for MRD detection8,23-25 and malignant
stem-cell compartment using CLL-1, its expression should be
stem-cell detection.26
stable in the course of the disease. Paired diagnosis/relapse samples
Figure 7 shows representative examples of follow-up analysis for 2
of 9 AML patients covering a large range of CLL-1 expression
patients with a different course of the disease. Patient 1 reached CR,
showed no significant differences (P ⫽ .9) and were significantly
which has lasted now for 3 years. Figure 7A shows that this patient had
correlated (R ⫽ 0.7, P ⫽ .04); median expression at diagnosis was
high CLL-1 expression on the CD34⫹CD38⫺ cells at diagnosis, but this
34% (range, 8%-91%) versus 42% (range, 5%-92%) at relapse.
expression decreased after chemo-therapy and remained absent; also the
CLL-1 expression of CD34ⴙCD38ⴚ cells in remission frequency of the CD34⫹CD38⫺CLL-1⫹ cells within the white blood
bone marrow cell (WBC) compartment (Figure 7B), which is a measure for the
amount of CLL-1⫹CD34⫹CD38⫺ cells, diminished and remained
Subsequently, the possibility to discriminate CLL-1⫹ malignant undetectable in time. Patient 2 reached CR, but relapsed after the second
CD34⫹CD38⫺ cells from CLL-1⫺ normal CD34⫹CD38⫺ cells in course of chemotherapy. CLL-1 was continuously expressed on the
remission bone marrow at different stages of treatment was CD34⫹CD38⫺ compartment, indicating that this compartment re-
investigated. Figure 6A,C,E shows representative flow cytometry mained predominantly malignant at all time points analyzed (Figure
pictures of 3 patients with high expression of CLL-1 on 7C). Also the frequency of the CD34⫹CD38⫺CLL-1⫹ cells within the
CD34⫹CD38⫺ cells at diagnosis. WBC compartment remained high and even increased toward relapse
Patient one (Figure 6A,B) reached complete remission (CR) (Figure 7D). These figures are representations of similar patients who
with resulting low frequency (0.01%) of minimal residual disease had a good remission (total n ⫽ 4) or a fast relapse (total n ⫽ 4).
(MRD) as measured using leukemia-associated phenotype (LAP) These results indicate that, using CLL-1, malignant
expression, which is prognostically highly favorable.8 In line with CD34⫹CD38⫺ cells can indeed be detected in remission bone
this, CLL-1 expression was not present on the CD34⫹CD38⫺ marrow and can be discriminated from the normal CD34⫹CD38⫺
compartment (Figure 6B). Patient 2 (Figure 6C,D) reached CR, but compartment. In a series of 13 patients, the putative possible role in
with relatively high MRD frequency (0.89%). This patient quickly prognosis was studied; in 8 of 9 cases with a quick relapse (ie,
relapsed (within one month). CD34⫹CD38⫺ cells remained largely median 6 months after diagnosis; range, 3-11 months), CLL-1
From www.bloodjournal.org by on December 17, 2010. For personal use only.
2664 VAN RHENEN et al BLOOD, 1 OCTOBER 2007 䡠 VOLUME 110, NUMBER 7

Figure 6. Examples of CLL-1 expression on


CD34ⴙCD38ⴚ cells in diagnosis and follow-up
samples. Gating was performed based on CD45dim/SSC
characteristics and CD34⫹ expression. Afterwards, CD38
and CLL-1 expression were determined. Similar to Figure
1, percentages shown concern the CLL-1⫹ and CLL-1⫺
populations within the CD34⫹CD38⫺ compartment. (A,B)
An example of a patient who remained in continuous
remission. This patient showed high CLL-1 expression on
CD34⫹CD38⫺ cells at diagnosis (A) but not after chemo-
therapy (B). Note the similarity with CLL-1 expression on
normal bone marrow CD34⫹CD38⫺ cells in Figure 1H.
(C,D) A diagnosis and MRD picture, as in panels A and B,
but now for a patient with a large proportion of
CD34⫹CD38⫺ cells being CLL-1⫹ in the MRD sample
(D). CLL-1 expression on CD34⫹CD38⫺ cells of an AML
patient diagnosed with t(8,21), at diagnosis (E) and after
first course of chemotherapy (F). In the remission sample,
the malignant character of the CD34⫹CD38⫺CLL-1⫹
population could be confirmed using FISH technique.

expression on the whole stem-cell compartment was high (median,


62%; range, 27%-91%). This is in contrast to the patients still in Discussion
remission (longest, 33 months), for which CLL-1⫹ stem cells were
In the present study, we show that CLL-1 is a marker of the
very low or absent in 4 of 5 cases (median, 0%; range, 0%-2%).
malignant CD34⫹CD38⫺ stem-cell compartment in the majority of
Moreover we were able to show that, irrespective of the time point
CD34⫹ AML patients. Transplantation of the CD34⫹CLL-1⫹ cells,
in disease/treatment, the frequency of CD34⫹CD38⫺ cells within
the WBC compartment significantly correlated with the frequency which putatively contain the CD34⫹CD38⫺ stem cells capable of
of MRD cells in these AML patients (n ⫽ 44 time points from initiating leukemia, resulted in the development of leukemia in
15 patients, R ⫽ 0.7, P ⬍ .001). MRD detection serves as the gold NOD/SCID mice in the 3 patients analyzed, with recovery of the
standard for risk assessment during treatment/disease, since it is a diagnosis CLL-1 expression. CLL-1 expression turned out to be
strong independent predictor for survival in AML patients as we specific for leukemic CD34⫹CD38⫺ cells, since it is absent both on
and others have shown before.8,23-25 The prognostic impact of MRD normal CD34⫹CD38⫺ resting bone marrow cells, on CD34⫹CD38⫺
frequency in the present patient cohort has been published by us cells in treated patients with other diseases, and in CD34⫹CD38⫺
before.17 cells obtained after G-CSF stimulation from non-AML patients.

Figure 7. Stem-cell parameters in AML patients with a


different course of the disease. Cells were analyzed at
diagnosis and at different time points during follow-up of
2 AML patients. (A,B) Patient 1. (C,D) Patient 2. (A,C)
Malignant CD34⫹CD38⫺ cells (defined by CLL-1) as a
fraction of the total CD34⫹CD38⫺ population. The solid
line represents the background expression in RBM for
CLL-1. (B,D) The frequency of CD34⫹CD38⫺CLL-1⫹
cells as percentage of the total WBC count. D indicates
diagnosis; R, relapse; 1st, after first course of chemo-
therapy; and 2nd and 3rd, after the second and third
course of chemotherapy, respectively. Patient 1 reached
complete remission after the first course of chemo-
therapy and has been in continuous complete remission
now for 3 years. Percentage of CLL-1 expression on the
total CD34⫹CD38⫺ compartment declined rapidly (A) as
did the frequency of CD34⫹CD38⫺CLL-1⫹cells (B). Pa-
tient 2 reached complete remission after the first course
of chemotherapy; however, a relapse occurred after the
second course of chemotherapy. Although CR was
reached, there was a continuous expression of CLL-1 on
the CD34⫹CD38⫺ cells (C). Also, the frequency of
CD34⫹CD38⫺CLL-1⫹ cells did not decrease after chemo-
therapy, but even increased (D).
From www.bloodjournal.org by on December 17, 2010. For personal use only.
BLOOD, 1 OCTOBER 2007 䡠 VOLUME 110, NUMBER 7 CLL-1 IS AN AML STEM CELL–ASSOCIATED ANTIGEN 2665

CLL-1 expression on the AML cells is likely stable, with no well-known inhibitor of NF-␬B, has been proposed for stem
difference found between samples at diagnosis and relapse. cell–targeted therapy.33 In vitro there is synergism between this
CLL-1 expression thus can be detected specifically on AML NF-␬B inhibitor and a conventional chemotherapeutic agent,
CD34⫹CD38⫺ cells present after chemotherapy in AML patients in idarubicin.34 Also, parthenolide, has been proposed to selec-
complete remission. In this way, CLL-1 expression may serve as tively target leukemic stem cells while sparing normal hemato-
marker for quantification of minimal residual stem-cell disease. poietic cells.35
The observation of quick relapses preceded by the presence of Internalization has been described for CLL-1 after incubation
AML stem cells might offer clinically highly relevant informa- with the antibody.12 However it is unlikely that the anti–CLL-1
tion—in addition to the classic immunophenotypical MRD detec- antibody as such will have antileukemic effect, since Moab binding
tion.8,23-25 Moreover, these results show that CLL-1 is a potential to the CLL-1⫹ HL60 cells had no effect on cell proliferation using a
target for antileukemia stem-cell therapy in remission bone marrow tritium-thymidine assay, while culturing normal CD34⫹ cells in a
with AML stem cells still present. Since thrombocytopenia is a long-term culture system in the presence of anti–CLL-1 antibody
major side effect of most therapies in the majority of AML patients, has no effect on CFU-E and CFU-GM colony formation (data not
and thus often a dose-limiting factor, it is of importance that CLL-1 shown). Also, CLL-1 labeling of cells did not influence engraft-
is absent on megakaryocytic progenitors. ment of AML CD34⫹ cells in NOD/SCID mice (not shown). It is
How would CLL-1 perform compared with CD123, CD33, and thus unlikely, although still unknown, that induction of antibody-
CD44? CD123 has been reported to be a stem cell–specific marker dependent cellular cytotoxicity and complement-dependent cytotox-
in AML.27 An advantage is the high staining intensity of icity via CLL-1 will be an effective targeting mechanism. However,
the putative function of CLL-1 in signal transduction13,14 might
CD34⫹CD38⫺ cells observed in the majority of AML,27 which we
offer possibilities that can be translated in therapeutic application-
were able to confirm (median expression, 98%; ranging from
s.Another promising therapeutic strategy would be to couple a
5%-100% in 36 cases). The median expression on NBM
toxic moiety to the anti–CLL-1 antibody; if such a complex is
CD34⫹CD38⫺ cells (n ⫽ 5) was higher compared with CLL-1
internalized after antigen binding this would result in cell death
(14.9%; ranging from 0%-18.8%), which was also found by
after antigen-mediated uptake.
others.11 To explore potential therapeutic use, a toxin-labeled IL-3
Anti–CLL-1 antibody–mediated therapy might be effective in CD34⫺
has been developed to target a functional IL3-receptor (consisting
AML as well, because CLL-1 expression is high in blast cells of CD34⫺
of both CD123 and CD131) and is currently being examined in AML (median, 96%; range, 15%-100%; n ⫽ 11; including 3 AML FAB
(pre-) clinical studies.28 However, important control experiments, M3). In the absence of a CD34⫹CD38⫺ compartment, the side
including possible staining of normal CD34⫹CD38⫺ cells in population (SP) defined by Hoechst staining is the most likely candi-
regenerating bone marrow (RBM) of patients treated with chemo- date36 to contain leukemic stem cells, which we indeed found in 6 of 9
therapy, have not yet been reported. Since we have found high CD34⫺ AML patients. Preliminary results indicate that CLL-1 is indeed
(median, 60%) CD123 expression on non–AML-regenerating bone expressed on SP cells of 6 of 6 patients (median, 64%; range,
marrow CD34⫹CD38⫺ cells in 5 such cases (not shown), this issue 46%-100%). Also, other myeloid malignancies, such as myelodysplas-
needs serious attention. In sharp contrast, we did not detect CLL-1 tic syndrome (MDS) and chronic myeloid leukemia (CML), show
on RBM CD34⫹CD38⫺ cells. CLL-1 expression on the CD34⫹ cells.12
CD33 is also expressed on CD34⫹CD38⫺ stem cells of AML In conclusion, CLL-1 is a marker that is expressed on AML
samples at diagnosis (median, 80%; range, 26%-100%; n ⫽ 13; CD34⫹CD38⫺ stem cells and not on normal CD34⫹CD38⫺ cells under
A.v.R., unpublished data, October 2005). In contrast to anti-CD123 all conditions of treatment and disease. In 3 patients tested, we found
antibody and similar to anti–CLL-1 antibody, internalization of that CD34⫹CLL-1⫹ cells repopulate in sublethally irradiated NOD/
anti-CD33 occurs upon binding to the receptor.29 However, CD33 SCID mice, indicating that they contain leukemia-initiating cells. The
is not specific for AML stem cells; expression has been shown on use of anti–CLL-1 for the detection of minimal residual disease would
normal stem cells.11 In our own experience too, CD33 is highly offer an attractive approach, in addition to the established MRD
expressed on normal CD34⫹CD38⫺ stem cells, both in resting frequency assessments, both as a prognostic marker and to guide time
NBM (median, 84%; range, 16%-100%; n ⫽ 9) and in RBM (61%, points for therapeutic intervention. Moreover, these unique properties
79%, and 95% in 3 different BM samples). These observations might be exploited for the development of effective antibody conjugates
might offer part of the explanation for the considerable hemato- for stem cell–directed therapy.
logic toxicity in patients undergoing anti-CD33 therapy,30 part of
which is certainly due to its effect on normal CD33⫹ progeni-
tors,31,32 and, maybe even more importantly, on CD34⫹CD38⫺ Acknowledgments
stem cells in normal bone marrow.11
We thank Floortje Kessler for assistance with the CFU-MK assay
CD44 has recently been described as a target on leukemic
and Rob Dee (Sanquin Research, Amsterdam, the Netherlands) for
CD34⫹CD38⫺ stem cells. It was shown that the activating antibody H90
performing qPCR of t(8;21). The Department of Cardiac Surgery
results in differentiation of cells and in a major reduction of engraftment
and the patients are acknowledged for providing healthy bone
in NOD/SCID mice.9 However, CD44 is also weakly expressed on
marrow samples.
normal CD34⫹CD38⫺ cells9 and on more differentiated hematopoietic
cells. Regenerating bone marrow was not evaluated. Moreover, the
different CD44 isoforms are expressed on many different tissues.10
An alternative approach to specifically target leukemic stem Authorship
cells is inhibition of the proteasome. Normal CD34⫹ progenitor/
stem cells do not express NF-␬B, whereas CD34⫹CD38⫺CD123⫹ Contribution: A.v.R. designed part of the research, performed the
cells in AML do.33 The proteasome inhibitor MG-132, a main part of the experiments and data analysis, and wrote the
From www.bloodjournal.org by on December 17, 2010. For personal use only.
2666 VAN RHENEN et al BLOOD, 1 OCTOBER 2007 䡠 VOLUME 110, NUMBER 7

paper; G.A.M.S.D. designed part of the research; A.K. performed samples and reviewed the paper; and G.J.S. designed and guided
part of the experiments; E.J.R. designed part of the research and the main part of the study and was the main reviewer of the paper.
performed part of the experiments; N.F. guided and carried out part Conflict-of-interest disclosure: The authors declare no compet-
of the research and data analysis and reviewed the paper; B.M. ing financial interests.
performed part of the experiments; M.S.W. performed part of the Correspondence: Gerrit Jan Schuurhuis, VU University Medi-
experiments; S.Z. guided part of the research and reviewed the cal Center, Department of Hematology, CCA 4.28, De Boelelaan
paper; G.J.O. was responsible for the availability of clinical 1117, 1081 HV Amsterdam; e-mail: gj.schuurhuis@vumc.nl.

References
1. Lowenberg B, Griffin JD, Tallman MS. Acute my- 14. Songyang Z, Shoelson SE, Chaudhuri M, et al. therapy predicts outcome in acute myeloid leuke-
eloid leukemia and acute promyelocytic leuke- SH2 domains recognize specific phosphopeptide mia. Blood. 2000;96:3948-3952.
mia. Hematology (Am Soc Hematol Educ Pro- sequences. Cell. 1993;72:767-778. 26. van Rhenen A, Moshaver B, Kelder A, et al. Aber-
gram). 2003:82-101. 15. van der Pol MA, Feller N, Roseboom M, et al. As- rant marker expression patterns on the
2. Cozzio A, Passegue E, Ayton PM, et al. Similar sessment of the normal or leukemic nature of CD34⫹CD38⫺ stem cell compartment in acute
MLL-associated leukemias arising from self-re- CD34⫹ cells in acute myeloid leukemia with low myeloid leukemia allows to distinguish the malig-
newing stem cells and short-lived myeloid pro- percentages of CD34 cells. Haematologica. nant from the normal stem cell compartment both
genitors. Genes Dev. 2003;17:3029-3035. 2003;88:983-993. at diagnosis and in remission. Leukemia. 2007;
3. Krivtsov AV, Twomey D, Feng Z, et al. Transfor- 16. Lowenberg B, Downing JR, Burnett A. Acute my- 21:1700-1707.
mation from committed progenitor to leukaemia eloid leukemia. N Engl J Med. 1999;341:1051- 27. Jordan CT, Upchurch D, Szilvassy SJ, et al. The
stem cell initiated by MLL-AF9. Nature. 2006;442: 1062. interleukin-3 receptor alpha chain is a unique
818-822.
17. van Rhenen A, Feller N, Kelder A, et al. High marker for human acute myelogenous leukemia
4. Huntly BJ, Shigematsu H, Deguchi K, et al. MOZ- stem cell frequency in acute myeloid leukemia at stem cells. Leukemia. 2000;14:1777-1784.
TIF2, but not BCR-ABL, confers properties of leu- diagnosis predicts high minimal residual disease
kemic stem cells to committed murine hematopoi- 28. Cohen KA, Liu TF, Cline JM, et al. Safety evalua-
and poor survival. Clin Cancer Res. 2005;11: tion of DT388IL3, a diphtheria toxin/interleukin 3
etic progenitors. Cancer Cell. 2004;6:587-596. 6520-6527. fusion protein, in the cynomolgus monkey. Can-
5. Bonnet D, Dick JE. Human acute myeloid leuke-
18. van der Pol MA, Broxterman HJ, Westra G, Os- cer Immunol Immunother. 2005;54:799-806.
mia is organized as a hierarchy that originates
senkoppele GJ, Schuurhuis GJ. Novel multipa- 29. van der Velden V, te Marvelde JG, Hoogeveen
from a primitive hematopoietic cell. Nat Med.
rameter flow cytometry assay using Syto16 for PG, et al. Targeting of the CD33-calicheamicin
1997;3:730-737.
the simultaneous detection of early apoptosis and immunoconjugate Mylotarg (CMA-676) in acute
6. Lapidot T, Sirard C, Vormoor J, et al. A cell initiat- apoptosis-corrected P-glycoprotein function in
ing human acute myeloid leukaemia after trans- myeloid leukemia: in vivo and in vitro saturation
clinical samples. Cytometry. 2003;55B:14-21. and internalization by leukemic and normal my-
plantation into SCID mice. Nature. 1994;367:645-
19. Lacombe F, Durrieu F, Briais A, et al. Flow cytom- eloid cells. Blood. 2001;97:3197-3204.
648.
etry CD45 gating for immunophenotyping of
7. Costello R, Mallet F, Chambost H, et al. The im- 30. Giles F, Estey E, O’Brien S. Gemtuzumab ozo-
acute myeloid leukemia. Leukemia. 1997;11:
munophenotype of minimally differentiated acute gamicin in the treatment of acute myeloid leuke-
1878-1886.
myeloid leukemia (AML-M0): reduced immunoge- mia. Cancer. 2003;98:2095-2104.
nicity and high frequency of CD34⫹/CD38⫺ leuke- 20. Sutherland H, Blair A, Vercauteren S, Zapf R. De-
31. Andrews RG, Torok-Storb B, Bernstein ID. My-
mic progenitors. Leukemia. 1999;13:1513-1518. tection and clinical significance of human acute
eloid-associated differentiation antigens on stem
myeloid leukaemia progenitors capable of long-
8. Feller N, van der Pol MA, van Stijn A, et al. MRD cells and their progeny identified by monoclonal
term proliferation in vitro. Br J Haematol. 2001;
parameters using immunophenotypic detection antibodies. Blood. 1983;62:124-132.
114:296-306.
methods are highly reliable in predicting survival 32. Griffin JD, Linch D, Sabbath K, Larcom P,
in acute myeloid leukaemia. Leukemia. 2004;18: 21. Zweegman S, Veenhof MA, Debili N, et al.
Megakaryocytic differentiation of human progeni- Schlossman SF. A monoclonal antibody reactive
1380-1390.
tor cells is negatively influenced by direct contact with normal and leukemic human myeloid pro-
9. Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. genitor cells. Leuk Res. 1984;8:521-534.
with stroma. Leukemia. 1999;13:935-943.
Targeting of CD44 eradicates human acute my-
eloid leukemic stem cells. Nat Med. 2006;12: 22. Pearce DJ, Taussig D, Zibara K, et al. AML en- 33. Guzman ML, Neering SJ, Upchurch D, et al.
1167-1174. graftment in the NOD/SCID assay reflects the Nuclear factor-kappaB is constitutively activated
outcome of AML: implications for our understand- in primitive human acute myelogenous leukemia
10. Ponta H, Sherman L, Herrlich PA. CD44: from
ing of the heterogeneity of AML. Blood. 2006;107: cells. Blood. 2001;98:2301-2307.
adhesion molecules to signalling regulators. Nat
Rev Mol Cell Biol. 2003;4:33-45. 1166-1173. 34. Guzman ML, Swiderski CF, Howard DS, et al.
23. San Miguel JF, Vidriales MB, Orfao A. Immuno- Preferential induction of apoptosis for primary
11. Taussig DC, Pearce DJ, Simpson C, et al. Hema-
logical evaluation of minimal residual disease human leukemic stem cells. Proc Natl Acad Sci
topoietic stem cells express multiple myeloid
markers: implications for the origin and targeted (MRD) in acute myeloid leukaemia (AML). Best U S A. 2002;99:16220-16225.
therapy of acute myeloid leukemia. Blood. 2005; Pract Res Clin Haematol. 2002;15:105-118. 35. Guzman ML, Rossi RM, Karnischky L, et al. The
106:4086-4092. 24. Kern W, Voskova D, Schoch C, et al. Determina- sesquiterpene lactone parthenolide induces apo-
12. Bakker AB, Van den Oudenrijn S, Bakker AQ, et tion of relapse risk based on assessment of mini- ptosis of human acute myelogenous leukemia
al. C-type lectin-like molecule-1: a novel myeloid mal residual disease during complete remission stem and progenitor cells. Blood. 2005;105:4163-
cell surface marker associated with acute myeloid by multiparameter flow cytometry in unselected 4169.
leukemia. Cancer Res. 2004;64:8443-8450. patients with acute myeloid leukemia. Blood. 36. Wulf GG, Wang RY, Kuehnle I, et al. A leukemic
13. Unkeless JC, Jin J. Inhibitory receptors, ITIM se- 2004;104:3078-3085. stem cell with intrinsic drug efflux capacity in
quences and phosphatases. Curr Opin Immunol. 25. Venditti A, Buccisano F, Del Poeta G, et al. Level acute myeloid leukemia. Blood. 2001;98:1166-
1997;9:338-343. of minimal residual disease after consolidation 1173.

Potrebbero piacerti anche