ArticlePDF Available

Abstract and Figures

Purpose: We studied the expression of the chemokine receptor CXCR4 on circulating CD34+ cells of patients with myelofibrosis with myeloid metaplasia (MMM), and examined its relationship to the severity of disease. Patients and methods: Surface and intracellular CXCR4 expression were measured flow cytometrically in 84 consecutive MMM patients, 16 patients with polycythemia vera (PV), and 20 healthy subjects. In 23 MMM patients, CXCR4 gene expression level was also quantitated by real time-RT-PCR in CD34+ cells. Results: The expression of CXCR4 on circulating CD34+ cells was significantly reduced in patients with MMM (P<0.001) as compared to normal controls and patients with PV (P=0.01). The levels of CXCR4 mRNA in CD34+ cells were lower in patients with MMM as compared with normal subjects, and were directly correlated with the degree of CXCR4 surface expression, demonstrating that transcriptional defects were the major cause for receptor down-regulation. No statistical association was found between JAK2V617F mutational status and the extent of CXCR4 down-regulation. CXCR4 expression on CD34+ cells inversely correlated with the number of circulating CD34+ cells (R=-0.55; P<0.001), and was severely down-regulated in high risk patients and patients with a high "myelodepletion severity index". CXCR4 down-regulation was associated with advanced patient age, the presence of severe anemia, thrombocytopenia, and degree of bone marrow fibrosis. Conclusions: Reduced expression of CXCR4 by CD34+ cells is a characteristic of MMM which is associated with the constitutive mobilization of CD34+ cells and occurs in patients with advanced forms of the disease.
Content may be subject to copyright.
The expression of CXCR4 is down-regulated on the CD34+ cells of patients
with myelofibrosis with myeloid metaplasia
Vittorio Rosti
a
, Margherita Massa
b
, Alessandro M. Vannucchi
c
, Gaetano Bergamaschi
d
,
Rita Campanelli
a
, Alessandro Pecci
e
, Gianluca Viarengo
f
, Valentina Meli
g
, Monia Marchetti
h
,
Paola Guglielmelli
c
, Edward Bruno
i,j
, Mingjiang Xu
i,j
, Ronald Hoffman
i,j
, Giovanni Barosi
h,j,
on behalf of the Investigators of the Italian Registry of Myelofibrosis with Myeloid Metaplasia and
the Myeloproliferative Disorders Research Consortium
a
Transplant Research Area, IRCCS Policlinico S. Matteo, Pavia, Italy
b
Laboratory of Biotechnology, IRCCS Policlinico S. Matteo, Pavia, Italy
c
Department of Hematology, Azienda Ospedaliera Universitaria Careggi, Florence, Italy
d
Unit of Internal Medicine I, IRCCS Policlinico S. Matteo, Pavia, Italy
e
Unit of Internal Medicine III, IRCCS Policlinico S. Matteo, Pavia, Italy
f
Unit of Clinical Immunology, Immunohematology, and Transfusion Service, IRCCS Policlinico S. Matteo, Pavia, Italy
g
Department of Pediatrics, IRCCS Policlinico S. Matteo, Pavia, Italy
h
Unit of Clinical Epidemiology, IRCCS Policlinico S. Matteo, Pavia, Italy
i
Section of Hematology/Oncology, Department of Medicine, University of Illinois College of Medicine, Chicago, IL, USA
j
MPD Research Consortium, Chicago, IL, USA
Submitted 8 January 2007
Available online 9 March 2007
(Communicated by R. Hoffman, M.D., 8 January 2007)
Abstract
Purpose: We studied the expression of the chemokine receptor CXCR4 on circulating CD34+ cells of patients with myelofibrosis with myeloid
metaplasia (MMM), and examined its relationship to the severity of disease.
Patients and methods: Surface and intracellular CXCR4 expression were measured flow cytometrically in 84 consecutive MMM patients, 16
patients with polycythemia vera (PV), and 20 healthy subjects. In 23 MMM patients, CXCR4 gene expression level was also quantitated by real
time-RT-PCR in CD34+ cells.
Results: The expression of CXCR4 on circulating CD34+ cells was significantly reduced in patients with MMM (P< 0.001) as compared to
normal controls and patients with PV (P= 0.01). The levels of CXCR4 mRNA in CD34+ cells were lower in patients with MMM as compared
with normal subjects, and were directly correlated with the degree of CXCR4 surface expression, demonstrating that transcriptional defects were
the major cause for receptor down-regulation. No statistical association was found between JAK2V617F mutational status and the extent of
CXCR4 down-regulation. CXCR4 expression on CD34+ cells inversely correlated with the number of circulating CD34+ cells (R=0.55;
P< 0.001), and was severely down-regulated in high risk patients and patients with a high myelodepletion severity index. CXCR4 down-
regulation was associated with advanced patient age, the presence of severe anemia, thrombocytopenia, and degree of bone marrow fibrosis.
Conclusions: Reduced expression of CXCR4 by CD34+ cells is a characteristic of MMM which is associated with the constitutive mobilization of
CD34+ cells and occurs in patients with advanced forms of the disease.
© 2007 Elsevier Inc. All rights reserved.
Keywords: Myelofibrosis; CXCR4; SDF-1; Mobilization
Blood Cells, Molecules, and Diseases 38 (2007) 280 286
www.elsevier.com/locate/ybcmd
Corresponding author. Laboratorio di Epidemiologia Clinica, IRCCS Policlinico S. Matteo, Viale Golgi 19, 27100 Pavia, Italy. Fax: +39 0382 503917.
E-mail address: barosig@smatteo.pv.it (G. Barosi).
1079-9796/$ - see front matter © 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.bcmd.2007.01.003
Introduction
Myelofibrosis with myeloid metaplasia (MMM) is a
Philadelphia chromosome-negative chronic myeloproliferative
disorder (Ph-CMPD). MMM is characterized by the con-
stitutive release of hematopoietic progenitor cells (HPCs) and
their accumulation in the blood and extramedullary sites
[1,2]. This biological feature distinguishes MMM from other
Ph-CMPD.
Mobilization of HPCs in humans is regulated by a network
of hematopoietic cytokines, chemokines and adhesion mole-
cules, among which stromal derived factor-1 (SDF-1,
CXCL12) and its receptor CXCR4 play a major role [35].
Increased protease activity in the bone marrow microenviron-
ment with SDF-1 degradation and CXCR4 inactivation has
been reported to be an indirect mechanism by which
chemotherapy or G-CSF mobilizes HPCs [6,7]. Recently, Xu
and coworkers provided evidence that plasma levels of
neutrophil elastase (NE) and matrix metalloproteinases were
increased in MMM patients, and that the absolute number of
CD34+ cells in the peripheral blood was correlated with the
levels of soluble VCAM-1 [8]. In addition, Passamonti et al.
documented that granulocytes were activated in patients with
MMM in a manner similar to that induced by G-CSF
administration [9]. These results support the hypothesis that a
G-CSF-like, protease-driven mechanism underlies HPCs
mobilization in MMM.
Down-regulation of CXCR4 and disruption of the CXCR4/
SDF-1 axis might also affect cell mobilization in MMM.
Blocking of CXCR4 by specific antibodies or peptides has
been reported to result in mobilization of tumor cells in
xenograft mouse models [10,11]. Furthermore down-regula-
tion of CXCR4 expression with a CXCR4 antagonist results
in mobilization of polymorphonuclear neutrophils and HPCs
into the peripheral blood of mice [12]. Moreover, transplanta-
tion of CXCR4-deficient HPCs into lethally irradiated
recipient mice, leads to a reconstituted marrow in which the
more mature myeloid elements are noticeably reduced due to
premature release of CXCR4-negative myeloid cells into the
blood stream [13]. The down regulation of CXCR4 expression
by CD34+ cells from patients in blast crisis of chronic
myeloid leukemia (CML), as compared with patients in the
chronic phase or control individuals has also been observed
[14]. More recently, G-CSF mobilization of HPCs has been
shown to be associated with the down-regulation of CXCR4
expression by bone marrow myeloid cells, probably in
association with the accumulation of neutrophil-derived
proteolytic enzymes [15].
The above mentioned results provide a rationale for studying
CXCR4 expression in MMM. These investigations are even
more compelling due to the recent reports that a Val617Phe
mutation in the exon 12 of the JAK2 gene (JAK2V617F) affects
approximately 50% of patients with MMM [1618]. The
possibility that the JAK2V617F mutation might lead to down
regulation of CXCR4 is not unprecedented. Down regulation of
CXCR4 in CML has been previously linked to the dose of BCR/
ABL in CML [14].
Materials and methods
Patients and study protocol
Eighty-four consecutive patients with a diagnosis of MMM,
referred between July, 2003 and April, 2006 to RIMM (Italian
Registry for Myelofibrosis) were included in this study. Patients
were included if they had not been previously treated with
cytoreductive drugs, thalidomide, interferon or corticosteroids,
or if these treatments had been stopped for at least 3 months. In
63 patients, the diagnosis of MMM was established according
to the Italian consensus conference criteria [19]. Nineteen cases
had been diagnosed with a prefibrotic form of myelofibrosis
according to the WHO classification system [20]. The study
protocol was approved by the Institutional Review Board of the
IRCCS Policlinico S. Matteo in Pavia, Italy, and all patients
provided written informed consent.
Patients were assigned a prognostic score based on the
scoring system developed by Dupriez et al. [21], and a
severityscore, as previously reported [22].
Sixteen patients with PV were also studied for comparison (8
females and 8 males with a median age of 46 years; range, 22 to
70 years). The diagnosis was made according to the WHO
criteria [20]. PV patients were studied at diagnosis, or after
diagnosis but before any drug therapy was started. Control
samples were obtained from 20 healthy individuals.
Flow cytometric enumeration of CD34+ circulating
hematopoietic progenitor cells
One hundred microliters of ethylenediaminetetraacetic acid
(EDTA) anticoagulated blood was stained with fluorescein
isothiocyanate (FITC)-, phycoerythrin (PE)-, and 7-amino-
actinomycin D (7-AAD)-conjugated monoclonal antibodies
(moAb). The CD45-FITC/CD34-PE/7-ADD antibodies were
obtained from Becton Dickinson (San Jose, CA, USA), and the
analyses were performed utilizing a FACScalibur (Becton
Dickinson) [23].
Surface and intracellular flow cytometric analysis
One hundred microliters of EDTA anticoagulated blood was
stained with anti-CD34-FITC and anti-CXCR4-PE (Becton
Dickinson, San Jose, CA, USA). After red cell lysis the samples
were centrifuged and the pellets resuspended in 300 μL of saline.
Cells (2 × 10
5
) were flow cytometrically analyzed using the
CellQuest software (Becton Dickinson, San Jose, CA, USA).
Results were expressed both as percentage of CD34+ cells co-
expressing CXCR4, or as mean fluorescence intensity (MFI).
MFI was calculated as ratio of geometric mean of CD34+/
CXCR4+ cells and geometric mean of CD34+ co-stained with
the appropriate isotype controls.
The cytofluorimetric surface expression of CXCR4 on
CD34+ cells was measured immediately after blood sampling
or no more than 6 h apart in samples maintained at +4 °C.
Since it has been reported that upregulation of CXCR4
expression on circulating CD34+ cells occurs after incubation
281V. Rosti et al. / Blood Cells, Molecules, and Diseases 38 (2007) 280286
at 37 °C [25], in 5 samples the intra-assay variance of the
CXCR4 expression was assessed by repeating the measure-
ment immediately after sampling, after 1 h, 2 h, 3 h and 6 h.
Maintaining the sample at +4 °C did not alter the signal
intensity (intra assay CV = 1.5%).
For intracellular analysis prior to fixation of CXCR4, red
cells lysis and surface staining with anti-CD34-FITC was
performed. Cells were then fixed with 4% paraformaldehyde for
30 min and permeabilized with 0.1% saponin in 1% FCS/PBS
for 10 min. CXCR4-PE was then added for 30 min in the dark,
and the samples washed twice with ice-cold 0.1% saponin in 1%
FCS/PBS. The percentage of CD34+/CXCR4+ cells was then
determined.
In order to investigate CXCR4 surface expression on mature
hematopoietic cells, flow cytometry studies on separated
granulocytes was performed using a whole blood lysis
technique (NH4Cl) as previously described [24].
Real time RT-PCR quantification of CXCR4 gene expression
level
For the gene expression analysis in CD34+ HPC, mono-
nuclear cells were separated over a Ficoll gradient (Lymphoprep,
Nycomed Pharma, Oslo, Norway), and processed through two
sequential steps utilizing immunomagnetic CD34
+
cell selection
devices (Miltenyi Biotech, Gladbach, Germany). The purity of
the CD34
+
cell preparation was assessed flow cytometrically
(FACScan, Becton Dickinson, San Jose, CA, USA) after
staining with CD34-PE/CD45-FITC antibodies. Aliquots of
CD34
+
cells were immediately lysed using a guanidine
thiocyanate/phenol solution (Trizol, Gibco BRL, Paisley, UK)
and total RNA purified in the presence of carrier glycogen,
according to the manufacturer's instructions. The level of
CXCR4 gene expression was determined by quantitative real-
time PCR using the Pre-Developed TaqMan Assay Reagent
(#Hs00607978_s1) from Applied Biosystems (Foster City, CA,
USA), concurrently with the housekeeping GAPDH gene. Total
RNA (1 μg) was reverse transcribed with random hexamers and
50 ng cDNA was amplified in a ABI PRISM 7300 Sequence
Detection System (Applied Biosystems). The SDS software was
used to analyze fluorescent signals and to calculate the cycle
threshold (C
T
). Quantitative normalization of cDNA in each
sample was obtained by the ΔC
T
method (CXCR4 C
T
GAPDH C
T
). Each sample was assayed in triplicate, and both
negative and positive controls were included in each assay.
For the analysis of CXCR4 gene expression levels in
separated granulocytes, cDNA was reverse transcribed and
processed for real-time PCR as described above, except that
RNAse P was used as the reference gene.
Plasma levels of SDF-1α
Plasma SDF-1αlevels were measured using commercially
available high-sensitivity ELISA kits (R&D Systems, Minnea-
polis, MN, USA) in platelet-depleted samples (centrifuged at
11,000×gfor 10 min) stored at 40 °C. The limit of detection of
the assay was 1560 pg/mL.
JAK2 V617F genotyping
DNA was extracted by standard procedures, after isolation of
total peripheral blood granulocytes. Screening for the JAK2
V617F mutation was initially performed by an allele-specific
PCR in which the mutated allele was specifically amplified
together with a fragment common to the mutated and wild type
genes, with a detection limit for the mutated allele of 5% to
10%. Samples positive for the mutation were subsequently
analysed by PCR amplification and digestion with the
restriction endonuclease BsaXI which allows for an estimation
of the ratio between the mutated and wild type alleles. Samples
were scored as homozygous if the proportion of the mutant
allele was greater than 50%.
Statistical methods
Skewed variables were logarithmically transformed before
entering a parametric analysis. Comparisons between groups
were performed by the Student t-test, the MannWhitney U-
test or Chi-squared test when appropriate. Associations between
patient characteristics (covariates) were assessed for pairs of
numerical variables by Spearman's correlation, and for
categorical and continuous variables by WilcoxonMann
Whitney statistics. Results were considered statistically sig-
nificant when Pvalues were less than 0.05. All calculations
were performed with STATISTICA software (Statsoft, Tulsa,
OK, USA).
Results
Patient characteristics
The clinical characteristics of the 84 MMM patients enrolled
in the study are summarized in Table 1. The JAK2 V617F
mutation was identified in 39 of 56 (69.6%) patients, 10
(25.6%) of whom were homozygous for the mutation. Patients
who were homozygous for the JAK2V617F mutation were
characterized by a larger spleen index (mean, 346.8 cm
2
) than
patients heterozygous for the mutation (mean, 202 cm
2
;
P= 0.010), or non-mutat ed (mean, 163 cm
2
;P= 0.007). JAK2
V617F homozygous patients had a higher number of circulating
CD34+ cells than patients who were heterozygous for
JAK2V617F (199.8 × 10
6
/L vs. 31.8× 10
6
/L; P= 0.0015).
CXCR4 expression in CD34+ circulating cells and
granulocytes
The percentage of CD34+ cells expressing CXCR4 was
significantly lower (P< 0.001) in MMM patients (mean, 41.6%,
range, 0.6393.2%) than in the normal controls (mean, 70.2%;
range, 3797%) (Fig. 1). The MFI of CXCR4 expression was
also significantly lower in MMM patients (mean, 1.45; range,
0.027.05) than in normal controls (mean, 3.50; range, 0.37
24.1; P= 0.003). The percentage of CXCR4-expressing CD34+
cells and MFI of CXCR4 staining cells were also statistically
lower in MMM patients than in PV patients (mean, 63.5% for
282 V. Rosti et al. / Blood Cells, Molecules, and Diseases 38 (2007) 280286
CD34/CXCR4 co-expression and 2.21 for MFI; P= 0.013 and
P= 0.02, respectively). In patients with PV, the percentage of
CD34+ cells co-expressing CXCR4 and the MFI of CXCR4
expression was not different from that observed in normal
controls.
CD34+ cells from MMM patients were also characterized by
a lower intracellular CXCR4 MFI than from the normal
unstimulated controls (6.48 vs. 12.4). A direct correlation was
evidenced between surface and intracellular CXCR4 MFI in
MMM patients (R= 0.48; P= 0.004) (Fig. 2).
In 16 patients with MMM, the surface CXCR4 expression
was also assayed on circulating granulocytes. MFI was 1.86
(mean; range 0.593.64) in contrast to 3.43 (mean; range 2.19
4.40) in normal subjects (N= 10; P< 0.001).
Correlation between plasma levels of SDF-1 and CXCR4
expression
The levels of SDF-1 in the plasma of the 84 MMM patients
(4160 pg/mL, range 156014875 pg/mL) was significantly
higher than that found in healthy controls (2830 pg/mL, range
15605530 pg/mL) (P< 0.005). The levels of circulating SDF-1
did not correlate however with the number of circulating CD34+
cells. Since it is known that high levels of SDF-1 concentration
result in CXCR4 internalization [25], we examined the relation-
ship between the surface CXCR4 expression (both as percentage
of CXCR4-expressing CD34+ cells and MFI) and the levels of
SDF-1 in plasma. No such correlation between plasma SDF-1
levels and the degree of CXCR4 expression by CD34+ cells was
documented.
CXCR4 mRNA levels in cells purified from MMM patients and
controls
In 23 MMM patients the level of CXCR4 mRNA present in
CD34+cellswasmeasuredbyReal-TimeRT-PCRand
compared to that in normal CD34+ cells. The median S.D.)
ΔC
T
value for MMM CD34+ cells was significantly higher
than for normal CD34+ cells, thus documenting signifi-
cantly reduced CXCR4 mRNA content (Table 2). CXCR4
mRNA levels were also significantly decreased in MMM
granulocytes (N= 28) compa red to healt hy controls (N= 15)
(Table 2).
In 5 patients with MMM, we documented an inverse
correlation between the CXCR4 surface expression on CD34+
cells and ΔC
T
measurement (R=0.93), documenting an
association between decreased mRNA level and decreased
protein expression on the surface of the CD34+ cells. In 16
Table 1
Clinical characteristics of the 84 patients with MMM at the time of enrollment into the study
Characteristics All patients
(N=84)
Patients with a fibrotic
stage of MMM (N= 63)
Patients with a prefibrotic
form of MMM (N= 21)
Age, years, median (range) 51 (2084) 57 (2084) 45 (2474)
Sex, male, no. of patients (%) 42 (50%) 37 (58.7%) 5 (23.8%)
Hemoglobin, g/L, median (range) 113 (59167) 110 (59167) 117 (86165)
White blood cell count, × 10
9
/L, median (range) 8.5 (2.359.7) 7.6 (2.359.7) 8.6 (4.417.8)
Circulating immature myeloid cells + blasts, %,
median (range)
4.5 (021) 5 (021) 4 (110)
Circulating erythroblasts, %, median (range) 1.0 (0120) 1.0 (0120) 1.0 (02)
Platelet count, ×10
9
/L, median (range) 275 (101331) 257 (101268) 507 (1261331)
Spleen volume index, cm
2
, median (range)
a
190 (80672) 229 (90672) 120 (80600)
Time from diagnosis to examination, months,
median (range)
26 (1311) 22.5 (1205) 41 (2311)
Dupriez prognostic score at the time of the study,
number of patients (%)
b
0 (low risk) 52 (61.9%) 34 (54.0%) 18 (85.7%)
1 (intermediate risk) 23 (27.4%) 20 (31.7%) 3 (14.3%)
2 (high risk) 9 (10.7%) 9 (14.3%) 0 (0%)
Severity score, number of patients (%)
c
03 52 (61.9%) 34 (54.0%) 18 (85.7%)
45 32 (38.1%) 29 (46.0%) 3 (14.3%)
a
Spleen size was measured by ultrasonography by recording the spleen index, i.e. the product of the longitudinal by the transverse spleen axis.
b
A Dupriez score of 0 was assigned to a hemoglobin concentration greater than 100 g/L and a white blood cell count between 4×10
9
/L and 30 × 10
9
/L, a score of 1
to either a hemoglobin concentration less than 100 g/L or a white blood cell count greater than 30× 10
9
/L or less than 4 × 10
9
/L, and a score of 2 was given if both the
hemoglobin concentration and white blood cell count were in the aberrant ranges.
c
The severity score is the sum of myeloproliferation index and myelodepletion index derived from the values of hemoglobin, white blood cell count, platelet count
and spleen volume. Myeloproliferation index scored leukocytosis (corrected for the circulating erythroblasts), thrombocytosis and splenomegaly (or hepatomegaly in
splenectomized patients), while myelodepletion index scored anemia, leukopenia and thrombocytopenia. The following parameters were scored according to 3 grades:
splenomegaly (0=non-palpable or removed, 1 = 10 cm or less below the costal margin, 2=more than 10 cm below the costal margin) and anemia (0 = hemoglobin
concentration greater or equal to 120 g/L; 1 = hemoglobin concentration from 100 to 120 g/L; and 2 =hemoglobin concentration lower than 100 g/L or transfusion-
dependency). Two grades were considered for leukocytosis (0 = leukocyte count lower than 15 × 10
9
/L; 1= leukocyte count higher or equal to 15 × 10
9
/L),
thrombocytosis (0 = platelet count lower than 500 × 10
9
/L; 1 = platelet count higher or equal to 500× 10
9
/L), thrombocytopenia (0 = platelet count higher than
150 × 10
9
/L; 1 = platelet count equal or lower than 150× 10
9
/L) and leukopenia (0 = leukocyte count higher or equal to 4 × 10
9
/L; 1 = leukocyte count equal or lower
than 10
9
/L). Myelodepletion and myeloproliferation indexes ranged from 0 to 4, while the overall severity score ranged from 0 to 6.
283V. Rosti et al. / Blood Cells, Molecules, and Diseases 38 (2007) 280286
patients with MMM a similar relationship was observed in
peripheral blood granulocytes (R=0.62; P= 0.01).
Correlation between CXCR4 expression and disease
characteristics
An inverse correlation was observed between the expres-
sion of CXCR4 by CD34+ cells and the number of circulating
CD34+ cells present in the peripheral blood (R=0.55;
P< 0.001 for percentage of CXCR4 expressing CD34+ cells,
and R=0.35, P= 0.001 for MFI) (Fig. 3).
The decreased intensity of CXCR4 expression by MMM
CD34+ cells, as measured by MFI, was associated with
lower Hb levels (R= 0.384; P= 0.001), older age (R=0.31;
P= 0.026), longer disease duration (R=0.37; P= 0.008), and
lower platelet count (R= 0.42; P= 0.007). Patients with a
prefibrotic form of myelofibrosishad higher levels of
CXCR4 expression at the time of the study than those with
fibrotic bone marrows (mean, 56.4% vs. 36.5%, P= 0.001 for
percentage of CXCR4-expressing CD34+ cells; 3.05 vs. 0.98,
P= 0.011 for MFI). Both the patient's Dupriez prognostic
score and severityscore were correlated with surface CXCR4
down-regulation (CXCR4-expressing CD34+ cell, percentage,
F= 4.04; P= 0.021 for Dupriez score; F= 2.208, P= 0.05 for
severity score). The myelodepletion index, which includes
leukopenia, thrombocytopenia and anemia, contributed to a
greater extent to the correlation between the severity score
and the degree of CXCR4 down-regulation (F= 2.78; P= 0.03)
(Fig. 4).
Correlation between CXCR4 expression and JAK2 V617F
mutational state
The expression of CXCR4 on CD34+ cells did not differ in
MMM patients heterozygous for JAK2V617F mutation as
compared to those with a wild-type genotype. Although a
Fig. 1. CXCR4 expression on circulating CD34+ cells from patients with MMM,
PVor control subjects. Patients with MMM include idiopathic myelofibrosis and
post-ET and post-PV myelofibrosis. CXCR4 expression levels (percent of
CXCR4-expressing CD34+ cells) are shown as a box plot where dots indicate the
mean value, bars indicate the standard deviation, while the lower and upper edges
of the boxes indicate the standard error. The Student t-test showed significant
differences between MMM patients and PV patients or normal subjects
(P<0.001).
Fig. 2. Relationship between circulating CD34+ intracellular CXCR4 mean
flurescence intensity (MFI) and CD34+ cells surface CXCR4 MFI. Linear
regression analysis demonstrated a significant correlation (R= 0.48; P= 0.004).
Table 2
Measurement of CXCR4 gene expression levels in MMM and normal cell
populations
ΔC
T
(mean ± S.D.) Range P
Cell source
MMM CD34+ cells (N= 23) 5.12 ± 2.42 1.159.77 0.003
Normal CD34+ cells (N= 7) 1.53 ± 0.79 0.312.68
MMM Granulocytes (N= 28) 2.36 ± 0.93 1.045.09 0.014
Normal Granulocytes (N= 15) 1.60 ± 0.91 0.32.77
CD34+ cells and granulocytes were purified from the peripheral blood (PB) of
MMM patients, or from peripheral blood of normal volunteer donors. CXCR4
mRNA levels were quantified by a real time RT-PCR assay, and expressed as
ΔC
T
using GAPDH or RNAseP (in case of granulocytes) as the housekeeping
reference gene.
Fig. 3. Relationship between the expression of CXCR4 on the surface of CD34+
cells of patients with MMM and the number of CD34+ hematopoietic progenitor
cells (HPCs) in peripheral blood. The CXCR4 expression was measured as
CXCR4-expressing CD34+ cells (percentage). Linear regression analysis
demonstrated a significant correlation (R=0.55; P< 0.001).
284 V. Rosti et al. / Blood Cells, Molecules, and Diseases 38 (2007) 280286
reduction of CXCR4 expression on CD34+ cells was evident in
JAK2V617F homozygous patients as compared to heterozy-
gous and wild-type patients (mean, CXCR4-expressing CD34+
cells, 29.6% vs. 41.6%; MFI, 0.56 vs. 1.68, respectively), these
differences did not, however, reach statistical significance.
Discussion
MMM has been shown to be characterized by an unusual
degree of mobilization of HPCs, stem cells as well as
endothelial cells into the peripheral blood [2,26]. The mechan-
ism underlying the constitutive mobilization of CD34+ cells in
MMM patients remains poorly understood. In this study, we
analysed the hypothesis that down-regulation of the chemokine
receptor CXCR4 might contribute to this process. SDF-1 is the
natural ligand for CXCR4. The CXCR4/SDF-1 axis has been
documented to play a role in the homing and retention of
primitive HPCs and stem cells within the bone marrow [27].
Disruption of this ligand/receptor interaction has been impli-
cated in cytokine mediated mobilization of CD34+ cells [28].In
CML cross talk between BCR/ABL and the CXCR4 pathway
has been shown to result in the disruption of chemokine
signalling and chemotaxis [2932]. Geay et al. have recently
reported that cells expressing high levels of BCR/ABL are
characterized by a significant signalling defect accompanied by
a reduction of CXCR4 expression at the transcriptional level
[14]. Furthermore the tyrosine kinase activity of BCR/ABL was
clearly shown to down regulate the expression and function of
CXCR4. We found that circulating CD34+ cells of MMM
patients, on average, displayed lower levels of CXCR4 than
CD34+ cells from patients with PV or normal individuals. We
documented that the CXCR4 mRNA levels in CD34+ cells and
granulocytes from MMM patients were lower as compared to
controls. Moreover, by correlating CXCR4 gene and receptor
expression, we showed a strict correlation between reduced
mRNA content and cell surface protein expression, demonstrat-
ing that the CXCR4 down-regulation in CD34+ cells and
granulocytes of MMM is the consequence of a transcriptional
event.
The down-regulation of CXCR4 on CD34+ cells was asso-
ciated with increasing numbers of circulating CD34+ cells,
suggesting that the down regulation of CXCR4 by CD34+ cells
may play a role in the constitutive HPC mobilization. Although
CXCR4 down-regulation on CD34+ cells is able to directly
influence cell mobilization via the disruption of the SDF-1 axis
in bone marrow, the constitutive mobilization of CD34+ cells in
MMM is more likely the result of multiple events affecting stem
cell trafficking.
The down-regulation of CXCR4 by CD34+ cells and
granulocytes were shown in this report to be due to reduced
gene expression. Both genetic and epigenetic mechanisms
have been documented to affect transcriptional regulation of
the CXCR4 gene [33,34]. We present evidence here to indicate
that the decreased expression of CXCR4 in MMM occurs
irrespective of the JAK2V617F mutational status of patients
with MMM, suggesting that other intracellular signals are
likely responsible for this event. Further studies are clearly
required to gain greater understanding of the precise molecular
mechanism responsible for this transcriptional silencing of
CXCR4.
In our series of cases, down-regulation of CXCR4 in MMM
peripheral blood CD34+ cells was associated with disease
characteristics that reflected the severity of the disease process,
including older age [35], overt bone marrow fibrosis, anemia,
leukopenia and thrombocytopenia. Since CXCR4 participates
in the regulation of bone marrow myelopoiesis, and in the long
term bone marrow repopulation by pluripotent stem cells [36
38], the down-modulation of CXCR4 receptor in MMM may
represent a crucial event in the progression of the disease that
may contribute to the aberrant trafficking of HPCs and the
depletion of bone marrow stem cells [39], leading to the
development of a bone marrow failure state that characterizes
the more advanced stages of the disease.
Acknowledgments
Research Grant Support: This work was supported by a grant
(CS30, 2003) from Istituto Superiore di Sanità; a grant from the
Italian Ministry of Health (Ricerca Finalizzata 2002), a grant
from MIUR (2003, #06488803); a grant from Associazione
Italiana per la Ricerca sul Cancro (Milano) and the MPD
Research Consortium funded by the National Cancer Institute
(CA P01 CA 108671-01A2).
References
[1] G. Barosi, R. Hoffman, Idiopathic myelofibrosis, Semin. Hematol. 42
(2005) 248258.
[2] G. Barosi, G. Viarengo, A. Pecci, et al., Diagnostic and clinical relevance
of the number of circulating CD34+ cells in myelofibrosis with myeloid
metaplasia, Blood 98 (2001) 32493255.
[3] A. Peled, I. Petit, O. Kollet, et al., Dependence of human stem cell
Fig. 4. CXCR4 expression on the surface of CD34+ cells of MMM patients
categorized according their myelodepletion index. CXCR4 expression levels
(percent of CXCR4-expressing CD34+ cells) are shown as a box plot where dots
indicate the mean value, bars indicate the standard deviation, while the lower
and upper edges of the boxes indicate the standard error. Myelodepletion index,
which scores leukopenia, thrombocytopenia and anemia was correlated with
surface CXCR4-down regulation (F=2.78; P=0.03).
285V. Rosti et al. / Blood Cells, Molecules, and Diseases 38 (2007) 280286
engraftment and repopulation of NOD/SCID mice on CXCR4, Science
283 (1999) 845848.
[4] T. Lapidot, I. Petit, Current understanding of stem cell mobilization: the
roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines,
and stromal cells, Exp. Hematol. 30 (2000) 973981.
[5] M. Kucia, R. Reca, K. Miekus, et al., Trafficking of normal stem cells and
metastasis of cancer stem cells involve similar mechanisms: pivotal role of
the SDF-1-CXCR4 axis, Stem Cells 23 (2005) 879894.
[6] L.M. Pelus, H. Bian, A.G. King, S. Fukuda, Neutrophil-derived MMP-9
mediates synergistic mobilization of hematopoietic stem and progenitor
cells by the combination of G-CSF and the chemokines GRObeta/CXCL2
and GRObetaT/CXCL2delta4, Blood 103 (2004) 110119.
[7] J.P. Levesque, J. Hendy, Y. Takamatsu, et al., Mobilization by either
cyclophosphamide or granulocyte colony-stimulating factor transforms the
bone marrow into a highly proteolytic environment, Exp. Hematol. 30
(2002) 440449.
[8] M. Xu, E. Bruno, J. Chao, et al., Constitutive mobilization of CD34+ cells
into the peripheral blood in idiopathic myelofibrosis may be due to the
action of a number of proteases, Blood 105 (2005) 45084515.
[9] F. Passamonti, E. Rumi, D. Pietra, et al., Relation between JAK2 (V617F)
mutation status, granulocyte activation, and constitutive mobilization of
CD34+ cells into peripheral blood in myeloproliferative disorders, Blood
107 (2006) 36763682.
[10] F. Bertolini, C. Dell'Agnola, P. Mancuso, et al., CXCR4 neutralization, a
novel therapeutic approach for non-Hodgkin's lymphoma, Cancer Res. 62
(2002) 31063112.
[11] A. Larochelle, A. Krouse, M. Metzger, et al., AMD3100 mobilizes
hematopoietic stem cells with long-term repopulating capacity in nonhu-
man primates, Blood 107 (2006) 37723778.
[12] L.M. Pelus, H. Bian, S. Fukuda, et al., The CXCR4 agonist peptide,
CTCE-0021, rapidly mobilizes polymorphonuclear neutrophils and
hematopoietic progenitor cells into peripheral blood and synergizes with
granulocyte colony-stimulating factor, Exp. Hematol. 33 (2005) 295307.
[13] Q. Ma, D. Jones, P.R. Bonghesan, et al., Impaired B-lymphopoiesis,
myelopoiesis, and derailed cerebellar neuron migration in CXCR4-
and SDF-1 deficient mice, Proc. Natl. Acad. Sci. U. S. A. 95 (1998)
94489453.
[14] J.F. Geay, D. Buet, Y. Zhang, et al., p210BCR-ABL inhibits SDF-1
chemotactic response via alteration of CXCR4 signaling and down-
regulation of CXCR4 expression, Cancer Res. 65 (2005) 26762683.
[15] H.K. Kim, M.D. Sierra, C.K. Williams, et al., G. G-CSF downregulation of
CXCR4 expression identified as a mechanism for mobilization of myeloid
cells, Blood prepublished online March 14, 2006.
[16] R.A. Mesa, H. Powell, T. Lasho, et al., A longitudinal study of the JAK2
(V617F) mutation in myelofibrosis with myeloid metaplasia: analysis at
two time points, Haematologica/THJ 91 (2006) 415416.
[17] P.J. Campbell, M. Griesshammer, K. Dohner, et al., V617F mutation in
JAK2 is associated with poorer survival in idiopathic myelofibrosis, Blood
107 (2006) 20982100.
[18] A. Tefferi, T.L. Lasho, S.M. Schwager, et al., The JAK2(V617F) tyrosine
kinase mutation in myelofibrosis with myeloid metaplasia: lineage
specificity and clinical correlates, Br. J. Haematol. 131 (2005) 320328.
[19] G. Barosi, A. Ambrosetti, C. Finelli, et al., The Italian Consensus
Conference on diagnostic criteria for myelofibrosis with myeloid
metaplasia, Br. J. Haematol. 104 (1999) 730737.
[20] J. Thiele, R. Pierre, M. Imbert, et al., in: E.S. Jaffe, N. Harris, N. Stan, J.W.
Vardiman (Eds.), Chronic Idiopathic Myelofibrosis in World Health
Organization of Tumors of Hematopoietic and Lymphoid Tissues, IARC
Press, Washington, DC, 2001, pp. 3538.
[21] B. Dupriez, P. Morel, J.L. Demory, et al., Prognostic factors in agnogenic
myeloid metaplasia: a report on 195 cases with a new scoring system,
Blood 88 (1996) 10131018.
[22] M. Marchetti, G. Barosi, F. Balestri, et al., Low-dose thalidomide
ameliorates cytopenias and splenomegaly in myelofibrosis with myeloid
metaplasia: a phase II trial, J. Clin. Oncol. 22 (2004) 424431.
[23] M. Keeney, I. Chin-Yee, K. Weir, et al., Single platform flow cytometric
absolute CD34+ cell counts based on the ISHAGE guidelines. Interna-
tional Society of Hematotherapy and Graft Engineering, Cytometry 34
(1998) 6170.
[24] G.T. Stelzer, G. Marti, A. Hurlly, et al., U.S.Canadian Consensus
recommendations on the immunophenotypic analysis of hematologic
neoplasia by flow cytometry: standardization and validation of laboratory
procedures, Cytometry 30 (1997) 214230.
[25] J.-J. Lataillade, D. Clay, C. Dupuy, et al., Chemokine SDF-1 enhances
circulating CD34+ cells proliferation in synergy with cytokines: possible
role in progenitor survival, Blood 95 (2000) 756768.
[26] M. Massa, V. Rosti, I. Ramajoli, R. Campanelli, A. Pecci, G. Viarengo, V.
Meli, M. Marchetti, R. Hoffman, G. Barosi, Circulating CD34+, CD133+,
and vascular endothelial growth factor receptor 2-positive endothelial
progenitor cells in myelofibrosis with myeloid metaplasia, J. Clin. Oncol.
23 (2005) 56885695.
[27] A. Aiuti, I.J. Webb, C. Bleul, T. Springer, J.C. Gutierrez-Ramos, The
chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic
progenitor cells and provides a new mechanism to explain the mobilization
of CD34+ progenitors to peripheral blood, J. Exp. Med. 185 (1997)
111120.
[28] T. Lapidot, I. Petit, Current understanding of stem cell mobilization: the
roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines,
and stromal cells, Exp. Hematol. 30 (2002) 973981.
[29] R. Salgia, E. Quackenbush, J. Lin, et al., The BCR/ABL oncogene alters
the chemotactic response to stromal-derived factor-1alpha, Blood 94
(1999) 42334246.
[30] A. Ptasznik, E. Urbanowska, S. Chinta, et al., Crosstalk between BCR/
ABL oncoprotein and CXCR4 signaling through a Src family kinase in
human leukemia cells, J. Exp. Med. 196 (2002) 667678.
[31] A. Peled, I. Hardan, L. Trakhtenbrot, et al., Immature leukemic
CD34+CXCR4+ cells from CML patients have lower integrin-depen-
dent migration and adhesion in response to the chemokine SDF-1,
Stem Cells 20 (2002) 259266.
[32] J. Durig, C. Rosenthal, A. Elmaagacli, et al., Biological effects of stroma-
derived factor-1 alpha on normal and CML CD34+ haemopoietic cells,
Leukemia 14 (2000) 16521660.
[33] T. Mori, J. Kim, T. Yamano, et al., Epigenetic up-regulation of CC
chemokine receptor 7 and CXC chemokine receptor 4 expression in
melanoma cells, Cancer Res. 65 (2005) 18001807.
[34] N. Sato, H. Matsubayashi, N. Fukushima, et al., The chemokine receptor
CXCR4 is regulated by DNA methylation in pancreatic cancer, Biol. Ther.
4 (2005) 7076.
[35] K. Strasser-Weippl, M. Steurer, M. Kees, et al., Age and hemoglobin level
emerge as most important clinical prognostic parameters in patients with
osteomyelofibrosis: introduction of a simplified prognostic score, Leuk.
Lymphoma 47 (2006) 441450.
[36] J. Kahn, T. Byk, L. Jansson-Sjostrand, et al., Overexpression of CXCR4 on
human CD34+ progenitors increases their proliferation, migration, and
NOD/SCID repopulation, Blood 103 (2004) 29422949.
[37] T. Nagasawa, S. Hirota, K. Tachibana, et al., Defects of B-cell
lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC
chemokine PBSF/SDF-1, Nature 382 (1996) 638653.
[38] Q. Ma, D. Jones, T.A. Springer, The chemokine receptor CXCR4 is
required for the retention of B lineage and granulocytic precursors within
the bone marrow microenvironment, Immunity 10 (1999) 463471.
[39] H. Ni, G. Barosi, D. Rondelli, R. Hoffman, Studies of the site and
distribution of CD34+ cells in idiopathic myelofibrosis, Am. J. Clin.
Pathol. 123 (2005) 833839.
286 V. Rosti et al. / Blood Cells, Molecules, and Diseases 38 (2007) 280286
... The changes in the eNAMPT concentration induce the activation of HPCs/HSCs mobilization, dependent on the chemokine (C-X-C Motif) receptor 4 (CXCR4). The mechanism is NAMPT-induced NAD-dependent Sirtuin1 activity, which leads to a reduced CXCR4 expression on hematopoietic cells in PMF, caused by a hyper-methylation of the CXCR4 gene promoter [107][108][109]. In 2007, we reported that the low membrane expression of CXCR4 on circulating CD34+ in PMF patients was correlated with the mobilization of cells [107]. ...
... The mechanism is NAMPT-induced NAD-dependent Sirtuin1 activity, which leads to a reduced CXCR4 expression on hematopoietic cells in PMF, caused by a hyper-methylation of the CXCR4 gene promoter [107][108][109]. In 2007, we reported that the low membrane expression of CXCR4 on circulating CD34+ in PMF patients was correlated with the mobilization of cells [107]. ...
Article
Full-text available
Primary myelofibrosis (PMF) is a myeloproliferative neoplasm due to the clonal proliferation of a hematopoietic stem cell. The vast majority of patients harbor a somatic gain of function mutation either of JAK2 or MPL or CALR genes in their hematopoietic cells, resulting in the activation of the JAK/STAT pathway. Patients display variable clinical and laboratoristic features, including anemia, thrombocytopenia, splenomegaly, thrombotic complications, systemic symptoms, and curtailed survival due to infections, thrombo-hemorrhagic events, or progression to leukemic transformation. New drugs have been developed in the last decade for the treatment of PMF-associated symptoms; however, the only curative option is currently represented by allogeneic hematopoietic cell transplantation, which can only be offered to a small percentage of patients. Disease prognosis is based at diagnosis on the classical International Prognostic Scoring System (IPSS) and Dynamic-IPSS (during disease course), which comprehend clinical parameters; recently, new prognostic scoring systems, including genetic and molecular parameters, have been proposed as meaningful tools for a better patient stratification. Moreover, new biological markers predicting clinical evolution and patient survival have been associated with the disease. This review summarizes basic concepts of PMF pathogenesis, clinics, and therapy, focusing on classical prognostic scoring systems and new biological markers of the disease.
... One consequence of changes in the extra-cellular NAMPT concentration in MPNs is activation of the chemokine (C-X-C Motif) Receptor 4 (CXCR4)-dependent mobilization of hematopoietic stem and progenitor cells. The mechanism of this effect is NAMPT-induced NAD-dependent SIRT1 activity, a potent repressor of CXCR4 expression [4], the consequence of which is that hematopoietic cells in MPN-associated myelofibrosis have consistently reduced membrane CXCR4 expression [6][7][8][9][10], because of hyper-methylation of the CXCR4 gene promoter [9]. ...
... In 2007 we reported decreased surface expression of CXCR4 on CD34-positive blood cells in MPN-associated myelofibrosis was associated with mobilization of the cells [6]. Because constitutive mobilization of CD34-positive blood cells correlates with disease activity [11], we hypothesized CXCR4 surface expression on CD34-positve cells might correlate with disease activity in MPNs. ...
Article
Full-text available
The expression of the CXCR4 chemokine receptor on CD34-positive blood cells is reduced in persons with primary myelofibrosis (PMF). We analyzed the relevance of cytofluorimetric assessment of the percentage of CD34-positive blood cells that had a positive CXCR4 surface expression (CD34/CXCR4-se) in a large cohort of subjects with myeloproliferative neoplasms. Mean CD34/CXCR4-se was lower in subjects with PMF compared with those with essential thrombocythemia (ET) or polycythemia vera (PV). A cutoff value of 39% was associated with a diagnosis of pre-fibrotic PMF vs. ET with a positive predictive value of 97%. In PMF male sex, older age, and MPL mutation were independent correlates of reduced CD34/CXCR4-se and associated with a briefer interval to development of severe anemia, large splenomegaly, thrombocytopenia, leukopenia, elevated CD34-positive blood cells, blast transformation and death. We constructed a prognostic model including age >65 years, hemoglobin < 100 g/L, CD34-positive blood cells > 50 × 106/L, and CD34/CXCR4-se <39% at diagnosis. The model identified three risk cohorts with greater accuracy compared with the International Prognostic Scoring System. In conclusion, CD34/CXCR4-se is a highly sensitive marker of disease activity and a new potential diagnostic and prognostic biomarker in PMF.
... Among the major biological parameters of disease progression, constitutive p-STAT5 MFI values were significantly inversely correlated with both the frequency of CD34 + CXCR4 + cells (R = -0.47 p = 0.0007), whose reduction marks the increase of the disease severity [18], and the serum cholesterol concentration (R = -0.51, p = 0.009); in addition, constitutive p-STAT5 MFI values were significantly directly correlated (R = 0.38, p = 0.005) with the number/L of circulating CD34 + cells. ...
... Abnormal CD34 + cell mobilization is a well known specific biological feature of patients with PMF [1]. We have also previously shown that in patients with MF a reduced number of CD34 + CXCR4 + circulating cells correlates with disease severity [18]. CXCR4 signaling, which occurs following the binding with its ligand SDF-1α/CXCL12, is known to involve p-STAT5 [34], thus the constitutive activation of p-STAT5 could result in a reduced expression of the CXCR4 on the cell membrane representing a potential mechanism of CD34 + cell mobilization from the BM into the PB. ...
Article
Full-text available
Primary Myelofibrosis (PMF) is a myeloproliferative disorder associated with JAK2V617F, Calreticulin (CALR) indels, and MPLW515L/K mutations activating the tyrosine kinase JAK2 and its downstream signaling pathway. The nature of signaling abnormalities in primary cells from PMF patients is poorly understood, since most of the work has been performed in cell lines or animal models. By flow cytometry we measured constitutive and cytokine induced phosphorylation of STAT5, STAT3, and ERK1/2 in circulating CD34+ cells from 57 patients with PMF (20 with prefibrotic-PMF) and 13 healthy controls (CTRLs). Levels of constitutive and TPO induced p-STAT5, and IL6 induced p-STAT3 were higher in patients than in CTRLs. Constitutive p-STAT5 values were lower in CALR than homozygous JAK2V617F mutated CD34+ cells from PMF patients. Moreover, constitutive p-STAT5 and IL6 induced p-STAT3 values correlated directly with circulating CD34+ cell number/L, and inversely with the frequency of circulating CD34+ cells expressing CXCR4. Constitutive p-STAT5 values of CD34+ cells were also inversely correlated with hemoglobin levels. When the patients were divided according with presence/absence of JAK2V617F mutation, all the correlations described characterized the JAK2V617F+ patients with prefibrotic-PMF (P-PMF). In conclusion, increased constitutive p-STAT5 and IL6 induced p-STAT3 values in circulating CD34+ cells characterize patients with PMF. Constitutive p-STAT5 and IL6 induced p-STAT3 values correlate with circulating CD34+ cell number/L, the frequency of circulating CD34+ cells expressing CXCR4 and hemoglobin levels within the prefibrotic JAK2V617F+ patient population. Our data point toward a complex activation of STAT5-dependent pathways in the stem/progenitor cell compartment, that characterize the phenotypic diversity of PMF.
... The expression of C-X-C motif chemokine ligand 12 (CXCL12), which is normally produced by osteoblasts and promotes the maintenance of HSCs, is increased in the spleen of patients with MF, leading to egress of hematopoietic elements from the bone marrow and localization in the spleen (43). Meanwhile, CXCR4-the canonical receptor for CXCL12-is downregulated in CD34 + cells from patients with MF secondary to hypermethylation, leading to decreased expression and likely egress from the HSC niche (44,45). However, AZA-treated primary myelofibrosis (PMF) CD34 + cells exhibit a relatively complete reversal of CpG1 island hypermethylation and increased CXCR4 expression (45). ...
Article
Full-text available
Background Atypical chronic myeloid leukemia (aCML) is a BCR::ABL1 negative myelodysplastic/myeloproliferative neoplasm with poor overall survival. Some patients can be treated by allogeneic hematopoietic stem cell transplantation (allo-HSCT) from suitable donors. The effectiveness of decitabine or azacitidine (AZA) has recently been reported; however, their combined efficacy with selinexor has not yet been reported. Case description In this study, we report the case of a patient with aCML who was successfully treated with selinexor combined with AZA. A 67-year-old man with a history of gastric mucosa-associated lymphoid tissue (MALT) lymphoma was admitted to the hospital with fatigue and emaciation. He was diagnosed with aCML and no longer responded to decitabine treatment after undergoing seven cycles. The patient was subsequently administered hydroxyurea (HU), selinexor, and AZA. After four courses of combination therapy, his blood cell counts improved; he no longer required transfusions and was able to discontinue HU. The patient continued receiving selinexor and AZA without severe complications. This case is the first to show that combinatorial selinexor and AZA therapy can effectively treat aCML. Conclusion Our case sheds light on the importance of selinexor and AZA combined therapy in the exploration of new treatment strategies for aCML. Moreover, this treatment approach offers the possibility of bridging with allo-HSCT.
... CXCR4 has an important role for homing and retention of hematopoietic stem cells (HSC) [82] and for maintaining HSC quiescence as well [83]. We and others have previously reported CXCR4 to be significantly downregulated in MPNs, in particular in myelofibrosis [43], consistent with previous studies displaying downregulation of CXCR4 in CD34+ cells [84]. Accordingly, upregulation of the CXCR4 gene during rIFNα treatment might prohibit egress of CD34 + positive stem cells from bone marrow niches and accordingly development of extramedullary hematopoiesis in the spleen and liver. ...
Article
Full-text available
Chronic inflammation is considered a major driving force for clonal expansion and evolution in the Philadelphia-negative myeloproliferative neoplasms, which include essential thrombocythemia, polycythemia vera and primary myelofibrosis (MPNs). One of the key mutation drivers is the JAK2V617F mutation, which has been shown to induce the generation of reactive oxygen species (ROS). Using whole blood gene expression profiling, deregulation of several oxidative stress and anti-oxidative defense genes has been identified in MPNs, including significant downregulation of TP53 , the NFE2L2 or NRF2 genes. These genes have a major role for maintaining genomic stability, regulation of the oxidative stress response and in modulating migration or retention of hematopoietic stem cells. Therefore, their deregulation might give rise to increasing genomic instability, increased chronic inflammation and disease progression with egress of hematopoietic stem cells from the bone marrow to seed in the spleen, liver and elsewhere. Interferon-alpha2 (rIFNα) is increasingly being recognized as the drug of choice for the treatment of patients with MPNs. Herein, we report the first gene expression profiling study on the impact of rIFNα upon oxidative stress and antioxidative defense genes in patients with MPNs (n = 33), showing that rIFNα downregulates several upregulated oxidative stress genes and upregulates downregulated antioxidative defense genes. Treatment with rIFNα induced upregulation of 19 genes in ET and 29 genes in PV including CXCR4 and TP53 . In conclusion, this rIFNα- mediated dampening of genotoxic damage to hematopoietic cells may ultimately diminish the risk of additional mutations and accordingly clonal evolution and disease progression towards myelofibrotic and leukemic transformation.
... Despite an increased plasma level and BM deposition of CXCL12 were described in MF patients [36], our findings reveal no difference in the CXCL12 mRNA levels and protein secretion between MSCs from HF-MF and HD. Abnormal HSC trafficking/homing in MF patients may be due to reduced expression of CXCR4 on HSCs through promoter hypermethylation and disfunction of CXCR4/CXCL12 axis by enhanced proteolysis [37][38][39]. ...
Article
Full-text available
Mesenchymal stromal cells (MSCs) represent an essential component of the bone marrow (BM) niche and display disease-specific alterations in several myeloid malignancies. The aim of this work was to study possible MSC abnormalities in Philadelphia-negative myeloproliferative neoplasms (MPNs) in relationship to the degree of BM fibrosis. MSCs were isolated from BM of 6 healthy donors (HD) and of 23 MPN patients, classified in 3 groups according to the diagnosis and the grade of BM fibrosis: polycythemia vera and essential thrombocythemia (PV/ET), low fibrosis myelofibrosis (LF-MF), and high fibrosis MF (HF-MF). MSC cultures were established from 21 of 23 MPN patients. MPN-derived MSCs did not exhibit any functional impairment in their adipogenic/osteogenic/chondrogenic differentiation potential and displayed a phenotype similar to HD-derived MSCs but with a decreased expression of CD146. All MPN-MSC lines were negative for the patient-specific hematopoietic clone mutations (JAK2, MPL, CALR). MSCs derived from HF-MF patients displayed a reduced clonogenic potential and a lower growth kinetic compared to MSCs from HD, LF-MF, and PV/ET patients. mRNA levels of hematopoiesis regulatory molecules were unaffected in MSCs from HF-MF compared to HD. Finally, in vitro ActivinA secretion by MSCs was increased in HF-MF compared to LF-MF patients, in association with a lower hemoglobin value. Increased ActivinA immunolabeling on stromal cells and erythroid precursors was also observed in HF-MF BM biopsies. In conclusion, higher grade of BM fibrosis is associated with functional impairment of MSCs and the increased secretion of ActivinA may represent a suitable target for anemia treatment in MF patients.
... The abnormally mobilized CD34+ cells in MF have decreased expression of CXCR4. Its level of expression has been correlated inversely with the number of circulating CD34+ cells as well as the severity index in PMF [12,13]. GATA-1 is a transcription factor that plays a role in erythroid and megakaryocytic cell differentiation [14]. ...
Article
Full-text available
Purpose of review: Myelofibrosis is a chronic myeloproliferative neoplasm which can lead to massive splenomegaly. Currently approved medical therapies do not improve splenomegaly in all patients and effects are not sustained. Thus, spleen-directed therapies (i.e., splenectomy and splenic irradiation) have been used in some cases to palliate the signs and symptoms of the disease. Here, we critically review the literature regarding palliative splenectomy and splenic irradiation in myelofibrosis, and discuss their position in the current treatment landscape. Recent findings: Retrospective studies have demonstrated that splenectomy improves symptoms of splenomegaly, decreases complications of portal hypertension, and decreases transfusion dependence. However, it carries a significant peri-operative and long-term morbidity and mortality rate. Splenic irradiation reduces splenic size but is limited by duration of response and myelosuppression. Spleen-directed therapies in myelofibrosis may be considered for refractory symptoms and complications of massive splenomegaly after carefully weighing the associated risks, though overall survival may not be impacted. Development of medical therapies that target and reverse the underlying disease pathophysiology is required in order to have a significant impact on the natural history of the disease process.
Article
Introduction: Myelofibrosis is a hematologic malignancy with a variety of clinical manifestations including splenomegaly, which is present in approximately 80% of newly diagnosed patients. JAK inhibitors are the mainstay of pharmacologic treatment for splenomegaly in myelofibrosis, although spleen size reduction is not universal, and the duration of benefit is only moderately durable. Areas covered: We first discuss the pathobiology of splenomegaly in myelofibrosis before detailing approved and novel pharmacotherapies that can reduce spleen size while also highlighting non-pharmacologic approaches. In this review, efficacy of these treatments is measured solely by spleen volume reduction, acknowledging that other outcome measures such as symptom improvement and survival are also critical. Expert opinion: Currently, ruxolitinib can be administered to the majority of frontline patients although those with severe thrombocytopenia should receive pacritinib to address spleen burden. Momelotinib may be particularly well suited for patients with significant anemia and novel combination treatments in clinical development may improve the depth and duration of spleen responses. After frontline treatment failure, fedratinib or pacritinib are commercial options for patients with persistent symptomatic splenomegaly. Novel agents given alone or in combination with a JAK inhibitor are being explored in trials which may ameliorate splenomegaly and ultimately improve disease progression.
Article
The myeloproliferative neoplasms (MPNs) are characterized by overproduction of mature blood cells and increased risk of transformation to frank leukemia. The acquired kinase mutation JAK2V617F plays a central role in a majority of patients with these diseases. As MPNs are clonal stem cell disorders (i.e. arise from a single stem cell which eventually expands), the hematopoietic stem/progenitor cell (HSPC) compartment in MPNs is heterogeneous with the presence of both JAK2 wild-type and JAK2V617F mutant cells. Mechanisms responsible for the mutant stem cell expansion in MPNs are not fully understood. Utilizing in vitro co-culture assays and in vivo competitive transplantation assays, we show that the presence of wild-type cells alters both the gene expression profile and cellular function of JAK2V617F mutant HSPCs and inhibits the expansion of co-existing JAK2V617F mutant cells in a normal microenvironment. In contrast, we found that a microenvironment bearing the mutant kinase promotes JAK2V617F mutant HSPC expansion over wild-type cells due in part to altered CXCL12/CXCR4 signaling. Further understanding of the molecular mechanisms controlling the competitive interactions between normal and JAK2V617F mutant cells, and how these mechanisms break down during MPN disease progression hold great potential for advances in treating patients with these diseases.
Article
Background: Leukemic transformation is an unfavorable event emerging in a minority of patients with myelofibrosis (MF) and carrying a poor prognosis. Patients with post-MF acute myeloid leukemia (AML) may be treated with curative or noncurative intent, but there is no standard of care. The use of hypomethylating agents has been correlated with limited activity and low overall survival rates, while ruxolitinib has been proven to have modest antileukemic activity. Case presentation: In this case study, we present the case of a 67-year-old female patient with post-polycythemia vera MF who developed AML and was successfully treated with a combination of 5-azacytidine and ruxolitinib, without any significant toxicity. Summary: To our knowledge, this is the first report of a patient with post-MF AML achieving a complete remission with a combination of ruxolitinib and a hypomethylating agent. This combination is actively studied in phase I/II clinical trials and may be proven effective in this hard-to-treat group of patients.
Article
Full-text available
In concert with the International Society of Hematotherapy and Graft Engineering (ISHAGE), we previously described a set of guidelines for detection of CD34+ cells based on a four-parameter flow cytometry method (CD45 FITC/CD34 PE staining, side and forward angle light scatter). With this procedure, an absolute CD34+ count is generated by incorporating the leukocyte count from an automated hematology analyser (two-platform method). In the present study, we modified the basic ISHAGE method with the addition of a known number of Flow-Count™ fluorospheres. To reduce errors inherent to sample washing/centrifugation, we implemented ammonium chloride lyse, no-wash no-fix sample processing. These modifications convert the basic protocol into a single-platform method to determine the absolute CD34 count directly from a flow cytometer and form the basis of the Stem-Kit from Coulter/Immunotech. A total of 72 samples of peripheral blood, apheresis packs, and cord blood were analysed and compared using the ISHAGE protocol with or without the addition of fluorescent microspheres. Comparison of methods showed a high correlation coefficient (r = 0.99), with no statistically significant difference or bias between methods (P > 0.05). Linearity of the absolute counting method generated an R² value of 1.00 over the range of 0–250/μl. Precision of the absolute counting method measured at three concentrations of CD34+-stabilised KG1a cells (Stem-Trol, COULTER®) generated a coefficient of variation (C.V.) ranging from 4% to 9.9%. In a further modification of the single-platform method, the viability dye 7-amino actinomycin D was included and demonstrated that both viable and nonviable CD34+ cells could be identified and quantitated. Together, these modifications combine the accuracy and sensitivity of the original ISHAGE method with the ability to produce an absolute count of viable CD34+ cells. It is the accurate determination of this value that is most clinically relevant in the transplant setting. These modifications may improve the interlaboratory reproducibility of CD34 determinations due to the reduction in sample handling and calculation of results. Cytometry (Comm. Clin. Cytometry) 34:61–70, 1998. © 1998 Wiley-Liss, Inc.
Article
Full-text available
CXCR4 receptor expression is required for the retention of granulocyte precursors and mature neutrophils within the bone marrow, and disruption of the SDF-1/CXCR4 axis in the bone marrow results in the mobilization of myeloid lineage cells to the peripheral circulation. We report that G-CSF down-regulates CXCR4 expression in bone marrow-derived murine and human myeloid lineage cells. When exposed to G-CSF, murine Gr1(+) bone marrow myeloid cells display a time-dependent reduction of cell-surface CXCR4 and respond poorly to SDF-1 in attachment and migration assays. Bone marrow-derived cells of nonmyeloid lineage display no change in surface CXCR4 expression upon exposure to G-CSF. Compared with controls, mice treated with G-CSF for mobilization of hematopoietic progenitor cells display reduced levels of CXCR4 selectively in bone marrow Gr1(+) myeloid cells. Since bone marrow myeloid cells express G-CSF receptors and G-CSF rapidly reduces CXCR4 expression in purified Gr1(+) cells populations, these results provide evidence that G-CSF acts directly on myeloid lineage cells to reduce CXCR4 expression. By down-regulating CXCR4 expression in bone marrow myeloid cells and attenuating their responsiveness to SDF-1, G-CSF promotes their mobilization from the bone marrow to the peripheral blood.
Article
We compared the biological effects of the CXC chemokine SDF-1α on immunomagnetically purified CD34+ cells isolated from human normal bone marrow (NBM), leukapheresis products (LP) and patients with chronic myeloid leukaemia (CML). LP CD34+ cells showed a significantly stronger migration response to SDF-1α (100 ng/ml) than CD34+ cells isolated from the peripheral blood (PB) of CML patients (P 0.05). Finally, no differences in the modulation of CXCR-4 levels in response to SDF-1α and serum were observed in CML and normal CD34+ cells. Our data suggest that the impaired chemotactic response of CML CD34+ cells to SDF-1α is not caused by a lack or complete uncoupling of CXCR-4, but may be due to an intracellular signalling defect downstream of the receptor. Leukemia (2000) 14, 1652–1660.
Article
THE chemokines are a large family of small, structurally related cytokines1,2. The physiological importance of most members of this family has yet to be elucidated, although some are inducible inflammatory mediators that determine leukocyte chemotaxis1–5. Pre-B-cell growth-stimulating factor/stromal cell-derived factor-1 (PBSF/SDF-1) is a member of the CXC group of chemokines6,7. PBSF/SDF-1 stimulates proliferation of B-cell progenitors in vitro 6 and is constitutively expressed in bone-marrow-derived stromal cells6,7. Here we investigate the physiological roles of PBSF/SDF-1 by generating mutant mice with a targeted disruption of the gene encoding PBSF/SDF-1. We found that mice lacking PBSF/SDF-1 died perinatally and that although the numbers of B-cell progenitors in mutant embryos were severely reduced in fetal liver and bone marrow, myeloid progenitors were reduced only in the bone marrow but not in the fetal liver, indicating that PBSF/SDF-1 is responsible for B-cell lymphopoiesis and bone-marrow myelopoiesis. In addition, the mutants had a cardiac ventricular septal defect. Hence, we have shown that the chemokine PBSF/SDF-1 has several essential functions in development.
Article
The purpose of this work was to develop a definition of myelofibrosis with myeloid metaplasia (MMM) using diagnostic criteria that would remain valid within the set of patients with chronic myeloproliferative disorders or myelodysplastic syndromes. A list of 12 names for the disease and 37 diagnostic criteria were proposed to a Consensus Panel of 12 Italian experts who ranked them in order so as to identify a core set of criteria. The Panel was then asked to score the diagnosis of 46 patient profiles as appropriate or not appropriate for MMM. Using the experts' consensus as the gold standard, the performance of 90 possible definitions of the disease obtained through the core set was evaluated. ‘Myelofibrosis with myeloid metaplasia’ ranked as the preferred name of the disease. Necessary criteria consisted of ‘diffuse bone marrow fibrosis’ and ‘absence of Philadelphia chromosome or BCR-ABL rearrangement in peripheral blood cells’. The six optional criteria in the core set consisted of: splenomegaly of any grade; anisopoikilocytosis with tear-drop erythrocytes; the presence of circulating immature myeloid cells; the presence of circulating erythroblasts; the presence of clusters of megakaryoblasts and anomalous megakaryocytes in bone marrow sections; myeloid metaplasia. The definition of the disease with the highest final score was as follows: necessary criteria plus any other two criteria when splenomegaly is present or any four when splenomegaly is absent. The use of this definition will help to standardize the conduct and reporting of clinical studies and should help practitioners in clinical practice.
Article
We studied the survival of 195 patients with agnogenic myeloid metaplasia (AMM) diagnosed between 1962 and 1992 in an attempt to stratify patients into risk groups. Median survival was 42 months. Adverse prognostic factors for survival were age > 60 years, hepatomegaly, weight loss, low hemoglobin level (Hb), low or very high leukocyte count (WBC), high percentage of circulating blasts, male sex, and low platelet count. A new scoring system based on two adverse prognostic factors, namely Hb < 10 g/dL and WBC < 4 or > 30 x 10(9)/L, was able to separate patients in three groups with low (0 factor), intermediate (1 factor), and high (2 factors) risks, associated with a median survival of 93, 26, and 13 months, respectively. An abnormal karyotype (32 cases of 94 tested patients) was associated with a short survival, especially in the low-risk group (median survival of 50 v 112 months in patients with normal karyotype). The prognostic factors for acute conversion were WBC > 30 x 10(9)/L and abnormal karyotype. Thus, hemoglobin level and leukocyte count provide a simple prognostic model for survival in AMM, and the adverse prognostic value of abnormal karyotype may be related to a higher rate of acute conversion.