Original Article Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Stem Cells. Oct 26, 2013; 5(4): 196-204
Published online Oct 26, 2013. doi: 10.4252/wjsc.v5.i4.196
Mobilization of CD34+CD38- hematopoietic stem cells after priming in acute myeloid leukemia
Adriana Plesa, Eve Mattei, Charles Dumontet, Laboratoire de Cytologie et d’Immunologie, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, 69495 Pierre Bénite, France
Youcef Chelghoum, Hélène Labussière, Giovanna Cannas, Mauricette Michallet, Xavier Thomas, Department of Hematology, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, 69495 Pierre Bénite, France
Mohamed Elhamri, Stéphane Morisset, Unité de Recherche Clinique, Service d’Hématologie, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, 69495 Pierre Bénite, France
Inès Tagoug, INSERM U590, Laboratoire de Cytologie Analytique, Faculté de Médecine Rockefeller, Université Lyon-I, 69495 Lyon, France
Author contributions: Plesa A was responsible for co-ordinating immunophenotyping data and participated in the analysis and interpretation of data; Mattei E and Tagoug I participated in the collection of immunophenotyping data; Morisset S performed statistical analyses; Chelghoum Y, Labussière H and Cannas G included patients and collected samples; Elhamri M collected the data and provided technical support; Michallet M and Dumontet C reviewed the manuscript and gave final approval; Thomas X included patients, collected samples, interpreted the data and wrote the manuscript.
Correspondence to: Xavier Thomas, MD, PhD, Department of Hematology, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Pavillon Marcel Bérard, Bât.1G, 165 chemin du Grand Revoyet, 69495 Pierre Bénite, France. xavier.thomas@chu-lyon.fr
Telephone: +33-478862235 Fax: +33-472678880
Received: May 2, 2013
Revised: June 23, 2013
Accepted: July 30, 2013
Published online: October 26, 2013
Processing time: 199 Days and 18 Hours

Abstract

AIM: To evaluate quantitatively and qualitatively the different CD34+ cell subsets after priming by chemotherapy granulocyte colony-stimulating factor (± G-CSF) in patients with acute myeloid leukemia.

METHODS: Peripheral blood and bone marrow samples were harvested in 8 acute myeloid leukemia patients during and after induction chemotherapy. The CD34/CD38 cell profile was analyzed by multi-parameter flow cytometry. Adhesion profile was made using CXC chemokine receptor 4 (CXCR4) (CD184), VLA-4 (CD49d/CD29) and CD47.

RESULTS: Chemotherapy ± G-CSF mobilized immature cells (CD34+CD38 population), while the more mature cells (CD34+CD38low and CD34+CD38+ populations) decreased progressively after treatment. Circulating CD34+ cells tended to be more sensitive to chemotherapy after priming with G-CSF. CD34+ cell mobilization was correlated with a gradual increase in CXCR4 and CD47 expression, suggesting a role in cell protection and the capacity of homing back to the marrow.

CONCLUSION: Chemotherapy ± G-CSF mobilizes into the circulation CD34+ bone marrow cells, of which, the immature CD34+CD38 cell population. Further manipulations of these interactions may be a means with which to control the trafficking of leukemia stem cells to improve patients’ outcomes.

Key Words: Acute myeloid leukemia, Leukemia stem cell, Immunophenotype, Priming, Timed sequential chemotherapy

Core tip: Timed sequential chemotherapy and priming with hematopoietic growth factors have been recently used in the treatment of acute myeloid leukemia in order to mobilize more leukemic cells in the cell cycle and therefore improve the cytotoxic effect of chemotherapy. In this paper, we looked the impact of this type of treatment in a small series of patients on the mobilization of different subsets of CD34+ cells involving “bulk” leukemic cells and more ‘‘immature’’ leukemic cells.



INTRODUCTION

Acute myeloid leukemia (AML) is a heterogeneous clonal disorder that originates from leukemia stem cells (LSCs) with the ability to generate an excessive amount of malignant myeloid blasts. Despite therapeutic advances in younger adults, relapses remain a major issue[1]. Chemotherapeutic regimens markedly reduce tumor burden, but only target the ‘‘bulk’’, non-clonogenic cells and spare LSCs, which allow for recrudescence of leukemia[2]. Interactions of the leukemic cells with the bone marrow (BM) microenvironment via specific receptor and adhesion molecules, such as the CXC chemokine receptor 4 (CXCR4)-stromal cell-derived factor-1 (SDF-1) axis, are in part responsible for chemotherapy resistance. AMLs with high CXCR4 cell surface expression and therefore a high tendency of stromal protection have been shown to have a poor prognosis[3,4].

The cell cycle is a critical regulator of the processes of cell proliferation and growth. One strategy to increase the cytotoxicity of cycle-dependent antileukemic agents is to enter more leukemic cells into the cell cycle. Timed sequential chemotherapy (TSC) is based on the findings that the initial cytoreductive drug induces the remaining malignant cell cohort to enter a proliferative state at a predictable time following drug administration[5]. Combination with hematopoietic growth factors (HGFs) has also been developed to enhance the efficacy of cytotoxic agents[6,7]. Granulocyte colony-stimulating factor (G-CSF) stimulates very immature progenitors and cleaves SDF-1[8], inducing stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating its receptor CXCR4[9]. Such an approach could theoretically target the self-renewal machinery of LSCs by inducing the quiescent LSCs into the cycle and circulation. However, a better understanding of those mechanisms is warranted.

Basically, low numbers of immature hematopoietic cells are released into the peripheral blood (PB). Treatments, including HGFs and/or TSC DNA-damaging agents, known as priming, can cause a marked increase in hematopoietic stem cell mobilization[10]. The aim of the present study was to investigate BM and PB leukemic blasts in the CD34 versus CD38 bidimensional space in order to evaluate quantitatively and qualitatively the different CD34+ cell subsets during and after priming by TSC ± G-CSF as induction chemotherapy in adult AML patients.

MATERIALS AND METHODS
Patients and treatments

PB and BM samples were procured from a total of 8 AML patients (Table 1). All studied patients were treated according to the Acute Leukemia French Association (ALFA)-0702 trial[11]. Diagnosis was morphologically proven according to the French-American-British classification[12]. The study protocol was approved by the Human Ethics Committee of our institution and was conducted in accordance with the Declaration of Helsinki. All patients gave written informed consent prior to registration on the study. This trial was registered at http://www.clinicaltrials.gov as No. NCT00932412. All patients received TSC induction and 5 of them were also primed by G-CSF (filgrastim). Morphological complete remission (CR)[13] and risk classification, based on cytogenetics and molecular marker analyses, were defined as previously described[7,14].

Table 1 Characteristics at diagnosis and outcome of the 8 acute myeloid leukemia patients.
PtsAge(yr)SexCytogeneticsCD34+(%)1FABMolecular Biology2WBC(x 109/l)2Risk-groupG-CSFprimingBM 15 dCRConsolidationand DFS
159MNormal85M5Flt3-ITD neg CEBPA pos55.1Favorable intermediateYesNo blastYesChemotherapy 15+ mo
235MNormal85M2Flt3-ITD neg CEBPA pos4.8Favorable intermediateYesNo blastYesChemotherapy 16+ mo
356F+8, del (20) (q11; q13)93M1Flt3-ITD pos12.6Poor intermediateYesNDYesChemotherapy 3 mo
419MNormal67M6Flt3-ITD pos MLL pos0.9UnfavorableYesNDYesAlloSCT 6 mo
555MNormal8M1Flt3-ITD pos NPM1 pos147.3Poor intermediateYesNDYesAlloSCT 14+ mo
655FNormal3M5Flt3-ITD neg Evi-1 pos3.7UnfavorableNo5% blastsYesAlloSCT 14+ mo
744MComplex94M0Flt3-ITD neg NPM1 neg1.4UnfavorableNo> 5% blastsNo3AlloSCT 11 mo
845F–7, +2183M4Flt3-ITD neg NPM1 neg2.4UnfavorableNoNo blastYesAlloSCT11+ mo
AML cells and flow cytometry analyses

PB and BM samples were harvested in all patients at different times: at diagnosis (T0), at the end of the first sequence of chemotherapy (4 d) (T4), at the beginning of the second sequence of chemotherapy (8 d) (T8), at the end of chemotherapy (10 d) (T10), during aplasia (15 d) (T15), and at the time of cell recovery (between 28 and 35 d) (TR).

Surface and intracellular antigen detection were performed by multi-parameter flow cytometry (MFC). Briefly, the CD34/CD38 cell profile was analyzed in one single tube by a multi-parameter combination using CD7, CD13, CD33, CD34, CD38, CD45 and CD19. Analyses were performed, using FACS Diva software (BD Bioscience). Instrument set-up was routinely optimized by analyzing Calibrite beads- Rainbows 8 picks beads and CST beads system. Adhesion profile was made using CXCR4 (CD184), VLA-4 (CD49d/CD29) and CD47. Cells were incubated with the appropriate combination of MoAbs (1 × 106 total cells per tube), washed and then analyzed by flow cytometry. The required minimal number of CD34+ events was set at 20 and the total number of events ranged from 100000 to 500000. Immunoglobulin G (IgG) isotype staining was used as a negative control for both CD38 and adhesion markers expression for ratio median fluorescence intensity (rMFI) evaluation. Discriminating the apoptotic/necrotic status of each subpopulation was made possible by using fluorescein isothiocyanate (FITC)-labelled annexin V and propidium iodide (PI), as previously described[15].

The gating strategy was based on CD45low/SSC total blast and CD34+CD45low cell populations gated from total FSC/SSC viable cells. Three populations of CD34+ cells were distinguished based on differential expression of the CD34 and CD38 antigens[16]: A first cell population, which expressed a great amount of the CD34 antigen, lack of CD38 (CD34+CD38) and often contained very few events requiring to be tightly clustered in a FSC/SSC and CD45/SSC plot; a second population characterized by a great amount of the CD34 antigen and by a low density of CD38 antigen (CD34+CD38low); and a third population characterized by a large density of CD38 and CD34 antigens (CD34+CD38+). Figures 1 and 2 are typical examples of an analysis. CD38 was expressed as percent positively stained cells within CD34+ populations as well as intensity of fluorescence signal quantified as rMFICD38 from CD34+ gated cells and from CD45low/SSC total blast cells. The adhesion markers CXCR4 and VLA4 were also expressed as rMFI. Regarding CD47 expression, a normalized rMFI was used and defined as followed: (median expression of CD47 in blasts-median expression of IgG1 in blasts)/(median expression of CD47 in lymphocytes-median expression of IgG1 in lymphocytes).

Figure 1
Figure 1 Multicolor flow cytometry (patient # 3): (left) Strategy used for gating the blast cell population and CD34+CD38 cell subpopulations (red: immature CD45low/SSC blast cells, blue: CD34+ cells, green: lymphocytes, blue cyan: monocytes, violet: granulocytes, black: CD45+ cells (erythroblasts); CD34+ cells were separated into different stem cell fractions based on their CD38 antigen expression: A first cell population expressing a great amount of the CD34 antigen and lacked of CD38 (CD34+CD38-), a second cell population characterized by a great amount of the CD34 antigen and by a low density of CD38 antigen (CD34+CD38low/), and a third cell population characterized by a large density of CD38 antigen and of CD34 antigen (CD34+CD38+); (right) The two density plots indicate CD45low/SSC blast cells (in red) and CD34+CD38 subpopulations (CD34+CD38- in orange, CD34+CD38low in green and CD34+CD38+ in blue).
Figure 2
Figure 2 Characterization of the different stem cell fractions. A typical example of analysis (patient 3) performed at different times: at diagnosis (T0), at the end of the first sequence of chemotherapy (4 d) (T4), at the beginning of the second sequence of chemotherapy (8 d) (T8), at the end of chemotherapy (10 d) (T10), during aplasia (15 d) (T15), and at the time of cell recovery (TR). CD34+ cells were separated into different stem cell fractions based on their CD38 antigen expression: A first cell population expressing a great amount of the CD34 antigen and lack of CD38 (CD34+CD38-); a second cell population characterized by a great amount of the CD34 antigen and by a low density of CD38 antigen (CD34+CD38low); and a third cell population characterized by a large density of CD38 antigen and of CD34 antigen (CD34+CD38+).
Statistical analysis

Descriptive statistical values included median, mean ± SD and range. For most of the analyses, BM cell samples from the 8 patients were pooled as well as all PB cell samples from the same 8 patients. Statistical significance was assessed using the ANOVA test, assuming equal variance for comparison of 2 groups. Correlations between the expressions of cell surface antigens were given by the linear Pearson test.

RESULTS
Expression of CD34 during and after chemotherapy

At T0, the average proportion of CD34+ cells in BM was 32.06% ± 28.96% (median, 21.47%) and 15.33% ± 23.89% (2.84%) in PB. At T0, most CD34+ cells corresponded to the “bulk” leukemia cell population. The CD34+ cell population decreased progressively in BM (4.6% ± 9.54%; 0.31% at T15) corresponding to the decrease in leukemic blasts after chemotherapy, and increased again lightly at the time of cell recovery (1.25% ± 0.84; 1.22%), corresponding mainly to the emergence of normal progenitor cells. In PB, a nadir of CD34+ cells was observed at T15 (0.51% ± 0.87%; 0.15%), while the percentage of CD34+ cells remained low at TR (0.10% ± 0.07%; 0.16%), confirming undetectable levels of immature cells in PB.

Expression of CD38 antigen on CD34+ cells

Three populations of CD34+ cells were distinguished on differential expression of the CD38 antigen. The CD34+CD38 population appeared in a restricted light-scattering region. When considering CD34 and CD45 expressions, these cells projected initially mainly in the CD34+CD45low/+, and then appeared in the CD34+CD45−/low area after cell mobilization. The CD34+CD38low cells were mainly found in the CD34+CD45low/+ area, but also involved the CD34+CD45−/low area. With respect to light-scattering properties, the cell population indicated by CD34+CD38+ was more heterogeneous but projected almost exclusively in the CD34+CD45low/+ area. At T0, the smallest population (CD34+CD38) lacked the CD38 antigen, while the larger population (CD34+CD38+) expressed the greatest amount of the CD38 antigen. The average proportions of the CD34+CD38, CD34+CD38low and CD34+CD38+ populations in BM and PB were 3.48% ± 5.66%, 12.62% ± 7.31%, 84.65% ± 11.91%, 15.40% ± 19.21%, 32.35% ± 20.41% and 53.81% ± 35.37%, respectively. Chemotherapy ± priming with G-CSF mobilized immature cells (CD34+CD38 population), while the more mature cells expressing CD38 (CD34+CD38low and CD34+CD38+ populations) decreased progressively after treatment. At T15, the average proportions of the CD34+CD38, CD34+CD38low and CD34+CD38+ cell populations in PB were 66.42% ± 20.18%, 27.33% ± 9.26%, and 8.05% ± 12.24%, respectively. In BM, the CD34+CD38 and CD34+CD38low populations were increased at T15, while the CD34+CD38+ population was decreased: 22.02% ± 17.11%, 48.96% ± 16.20%, and 29.91% ± 26.69%, respectively. Proportions of CD34CD38 cell fractions returned to baseline at TR (Table 2).

Table 2 Circulating CD34+ cell subsets before, during and after intensive chemotherapy.
TimeCD34 vs CD38
CD34+CD38-CD34+CD38lowCD34+CD38+
At diagnosis (T0)
Median7.32%35.45%56.74%
Mean ± SD15.40% ± 19.21%32.35% ± 20.41%53.81% ± 35.37%
At 4 d (T4)
Median12.82%39.15%37.30%
Mean ± SD21.17% ± 22.68%34.01% ± 23.08%45.11% ± 37.48%
At 8 d (T8)
Median15.99%26.21%36.81%
Mean ± SD30.14% ± 29.51%30.28% ± 20.87%39.06% ± 32.98%
At 10 d (T10)
Median41.35%22.99%18.90%
Mean ± SD48.18% ± 26.79%27.86% ± 18.65%24.29% ± 22.73%
At 15 d (T15)
Median73.56%26.18%3.93%
Mean ± SD66.42% ± 20.18%27.33% ± 9.26%8.05% ± 12.24%
At recovery (TR)
Median5.37%32.76%63.42%
Mean ± SD10.63% ± 13.81%29.53% ± 11.23%61.79% ± 18.39%
Effect of priming with G-CSF on CD34+ cell populations

CD34+ cells in PB tended to be more sensitive to chemotherapy after priming with G-CSF (mean ± SD: 0.21% ± 0.18%; median: 0.17% at T10) than without G-CSF (5.17% ± 8.47%; 0.31%). The evolution of the three subsets of CD34+ cell populations over time were confirmed when considering the absolute number of cells. The CD34+CD38+/low cell population, containing the leukemia bulky cell population, followed the evolution of white blood cell count with a massive decrease during and after chemotherapy corresponding to cell lysis and a slight increase at the time of evaluation corresponding to cell recovery. At T15, the absolute number of the more mature cell population (CD34+CD38+) tended to be lower after priming with G-CSF (5.1 × 106/l ± 4.1) than without G-CSF (19.3 × 106/l ± 15.2), suggesting a lytic impact of priming on leukemic cells. Reversely, the most immature subset (CD34+CD38) increased progressively during and after chemotherapy corresponding to mobilization with the highest absolute count for the CD34+CD38 cell subset in PB at T15 (179.4 × 106/l ± 79.7) and decreased thereafter suggesting partial lysis, migration to BM and/or differentiation through the CD34+ CD38+ cell pool. When considering the CD34+CD38 population, the increase in cell percentage between T0 and T15 was 24 fold higher after priming with G-CSF than without priming with G-CSF, suggesting a higher level of cell mobilization.

Expression of CXCR4 and adhesion molecules on CD34+ cell subsets

Figure 3 summarizes the evolution of CXCR4 (Figure 3A), CD47 (Figure 3B) and VLA-4 (Figure 3C) rMFI on CD34+ cells and on CD34+ CD38 cell sub-populations in BM and PB during the induction course. CXCR4 and CD47 rMFI on CD34+ BM and PB cells increased with chemotherapy ± priming by G-CSF, while VLA-4 remained stable. The evolution of CXCR4 and CD47 expression was correlated in both BM (r = 0.64) and PB (r = 0.55). From T0 to T15, CD47 expression increased 10 fold more on PB CD34+ cells than on BM cells with a mean expression of 280.2 vs 43.6, while increased expression of CXCR4 was similar. When considering CD34+ cell subsets according to CD38 expression, CD47 significantly increased at T15 mainly in PB for the CD34+CD38 and CD34+CD38low populations, while mean rMFI did not change for the CD34+CD38+ population. CXCR4 significantly increased at T15 in both PB and BM mainly for the CD34+CD38low cell population, while mean rMFI did not change for the CD34+CD38 and CD34+CD38+ populations.

Figure 3
Figure 3 Evolution of CXCR4, CD47, and VLA-4 ratio median fluorescence intensity on CD34+ cells and CD34+CD38 cell sub-populations in bone marrow and peripheral blood. A: CXCR4, B: CD47, C: VLA-4. PB: Peripheral blood; BM: Bone marrow; rMFI: Ratio median fluorescence intensity.
Identification of apoptotic and necrotic cells

The fluorescence parameters allowed characterization of necrotic (PI+ annexin V-FITC+ cells), apoptotic (PI annexin V-FITC+ cells) and viable cells (PI annexin V-FITC cells) (Table 3). The evolution over time of mean proportions of apoptotic and necrotic cells was similar in BM and PB for all cell subsets: a decrease of viable cells was noted after chemotherapy, while a massive increase in apoptotic and necrotic cell populations was observed (Figure 4). However, the percentage of cells with apoptotic/necrotic status tended to be higher in PB. At T15, the percentage of necrotic cells in both PB and BM was higher in the more mature cell populations (CD34+CD38lowand CD34+CD38+) than in the immature CD34+CD38 population. However, survival tended to be promoted by G-CSF, as indicated by a decrease of annexin V-FITC+ cells in patients primed with G-CSF. The percentage of apoptotic cells did not differ among patients primed with G-CSF and those not primed: 51% ± 21% vs 48% ± 21% in the whole CD34+ cell population. Although not statistically significant, the percentage of necrotic cells tended to be lower in patients primed with G-CSF: 12% ± 6% vs 27% ± 21% in the whole CD34+ cell population (P = 0.2).

Table 3 Mean percentage of viable, apoptotic and necrotic cells in peripheral blood and bone marrow samples at T0, T15 and time of cell recovery.
Cell populationT0 (BM)T0 (PB)T15 (BM)T15 (PB)TR (BM)TR (PB)
Immature cells
Viable cells75.5 ± 17.652.0 ± 21.218.5 ± 3.514.5 ± 12.037.0 ± 16.934.0 ± 14.1
Apoptotic cells17.5 ± 17.619.5 ± 21.933.0 ± 7.027.5 ± 23.334.5 ± 13.424.5 ± 7.7
Necrotic cells3.0 ± 1.428.0 ± 0.045.0 ± 11.354.0 ± 38.125.0 ± 2.838.5 ± 24.7
CD34+ cells
Viable cells77.5 ± 20.559.5 ± 33.234.0 ± 19.724.5 ± 3.565.0 ± 25.429.0 ± 19.7
Apoptotic cells20.5 ± 20.534.5 ± 28.931.0 ± 29.638.0 ± 15.532.0 ± 22.644.0 ± 1.4
Necrotic cells2.5 ± 0.75.0 ± 2.835.0 ± 8.434.0 ± 21.23.0 ± 2.824.0 ± 19.7
CD34+CD38- population
Viable cells68.0 ± 19.759.5 ± 34.652.5 ± 24.721.5 ± 10.65132.5 ± 30.4
Apoptotic cells27.5 ± 24.738.0 ± 33.933.5 ± 23.360.0 ± 2.84752.5 ± 16.2
Necrotic cells0.5 ± 0.72.0 ± 1.413.5 ± 2.118.0 ± 14.1122.5 ± 0.7
CD34+CD38low population
Viable cells72.5 ± 26.148.0 ± 43.815.5 ± 6.315.0 ± 7.033.5 ± 2.130
Apoptotic cells25.5 ± 26.145.0 ± 38.134.5 ± 28.938.5 ± 20.554.0 ± 5.649
Necrotic cells2.0 ± 1.47.0 ± 5.649.5 ± 33.246.5 ± 27.512.5 ± 3.518
CD34+CD38+ population
Viable cells78.0 ± 19.770.5 ± 30.421.5 ± 7.727.5 ± 6.367.0 ± 25.444.5 ± 36.0
Apoptotic cells19.0 ± 19.723.5 ± 27.526.0 ± 26.820.5 ± 20.531.0 ± 24.030.5 ± 0.7
Necrotic cells2.0 ± 0.05.0 ± 2.853.0 ± 18.342.0 ± 43.81.5 ± 0.728.0 ± 39.5
Figure 4
Figure 4 Kinetics of viable, apoptotic and necrotic cells in peripheral blood according to the different times of study (patient 8). Values were given comparatively to those observed at the time of diagnosis, assuming that the observed value at T0 corresponded to 100%.
DISCUSSION

The pursuit of the best chemotherapy regimen for AML continues in an attempt to improve CR proportions and long-term survival. The LSC model has implications for the development of new therapeutic strategies. LSCs respond to depletion of the leukemia cell mass that occurs when anti-proliferative drugs are administered. It is therefore suggested that one way to eliminate dormant LSCs is to find the window in which they cycle and then kill them[17]. Priming may modulate cell cycle kinetics of AML blasts and render them more susceptible to phase-specific agents. This has been shown in vitro[18] and in vivo in leukemic mice[19], and more recently in clinical trials for younger adults with AML[6,7].

Our study showed that CD34+ cells were significantly affected by chemotherapy in both BM and PB. However, treatment mainly impacted on the more mature cell populations, as demonstrated by the decrease of the CD34+CD38+/low cell population corresponding to “bulk’’ leukemic blasts. LSCs represent only a small fraction of malignant cells[20] and are believed to be restricted to the LinCD34+CD38 fraction[21,22], which was insignificant at T0 in most of our patients. However, this cell subset was able to proliferate and to egress from BM to PB after chemotherapy ± G-CSF, and partly differentiates through the more mature CD34+CD38+/low or CD34+CD38+ cell phenotypes, before returning to its initial value after leukocyte recovery. However, LSCs can arise from the malignant transformation of a normal stem cell that has accumulated oncogenic insults over time, or from a more differentiated cell that develops the capability for continual self-renewal[23]. The phenotype of LSCs is therefore heterogeneous and can vary even within a single sample. Some AML have LSCs exclusively in the CD34 fraction[24]. Similarly, CD34+CD38+ fraction of certain AML samples contains all or at least most LSCs[25]. Furthermore, CD38 is reversibly expressed on CD34+ repopulating cells between negative and low levels[26]. Characteristics that are relevant to therapy may then differ based on the origin of the malignant cell with sensitive AMLs derived from more differentiated stem cells and resistant AMLs derived from earlier stages.

Resistance may in part be provoked by cell adherence to the stromal environment. Inhibition of the CXCR4-SDF-1 axis induces mobilization of cells into circulation and enhances anti-leukemic effects of chemotherapy[27]. CXCR4 plays a dominant role in cell-trafficking, as confirmed here by up-regulation after CD34+ cells egress to the circulation. This was observed for all PB CD34+ cells. However, the more mature CD34+ cells mobilized earlier than immature CD34+ cells, suggesting different degrees of sensitivity. CXCR4 expression favors the enrichment of a non-cycling population of AML cells, which represent dormant leukemia progenitors serving as a reservoir for minimal residual disease. G-CSF results in a decreased expression of SDF-1 in the BM, resulting in premature release of immature cells[9]. This was in accordance with our results showing an increased expression of CXCR4 on immature CD34+ cells after reaching the PB circulation. The peak number of labeled cells was shown after 72 h preventing an early homing back to the BM[28]. CXCR4 may contribute to immature cell clearance from the blood observed after T15 by directing cells again to the BM.

Many other proteins, including the VLA-4 integrins, are regarded as essential for AML cell adhesion to stromal cells and their protection from drug-induced apoptosis[29], but only slight variations in their expression level were noted during and after therapy, suggesting a minor involvement in immature CD34+ cell trafficking.

Mobilizing treatments cause CD47 to be transiently up-regulated on progenitors just prior to and during their migratory phase[30]. This was confirmed by our study, showing an increased expression of CD47 most particularly in the circulating CD34+CD38 cell subgroup after chemotherapy ± G-CSF. Overexpression of CD47 on AML cells is known to increase their pathogenicity by allowing them to evade macrophage phagocytosis[31,32]. This could explain the strong correlation found between the evolution of CD47 expression and that of CXCR4 on immature CD34+ cells.

Circulating CD34+ cells tended to be more sensitive to chemotherapy after priming with G-CSF. However, the impact of priming mainly concerned the more mature CD34+ cells containing “bulk” leukemic cells, while it was limited on immature CD34+ cells susceptible to contain leukemia-initiating cells. All CD34+ annexin V+ cell populations were involved in the apoptotic/necrotic process, which concerned a larger proportion of PB cells than BM cells. Cell survival seemed to be promoted by G-CSF, as indicated by a decrease in annexin V+ cells. At T15, the necrotic process involved preferentially the more mature cell subsets (CD34+CD38low and CD34+CD38+ populations), suggesting a higher protection of CD34+CD38 cells from the action of cytotoxic drugs.

Despite disappointing results regarding priming of the most immature CD34+ cells, our study represents a first step towards further explorations. Mobilization of LSCs is a concept that is presently being revisited by novel targeted therapies. Inhibition of CXCR4, based on the ability of the CXCR4 antagonist plerixafor to push LSCs out of their BM niches, induces the rapid mobilization of stem cells from BM to PB[33], and has been shown to sensitize leukemic blasts to chemotherapy[27,34-36]. A mobilization of up to 80% leukemic cells, including the more primitive CD34+CD38 cell subset, was observed when HGF was followed by anti-CXCR4[37]. Accompanied by chemotherapy, this might lead to a better eradication of LSCs. From that perspective, plerixafor is currently being tested by French centers for the mobilization of dormant LSCs. Consequently, new paradigms must be devised for evaluating the therapeutic agents. For instance, clinical trial design can use intermediate end points such as time to progression following the administration of an agent that can target LSCs.

COMMENTS
Background

Acute myeloid leukemia (AML) is a heterogeneous clonal disorder that originates from leukemia stem cells (LSCs) with the ability to generate an excessive amount of malignant myeloid blasts. Interactions of the leukemic cells with the bone marrow (BM) microenvironment via specific receptor and adhesion molecules, such as the CXC chemokine receptor 4 (CXCR4)-stromal cell-derived factor-1 (SDF-1) axis, are in part responsible for chemotherapy resistance. AMLs with high CXCR4 cell surface expression and therefore a high tendency of stromal protection have been shown to have a poor prognosis.

Research frontiers

The cell cycle is a critical regulator of the processes of cell proliferation and growth. One strategy to increase the cytotoxicity of cycle-dependent antileukemic agents is to enter more leukemic cells into cell cycle. Timed sequential chemotherapy is based on the findings that the initial cytoreductive drug induces the remaining malignant cell cohort to enter a proliferative state at a predictable time following drug administration. Combination with hematopoietic growth factors (HGFs) has also been developed to enhance the efficacy of cytotoxic agents.

Innovations and breakthroughs

The study represents a first step towards further explorations. Mobilization of LSCs is a concept that is presently being revisited by novel targeted therapies. Inhibition of CXCR4, based on the ability of the CXCR4 antagonist plerixafor to push LSCs out of their BM niches, induces the rapid mobilization of stem cells from BM to PB and has been shown to sensitize leukemic blasts to chemotherapy. A mobilization of up to 80% leukemic cells, including the more primitive CD34+CD38 cell subset, was observed when HGF was followed by anti-CXCR4. Accompanied by chemotherapy, this might lead to a better eradication of LSCs. From that perspective, plerixafor is currently being tested by French centers for the mobilization of dormant LSCs. Consequently, new paradigms must be devised for evaluating the therapeutic agents. For instance, clinical trial design can use intermediate end points such as time to progression following the administration of an agent that can target LSCs.

Applications

The study was to evaluate quantitatively and qualitatively the different CD34+ cell subsets after priming by chemotherapy and granulocyte colony-stimulating factor (± G-CSF) in patients with acute myeloid leukemia.

Peer review

The authors studied the mobilization of CD34+ bone marrow stem cells into the circulation in AML patients. The results show that CD34+ cells are more sensitive to chemotherapy after priming with GM-CSF associated with an increase in CXCR4 and CD47 expression.

Footnotes

P- Reviewers Bright JJ, Sloan AJ S- Editor Zhai HH L- Editor Roemmele A E- Editor Wu HL

References
1.  Döhner H, Estey EH, Amadori S, Appelbaum FR, Büchner T, Burnett AK, Dombret H, Fenaux P, Grimwade D, Larson RA. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010;115:453-474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2353]  [Cited by in F6Publishing: 2498]  [Article Influence: 178.4]  [Reference Citation Analysis (0)]
2.  Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105-111.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Rombouts EJ, Pavic B, Löwenberg B, Ploemacher RE. Relation between CXCR-4 expression, Flt3 mutations, and unfavorable prognosis of adult acute myeloid leukemia. Blood. 2004;104:550-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 219]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
4.  Konoplev S, Rassidakis GZ, Estey E, Kantarjian H, Liakou CI, Huang X, Xiao L, Andreeff M, Konopleva M, Medeiros LJ. Overexpression of CXCR4 predicts adverse overall and event-free survival in patients with unmutated FLT3 acute myeloid leukemia with normal karyotype. Cancer. 2007;109:1152-1156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 119]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
5.  Karp JE, Donehower RC, Enterline JP, Dole GB, Fox MG, Burke PJ. In vivo cell growth and pharmacologic determinants of clinical response in acute myelogenous leukemia. Blood. 1989;73:24-30.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Löwenberg B, van Putten W, Theobald M, Gmür J, Verdonck L, Sonneveld P, Fey M, Schouten H, de Greef G, Ferrant A. Effect of priming with granulocyte colony-stimulating factor on the outcome of chemotherapy for acute myeloid leukemia. N Engl J Med. 2003;349:743-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 297]  [Cited by in F6Publishing: 289]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
7.  Thomas X, Raffoux E, Renneville A, Pautas C, de Botton S, Terre C, Gardin C, Hayette S, Preudhomme C, Dombret H. Which AML subsets benefit from leukemic cell priming during chemotherapy? Long-term analysis of the ALFA-9802 GM-CSF study. Cancer. 2010;116:1725-1732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 21]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
8.  Carter BZ, Milella M, Altieri DC, Andreeff M. Cytokine-regulated expression of survivin in myeloid leukemia. Blood. 2001;97:2784-2790.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 146]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
9.  Petit I, Szyper-Kravitz M, Nagler A, Lahav M, Peled A, Habler L, Ponomaryov T, Taichman RS, Arenzana-Seisdedos F, Fujii N. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol. 2002;3:687-694.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  To LB, Haylock DN, Simmons PJ, Juttner CA. The biology and clinical uses of blood stem cells. Blood. 1997;89:2233-2258.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Thomas X, Raffoux E, Elhamri M, Lobe I, Cannas G, Dombret H. Clofarabine for the treatment of adult acute myeloid leukemia. Future Oncol. 2009;5:1197-1210.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR, Sultan C. Criteria for the diagnosis of acute leukemia of megakaryocyte lineage (M7). A report of the French-American-British Cooperative Group. Ann Intern Med. 1985;103:460-462.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Cheson BD, Bennett JM, Kopecky KJ, Büchner T, Willman CL, Estey EH, Schiffer CA, Doehner H, Tallman MS, Lister TA. Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. J Clin Oncol. 2003;21:4642-4649.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Thomas X, Elhamri M, Raffoux E, Renneville A, Pautas C, de Botton S, de Revel T, Reman O, Terré C, Gardin C. Comparison of high-dose cytarabine and timed-sequential chemotherapy as consolidation for younger adults with AML in first remission: the ALFA-9802 study. Blood. 2011;118:1754-1762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
15.  Vermes I, Haanen C, Steffens-Nakken H, Reutelingsperger C. A novel assay for apoptosis. Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V. J Immunol Methods. 1995;184:39-51.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Terstappen LW, Huang S, Safford M, Lansdorp PM, Loken MR. Sequential generations of hematopoietic colonies derived from single nonlineage-committed CD34+CD38- progenitor cells. Blood. 1991;77:1218-1227.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Guzman ML, Swiderski CF, Howard DS, Grimes BA, Rossi RM, Szilvassy SJ, Jordan CT. Preferential induction of apoptosis for primary human leukemic stem cells. Proc Natl Acad Sci. USA. 2002;99:16220-16225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 332]  [Cited by in F6Publishing: 313]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
18.  te Boekhorst PA, Löwenberg B, Vlastuin M, Sonneveld P. Enhanced chemosensitivity of clonogenic blasts from patients with acute myeloid leukemia by G-CSF, IL-3 or GM-CSF stimulation. Leukemia. 1993;7:1191-1198.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Ben-Ishay Z, Prindull G, Sharon S. Improved prognosis in mice with advanced myeloid leukemia following administration of GM-CSF and cytosine arabinoside. Leuk Res. 1991;15:321-325.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, Minden M, Paterson B, Caligiuri MA, Dick JE. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature. 1994;367:645-648.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997;3:730-737.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Ishikawa F, Yoshida S, Saito Y, Hijikata A, Kitamura H, Tanaka S, Nakamura R, Tanaka T, Tomiyama H, Saito N. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol. 2007;25:1315-1321.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Cozzio A, Passegué E, Ayton PM, Karsunky H, Cleary ML, Weissman IL. Similar MLL-associated leukemias arising from self-renewing stem cells and short-lived myeloid progenitors. Genes Dev. 2003;17:3029-3035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 507]  [Cited by in F6Publishing: 472]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
24.  Taussig DC, Vargaftig J, Miraki-Moud F, Griessinger E, Sharrock K, Luke T, Lillington D, Oakervee H, Cavenagh J, Agrawal SG. Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction. Blood. 2010;115:1976-1984.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 279]  [Article Influence: 19.9]  [Reference Citation Analysis (0)]
25.  Taussig DC, Miraki-Moud F, Anjos-Afonso F, Pearce DJ, Allen K, Ridler C, Lillington D, Oakervee H, Cavenagh J, Agrawal SG. Anti-CD38 antibody-mediated clearance of human repopulating cells masks the heterogeneity of leukemia-initiating cells. Blood. 2008;112:568-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 298]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
26.  McKenzie JL, Gan OI, Doedens M, Dick JE. Reversible cell surface expression of CD38 on CD34-positive human hematopoietic repopulating cells. Exp Hematol. 2007;35:1429-1436.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Zeng Z, Shi YX, Samudio IJ, Wang RY, Ling X, Frolova O, Levis M, Rubin JB, Negrin RR, Estey EH. Targeting the leukemia microenvironment by CXCR4 inhibition overcomes resistance to kinase inhibitors and chemotherapy in AML. Blood. 2009;113:6215-6224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 390]  [Cited by in F6Publishing: 389]  [Article Influence: 25.9]  [Reference Citation Analysis (0)]
28.  Eash KJ, Means JM, White DW, Link DC. CXCR4 is a key regulator of neutrophil release from the bone marrow under basal and stress granulopoiesis conditions. Blood. 2009;113:4711-4719.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 215]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
29.  Wright DE, Bowman EP, Wagers AJ, Butcher EC, Weissman IL. Hematopoietic stem cells are uniquely selective in their migratory response to chemokines. J Exp Med. 2002;195:1145-1154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 379]  [Cited by in F6Publishing: 357]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
30.  Jaiswal S, Jamieson CH, Pang WW, Park CY, Chao MP, Majeti R, Traver D, van Rooijen N, Weissman IL. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009;138:271-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 972]  [Cited by in F6Publishing: 1121]  [Article Influence: 74.7]  [Reference Citation Analysis (0)]
31.  Ritchie DS, Smyth MJ. A new therapeutic target for leukemia comes to the surface. Cell. 2009;138:226-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 9]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
32.  Majeti R, Chao MP, Alizadeh AA, Pang WW, Jaiswal S, Gibbs KD, van Rooijen N, Weissman IL. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138:286-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1015]  [Cited by in F6Publishing: 1223]  [Article Influence: 81.5]  [Reference Citation Analysis (0)]
33.  Broxmeyer HE, Kohli L, Kim CH, Lee Y, Mantel C, Cooper S, Hangoc G, Shaheen M, Li X, Clapp DW. Stromal cell-derived factor-1/CXCL12 directly enhances survival/antiapoptosis of myeloid progenitor cells through CXCR4 and G(alpha)i proteins and enhances engraftment of competitive, repopulating stem cells. J Leukoc Biol. 2003;73:630-638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 150]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
34.  Liesveld JL, Bechelli J, Rosell K, Lu C, Bridger G, Phillips G, Abboud CN. Effects of AMD3100 on transmigration and survival of acute myelogenous leukemia cells. Leuk Res. 2007;31:1553-1563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 56]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
35.  Juarez J, Dela Pena A, Baraz R, Hewson J, Khoo M, Cisterne A, Fricker S, Fujii N, Bradstock KF, Bendall LJ. CXCR4 antagonists mobilize childhood acute lymphoblastic leukemia cells into the peripheral blood and inhibit engraftment. Leukemia. 2007;21:1249-1257.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Nervi B, Ramirez P, Rettig MP, Uy GL, Holt MS, Ritchey JK, Prior JL, Piwnica-Worms D, Bridger G, Ley TJ. Chemosensitization of acute myeloid leukemia (AML) following mobilization by the CXCR4 antagonist AMD3100. Blood. 2009;113:6206-6214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 371]  [Cited by in F6Publishing: 393]  [Article Influence: 24.6]  [Reference Citation Analysis (0)]
37.  Andreeff M, Konoplev S, Wang RY, Zeng Z, McQueen T, Shi YX, Medeiros LJ, Estey E, McCarty JM, Elkins S. Massive mobilization of AML cells into circulation by disruption of leukemia/stroma cell interactions using CXCR4 antagonist AMD3100: First evidence in patients and potential for abolishing bone marrow microenvironment-mediated resistance. Blood. 2006;108 suppl1:171a.  [PubMed]  [DOI]  [Cited in This Article: ]