Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Jan 13.
Published in final edited form as: Curr Opin Hematol. 2008 Jul;15(4):285–292. doi: 10.1097/MOH.0b013e328302f43a

Peripheral blood stem cell mobilization: new regimens, new cells, where do we stand

Louis M Pelus 1
PMCID: PMC2806229  NIHMSID: NIHMS168557  PMID: 18536564

Abstract

Purpose of review

Granulocyte colony-stimulating factor-mobilized peripheral blood stem cells are widely used to reconstitute hematopoiesis; however, preclinical and clinical studies show that improvements to this mobilization can be achieved. We discuss the development of new mobilizing regimens and evaluation of new findings on mobilized stem cell populations that may improve the utility and convenience of peripheral blood stem cell transplant.

Recent findings

Chemokines and their receptors regulate leukocyte trafficking, and altering chemokine signaling pathways mobilizes stem cells. In recent trials, combination use of the chemokine (C-X-C motif) receptor 4 antagonist AMD3100 and granulocyte colony-stimulating factor mobilized more CD34+ cells in fewer days than granulocyte colony-stimulating factor alone and allowed more patients to proceed to autotransplant. In preclinical studies the chemokine GROβ synergizes with granulocyte colony-stimulating factor and when used alone or with granulocyte colony-stimulating factor mobilizes more primitive hematopoietic stem cells with less apoptosis, higher integrin activation, lower CD26 expression and enhanced marrow homing compared with granulocyte colony-stimulating factor. Hematopoietic stem cells mobilized by GROβ or AMD3100 demonstrate superior engraftment and contribution to chimerism in primary and secondary transplant studies in mice, and peripheral blood stem cells mobilized by AMD3100 and granulocyte colony-stimulating factor in patients demonstrate enhanced engraftment capabilities in immunodeficient mice.

Summary

Alternate regimens differentially mobilize stem cell populations with unique intrinsic properties with the potential to expand the utility of hematopoietic transplantation. Continued mechanistic evaluation will be critical to our understanding of mechanisms of mobilization and their use in regenerative medicine.

Keywords: hematopoietic transplantation, peripheral blood stem cell mobilization, stem cells

Introduction

Adult bone marrow is the primary source of stem cells that regenerate hematopoiesis. Bone marrow also harbors other stem and progenitor cells, including endothelial progenitor cells (EPCs) that can contribute to revascularization [1] and mesenchymal stem cells (MSCs), which can differentiate into adipocytes, chondrocytes and osteo-blasts [24]. Normally, most hematopoietic stem cells (HSCs), EPCs and MSCs reside in marrow, but HSCs and EPCs also circulate in peripheral blood at low frequency. Early studies [57] showed that hematopoietic progenitor cells (HPCs) were elevated in blood of patients recovering from chemotherapy, which led to the realization that HSCs and HPCs could be induced to exit bone marrow, a phenomenon called ‘mobilization’, and that these mobilized cells can be collected and will re-home to marrow and repopulate hematopoiesis. Currently, mobilized peripheral blood is the primary source of hematopoietic cells used for hematopoietic transplantation [8]. In this review, new mobilizing regimens and studies demonstrating differential mobilization of stem cell populations with different quantitative and qualitative traits, and their potential to improve convenience, efficiency and outcome of peripheral blood stem cell transplantation will be discussed.

Peripheral blood stem cell mobilization by granulocyte colony-stimulating factor: the gold standard

Multiple agents induce peripheral blood stem cell mobilization, each with different kinetics and efficiencies [911] (Table 1), which suggests multiple mechanisms of action or multiple interventions along a common pathway. Combinations of agents can be additive or synergistic, and can produce grafts with quantitative and/or qualitative differences. Granulocyte colony-stimulating factor (G-CSF) is the predominant mobilizer used clinically. G-CSF-mobilized peripheral blood stem cells (PBSCs) engraft better than bone marrow or umbilical cord blood [1215], with faster neutrophil and platelet recoveries, fewer platelet transfusions, faster lymphocyte reconstitution, fewer febrile episodes and lower regimen related mortality [12,1621]. Recovery of blood counts using chemotherapy-mobilized PBSC takes longer and febrile complications and transfusion requirements are higher than G-CSF-mobilized PBSC [22], and chemotherapy-based mobilization regimens are not viable for allogeneic transplantation. PBSC collection is less invasive than bone marrow harvest and avoids risks associated with general anesthesia. Target yields are usually achieved with 1–3 aphereses, which is adequate for human leukocyte antigen (HLA)-identical and matched-related transplant, but may be suboptimal in the haploidentical setting [23,24]. G-CSF-based mobilization is generally well tolerated, although rare cases of splenic rupture have been documented [25,26]. Most donors complete the procedure with collection of sufficient cells for transplantation. Although there is no clear minimum threshold of mobilized PBSC for successful transplantation, infusion below 2 × 106 CD34+ cells/kg may be associated with higher risk of graft failure. Higher CD34+ cell doses may negatively impact the rate of acute or chronic graft-versus-host disease (GVHD) [2729] but positively impact overall patient survival [3032]. Cellular subsets, particularly T cells and CD14+ monocytes, within the graft can impact outcome [30,33]. Overall, alterations in the quantitative and qualitative attributes of the PBSC graft can significantly impact engraftment and transplant outcome.

Table 1.

Peripheral blood stem cell mobilizing agents

Agent class Examples Time of maximal mobilization
Chemotherapy Cytoxan, 5-fluorouracil 1–3 Weeks
Growth factors G-CSF, GM-CSF, stem cell factor, thrombopoietin, erythropoietin, growth hormone, IL-3, IL-17, parathyroid hormone, VEGF, angiopoietin-1 4–6 Days
Antibodies Anti-VLA-4, anti-VCAM-1 1–2 Days
Polyanions Fucoidan, dextran sulfate 1–2 h
Chemokines/chemokine mimetics GROβ, KC (murine GRO), MIP1α, Met-SDF-1β, CTCE0021, CTCE0214 15min–2 h
Receptor antagonists/signaling pathway inhibitors AMD3100, Rho GTPase inhibitor, β2-agonists 1–6 h

G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor; IL-3, interleukin-3; IL-17, interleukin-17; VCAM, vascular cell adhesion molecule-1; VEGF, vascular endothelial growth factor; VLA-4, very late antigen-4.

Poor mobilization to G-CSF occurs in 25% of patients, particularly in those with lymphomas [34,35], multiple myeloma [36,37] and acute leukemia [3840], and in 10–20% of normal volunteers [41,42], requiring extended aphereses [43]. Poor mobilization is also seen in heavily treated cancer patients [44,45] and genetic disorders, such as Fanconi's anemia [46]. In a recent study [47••] of the feasibility of PBSC mobilization and autologous transplant in recovered aplastic anemia patients, PBSC collection using G-CSF was feasible in only a small percentage of patients. There is a broad individual variability in mobilization responsiveness of donors and patients to G-CSF [4850]. Expanded application of allogeneic transplantation, particularly to patients with malignant disease receiving matched-unrelated grafts, or to patients receiving tolerizing grafts for organ transplant or for treatment of genetic disorders that do not require lethal conditioning, will require larger doses of mobilized cells to overcome HLA barriers and provide durable engraftment, acceptable leukocyte recovery kinetics and low incidence of GVHD [23,24,51]. Thus despite the success of G-CSF, there are areas for improvement that can have significant clinical impact: specifically, multiple dosing, variable and suboptimal mobilization in some groups, number of aphereses, inability to predict optimal mobilization times and graft composition.

Alternative mobilization strategies

Clinical trials continue to affirm the use and benefit of G-CSF-mobilized PBSC. Longer acting G-CSF, Pegfilgrastim, may address the multidosing requirement of conventional G-CSF. In patients with multiple myeloma [52,53], aggressive lymphoma [54] and previously treated lymphoma patients [55,56], a single dose of Pegfilgrastim in combination with chemotherapy was as effective as G-CSF in mobilizing sufficient CD34+ cells for auto-transplant, with sustained engraftment and rapid absolute neutrophil count (ANC) recovery. In patients with multiple myeloma, Pegfilgrastim used alone was effective in mobilizing adequate PBSCs with efficacy and toxicities similar to a standard G-CSF regimen [57]. Similar to G-CSF, splenic rupture in a patient following Pegfilgrastim mobilization has been reported [58]. In the allogeneic setting, there is little information available for Pegfilgrastim mobilization. In one study in normal donors [59], 80% achieved target CD34+ cell counts in one apheresis with a single Pegfilgrastim dose, and increased leukocytosis was demonstrated in a pharmacokinetic/pharmacodynamic study in normal volunteers [60]. Although Pegfilgrastim studies are at early stages, it appears that it may address shortcomings of the multiday G-CSF regimen. Poor mobilization response in some patients and donors, however, remains an area of unmet medical need.

Other agents have been used to try to enhance the G-CSF-induced peripheral blood stem cell mobilization (PBSCM) response, including stem cell factor (SCF) and thrombopoietin, the respective ligands for the c-kit and c-mpl receptors expressed on HSCs and HPCs; however, lack of efficacy or toxicity has prevented their development [34,61,62]. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is approved for PBSCM; however, it mobilizes less CD34+ cells than G-CSF [22,63] and little overall benefit was seen in combination trials [37,64].

Chemokines and their receptors direct leukocyte movement, including HSCs [6567], and have been implicated in PBSCM. Macrophage inflammatory protein-1α (MIP1α/CCL3) mobilizes HPCs and short-term repopulating cells and shows greater than additive effect with GCSF [6870,71]. The chemokine (C-X-C motif) receptor 2 (CXCR2) ligands GROβ and GROβΔ4 [70,72,73] and interleukin-8 (IL-8) [74] mobilize HSCs and HPCs and synergize with G-CSF [70,72]. Stromal cell derived factor-1α [SDF-1α/chemokine (C-X-C motif) receptor 12 (CXCL12)] and its receptor chemokine (C-X-C motif) receptor 4 (CXCR4) are an important axis for movement into and retention in the marrow [75]. Mobilization is observed using SDF-1α peptide analogs, CTCE-0021 and CTCE-0214 [76,77], the CXCR4 antagonist AMD3100 [78,79] and the SDF1 analog Met-SDF-1β [80], suggesting that altering SDF-1/CXCR4 signaling produces the mobilization response, most likely by CXCR4 receptor down modulation [47••,77,80].

The bicyclam AMD3100 induces HSC and HPC mobilization in mice [78], dogs [81], monkeys [82] and humans [78,79,83,84] and synergizes with G-CSF [71,78,85, 86••,87]. In a phase II trial, AMD3100 and G-CSF mobilized more CD34+ cells than G-CSF in 84% of patients with multiple myeloma and non-Hodgkin's lymphoma with a higher yield of CD34+ cells [86••]. In two recent randomized placebo-controlled phase III trials, AMD3100 (plerixafor) and G-CSF mobilized more CD34+ cells in multiple myeloma [88] and non-Hodgkin's lymphoma [89] patients in fewer days and allowed more patients to proceed to successful hematopoietic transplantation. In a third trial, AMD3100 (plerixafor) and G-CSF successfully mobilized sufficient cells for transplant in a large proportion of patients who previously failed G-CSF mobilization with G-CSF [90].

The success of AMD3100 and G-CSF significantly addresses a number of areas of improvement in PBSCM; however, it still requires multiple dosing. It will be interesting to determine if AMD3100 can be successfully used in combination with single dosing with Pegfilgrastim or in combination with other chemokines or used as a standa-lone mobilizer, taking advantage of the rapid mobilization that is the hallmark of chemokine axis mobilization.

Not all mobilized hematopoietic stem cells are created equal

Stem cell number, cell cycle status [91,92], facilitating or accessory cell content [93,94] or intrinsic differences in homing or proliferative properties [67,95,96] affect the ultimate function of the HSC graft. The superiority of GCSF-mobilized PBSCs raises questions concerning stem cell quantity versus quality. Although total clonogenic cells in the PBSC graft often exceed that of marrow grafts [97,98], earlier engraftment has also been seen when equivalent numbers of stem/progenitor cells were transplanted, suggesting that stem cell quality may impact efficacy [99]. Differences in gene and protein expression patterns and function have been described, at least in vitro, but have not led to an understanding of the enhanced engraftment properties of G-CSF-mobilized PBSCs. Recently, however, differential homing of G-CSF-mobilized stem cells resulting from enhanced motility and low CD26 expression relative to bone marrow stem cells have been suggested as contributing factors for enhanced engraftment [100]. In addition, detailed analysis of PBSCs mobilized by alternative regimens has begun to shed significant light on the importance of the HSC populations mobilized.

In contrast to G-CSF, chemokine mobilization occurs rapidly. In mice, PBSC mobilization by MIP1α, CTCE0021 and AMD3100 is maximal within 60 min, while mobilization by GROβ is maximal at 15 min. Synergistic PBSCM is seen when MIP1α, CTCE0021, AMD3100 or GROβ are combined with G-CSF (Table 2) and combinations of chemokines alone without G-CSF are synergistic [71]. Even more dramatic synergy is seen using chemokine combinations and G-CSF [71]. In competitive transplant studies G-CSF, AMD3100 and GROβ-mobilized cells contribute significantly to donor repopulation, with GROβ-mobilized cells producing significantly higher chimerism than G-CSF or AMD3100-mobilized cells (Table 2) [71,101••]. Significantly higher chimerism is observed using GROβ and G-CSF or AMD3100 and G-CSF-mobilized cells. In general, the GROβ and GROβ plus G-CSF-mobilized grafts contain twice the number of Sca-1+, c-kit+, lineageneg (SKL) cells and three times the number of CD34neg SKL cells, compared with G-CSF mobilized grafts [101••], suggesting that increased HSC frequency contributes to superior performance. In secondary transplant studies that test for HSC self-renewal, one expects chimerism in the second host to be identical to that in the primary host, and is in fact what is observed for G-CSF-mobilized cells (Table 2). The marrow cells from mice receiving grafts mobilized by GROβ or AMD3100 alone, or GROβ or AMD3100 plus G-CSF, however, show significantly higher contribution to chimerism than observed in the primary transplant (Table 2), suggesting chemokine-mobilized HSCs are inherently more competitive than bone marrow or G-CSF-mobilized HSCs [71,78,101••]. The enhanced engraftment properties of GROβ-mobilized PBSCs are also apparent in that recipients recover white blood cell counts faster than G-CSF-mobilized PBSCs [72,73,101••].

Table 2.

Quantitative versus qualitative peripheral blood stem cell mobilization

Mobilization fold increase over baseline
% Donor chimerisma
Mobilizer CFU-GM SKL Primary Secondary
G-CSFb 24 ± 5 12 ± 2 18 ± 2 23 ± 6
GROβc 12 ± 2 18 ± 3 38 ± 3 58 ± 5
AMD3100c 7 ± 2 6 ± 1 19 ± 2 51 ± 3
MIP1αc 3 ± 1
CTCE0021c 14 ± 4 5 ± 2
GROβ + G-CSFd 141 ± 12 54 ± 6 53 ± 6 85 ± 6
AMD3100 + G-CSFd 51 ± 5 63 ± 2 50 ± 2 84 ± 3
MIP1α + G-CSFd 39 ± 6
CTCE0021 + G-CSFd 123 ± 14

CFU-GM, colony-forming units granulocyte-macrophage; G-CSF, granulocyte colony-stimulating factor; MIP1α, macrophage inflammatory protein-1α; SKL, Sca-1+, c-kit+, lineage neg cells.

a

Competitive transplantation was performed comparing mobilized PBSCs to congenic bone marrow. In primary transplanted mice, donor chimerism is peripheral blood was analyzed at 6 months post-transplant. Bone marrow was obtained from primary mice at 6 months and transplanted in noncompetitive fashion into secondary lethally irradiated hosts and donor chimerism is peripheral blood was analyzed at 6 months post-transplant.

b

G-CSF was administered at a dose of 50 μg/kg; SC; bid × 4 days.

c

GROβ (2.5 mg/kg), CTCE0021 (25 mg/kg), MIP1α (50 μg/kg), AMD3100 (5 mg/kg), were administered as single bolus SC injections and peripheral blood cells harvested at 15 min (GROβ) or 60 min (MIP1α, CTCE0021, AMD3100) after administration.

d

GROβ, CTCE0021, MIP1α, AMD3100, were administered as single bolus injections approximately 16 h after the last dose of G-CSF (50 μg/kg; SC; bid × 4 days) and peripheral blood harvested 15 min (GROβ) or 60 min (CTCE0021, MIP1α, AMD3100) later.

Extensive characterization of PBSCs mobilized by GROβ and G-CSF indicates that GROβ-mobilized HSCs show significantly lower levels of apoptosis, significantly enhanced marrow homing that is less dependent on the SDF-1/CXCR4 axis and express more activated adhesion receptors, lower expression of CD26 and twice the number of CD26+ SKL cells compared with G-CSF-mobilized cells [101••], which are likely responsible for their superior engraftment capacity.

In preclinical studies, the frequency of severe combined immunodeficient (SCID) reconstituting cells (SRCs) was higher in AMD3100-mobilized PBSCs compared with G-CSF [78]. In rhesus macaques, AMD3100-mobilized CD34+ cells showed long-term reconstituting capacity, with AMD3100-mobilized PBSCs containing more G1 cells and higher CXCR4 and very late antigen-4 (VLA-4) expression, which likely contribute to enhanced engraftment [82]. Higher expression of genes associated with antiapoptosis, cell motility and cell cycle were found in CD34+ cells mobilized by AMD3100 and G-CSF compared with G-CSF alone, which may support their superior repopulating ability [102]; however, further functional studies are needed. In a clinical study [103•• ], AMD3100-mobilized PBSCs demonstrated a three-fold higher repopulating frequency in NOD/SCID mice despite two-fold lower CD34+ cell counts compared with G-CSF mobilized PBSCs, although only marginally better repopulating frequency was observed when purified CD34+ cells were used. In matched sibling donors, AMD3100-mobilized sufficient CD34+ cells for transplant in one leukapheresis in 10/11 patients, and although containing fewer CD34+ cells and more T cells than with mobilization by G-CSF, engraftment kinetics were similar to G-CSF with no appreciable increase in GVHD [104], suggesting that the AMD3100-mobilized HSCs have enhanced intrinsic HSC function.

Mobilization beyond hematopoietic stem cell

The emerging role of marrow as a site of adult stem cells that contribute to tissue repair raises the question of whether mobilizing agents can mobilize stem cells other than HSCs or whether new agents can be developed that differentially mobilize adult stem cells. Reestablishment of circulation to limit tissue damage is critical following myocardial infarction (MI) or limb ischemia. Cytokines including vascular endothelial growth factor (VEGF), angiopoietin, G-CSF and GM-CSF mobilize EPCs with high proliferative capacity [1,105108] and circulating angiogenic cells (CACs) that contribute to neovascularization through paracrine mechanisms [109111]. In animal models, autologous mobilization showed improved neovascularization and ventricular function; however, in randomized double-blind trials, no significant improvement in cardiac function was observed in acute MI patients mobilized by G-CSF [112,113]. Likely, the kinetics of EPC mobilization by G-CSF may not be optimal for this application since peak APC mobilization is not achieved until days 4–5 [114]. However, in two recent studies, intracoronary cell infusion with G-CSF-mobilized PBSCs showed a trend towards efficacy over systemic G-CSF administration in patients with MI [115], and intracardiac injection of autologous G-CSF-mobilized CD34+ cells in patients with intractable angina provided evidence for feasibility, safety and efficacy [116]. A recent study [117] shows that transplantation of autologous G-CSF-mobilized CD34+ cells into the muscle of ischemic limbs demonstrated therapeutic angiogenesis, and in a report [118] of a single patient, injection of autologous G-CSF-mobilized CD133+ EPCs produced long-term limb salvage, symptomatic relief and functional improvement. Recently, single administration of AMD3100 was shown to rapidly and significantly mobilize EPCs and CACs and that these cells could be harvested by leukapheresis after 4 h [119••]. This exciting finding offers potential strategies to rapidly mobilize cells to sites of vascular injury in an acute setting and to collect cells that can be directly transplanted into ischemic tissue.

MSCs can be isolated from umbilical blood [120122]; however, isolation from peripheral blood and the ability to mobilize MSCs are not clear. The presence [123126] and absence [127,128] of MSCs in peripheral blood have been reported. In general, this controversy stems from variation in the volume of blood used as a source of cells, differences in the methods used to identify MSCs and the stringency used to establish the criteria of stem cell activity. However, in two studies [126,129] using mobilized blood and adherence to fibrin microbeads, MSCs were isolated from normal donors and cancer patients. In primates, circulating fibroblastoid colony-forming units (CFU-F) that demonstrated both adipogeneic and osteogenic differentiation were detected in animals treated with G-CSF and SCF [130]. Although the controversy concerning detection of circulating MSCs needs resolution, it is becoming apparent that with appropriate stimuli they can be mobilized. From a therapeutic standpoint, however, it needs to be determined what role mobilized MSCs may play versus recruitment locally at sites of injury.

Conclusion

It is becoming clear that different agents mobilize HSC populations with overlapping stem cell function, but also mobilize HSCs that have unique intrinsic characteristics that may facilitate and expand our use of hematopoietic transplantation for curative purposes. HSCs that have enhanced homing and engraftment properties will certainly have utility in patients who mobilize poorly but may also be useful for gene therapy approaches to increase overall gene expression levels. In addition, it is clear that the cell composition of the mobilized graft may also significantly impact outcomes, and increased understanding of the mobilization process may allow us to engineer graft composition in ways that will alter the likelihood of graft failure, GVHD and impaired immune reconstitution. Current mobilization agents and combinations have their advantages and disadvantages; however, preclinical mechanistic studies on the action of these agents as well as continued mechanism driven investigation of new mobilization targets will be critical to our ultimate understanding of stem cell function and their use in regenerative medicine.

Acknowledgement

Supported by grants HL69669 and HL079654 from the National Institutes of Health.

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

• of special interest

• • of outstanding interest

Additional references related to this topic can also be found in the Current World Literature section in this issue (pp. 424–425).

  • 1.Asahara T, Takahashi T, Masuda H, et al. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J. 1999;18:3964–3972. doi: 10.1093/emboj/18.14.3964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Friedenstein AJ, Petrakova KV, Kurolesova AI, et al. Heterotopic transplants of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation. 1968;6:230–247. [PubMed] [Google Scholar]
  • 3.Grigoriadis AE, Heersche JN, Aubin JE. Differentiation of muscle, fat, cartilage, and bone from progenitor cells present in a bone-derived clonal cell population: effect of dexamethasone. J Cell Biol. 1988;106:2139–2151. doi: 10.1083/jcb.106.6.2139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Caplan AI. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J Cell Physiol. 2007;213:341–347. doi: 10.1002/jcp.21200. [DOI] [PubMed] [Google Scholar]
  • 5.McCredie KB, Hersh EM, Freireich EJ. Cells capable of colony formation in the peripheral blood of man. Science. 1971;171:293–294. doi: 10.1126/science.171.3968.293. [DOI] [PubMed] [Google Scholar]
  • 6.Kurnick JE, Robison WA. Colony growth of human peripheral white blood cells in vitro. Blood. 1971;37:136–141. [PubMed] [Google Scholar]
  • 7.Chervenick PA, Boggs DR. In vitro growth of granulocytic and mononuclear cell colonies from blood of normal individuals. Blood. 1971;37:131–135. [PubMed] [Google Scholar]
  • 8.Gratwohl A, Baldomero H, Horisberger B, et al. Current trends in hematopoietic stem cell transplantation in Europe. Blood. 2002;100:2374–2386. doi: 10.1182/blood-2002-03-0675. [DOI] [PubMed] [Google Scholar]
  • 9.Papayannopoulou T. Current mechanistic scenarios in hematopoietic stem/progenitor cell mobilization. Blood. 2004;103:1580–1585. doi: 10.1182/blood-2003-05-1595. [DOI] [PubMed] [Google Scholar]
  • 10.Fruehauf S, Seggewiss R. It's moving day: factors affecting peripheral blood stem cell mobilization and strategies for improvement. Br J Haematol. 2003;122:360–375. doi: 10.1046/j.1365-2141.2003.04483.x. [DOI] [PubMed] [Google Scholar]
  • 11.Thomas J, Liu F, Link DC. Mechanisms of mobilization of hematopoietic progenitors with granulocyte colony-stimulating factor. Curr Opin Hematol. 2002;9:183–189. doi: 10.1097/00062752-200205000-00002. [DOI] [PubMed] [Google Scholar]
  • 12.To LB, Haylock DN, Simmons PJ, et al. The biology and clinical uses of blood stem cells. Blood. 1997;89:2233–2258. [PubMed] [Google Scholar]
  • 13.Goldman JM, Horowitz MM. The international bone marrow transplant registry. Int J Hematol. 2002;76(Suppl 1):393–397. doi: 10.1007/BF03165291. [DOI] [PubMed] [Google Scholar]
  • 14.Ringden O, Remberger M, Runde V, et al. Faster engraftment of neutrophils and platelets with peripheral blood stem cells from unrelated donors: a comparison with marrow transplantation. Bone Marrow Transplant. 2000;25(Suppl 2):S6–S8. doi: 10.1038/sj.bmt.1702343. [DOI] [PubMed] [Google Scholar]
  • 15.Benito AI, Diaz MA, Gonzalez-Vicent M, et al. Hematopoietic stem cell transplantation using umbilical cord blood progenitors: review of current clinical results. Bone Marrow Transplant. 2004;33:675–690. doi: 10.1038/sj.bmt.1704405. [DOI] [PubMed] [Google Scholar]
  • 16.Bensinger W, Singer J, Appelbaum F, et al. Autologous transplantation with peripheral blood mononuclear cells collected after administration of recombinant granulocyte stimulating factor. Blood. 1993;81:3158–3163. [PubMed] [Google Scholar]
  • 17.Blume KG, Thomas ED. A review of autologous hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2000;6:1–12. doi: 10.1016/s1083-8791(00)70047-4. [DOI] [PubMed] [Google Scholar]
  • 18.Kennedy J. Peripheral blood progenitor cell mobilization: a clinical review. Pharmacotherapy. 1998;18:3–8. [PubMed] [Google Scholar]
  • 19.Welte K, Gabrilove J, Bronchud MH, et al. Filgrastim (r-metHuG-CSF): the first 10 years. Blood. 1996;88:1907–1929. [PubMed] [Google Scholar]
  • 20.Heldal D, Tjonnfjord G, Brinch L, et al. A randomized study of allogeneic transplantation with stem cells from blood or bone marrow. Bone Marrow Transplant. 2000;25:1129–1136. doi: 10.1038/sj.bmt.1702422. [DOI] [PubMed] [Google Scholar]
  • 21.Bensinger WI, Martin PJ, Storer B, et al. Transplantation of bone marrow as compared with peripheral-blood cells from HLA-identical relatives in patients with hematologic cancers. N Engl J Med. 2001;344:175–181. doi: 10.1056/NEJM200101183440303. [DOI] [PubMed] [Google Scholar]
  • 22.Desikan KR, Barlogie B, Jagannath S, et al. Comparable engraftment kinetics following peripheral-blood stem-cell infusion mobilized with granulocyte colony-stimulating factor with or without cyclophosphamide in multiple myeloma. J Clin Oncol. 1998;16:1547–1553. doi: 10.1200/JCO.1998.16.4.1547. [DOI] [PubMed] [Google Scholar]
  • 23.Aversa F, Tabilio A, Velardi A, et al. Treatment of high-risk acute leukemia with T-cell-depleted stem cells from related donors with one fully mismatched HLA haplotype. N Engl J Med. 1998;339:1186–1193. doi: 10.1056/NEJM199810223391702. [DOI] [PubMed] [Google Scholar]
  • 24.Reisner Y, Martelli MF. Transplantation tolerance induced by ‘mega dose’ CD34+ cell transplants. Exp Hematol. 2000;28:119–127. doi: 10.1016/s0301-472x(99)00132-0. [DOI] [PubMed] [Google Scholar]
  • 25.Falzetti F, Aversa F, Minelli O, et al. Spontaneous rupture of spleen during peripheral blood stem-cell mobilisation in a healthy donor. Lancet. 1999;353:555. doi: 10.1016/S0140-6736(99)00268-8. [DOI] [PubMed] [Google Scholar]
  • 26.Balaguer H, Galmes A, Ventayol G, et al. Splenic rupture after granulocyte-colony-stimulating factor mobilization in a peripheral blood progenitor cell donor. Transfusion. 2004;44:1260–1261. doi: 10.1111/j.1537-2995.2004.00413.x. [DOI] [PubMed] [Google Scholar]
  • 27.Mohty M, Bilger K, Jourdan E, et al. Higher doses of CD34+ peripheral blood stem cells are associated with increased mortality from chronic graft-versus-host disease after allogeneic HLA-identical sibling transplantation. Leukemia. 2003;17:869–875. doi: 10.1038/sj.leu.2402909. [DOI] [PubMed] [Google Scholar]
  • 28.Przepiorka D, Smith TL, Folloder J, et al. Risk factors for acute graft-versus-host disease after allogeneic blood stem cell transplantation. Blood. 1999;94:1465–1470. [PubMed] [Google Scholar]
  • 29.Heimfeld S. HLA-identical stem cell transplantation: is there an optimal CD34 cell dose? Bone Marrow Transplant. 2003;31:839–845. doi: 10.1038/sj.bmt.1704019. [DOI] [PubMed] [Google Scholar]
  • 30.Panse JP, Heimfeld S, Guthrie KA, et al. Allogeneic peripheral blood stem cell graft composition affects early T-cell chimaerism and later clinical outcomes after nonmyeloablative conditioning. Br J Haematol. 2005;128:659–667. doi: 10.1111/j.1365-2141.2005.05363.x. [DOI] [PubMed] [Google Scholar]
  • 31.Heimfeld S. Bone marrow transplantation: how important is CD34 cell dose in HLA-identical stem cell transplantation? Leukemia. 2003;17:856–858. doi: 10.1038/sj.leu.2402893. [DOI] [PubMed] [Google Scholar]
  • 32.Sierra J, Storer B, Hansen JA, et al. Transplantation of marrow cells from unrelated donors for treatment of high-risk acute leukemia: the effect of leukemic burden, donor HLA-matching, and marrow cell dose. Blood. 1997;89:4226–4235. [PubMed] [Google Scholar]
  • 33.Baron F, Maris MB, Storer BE, et al. High doses of transplanted CD34+ cells are associated with rapid T-cell engraftment and lessened risk of graft rejection, but not more graft-versus-host disease after nonmyeloablative conditioning and unrelated hematopoietic cell transplantation. Leukemia. 2005;19:822–828. doi: 10.1038/sj.leu.2403718. [DOI] [PubMed] [Google Scholar]
  • 34.Stiff P, Gingrich R, Luger S, et al. A randomized phase 2 study of PBPC mobilization by stem cell factor and filgrastim in heavily pretreated patients with Hodgkin's disease or non-Hodgkin's lymphoma. Bone Marrow Transplant. 2000;26:471–481. doi: 10.1038/sj.bmt.1702531. [DOI] [PubMed] [Google Scholar]
  • 35.Sugrue MW, Williams K, Pollock BH, et al. Characterization and outcome of ‘hard to mobilize’ lymphoma patients undergoing autologous stem cell transplantation. Leuk Lymphoma. 2000;39:509–519. doi: 10.3109/10428190009113381. [DOI] [PubMed] [Google Scholar]
  • 36.Koenigsmann M, Jentsch-Ullrich K, Mohren M, et al. The role of diagnosis in patients failing peripheral blood progenitor cell mobilization. Transfusion. 2004;44:777–784. doi: 10.1111/j.0041-1132.2004.03321.x. [DOI] [PubMed] [Google Scholar]
  • 37.Gazitt Y. Comparison between granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor in the mobilization of peripheral blood stem cells. Curr Opin Hematol. 2002;9:190–198. doi: 10.1097/00062752-200205000-00003. [DOI] [PubMed] [Google Scholar]
  • 38.Carral A, de la RJ, Martin G, et al. Factors influencing the collection of peripheral blood stem cells in patients with acute myeloblastic leukemia and nonmyeloid malignancies. Leuk Res. 2003;27:5–12. doi: 10.1016/s0145-2126(02)00068-1. [DOI] [PubMed] [Google Scholar]
  • 39.Keating S, Suciu S, de Witte T, et al. The stem cell mobilizing capacity of patients with acute myeloid leukemia in complete remission correlates with relapse risk: results of the EORTC-GIMEMA AML-10 trial. Leukemia. 2003;17:60–67. doi: 10.1038/sj.leu.2402782. [DOI] [PubMed] [Google Scholar]
  • 40.Mele A, Leopardi G, Sparaventi G, et al. Mini-ICE effectively mobilises peripheral blood stem cells after fludarabine-based regimens in acute myeloid leukaemia. Eur J Haematol. 2005;74:277–281. doi: 10.1111/j.1600-0609.2004.00383.x. [DOI] [PubMed] [Google Scholar]
  • 41.Anderlini P, Przepiorka D, Seong C, et al. Factors affecting mobilization of CD34+ cells in normal donors treated with filgrastim. Transfusion. 1997;37:507–512. doi: 10.1046/j.1537-2995.1997.37597293882.x. [DOI] [PubMed] [Google Scholar]
  • 42.Holm M. Not all healthy donors mobilize hematopoietic progenitor cells sufficiently after G-CSF administration to allow for subsequent CD34 purification of the leukapheresis product. J Hematother. 1998;7:111–113. doi: 10.1089/scd.1.1998.7.111. [DOI] [PubMed] [Google Scholar]
  • 43.Schmitz N, Dreger P, Suttorp M, et al. Primary transplantation of allogeneic peripheral blood progenitor cells mobilized by filgrastim (granulocyte colony-stimulating factor). Blood. 1995;85:1666–1672. [PubMed] [Google Scholar]
  • 44.Ng-Cashin J, Shen T. Mobilization of autologous peripheral blood hematopoietic cells for support of high-dose cancer therapy. In: Blume KG, Forman SJ, Appelbaum FR, editors. Thomas’ hematopoietic cell transplantation. 3rd ed Blackwell Publishing Ltd; Malden: 2004. pp. 576–587. [Google Scholar]
  • 45.Schmitz N. Peripheral blood hematopoietic cells for allogeneic transplantation. In: Blume KG, Forman SJ, Appelbaum FR, editors. Thomas’ hematopoietic cell transplantation. 3rd ed Blackwell Publishing Ltd; Malden: 2004. pp. 588–598. [Google Scholar]
  • 46.Croop JM, Cooper R, Fernandez C, et al. Mobilization and collection of peripheral blood CD34+ cells from patients with Fanconi anemia. Blood. 2001;98:2917–2921. doi: 10.1182/blood.v98.10.2917. [DOI] [PubMed] [Google Scholar]
  • 47••.Sloand EM, Read EJ, Scheinberg P, et al. Mobilization, collection, and immunomagnetic selection of peripheral blood CD34 cells in recovered aplastic anemia patients. Transfusion. 2007;47:1250–1253. doi: 10.1111/j.1537-2995.2007.01258.x. [PBSC collection was feasible only in 2/9 recovered patients with aplastic anemia, clearly pointing to the need for development of alternate regimens for expansion of the utility of PBSC transplantation] [DOI] [PubMed] [Google Scholar]
  • 48.Roberts AW, DeLuca E, Begley CG, et al. Broad inter-individual variations in circulating progenitor cell numbers induced by granulocyte colony-stimulating factor therapy. Stem Cells. 1995;13:512–516. doi: 10.1002/stem.5530130508. [DOI] [PubMed] [Google Scholar]
  • 49.Grigg AP, Roberts AW, Raunow H, et al. Optimizing dose and scheduling of filgrastim (granulocyte colony-stimulating factor) for mobilization and collection of peripheral blood progenitor cells in normal volunteers. Blood. 1995;86:4437–4445. [PubMed] [Google Scholar]
  • 50.de la RJ, Arbona C, de Arriba F, et al. Analysis of factors associated with low peripheral blood progenitor cell collection in normal donors. Transfusion. 2002;42:4–9. doi: 10.1046/j.1537-2995.2002.00010.x. [DOI] [PubMed] [Google Scholar]
  • 51.Bachar-Lustig E, Li HW, Gur H, et al. Induction of donor-type chimerism and transplantation tolerance across major histocompatibility barriers in subleth-ally irradiated mice by Sca-1(+)Lin(−) bone marrow progenitor cells: synergism with nonalloreactive (host donor)F(1) T cells. Blood. 1999;94:3212–3221. [PubMed] [Google Scholar]
  • 52.Fruehauf S, Klaus J, Huesing J, et al. Efficient mobilization of peripheral blood stem cells following CAD chemotherapy and a single dose of pegylated G-CSF in patients with multiple myeloma. Bone Marrow Transplant. 2007;39:743–750. doi: 10.1038/sj.bmt.1705675. [DOI] [PubMed] [Google Scholar]
  • 53.Bruns I, Steidl U, Kronenwett R, et al. A single dose of 6 or 12 mg of pegfilgrastim for peripheral blood progenitor cell mobilization results in similar yields of CD34+ progenitors in patients with multiple myeloma. Transfusion. 2006;46:180–185. doi: 10.1111/j.1537-2995.2006.00699.x. [DOI] [PubMed] [Google Scholar]
  • 54.Nosari A, Cairoli R, Ciapanna D, et al. Efficacy of single dose pegfilgrastim in enhancing the mobilization of CD34+ peripheral blood stem cells in aggressive lymphoma patients treated with cisplatin–aracytin-containing regimens. Bone Marrow Transplant. 2006;38:413–416. doi: 10.1038/sj.bmt.1705459. [DOI] [PubMed] [Google Scholar]
  • 55•.Russell N, Mesters R, Schubert J, et al. A phase 2 pilot study of pegfilgrastim and filgrastim for mobilizing peripheral blood progenitor cells in patients with non-Hodgkin's lymphoma receiving chemotherapy. Haematologica. 2008;93:405–412. doi: 10.3324/haematol.11287. [In a randomized double-blind study a single dose of Pegfilgrastim following chemotherapy effectively mobilized sufficient CD34+ cells for transplant in the majority of patients, with rapid ANC recovery posttransplant. The trial lays the foundation for use of a single dose of Pegfilgrastim for PBSC mobilization and transplantation] [DOI] [PubMed] [Google Scholar]
  • 56.Isidori A, Tani M, Bonifazi F, et al. Phase II study of a single pegfilgrastim injection as an adjunct to chemotherapy to mobilize stem cells into the peripheral blood of pretreated lymphoma patients. Haematologica. 2005;90:225–231. [PubMed] [Google Scholar]
  • 57.Hosing C, Qazilbash MH, Kebriaei P, et al. Fixed-dose single agent pegfilgrastim for peripheral blood progenitor cell mobilisation in patients with multiple myeloma. Br J Haematol. 2006;133:533–537. doi: 10.1111/j.1365-2141.2006.06054.x. [DOI] [PubMed] [Google Scholar]
  • 58.Kuendgen A, Fenk R, Bruns I, et al. Splenic rupture following administration of pegfilgrastim in a patient with multiple myeloma undergoing autologous peripheral blood stem cell transplantation. Bone Marrow Transplant. 2006;38:69–70. doi: 10.1038/sj.bmt.1705382. [DOI] [PubMed] [Google Scholar]
  • 59.Kroschinsky F, Holig K, Poppe-Thiede K, et al. Single-dose pegfilgrastim for the mobilization of allogeneic CD34+ peripheral blood progenitor cells in healthy family and unrelated donors. Haematologica. 2005;90:1665–1671. [PubMed] [Google Scholar]
  • 60.Roskos LK, Lum P, Lockbaum P, et al. Pharmacokinetic/pharmacodynamic modeling of pegfilgrastim in healthy subjects. J Clin Pharmacol. 2006;46:747–757. doi: 10.1177/0091270006288731. [DOI] [PubMed] [Google Scholar]
  • 61.Shpall EJ, Wheeler CA, Turner SA, et al. A randomized phase 3 study of peripheral blood progenitor cell mobilization with stem cell factor and filgrastim in high-risk breast cancer patients. Blood. 1999;93:2491–2501. [PubMed] [Google Scholar]
  • 62.Linker C, Anderlini P, Herzig R, et al. Recombinant human thrombopoietin augments mobilization of peripheral blood progenitor cells for autologous transplantation. Biol Blood Marrow Transplant. 2003;9:405–413. doi: 10.1016/s1083-8791(03)00101-0. [DOI] [PubMed] [Google Scholar]
  • 63.Lane TA, Law P, Maruyama M, et al. Harvesting and enrichment of hematopoietic progenitor cells mobilized into the peripheral blood of normal donors by granulocyte-macrophage colony-stimulating factor (GM-CSF) or G-CSF: potential role in allogeneic marrow transplantation. Blood. 1995;85:275–282. [PubMed] [Google Scholar]
  • 64.Spitzer G, Adkins D, Mathews M, et al. Randomized comparison of G-CSF + GM-CSF vs. G-CSF alone for mobilization of peripheral blood stem cells: effects on hematopoietic recovery after high-dose chemotherapy. Bone Marrow Transplant. 1997;20:921–930. doi: 10.1038/sj.bmt.1700999. [DOI] [PubMed] [Google Scholar]
  • 65.Kim CH, Broxmeyer HE. In vitro behavior of hematopoietic progenitor cells under the influence of chemoattractants: stromal cell-derived factor-1, steel factor, and the bone marrow environment. Blood. 1998;91:100–110. [PubMed] [Google Scholar]
  • 66.Mohle R, Bautz F, Rafii S, et al. The chemokine receptor CXCR-4 is expressed on CD34+ hematopoietic progenitors and leukemic cells and mediates transendothelial migration induced by stromal cell-derived factor-1. Blood. 1998;91:4523–4530. [PubMed] [Google Scholar]
  • 67.Lapidot T, Kollet O. The essential roles of the chemokine SDF-1 and its receptor CXCR4 in human stem cell homing and repopulation of transplanted immune-deficient NOD/SCID and NOD/SCID/B2m(null) mice. Leukemia. 2002;16:1992–2003. doi: 10.1038/sj.leu.2402684. [DOI] [PubMed] [Google Scholar]
  • 68.Lord BI, Woolford LB, Wood LM, et al. Mobilization of early hematopoietic progenitor cells with BB-10010: a genetically engineered variant of human macrophage inflammatory protein-1 alpha. Blood. 1995;85:3412–3415. [PubMed] [Google Scholar]
  • 69.Broxmeyer HE, Cooper S, Hangoc G, et al. Dominant myelopoietic effector functions mediated by chemokine receptor CCR1. J Exp Med. 1999;189:1987–1992. doi: 10.1084/jem.189.12.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Pelus LM, Bian H, King AG, et al. 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. 2004;103:110–119. doi: 10.1182/blood-2003-04-1115. [DOI] [PubMed] [Google Scholar]
  • 71•.Pelus LM, Fukuda S. Chemokine-mobilized adult stem cells; defining a better hematopoietic graft. Leukemia. 2008;22:466–473. doi: 10.1038/sj.leu.2405021. [Compares the engraftment potential of different mobilized PBSC grafts in mice, showing qualitative different engraftment potentials] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.King AG, Horowitz D, Levin R, et al. Rapid mobilization of murine hematopoietic stem cells with enhanced engraftment properties and evaluation of hematopoietic progenitor cell mobilization in rhesus monkeys by a single injection of SB-251353, a specific truncated form of the human CXC chemokine GRObeta. Blood. 2001;97:1534–1542. doi: 10.1182/blood.v97.6.1534. [DOI] [PubMed] [Google Scholar]
  • 73.Pelus LM, Horowitz D, Cooper SC, et al. Peripheral blood stem cell mobilization. A role for CXC chemokines. Crit Rev Oncol Hematol. 2002;43:257–275. doi: 10.1016/s1040-8428(01)00202-5. [DOI] [PubMed] [Google Scholar]
  • 74.Fibbe WE, Pruijt JF, Velders GA, et al. Biology of IL-8-induced stem cell mobilization. Ann N Y Acad Sci. 1999;872:71–82. doi: 10.1111/j.1749-6632.1999.tb08454.x. [DOI] [PubMed] [Google Scholar]
  • 75.Pelus LM, Broxmeyer HE. Chemokine axes in hematopoietic stem cell mobilization. In: Neote K, Letts GL, Moser B, editors. Chemokine biology-basic research and clinical application. Vol II. Pathophysiology of chemokines. Birkhauser Verlag AG; Basel: 2007. pp. 125–144. [Google Scholar]
  • 76.Merzouk A, Wong D, Salari H, et al. Rational design of chemokine SDF-1 analogs with agonist activity for the CXCR4 receptor and the capacity to rapidly mobilize PMN and hematopoietic progenitor cells in mice. Lett Drug Design Discov. 2004;1:126–134. [Google Scholar]
  • 77.Pelus LM, Bian H, Fukuda S, 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. 2005;33:295–307. doi: 10.1016/j.exphem.2004.11.008. [DOI] [PubMed] [Google Scholar]
  • 78.Broxmeyer HE, Orschell CM, Clapp DW, et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med. 2005;201:1307–1318. doi: 10.1084/jem.20041385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Liles WC, Broxmeyer HE, Rodger E, et al. Mobilization of hematopoietic progenitor cells in healthy volunteers by AMD3100, a CXCR4 antagonist. Blood. 2003;102:2728–2730. doi: 10.1182/blood-2003-02-0663. [DOI] [PubMed] [Google Scholar]
  • 80.Shen H, Cheng T, Olszak I, et al. CXCR-4 desensitization is associated with tissue localization of hemopoietic progenitor cells. J Immunol. 2001;166:5027–5033. doi: 10.4049/jimmunol.166.8.5027. [DOI] [PubMed] [Google Scholar]
  • 81.Burroughs L, Mielcarek M, Little MT, et al. Durable engraftment of AMD3100-mobilized autologous and allogeneic peripheral-blood mononuclear cells in a canine transplantation model. Blood. 2005;106:4002–4008. doi: 10.1182/blood-2005-05-1937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Larochelle A, Krouse A, Metzger M, et al. AMD3100 mobilizes hematopoietic stem cells with long-term repopulating capacity in nonhuman primates. Blood. 2006;107:3772–3778. doi: 10.1182/blood-2005-09-3592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Devine SM, Flomenberg N, Vesole DH, et al. Rapid mobilization of CD34+ cells following administration of the CXCR4 antagonist AMD3100 to patients with multiple myeloma and non-Hodgkin's lymphoma. J Clin Oncol. 2004;22:1095–1102. doi: 10.1200/JCO.2004.07.131. [DOI] [PubMed] [Google Scholar]
  • 84.Grignani G, Perissinotto E, Cavalloni G, et al. Clinical use of AMD3100 to mobilize CD34+ cells in patients affected by non-Hodgkin's lymphoma or multiple myeloma. J Clin Oncol. 2005;23:3871–3872. doi: 10.1200/JCO.2005.55.250. [DOI] [PubMed] [Google Scholar]
  • 85.Liles WC, Rodger E, Broxmeyer HE, et al. Augmented mobilization and collection of CD34+ hematopoietic cells from normal human volunteers stimulated with granulocyte-colony-stimulating factor by single-dose administration of AMD3100, a CXCR4 antagonist. Transfusion. 2005;45:295–300. doi: 10.1111/j.1537-2995.2005.04222.x. [DOI] [PubMed] [Google Scholar]
  • 86••.Flomenberg N, Devine SM, DiPersio JF, et al. The use of AMD3100 plus G-CSF for autologous hematopoietic progenitor cell mobilization is superior to G-CSF alone. Blood. 2005;106:1867–1874. doi: 10.1182/blood-2005-02-0468. [This was the first study in humans to test the hypothesis that the hematopoietic graft mobilized by the combination of AMD3100 and G-CSF is superior to G-CSF alone. In a randomized trial in patients with multiple myeloma and non-Hodgkin's lymphoma, AMD3100 and G-CSF produced higher yields of CD34+ cells, effectively mobilized patients who failed G-CSF mobilization and reduced time for collection of sufficient cells for transplant. This study laid the groundwork for the three randomized phase III trials recently reported [88–90]] [DOI] [PubMed] [Google Scholar]
  • 87.Cashen A, Calandra G, Macfarland R, et al. A mobilizing regimen of AMD3100 and G-CSF increases stem cell collection in patients with Hodgkin's disease, and PK is similar to that of non-cancer patients. Blood. 2006;108:869a. [Google Scholar]
  • 88.DiPersio JF, Stadtmauer EA, Nademanee AP, et al. A phase III, multicenter, randomized, double-blind, placebo-controlled, comparative trial of AMD3100 (Perixafor) + G-CSF vs. G-CSF + placebo for mobilization in multiple myeloma (MM) patients for autologous hematopoietic stem cell (aHSC) transplantation. Blood. 2007;110:137a. [Google Scholar]
  • 89.Di Persio JF, Micallef I, Stiff P, et al. A phase III, multicenter, randomized, double-blind, placebo controlled, comparative trial of AMD3100 (Perixafor) + G-CSF vs. placebo + G-CSF in non-Hodgkins lymphoma (NHL) patients for autologous hematopoietic stem cell (aHSC) transplantation. Blood. 2007;110:185a. [Google Scholar]
  • 90.Micallef I, Stiff P, DiPersio JF, et al. Successful stem cell mobilization rescue by AMD3100 (Perixafor) + G-CSF for patients who failed primary mobilization: results from the phase III (3101-NHL) study. Blood. 2007;110:185a–186a. [Google Scholar]
  • 91.Gothot A, van der Loo JC, Clapp DW, et al. Cell cycle-related changes in repopulating capacity of human mobilized peripheral blood CD34(+) cells in nonobese diabetic/severe combined immune-deficient mice. Blood. 1998;92:2641–2649. [PubMed] [Google Scholar]
  • 92.Ramshaw HS, Rao SS, Crittenden RB, et al. Engraftment of bone marrow cells into normal unprepared hosts: effects of 5-fluorouracil and cell cycle status. Blood. 1995;86:924–929. [PubMed] [Google Scholar]
  • 93.Schuchert MJ, Wright RD, Colson YL. Characterization of a newly discovered T-cell receptor beta-chain heterodimer expressed on a CD8+ bone marrow subpopulation that promotes allogeneic stem cell engraftment. Nat Med. 2000;6:904–909. doi: 10.1038/78667. [DOI] [PubMed] [Google Scholar]
  • 94.Gandy KL, Domen J, Aguila H, et al. CD8+TCR+ and CD8+TCR− cells in whole bone marrow facilitate the engraftment of hematopoietic stem cells across allogeneic barriers. Immunity. 1999;11:579–590. doi: 10.1016/s1074-7613(00)80133-8. [DOI] [PubMed] [Google Scholar]
  • 95.Christopherson KW, Hangoc G, Mantel CR, et al. Modulation of hematopoietic stem cell homing and engraftment by CD26. Science. 2004;305:1000–1003. doi: 10.1126/science.1097071. [DOI] [PubMed] [Google Scholar]
  • 96.Peled A, Petit I, Kollet O, et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999;283:845–848. doi: 10.1126/science.283.5403.845. [DOI] [PubMed] [Google Scholar]
  • 97.Hassan HT, Zeller W, Stockschlader M, et al. Comparison between bone marrow and G-CSF-mobilized peripheral blood allografts undergoing clinical scale CD34+ cell selection. Stem Cells. 1996;14:419–429. doi: 10.1002/stem.140419. [DOI] [PubMed] [Google Scholar]
  • 98.Tricot G, Jagannath S, Vesole D, et al. Peripheral blood stem cell transplants for multiple myeloma: identification of favorable variables for rapid engraftment in 225 patients. Blood. 1995;85:588–596. [PubMed] [Google Scholar]
  • 99.Verfaillie CM, Almeida-Porada G, Wissink S, et al. Kinetics of engraftment of CD34(−) and CD34(+) cells from mobilized blood differs from that of CD34(−) and CD34(+) cells from bone marrow. Exp Hematol. 2000;28:1071–1079. doi: 10.1016/s0301-472x(00)00506-3. [DOI] [PubMed] [Google Scholar]
  • 100•.Bonig H, Priestley GV, Oehler V, et al. Hematopoietic progenitor cells (HPC) from mobilized peripheral blood display enhanced migration and marrow homing compared to steady-state bone marrow HPC. Exp Hematol. 2007;35:326–334. doi: 10.1016/j.exphem.2006.09.017. [The authors show that differential enhanced homing of G-CSF mobilized PBSC, that results in their enhanced engraftment compared to bone marrow, results from enhanced motility and lower expression levels of CD26] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101••.Fukuda S, Bian H, King AG, et al. The chemokine GROβ mobilizes early hematopoietic stem cells characterized by enhanced homing and engraftment. Blood. 2007;110:860–869. doi: 10.1182/blood-2006-06-031401. [The authors offer proof that PBSC mobilized by the chemokine GROβ are highly enriched for more primitive stem cells and that their enhanced homing and engraftment properties compared to stem cells mobilized by G-CSF is not dependent on the CXCR4/SDF-1 axis] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Fruehauf S, Seeger T, Maier P, et al. The CXCR4 antagonist AMD3100 releases a subset of G-CSF-primed peripheral blood progenitor cells with specific gene expression characteristics. Exp Hematol. 2006;34:1052–1059. doi: 10.1016/j.exphem.2006.06.003. [DOI] [PubMed] [Google Scholar]
  • 103••.Hess DA, Bonde J, Craft TP, et al. Human progenitor cells rapidly mobilized by AMD3100 repopulate NOD/SCID mice with increased frequency in comparison to cells from the same donor mobilized by granulocyte colony stimulating factor. Biol Blood Marrow Transplant. 2007;13:398–411. doi: 10.1016/j.bbmt.2006.12.445. [The authors used a pairwise approach, collecting PBSCs mobilized by G-CSF and AMD3100 in the same normal human donors. Their results demonstrate in head to head comparisons that the PBSCs mobilized by AMD3100 showed enhanced repopulating frequency in NOD/SCID mice] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Devine SM, Andritsos L, Todt L, et al. A pilot study evaluating the safety and efficacy of AMD3100 for the mobilization and transplantation of HLA-matched sibling donor hematopoietic stem cells in patients with advanced hematological malignancies. Blood. 2006;106:299a. [Google Scholar]
  • 105.Hattori K, Dias S, Heissig B, et al. Vascular endothelial growth factor and angiopoietin-1 stimulate postnatal hematopoiesis by recruitment of vasculogenic and hematopoietic stem cells. J Exp Med. 2001;193:1005–1014. doi: 10.1084/jem.193.9.1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Hu J, Takatoku M, Sellers SE, et al. Analysis of origin and optimization of expansion and transduction of circulating peripheral blood endothelial progenitor cells in the rhesus macaque model. Hum Gene Ther. 2002;13:2041–2050. doi: 10.1089/10430340260395893. [DOI] [PubMed] [Google Scholar]
  • 107.Orlic D, Kajstura J, Chimenti S, et al. Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc Natl Acad Sci U S A. 2001;98:10344–10349. doi: 10.1073/pnas.181177898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med. 2003;9:702–712. doi: 10.1038/nm0603-702. [DOI] [PubMed] [Google Scholar]
  • 109.Ingram DA, Caplice NM, Yoder MC. Unresolved questions, changing definitions, and novel paradigms for defining endothelial progenitor cells. Blood. 2005;106:1525–1531. doi: 10.1182/blood-2005-04-1509. [DOI] [PubMed] [Google Scholar]
  • 110.Rehman J, Li J, Orschell CM, et al. Peripheral blood ‘endothelial progenitor cells’ are derived from monocyte/macrophages and secrete angiogenic growth factors. Circulation. 2003;107:1164–1169. doi: 10.1161/01.cir.0000058702.69484.a0. [DOI] [PubMed] [Google Scholar]
  • 111.De Palma M, Venneri MA, Roca C, et al. Targeting exogenous genes to tumor angiogenesis by transplantation of genetically modified hematopoietic stem cells. Nat Med. 2003;9:789–795. doi: 10.1038/nm871. [DOI] [PubMed] [Google Scholar]
  • 112.Zohlnhofer D, Ott I, Mehilli J, et al. Stem cell mobilization by granulocyte colony-stimulating factor in patients with acute myocardial infarction: a randomized controlled trial. JAMA. 2006;295:1003–1010. doi: 10.1001/jama.295.9.1003. [DOI] [PubMed] [Google Scholar]
  • 113.Ripa RS, Jorgensen E, Wang Y, et al. Stem cell mobilization induced by subcutaneous granulocyte-colony stimulating factor to improve cardiac regeneration after acute ST-elevation myocardial infarction: result of the double-blind, randomized, placebo-controlled stem cells in myocardial infarction (STEMMI) trial. Circulation. 2006;113:1983–1992. doi: 10.1161/CIRCULATIONAHA.105.610469. [DOI] [PubMed] [Google Scholar]
  • 114.Stroncek DF, Clay ME, Herr G, et al. The kinetics of G-CSF mobilization of CD34+ cells in healthy people. Transfus Med. 1997;7:19–24. doi: 10.1046/j.1365-3148.1997.d01-75.x. [DOI] [PubMed] [Google Scholar]
  • 115•.Kang HJ, Kim HS, Koo BK, et al. Intracoronary infusion of the mobilized peripheral blood stem cell by G-CSF is better than mobilization alone by G-CSF for improvement of cardiac function and remodeling: 2-year follow-up results of the Myocardial Regeneration and Angiogenesis in Myocardial Infarction with G-CSF and Intra-Coronary Stem Cell Infusion (MAGIC Cell) 1 trial. Am Heart J. 2007;153:237–238. doi: 10.1016/j.ahj.2006.11.004. [This is one of two papers that suggest that direct administration of PBSCs mobilized by G-CSF directly to site of injury improves cardiac function] [DOI] [PubMed] [Google Scholar]
  • 116•.Losordo DW, Schatz RA, White CJ, et al. Intramyocardial transplantation of autologous CD34+ stem cells for intractable angina: a phase I/IIa double-blind, randomized controlled trial. Circulation. 2007;115:3165–3172. doi: 10.1161/CIRCULATIONAHA.106.687376. [This is one of two papers that suggest that direct administration of PBSCs mobilized by G-CSF directly to site of injury improves cardiac function] [DOI] [PubMed] [Google Scholar]
  • 117•.Kawamura A, Horie T, Tsuda I, et al. Clinical study of therapeutic angiogenesis by autologous peripheral blood stem cell (PBSC) transplantation in 92 patients with critically ischemic limbs. J Artif Organs. 2006;9:226–233. doi: 10.1007/s10047-006-0351-2. [The authors show in a clinical study that transplant of autologous CD34neg PBSC mobilized by G-CSF into muscles of ischemic limbs produced therapeutic angiogenesis in all but Fontaine stage IV patients] [DOI] [PubMed] [Google Scholar]
  • 118•.Canizo MC, Lozano F, Gonzalez-Porras JR, et al. Peripheral endothelial progenitor cells (CD133+) for therapeutic vasculogenesis in a patient with critical limb ischemia. One year follow-up. Cytotherapy. 2007;9:99–102. doi: 10.1080/14653240601034708. [First demonstration of therapeutic efficacy of autologous CD133+ mobilized by G-CSF in a patient with critical limb ischemia] [DOI] [PubMed] [Google Scholar]
  • 119••.Shepherd RM, Capoccia BJ, Devine SM, et al. Angiogenic cells can be rapidly mobilized and efficiently harvested from the blood following treatment with AMD3100. Blood. 2006;108:3662–3667. doi: 10.1182/blood-2006-06-030577. [The authors show that single administration of AMD3100 rapidly mobilized EPC and CAC and that these cells could be harvested by leukapheresis after 4 h and cryopreserved. This exciting finding offers potential strategies to rapidly mobilize cells to sites of vascular injury in an acute setting and to collect cells that can be directly transplanted into ischemic tissue] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109:235–242. doi: 10.1046/j.1365-2141.2000.01986.x. [DOI] [PubMed] [Google Scholar]
  • 121.Bieback K, Kern S, Kluter H, et al. Critical parameters for the isolation of mesenchymal stem cells from umbilical cord blood. Stem Cells. 2004;22:625–634. doi: 10.1634/stemcells.22-4-625. [DOI] [PubMed] [Google Scholar]
  • 122.Lee OK, Kuo TK, Chen WM, et al. Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood. 2004;103:1669–1675. doi: 10.1182/blood-2003-05-1670. [DOI] [PubMed] [Google Scholar]
  • 123.Fernandez M, Simon V, Herrera G, et al. Detection of stromal cells in peripheral blood progenitor cell collections from breast cancer patients. Bone Marrow Transplant. 1997;20:265–271. doi: 10.1038/sj.bmt.1700890. [DOI] [PubMed] [Google Scholar]
  • 124.Zvaifler NJ, Marinova-Mutafchieva L, Adams G, et al. Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res. 2000;2:477–488. doi: 10.1186/ar130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kuznetsov SA, Mankani MH, Gronthos S, et al. Circulating skeletal stem cells. J Cell Biol. 2001;153:1133–1140. doi: 10.1083/jcb.153.5.1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Kassis I, Zangi L, Rivkin R, et al. Isolation of mesenchymal stem cells from G-CSF-mobilized human peripheral blood using fibrin microbeads. Bone Marrow Transplant. 2006;37:967–976. doi: 10.1038/sj.bmt.1705358. [DOI] [PubMed] [Google Scholar]
  • 127.Lazarus HM, Haynesworth SE, Gerson SL, et al. Human bone marrow-derived mesenchymal (stromal) progenitor cells (MPCs) cannot be recovered from peripheral blood progenitor cell collections. J Hematother. 1997;6:447–455. doi: 10.1089/scd.1.1997.6.447. [DOI] [PubMed] [Google Scholar]
  • 128.Wexler SA, Donaldson C, Denning-Kendall P, et al. Adult bone marrow is a rich source of human mesenchymal ‘stem’ cells but umbilical cord and mobilized adult blood are not. Br J Haematol. 2003;121:368–374. doi: 10.1046/j.1365-2141.2003.04284.x. [DOI] [PubMed] [Google Scholar]
  • 129.Rochefort GY, Delorme B, Lopez A, et al. Multipotential mesenchymal stem cells are mobilized into peripheral blood by hypoxia. Stem Cells. 2006;24:2202–2208. doi: 10.1634/stemcells.2006-0164. [DOI] [PubMed] [Google Scholar]
  • 130.Levesque JP, Winkler IG, Larsen SR, et al. Mobilization of bone marrow-derived progenitors. Handb Exp Pharmacol. 2007:3–36. doi: 10.1007/978-3-540-68976-8_1. [DOI] [PubMed] [Google Scholar]

RESOURCES