Gene transfer experiments in nonhuman primates have been shown to be predictive of success in human clinical gene therapy trials. In most nonhuman primate studies, hematopoietic stem cells (HSCs) collected from the peripheral blood or bone marrow after administration of granulocyte colony-stimulating factor (G-CSF) + stem cell factor (SCF) have been used as targets, but this cytokine combination is not generally available for clinical use, and the optimum target cell population has not been systematically studied. In our current study we tested the retroviral transduction efficiency of rhesus macaque peripheral blood CD34+ cells collected after administration of different cytokine mobilization regimens, directly comparing G-CSF+SCF versus G-CSF alone or G-CSF+Flt3-L in competitive repopulation assays. Vector supernatant was added daily for 96 hours in the presence of stimulatory cytokines. The transduction efficiency of HSCs as assessed by in vitro colony-forming assays was equivalent in all 5 animals tested, but the in vivo levels of mononuclear cell and granulocyte marking was higher at all time points derived from target CD34+ cells collected after G-CSF+SCF mobilization compared with target cells collected after G-CSF (n = 3) or G-CSF+Flt3-L (n = 2) mobilization. In 3 of the animals long-term marking levels of 5% to 25% were achieved, but originating only from the G-CSF+SCF–mobilized target cells. Transduction efficiency of HSCs collected by different mobilization regimens can vary significantly and is superior with G-CSF+SCF administration. The difference in transduction efficiency of HSCs collected from different sources should be considered whenever planning clinical gene therapy trials and should preferably be tested directly in comparative studies.

Nonhuman primates are invaluable models for evaluation of gene therapy protocols prior to their implementation in human studies. The ability to transduce primitive human hematopoietic stem cells (HSCs) has been poorly predicted by murine and in vitro models, but has been more closely correlated with results in rhesus macaque and other nonhuman primate autologous transplantation assays. This is likely due to similarities in hematopoietic stem cell kinetics, cytokine responsiveness, and retroviral receptor levels between humans and nonhuman primates.1 Performance of transduction on a surface coated with the carboxy terminal fragment of fibronectin (FN) and inclusion of a combination of cytokines including stem cell factor (SCF), megakaryocyte growth and development factor (MGDF), andfms-like tyrosine kinase 3 ligand (Flt3-L) has been shown to greatly improve retroviral gene transduction in nonhuman primates, with long-term marking levels of 15% to 20% possible.2,3 Most importantly, advances in nonhuman primate gene transfer strategies have been translated into the first successful gene therapy clinical trial in patients with X-linked severe combined immunodeficiency syndrome (SCID).4,5 

One aspect of gene transfer into hematopoietic stem cells that has received less attention since the advent of more efficient retroviral transduction protocols is the source of target HSCs used in therapeutic or marking clinical trials. So far target HSCs for clinical gene transfer or therapy protocols have been collected via bone marrow (BM) harvest,4-10 granulocyte colony-stimulating factor (G-CSF)–mobilized peripheral blood (PB) leukapheresis,8,11-13 or umbilical cord blood collection.14,15 In 1994, Bodine et al showed that mobilized peripheral blood cells collected after administration of G-CSF+SCF were transduced better with retroviral vectors compared with the BM cells of mice treated with 5-fluorouracil (5-FU).16 We later extended these studies to the rhesus macaque model and found that the level of retroviral transduction was comparable in long-term engrafting cells collected from PB after 5 days of G-CSF+SCF administration or collected from BM 14 days after G-CSF+SCF administration.17 In the same set of experiments we also showed that CD34+ cells collected from BM primed with G-CSF+SCF administration were superior targets for retroviral transduction compared with steady state BM CD34+ cells, using transduction conditions without Flt3-L, MGDF, or fibronectin fragment support, now known to be suboptimal.

The combination of G-CSF+SCF has proven to be a more effective mobilizing regimen compared with G-CSF alone in murine,18-20 canine,21 and nonhuman primate22-25 models, and in several randomized clinical trials the combined use of G-CSF+SCF led to increased median yield of CD34+ cells per leukapheresis.26-30 It has been suggested that these cells might be better targets for gene therapy compared with G-CSF–mobilized cells,31 but to date this regimen has not been used in any clinical gene therapy trial.

Due to serious anaphylactic reactions occurring in patients treated with SCF in combination with other agents for aplastic anemia, this cytokine is no longer available, even for approved clinical protocols in the United States, although it is marketed in several other countries. In contrast, G-CSF has an extensive safety profile, both as an agent for accelerating recovery from neutropenia after myelotoxic treatments (chemotherapy or radiation) and as the primary agent for peripheral blood stem cell mobilization for transplantation purposes. It also has been reported recently that Flt3-L administered in combination with G-CSF for 5 days is a potent mobilizer of HSCs into the peripheral blood of mice32 and nonhuman primates,33 and more recently Rosenzweig et al used G-CSF+Flt3-L–mobilized PB CD34+ cells for retroviral gene transfer in nonhuman primate experiments, although in the latter study maximum number of colony-forming cells in the blood were obtained after 7 days of administration of the cytokines.34 In this study we directly compared the levels of long-term gene marking achievable in the nonhuman primate model using G-CSF+SCF–mobilized PB CD34+ HSC targets versus CD34+ cells obtained by mobilization with clinically more practical and available mobilization regimens, including G-CSF alone or the combination of G-CSF+Flt3-L, each regimen administered for 5 doses.

Collection of PB HSCs from rhesus macaques

Young rhesus macaques (Macaca mulatta) used in these studies were housed and handled in accordance with the guidelines set by the Committee on Care and Use of Laboratory Animals of the Institute of Laboratory Animal Resources, National Research Council (DHHS publication No. NIH 85-23), and the protocol was approved by the Animal Care and Use Committee of the National Heart, Lung, and Blood Institute. The animals received recombinant human (rhu) G-CSF (10 μg/kg; Amgen, Thousand Oaks, CA) alone or in combination with either recombinant polyethylene glycol conjugated human SCF (200 μg/kg; Amgen) or recombinant human Flt3-L (200 μg/kg; Immunex, Seattle, WA) as daily subcutaneous injections for 3 days, and twice daily on the fourth day. Mobilized PB cells were collected by leukapheresis on day 5 as described.25 Mononuclear cells were isolated using density gradient centrifugation over lymphocyte separation media (ICN Biomedicals/Cappel, Aurora, OH). CD34+ enrichment was performed using the 12.8 Immunoglobulin-M (IgM) anti-CD34 biotinylated antibody and MACS streptavidin microbeads (Miltenyi Biotec, Auburn, CA). The degree of progenitor enrichment was calculated from CFU assays performed before and after column purification.

Vectors and transduction procedures

G1Na and LNL6 are Moloney murine leukemia virus–derived retroviral vectors that carry an identical bacterial neomycin phosphotransferase (neo) gene.35 A 16-bp polylinker insertion 5′ of the neo gene allows quantitative assessment of marking from the 2 vectors within one polymerase chain reaction (PCR).36 The biologic titers of these vectors were assessed by making serial dilutions of the vectors and then monitoring the transfer of G418 resistance to HeLa cells. The biologic titer of both vectors were equivalent and between 2-5 × 105 biologically active vector particles/mL. For transduction, retroviral supernatant was harvested from subconfluent producer cells cultured for 12-18 hours in Dulbecco modified Eagle medium (DMEM) (Mediatech, Herndon, VA) supplemented with 10% fetal bovine serum (FBS) (Atlanta Biologicals, Norcross, GA), 4 mMl-glutamine, penicillin (50 mg/mL), and streptomycin (50 mg/mL) at 37°C in 5% CO2. Fresh vector supernatant was passed through a 0.22-mm filter (Millipore, Bedford, MA) to remove cellular debris before transduction.

CD34-enriched cells were cultured at a starting concentration of 1-2 × 105 cells/mL in filtered vector supernatant, supplemented with 100 ng/mL rhuSCF, 100 ng/mL rhuFlt-3 L, and 100 ng/mL rhuMGDF (Amgen), in flasks previously coated with the CH-296 fragment of fibronectin (Retronectin, TaKara, Shiga, Japan) per manufacturer's instructions. Every 24 hours, nonadherent cells were collected, spun down, and resuspended in fresh vector supernatant and cytokines, and added back to the same fibronectin-coated flask. At the end of 96 hours cultured cells were removed from the plates with trypsin, collected, frozen viably in 50% autologous serum mixed with 40% DMEM and 10% dimethyl sulfoxide (DMSO), placed on dry ice, and then transferred to a liquid nitrogen container.

Each animal was remobilized after a period of 6 weeks, with the alternative cytokine regimen, and CD34+ cells were obtained and purified using the same procedures. This remobilization interval was chosen as likely beyond the time period impacted by a prior mobilization cycle.37 CD34+ cells were transduced under the same conditions as described above, but with the alternative vector, and then cryopreserved. One week later, the animals received 500 cGy × 2 total body irradiation, and both aliquots of transduced cells were thawed and reinfused via a central venous catheter. Twenty-four hours later, the animals were started on G-CSF at 5 μg/kg intravenously daily until the total white blood cell count reached 6000/μL. Standard supportive care included blood product transfusions, fluid and electrolyte management, and antibiotic administration as needed. Hematopoietic recovery was monitored by daily complete blood counts.

Sample collection

PB samples were collected at the time of recovery, monthly through 6 months after transplantation, and then every 3 months. From each blood sample, mononuclear cells were isolated by density gradient centrifugation over lymphocyte separation medium, and granulocytes were obtained as previously described.38 

CFU assays

PB CD34-enriched cells from before and after each transduction and mononuclear cells obtained from bone marrow samples after transplantation were plated and analyzed by CFU assays using MethoCult M4230 methylcellulose (MC) media (StemCell Technologies, Vancouver, BC, Canada) supplemented with 5 U/mL rhu erythropoietin (Amgen), 10 ng/mL rhuGM-CSF (Sandoz, East Hanover, NJ), 10 ng/mL rhuIL-3, (Sandoz), and 100 ng/mL rhuSCF (Amgen) at 37°C in 5% CO2. Between days 10 and 14, colonies of more than 50 cells were counted, and between 16 and 32 individual CFUs were plucked from the plates at each time point for PCR analysis. Colonies were plucked into 50 μL distilled water, digested with 20 μg/mL proteinase K (Qiagen, Valencia, CA) at 55°C for 1 hour, followed by 99°C for 10 minutes, and later assessed for the presence of vector sequences by PCR as described below.

PCR analysis

Genomic DNA was extracted using the QIAamp DNA blood Midi kit (Qiagen). The primers and conditions used for neo PCR andβ-actin PCR have been previously described.8All neo and β-actin PCR reactions were run under conditions optimized to yield linear results in the range of the intensity of the in vivo samples. For the outer reaction, 100-200 ng DNA was used, and 18-20 cycles were performed for the inner reaction, based on the level of in vivo marking. For every PCR analysis, negative controls included DNA from normal rhesus PB samples extracted with the same methodology and a reagent control. Serial dilutions of G1Na DNA (containing 2 copies of integrated vector per cell) into normal rhesus PB DNA were used as positive controls for generating the control regression curve. Band intensity was quantitated using a PhosphorImager (Molecular Dynamics, Sunnyvale, CA). neo band intensity was normalized for amplifiable DNA content based on theβ-actin signal, and the overall contribution of each vector to in vivo marking was calculated by plotting the signal intensity of each band on a standard curve derived from known copy number controls amplified concurrently.

Southern blot analysis

Ten micrograms of genomic DNA was digested with KpnI (Boehringer-Mannheim, Indianapolis, IN), which cuts once within each viral long-terminal repeat, and thus can distinguish integrated retroviral G1Na and LNL6 DNA. The DNA fragments were transferred to a nylon membrane and hybridized with a radiolabeledneo gene–specific probe generated by PCR using the following primers: forward primer 5′-TCC ATC ATG GCT GAT GCA ATG CGG C-3′ and reverse primer 5′-GAT AGA AGG CGA TGC GCT GCG AAT CG-3′.39 

Statistical analysis

Analysis of significance using the 2-tailed Student ttest and regression analysis were carried out using SigmaPlot (SPSS Science, Chicago, IL) and Excel software (Microsoft, Seattle, WA).

The experimental design for comparison of G-CSF+SCF versus either G-CSF alone or G-CSF+Flt3-L–mobilized CD34+ cells as targets for retroviral transduction is shown in Figure1 and explained in “Materials and methods.” Three animals were used to compare G-CSF+SCF with G-CSF alone, and 2 animals were used to compare G-CSF+SCF with G-CSF+Flt3-L. Table 1 summarizes the order in which each mobilization regimen was administered and the marking vector used to transduce each population of CD34+ cells. The order of administration of cytokines was reversed in each consecutive animal to control for any impact of prior mobilization on remobilization.37 The G1Na and LNL6 vectors have comparable titers on HeLa cells, and in multiple prior studies have been shown to have equivalent transduction efficiency of rhesus hematopoietic stem and progenitor cells.3,39,40 

Fig. 1.

Experimental design.

Each animal initially received 5 doses of either G-CSF+SCF or G-CSF before leukapheresis. CD34+ cells were transduced with either G1Na or LNL6 vectors for 96 hours. Each animal was mobilized with the alternative regimen 6 weeks later, but this time transduction was done with the other vector. Both transduced aliquots were frozen at the end of transduction and then thawed and reinfused to the monkey after 500 cGy × 2 total body irradiation. The same experimental design was used to compare G-CSF+SCF– versus G-CSF+Flt3-L–mobilized cells.

Fig. 1.

Experimental design.

Each animal initially received 5 doses of either G-CSF+SCF or G-CSF before leukapheresis. CD34+ cells were transduced with either G1Na or LNL6 vectors for 96 hours. Each animal was mobilized with the alternative regimen 6 weeks later, but this time transduction was done with the other vector. Both transduced aliquots were frozen at the end of transduction and then thawed and reinfused to the monkey after 500 cGy × 2 total body irradiation. The same experimental design was used to compare G-CSF+SCF– versus G-CSF+Flt3-L–mobilized cells.

Close modal
Table 1.

Summary of CD34+ enrichment, CFU transduction efficiency, cell expansion, and engraftment kinetics of animals

Cytokine regimenVectorCD34+
enrichment*
Transduction
efficiency
Initial no.of cellsNo. of cells
at the end
of culture
ExpNo. of
infused
cells/kg
Eng1-153 (d)F/U1-155 (mo)
Animal RQ2314        18 
 G-CSF G1Na 100 89%  (25 of 28) 1.4 × 107 2.6 × 107 1.8 6.2 × 106   
 G-CSF+SCF LNL6 151 96%  (27 of 28) 2.1 × 107 2.6 × 108 12.4 6.2 × 107   
Animal RQ2284        15 
 G-CSF+SCF G1Na 73 100%  (16 of 16) 3.0 × 107 3.5 × 107 1.2 7.9 × 106   
 G-CSF LNL6 93 90%  (27 of 30) 2.2 × 107 2.2 × 107 1.0 4.8 × 106   
Animal RQ2237        12 
 G-CSF LNL6 113 100%  (32 of 32) 1.9 × 107 2.4 × 107 1.3 4.9 × 106   
 G-CSF+SCF G1Na 95 100%  (32 of 32) 4.5 × 107 1.1 × 108 2.5 2.2 × 107   
Animal RQ2277        12 
 G-CSF+SCF G1Na 155 100%  (32 of 32) 4.0 × 107 2.1 × 108 5.2 5.0 × 107   
 G-CSF+Flt3-L LNL6 95 100%  (32 of 32) 1.6 × 107 3.1 × 107 1.9 7.3 × 106   
Animal RQ2265        14 12 
 G-CSF+Flt3-L G1Na 127 100%  (32 of 32) 2.9 × 107 4.6 × 107 1.6 1.0 × 107   
 G-CSF+SCF LNL6 72 100%  (32 of 32) 3.9 × 107 9.2 × 107 2.3 2.1 × 107   
Cytokine regimenVectorCD34+
enrichment*
Transduction
efficiency
Initial no.of cellsNo. of cells
at the end
of culture
ExpNo. of
infused
cells/kg
Eng1-153 (d)F/U1-155 (mo)
Animal RQ2314        18 
 G-CSF G1Na 100 89%  (25 of 28) 1.4 × 107 2.6 × 107 1.8 6.2 × 106   
 G-CSF+SCF LNL6 151 96%  (27 of 28) 2.1 × 107 2.6 × 108 12.4 6.2 × 107   
Animal RQ2284        15 
 G-CSF+SCF G1Na 73 100%  (16 of 16) 3.0 × 107 3.5 × 107 1.2 7.9 × 106   
 G-CSF LNL6 93 90%  (27 of 30) 2.2 × 107 2.2 × 107 1.0 4.8 × 106   
Animal RQ2237        12 
 G-CSF LNL6 113 100%  (32 of 32) 1.9 × 107 2.4 × 107 1.3 4.9 × 106   
 G-CSF+SCF G1Na 95 100%  (32 of 32) 4.5 × 107 1.1 × 108 2.5 2.2 × 107   
Animal RQ2277        12 
 G-CSF+SCF G1Na 155 100%  (32 of 32) 4.0 × 107 2.1 × 108 5.2 5.0 × 107   
 G-CSF+Flt3-L LNL6 95 100%  (32 of 32) 1.6 × 107 3.1 × 107 1.9 7.3 × 106   
Animal RQ2265        14 12 
 G-CSF+Flt3-L G1Na 127 100%  (32 of 32) 2.9 × 107 4.6 × 107 1.6 1.0 × 107   
 G-CSF+SCF LNL6 72 100%  (32 of 32) 3.9 × 107 9.2 × 107 2.3 2.1 × 107   
*

Degree of progenitor cell enrichment by CD34 selection was calculated by enumerating total CFUs before and after column purification.

Transduction efficiency calculated by the percentage ofβ-actin-positive colonies that were also neopositive at the end of transduction, as determined by PCR of individual colonies.

Expansion: ratio of the number of cells at the end of 4 days of transduction (number of infused cells) to the number of cells plated on the first day.

F1-153

Engraftment: days to reach absolute neutrophil count greater than 500/μL.

F1-155

Follow-up: duration (months) of follow-up after transplantation.

There were no significant differences in the efficiency of transduction of committed progenitors, as defined using the in vitro CFU assay, found in G-CSF+SCF–mobilized CD34+ cells compared with G-CSF alone or G-CSF+Flt3-L (Table 1), and no overall discrepancies between results achieved using the G1Na versus the LNL6 vector. Comparable with our recent experiments,40 the transduction of committed progenitors was extremely efficient in all CD34+ populations, but our prior studies have indicated that CFU transduction may not correlate with transduction of more primitive in vivo repopulating cells.38-40 

The number of CD34+ cells collected by leukapheresis was higher in each experimental group after mobilization with G-CSF+SCF (3.20 ± 1.21 × 107) versus mobilization with G-CSF alone (1.83 ± 0.40 × 107; P = .157) or G-CSF+SCF (3.95 ± 0.07 × 107) versus mobilization with G-CSF+Flt3-L (2.25 ± 0.91 × 107;P = .248), although the differences did not reach statistical significance given the small numbers of animals in each group. Cells collected following G-CSF+SCF administration expanded more robustly during the 4-day transduction period compared with cells mobilized with G-CSF alone (5.4 versus 1.4, respectively;P = .350) or with G-CSF+Flt3-L (3.7 versus 1.7, respectively; P = .366). Consequently, all 5 animals received a higher number of transduced G-CSF+SCF–mobilized cells compared with transduced G-CSF–mobilized cells (3.06 ± 2.80 × 107 versus 0.53 ± 0.08 × 107, respectively; P = .249) or G-CSF+SCF–mobilized cells compared with G-CSF+Flt3-L–mobilized cells (3.55 ± 2.05 × 107 versus 0.86 ± 0.19 × 107, respectively; P = .339) per kilogram of body weight. Due to a small number of animals in each experimental group, a limitation inherent to these types of primate experiments, statistical significance was not achieved in these comparisons despite the clear observed trends.

All animals recovered their peripheral blood counts without significant morbidity, and there was a trend toward faster engraftment in the animals that received higher total cell doses, as we have previously observed.38 

After transplantation, semiquantitative PCR analysis of peripheral blood samples allowed assessment of the relative contribution of marked cells derived from CD34+ target cells collected using different cytokine regimens. Figure 2shows representative PCR gels from animals RQ2314, RQ2277, and RQ2237. In animals RQ2314 and RQ2277, there was detectable neomarking derived only from the G-CSF+SCF–mobilized target cells and not from the G-CSF or the G-CSF+Flt3-L fractions. In animal RQ2237, the presence of marked cells derived from both G-CSF+SCF targets and G-CSF alone targets can be appreciated in both mononuclear cells and in granulocytes collected from peripheral blood at different time points after transplantation, but the level of marking from G-CSF+SCF–mobilized cells is on average 206 and 48 times higher, respectively, in mononuclear cells and granulocytes, compared with marking from G-CSF–mobilized cells. A summary of marking levels in PB mononuclear cells and granulocytes over time, assuming one vector integration per cell, is depicted on a logarithmic scale in Figure3. Animals RQ2284 and RQ2265 had overall lower marking levels, but the G-CSF+SCF target cells still resulted in better marking levels compared with G-CSF alone or G-CSF+Flt3-L.

Fig. 2.

PCR analysis of in vivo marking.

Representative PCR of neo and β-actin sequences in PB mononuclear cells (MNCs) and granulocytes (Gran) at different time points after engraftment in 3 animals. Concurrentβ-actin PCR was performed for each sample to quantitate the amount of amplifiable template DNA. Serial dilutions of G1Na DNA (2 copies of integrated vector per cell) into normal rhesus PB DNA were used as positive controls. In animals RQ2314 and RQ2277 the only amplified band corresponds to the vector that was used to transduce G-CSF+SCF–mobilized cells. In animal RQ2237 in addition to the band corresponding to G-CSF+SCF aliquot, a very faint band can be seen that corresponds to transduced G-CSF–mobilized cells.

Fig. 2.

PCR analysis of in vivo marking.

Representative PCR of neo and β-actin sequences in PB mononuclear cells (MNCs) and granulocytes (Gran) at different time points after engraftment in 3 animals. Concurrentβ-actin PCR was performed for each sample to quantitate the amount of amplifiable template DNA. Serial dilutions of G1Na DNA (2 copies of integrated vector per cell) into normal rhesus PB DNA were used as positive controls. In animals RQ2314 and RQ2277 the only amplified band corresponds to the vector that was used to transduce G-CSF+SCF–mobilized cells. In animal RQ2237 in addition to the band corresponding to G-CSF+SCF aliquot, a very faint band can be seen that corresponds to transduced G-CSF–mobilized cells.

Close modal
Fig. 3.

Summary of in vivo gene marking.

Level of peripheral blood mononuclear cell and granulocyteneo gene marking as assessed by semiquantitative PCR at certain time points after transplantation, shown on a logarithmic scale. In each animal black bars (▪) represent the marking level originating from G-CSF+SCF–mobilized target cells; white bars (■), G-CSF–mobilized target cells; and gray bars (░), G-CSF+Flt3-L–mobilized cells.

Fig. 3.

Summary of in vivo gene marking.

Level of peripheral blood mononuclear cell and granulocyteneo gene marking as assessed by semiquantitative PCR at certain time points after transplantation, shown on a logarithmic scale. In each animal black bars (▪) represent the marking level originating from G-CSF+SCF–mobilized target cells; white bars (■), G-CSF–mobilized target cells; and gray bars (░), G-CSF+Flt3-L–mobilized cells.

Close modal

In Figure 4 we show the comparison between the ratio (G-CSF+SCF/G-CSF alone or G-CSF+SCF/G-CSF+Flt3-L fraction) of the number of infused cells (ie, number of cells collected at the end of 4 days of transduction) per kilogram weight of the animals versus the ratio of the marking achieved from each target cell group in each animal. Taking into account the number of infused cells, the ratio of marking remains significantly higher originating from the G-CSF+SCF–mobilized CD34+ cells compared with the ratio of infused cells, especially in the 3 animals that had high level gene marking.

Fig. 4.

Comparison of the ratio of in vivo gene marking versus the ratio of infused cells on a logarithmic scale.

In each animal the ratio of the average neo in vivo marking of first-year PB samples originating from G-CSF+SCF–mobilized cells to G-CSF alone or G-CSF+Flt3-L–mobilized cells were calculated (▪). For each animal this ratio was compared with the ratio of infused cells (ie, number of cells collected at the end of transduction) from G-CSF+SCF–mobilized cells to G-CSF alone or G-CSF+Flt3-L–mobilized cells (▨). Note that although the number of cells infused from the G-CSF+SCF aliquot was always higher in each animal compared with the other aliquot, the difference in gene marking levels between the aliquots was in every case significantly greater, except in animal RQ2265, which overall had low level marking. Monkeys RQ2314, RQ2284, and RQ2237 were mobilized with G-CSF+SCF versus G-CSF alone, and monkeys RQ2277 and RQ2265 were mobilized with G-CSF+SCF versus G-CSF + Flt3-L.

Fig. 4.

Comparison of the ratio of in vivo gene marking versus the ratio of infused cells on a logarithmic scale.

In each animal the ratio of the average neo in vivo marking of first-year PB samples originating from G-CSF+SCF–mobilized cells to G-CSF alone or G-CSF+Flt3-L–mobilized cells were calculated (▪). For each animal this ratio was compared with the ratio of infused cells (ie, number of cells collected at the end of transduction) from G-CSF+SCF–mobilized cells to G-CSF alone or G-CSF+Flt3-L–mobilized cells (▨). Note that although the number of cells infused from the G-CSF+SCF aliquot was always higher in each animal compared with the other aliquot, the difference in gene marking levels between the aliquots was in every case significantly greater, except in animal RQ2265, which overall had low level marking. Monkeys RQ2314, RQ2284, and RQ2237 were mobilized with G-CSF+SCF versus G-CSF alone, and monkeys RQ2277 and RQ2265 were mobilized with G-CSF+SCF versus G-CSF + Flt3-L.

Close modal

The level and ratio of marking originating from the 2 transduced fractions were confirmed by genomic Southern blotting of peripheral blood samples from selected time points in the 2 animals with the highest level of marking (RQ2314 and RQ2277). In the blot shown in Figure 5 the only detectableneo marking in each of these 2 animals corresponds to the vector that was used to transduce the G-CSF+SCF–mobilized cells, and there is no detectable level of marking from the other fraction.

Fig. 5.

Southern blot analysis of genomic marking.

In animals RQ2277 and RQ2314 the Southern blot analysis of PB mononuclear cells and granulocytes after engraftment shows only the vector that was used to transduce the G-CSF+SCF fraction (G1Na in animal RQ2277, and LNL6 in animal RQ2314). LNL6 DNA and serial dilutions of G1Na DNA into normal rhesus DNA were used as controls. Due to residual env sequences in LNL6 that are not present in G1Na, the expected fragment size is 3.0 kb for LNL6 and 2.3 kb for G1Na.

Fig. 5.

Southern blot analysis of genomic marking.

In animals RQ2277 and RQ2314 the Southern blot analysis of PB mononuclear cells and granulocytes after engraftment shows only the vector that was used to transduce the G-CSF+SCF fraction (G1Na in animal RQ2277, and LNL6 in animal RQ2314). LNL6 DNA and serial dilutions of G1Na DNA into normal rhesus DNA were used as controls. Due to residual env sequences in LNL6 that are not present in G1Na, the expected fragment size is 3.0 kb for LNL6 and 2.3 kb for G1Na.

Close modal

We also confirmed the functional high level of marking in these 2 animals long after transplantation by PCR analysis of CFU assays of BM mononuclear cells plated with and without G418 at 1.0 mg/mL active concentration, which is effective in inhibiting 98%-100% untransduced control rhesus cells plated in parallel.3 We counted the number of colonies from MC plates that were each plated with 105/mL of BM mononuclear cells. In animal RQ2314, 20 months after transplantation there were on average 78 colonies in each G418 plate and 6 colonies in each G418+plate; among the colonies plucked from G418+ plates, 27 of 29 were positive for LNL6 vector by PCR (G-CSF+SCF fraction) and only 2 of 29 colonies were positive for G1Na (G-CSF fraction). In animal RQ2277, 16 months after transplantation there were on average 51 colonies in each G418 plate and one colony in each G418+ plate; among colonies plucked from G418+plates, 16 of 18 were positive for G1Na (G-CSF+SCF fraction) and only 2 of 18 colonies were positive for LNL6 (G-CSF+Flt3-L fraction).

In this study we directly compared the level of in vivo marking of long-term repopulating cells obtained using 3 different mobilization regimens for collection of target CD34+ cells. As expected, based on previous nonhuman primate23-25 and human clinical studies,26-30 the number of CD34+ cells collected after G-CSF+SCF mobilization was higher than the number of cells collected using G-CSF alone. There is less prior information on the relative efficacy of G-CSF+Flt3-L mobilization, but we also found G-CSF+SCF to result in a higher number of cells collected compared with G-CSF+Flt3-L in the 2 animals tested when each combination was administered for 5 doses. The cells collected following G-CSF+SCF mobilization expanded more in vitro. Despite a similar level of transduction efficiency of progenitors as assessed by PCR for vector in CFUs present at the end of the 4-day culture period, the in vivo results using the different target cell mobilization regimens were quite different. In all 5 animals the marking of circulating mononuclear cells and granulocytes was significantly higher from the G-CSF+SCF–mobilized cells compared with either G-CSF alone or G-CSF+Flt3-L, and in 3 animals we achieved stable long-term marking of 5%-25%.

There are at least 3 possible explanations for the much higher marking levels achieved using G-CSF+SCF–mobilized target cells. The first 2 are simply numeric: a larger number of HSCs mobilized with this regimen, or improved survival or expansion of these cells in vitro following transduction, if the actual transduction frequency remains the same, would result in higher in vivo marking given the competitive repopulation design. The third explanation would be actual differences in transduction efficiency of primitive HSCs. Although a higher number of starting cells or a higher number of infused cells from G-CSF+SCF aliquot collected at the end of 4 days of transduction might have contributed in part to the differences observed in marking levels, we believe that this is unlikely to be the only reason for the improved in vivo marking. As we demonstrated in Figure 4, even if we normalize the marking level achieved for the number of infused CD34+cells per kilogram weight of the animals, the level of marking remains significantly higher originating from the G-CSF+SCF–mobilized CD34+ cells. The relative frequency of actual repopulating stem cells present within the total CD34+ population may have been higher within G-CSF+SCF–mobilized cells and account for the differences seen,41 but in vivo NOD/SCID engraftment assays do not suggest this to be the case.42 Therefore, we suggest that there is some intrinsic difference in the target HSCs mobilized with G-CSF+SCF that permits more efficient transduction.

One of the major obstacles in retroviral gene transduction of HSCs is the quiescent nature of these cells, because successful transduction of HSCs by retroviral vectors requires mitotic divisions to allow entry of preintegration viral complexes into the nucleus.43-45Primitive CD34+ hematopoietic stem cells present in mobilized peripheral blood, whether with G-CSF or G-CSF+SCF, are primarily in the G0 phase of the cell cycle.39,46 Using different combinations of stimulatory cytokines in culture media allows primitive hematopoietic cells to enter cell cycle and hence become susceptible to retroviral transduction, but this occurs at the cost of differentiation and loss of engraftment potential.47,48 Gene transfer investigators have spent years testing many different combinations of cytokines, and recent improvements in gene transfer efficiency into primitive repopulating cells assayed in murine xenograft models or nonhuman primates have occurred using combinations including stem cell factor and Flt3-L, along with traditional cytokines such as interleukin-3 (IL-3) and IL-6 or, most recently, thrombopoietin/MGDF.2,38,49 Presumably these combinations induce the most primitive cells to cycle without initiating terminal differentiation.

Duarte and Frank50 transduced the MO7e primitive hematopoietic cell line, known to express the SCF receptor, with the G-CSF receptor gene. Treating these cells with a combination of G-CSF and SCF resulted in a significant reduction in the percentage of cells in G0/G1 compared with cells treated with G-CSF alone.50 They also observed a marked shortening of the duration of the G0/G1 phase of the cell cycle following G-CSF+SCF cotreatment compared with cells treated with G-CSF alone, mediated in part by decreased expression of p27kip-1. It is possible that primitive cells obtained from different sources vary in an intrinsic program governing cell division in vitro or are differentially susceptible to extrinsic cytokine stimulation. This has been demonstrated for primitive cells in cord blood or fetal liver compared with adult marrow or mobilized blood.51 We hypothesize that stem cells present in the G-CSF+SCF–mobilized blood might enter the cell cycle more frequently or rapidly than stem cells present after G-CSF or G-CSF+Flt3-L administration, and thus be susceptible to transduction by retroviral vectors.

The induction of cycling in vitro can be a double-edged sword. A number of investigators reported loss of engraftment potential with cytokine stimulation in vitro and linked this loss specifically to cells that were actively cycling.46,47,52,53 The potential reversibility of this engraftment defect was first shown by Habibian and coworkers, who reported dramatic fluctuations in the engraftment phenotype of murine HSCs linked to their progress through and then out of the cell cycle.54 We have recently extended this observation to the nonhuman primate model and gene transfer applications by demonstrating that cells transduced for 4 days in the presence of stimulatory cytokines regained engraftment ability if they were allowed to exit S/G2/M by maintenance in SCF alone for 48 hours on fibronectin-coated plates.39 One potential factor explaining our current results could be that G-CSF+SCF–mobilized stem cells enter cell cycle more rapidly in vitro, permitting transduction, but then come back out of cycle and thus by 96 hours have regained some engraftment potential. HSCs mobilized with G-CSF alone or G-CSF+Flt3-L instead perhaps enter cell cycle more slowly and are therefore both more difficult to transduce and could even be hindered in engraftment because at the time of reinfusion they may have finally entered cell cycle. Our ability to test these hypotheses is limited by the poor phenotypic characterization of rhesus primitive engrafting cells, thus cell cycle analysis on purified rhesus HSCs has not been feasible, but some of these issues could be explored in the NOD/SCID model using human cells.42 

One other factor that might contribute to our results is the level of amphotropic retrovirus receptor (amphoR) on target cells from different sources. Orlic et al55 have shown that a low level of amphoR mRNA in mouse HSCs correlated with a low level of amphotropic retrovirus transduction of these cells. They also reported that primitive c-KitHi HSCs have a higher level ofamphoR mRNA when marrow was collected after treatment with G-CSF+SCF compared with steady state, and that this higher level correlated with improved transduction efficiency. They found very low levels of amphoR mRNA in adult bone marrow LinCD34+ CD38 cells, compared with cord blood or fetal liver,56 but receptor levels have not been studied in the context of cytokine therapy. Although this phenomenon has not been tested directly in the human and nonhuman primate experiments regarding actual repopulating cell transduction, it seems likely that it may be at least partially responsible for some of the differences observed in the in vivo results using different sources of HSCs.

These studies all point to the fact that HSCs collected from alternative sources might have different properties both at the time of collection and at the end of transduction and, consequently, variable abilities to be transduced or to engraft at the end of culture. Highly proliferative characteristics of umbilical cord blood and increased amphotropic receptor levels, along with an in vivo advantage for corrected lymphoid cells, might explain the relatively encouraging results obtained in a trial for adenosine deaminase deficiency using autologous cord blood CD34+ cells.14,15The recent reports of successful gene therapy for X-linked SCID used marrow target cells, but from very young children,4,5,10and these cells may also have improved transduction capabilities based on the properties discussed above.

We believe our findings might provide an explanation for the low level of in vivo gene transduction results obtained so far in human clinical gene therapy trials using primitive hematopoietic stem cell targets in G-CSF–mobilized peripheral blood. Over the last several years advances in vector design, culture conditions, and cytokines used in vitro in gene transduction protocols have significantly improved the level of gene transfer using G-CSF+SCF–primed blood or bone marrow, or bone marrow from young children, but based on our results, it seems that more research is needed in elucidating the biology of CD34+cells collected through different sources and the effect it might have on the final results. The importance of reconsidering the use of SCF in mobilization regimens, at least in gene therapy applications, seems warranted, because the mast cell release toxicity generally can be avoided by premedication with antihistamines before SCF administration, and patients undergoing a brief treatment mobilization could receive all SCF doses in a monitored setting.

We thank Dr David Woo at Amgen for supplying rhuG-CSF and rhuSCF, Dr Hiroshi Miyazaki at Kirin for supplying rhuMGDF, and Dr Stewart Lyman at Immunex for supplying Flt3-L.

Prepublished online as Blood First Edition Paper, November 7, 2002; DOI 10.1182/blood-2002-08-2663.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Van Beusechem
VW
Valerio
D
Gene transfer into hematopoietic stem cells of nonhuman primates.
Hum Gene Ther.
7
1996
1649
1668
2
Kiem
HP
Andrews
RG
Morris
J
et al
Improved gene transfer into baboon marrow repopulating cells using recombinant human fibronectin fragment CH-296 in combination with interleukin-6, stem cell factor, FLT-3 ligand, and megakaryocyte growth and development factor.
Blood.
92
1998
1878
1886
3
Wu
T
Kim
HJ
Sellers
SE
et al
Prolonged high-level detection of retrovirally marked hematopoietic cells in non-human primates after transduction of CD34+ progenitors using clinically feasible methods.
Mol Ther.
1
2000
285
293
4
Cavazzana-Calvo
M
Hacein-Bey
S
de Saint Basile
G
et al
Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease.
Science.
288
2000
669
672
5
Hacein-Bey-Abina
S
Le Deist
F
Carlier
F
et al
Sustained correction of X-linked severe combined immunodeficiency by ex vivo gene therapy.
N Engl J Med.
346
2002
1185
1193
6
Hoogerbrugge
PM
Vossen
JM
Van Beusechem
VW
Valerio
D
Treatment of patients with severe combined immunodeficiency due to adenosine deaminase (ADA) deficiency by autologous transplantation of genetically modified bone marrow cells.
Hum Gene Ther.
3
1992
553
558
7
Brenner
MK
Rill
DR
Moen
RC
et al
Gene-marking to trace origin of relapse after autologous bone marrow transplantation.
Lancet.
341
1993
85
86
8
Dunbar
CE
Cottler-Fox
M
O'Shaughnessy
J
et al
Retrovirally marked CD34-enriched peripheral blood and bone marrow cells contribute to long-term engraftment after autologous transplantation.
Blood.
85
1995
3048
3057
9
Hoogerbrugge
PM
Van Beusechem
VW
Fisher
A
et al
Bone marrow gene transfer in three patients with adenosine deaminase deficiency.
Gene Ther.
3
1996
179
183
10
Aiuti
A
Slavin
S
Aker
M
et al
Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning.
Science.
296
2002
2410
2413
11
Malech
HL
Maples
PB
Whiting-Theobald
N
et al
Prolonged production of NADPH oxidase-corrected granulocytes after gene therapy of chronic granulomatous disease.
Proc Natl Acad Sci U S A.
94
1997
12133
12138
12
Liu
JM
Kim
S
Read
EJ
et al
Engraftment of hematopoietic progenitor cells transduced with the Fanconi Anemia Group C gene (FANCC).
Hum Gene Ther.
10
1999
2337
2346
13
Abonour
R
Williams
DA
Einhorn
L
et al
Efficient retrovirus-mediated transfer of the multidrug resistance 1 gene into autologous human long-term repopulating hematopoietic stem cells.
Nat Med.
6
2000
652
658
14
Kohn
DB
Weinberg
KI
Nolta
JA
et al
Engraftment of gene-modified umbilical cord blood cells in neonates with adenosine deaminase deficiency.
Nat Med.
1
1995
1017
1023
15
Kohn
DB
Hershfield
MS
Carbonaro
D
et al
T lymphocytes with a normal ADA gene accumulate after transplantation of transduced autologous umbilical cord blood CD34+ cells in ADA-deficient SCID neonates.
Nat Med.
4
1998
775
780
16
Bodine
DM
Seidel
NE
Gale
MS
Nienhuis
AW
Orlic
D
Efficient retrovirus transduction of mouse pluripotent hematopoietic stem cells mobilized into the peripheral blood by treatment with granulocyte colony-stimulating factor and stem cell factor.
Blood.
84
1994
1482
1491
17
Dunbar
CE
Seidel
NE
Doren
S
et al
Improved retroviral gene transfer into murine and rhesus peripheral blood or bone marrow repopulating cells primed in vivo with stem cell factor and granulocyte colony-stimulating factor.
Proc Natl Acad Sci U S A.
93
1996
11871
11876
18
Briddell
RA
Hartley
CA
Smith
KA
McNiece
IK
Recombinant rat stem cell factor synergizes with recombinant human granulocyte colony-stimulating factor in vivo in mice to mobilize peripheral blood progenitor cells that have enhanced repopulating potential.
Blood.
82
1993
1720
1723
19
Yan
XQ
Briddell
R
Hartley
C
Stoney
G
Samal
B
McNiece
I
Mobilization of long-term hematopoietic reconstituting cells in mice by the combination of stem cell factor plus granulocyte colony-stimulating factor.
Blood.
84
1994
795
799
20
Yan
XQ
Hartley
C
McElroy
P
Chang
A
McCrea
C
McNiece
I
Peripheral blood progenitor cells mobilized by recombinant granulocyte colony-stimulating factor plus recombinant rat stem cell factor contain long-term engrafting cells capable of cellular proliferation for more than two years as shown by serial transplantation in mice.
Blood.
85
1995
2303
2307
21
de Revel
T
Appelbaum
FR
Storb
R
et al
Effects of granulocyte colony-stimulating factor and stem cell factor, alone and in combination, on the mobilization of peripheral blood cells that engraft lethally irradiated dogs.
Blood.
83
1994
3795
3799
22
Andrews
RG
Bensinger
WI
Knitter
GH
et al
The ligand for c-kit, stem cell factor, stimulates the circulation of cells that engraft lethally irradiated baboons.
Blood.
80
1992
2715
2720
23
Andrews
RG
Briddell
RA
Knitter
GH
et al
In vivo synergy between recombinant human stem cell factor and recombinant human granulocyte colony-stimulating factor in baboons enhanced circulation of progenitor cells.
Blood.
84
1994
800
810
24
Andrews
RG
Briddell
RA
Knitter
GH
Rowley
SD
Appelbaum
FR
McNiece
IK
Rapid engraftment by peripheral blood progenitor cells mobilized by recombinant human stem cell factor and recombinant human granulocyte colony-stimulating factor in nonhuman primates.
Blood.
85
1995
15
20
25
Donahue
RE
Kirby
MR
Metzger
ME
Agricola
BA
Sellers
SE
Cullis
HM
Peripheral blood CD34+ cells differ from bone marrow CD34+ cells in Thy-1 expression and cell cycle status in nonhuman primates mobilized or not mobilized with granulocyte colony-stimulating factor and/or stem cell factor.
Blood.
87
1996
1644
1653
26
Moskowitz
CH
Stiff
P
Gordon
MS
et al
Recombinant methionyl human stem cell factor and filgrastim for peripheral blood progenitor cell mobilization and transplantation in non-Hodgkin's patients—results of a phase I/II trial.
Blood.
89
1997
3136
3147
27
Glaspy
JA
Shpall
EJ
LeMaistre
CF
et al
Peripheral blood progenitor cell mobilization using stem cell factor in combination with filgrastim in breast cancer patients.
Blood.
90
1997
2939
2951
28
Shpall
EJ
Wheeler
CW
Turner
SA
et al
A randomized phase 3 study of peripheral blood progenitor cell mobilization with stem cell factor and Filigrastim in patients with high-risk breast cancer.
Blood.
93
1999
2491
2501
29
Weaver
A
Change
J
Wrigley
E
et al
Randomized comparison of progenitor-cell mobilization using chemotherapy, stem-cell factor, and filgrastim or chemotherapy plus filgrastim alone in patients with ovarian cancer.
J Clin Oncol.
16
1998
2601
2602
30
Facon
T
Harousseau
JL
Maloisel
F
et al
Stem cell factor in combination with filgrastim after chemotherapy improves peripheral blood progenitor cell yield and reduces apheresis requirements in multiple myeloma patients: a randomized, controlled trial.
Blood.
94
1999
1218
1225
31
Lyman
SD
Jacobsen
SEW
c-kit ligand and Flt3 ligand: stem/progenitor cell factors with overlapping yet distinct activities.
Blood.
91
1998
1101
1134
32
Sudo
Y
Shimazaki
C
Ashihara
E
et al
Synergistic effect of FLT-3 ligand on the granulocyte colony-stimulating factor-induced mobilization of hematopoietic stem cells and progenitor cells into blood in mice.
Blood.
89
1997
3186
3191
33
Papayannopoulou
T
Nakamoto
B
Andrews
RG
Lyman
SD
Lee
MY
In vivo effects of Flt3/Flk2 ligand on mobilization of hematopoietic progenitors in primates and potent synergistic enhancement with granulocyte colony-stimulating factor.
Blood.
90
1997
620
629
34
Rosenzweig
M
MacVittie
TJ
Harper
D
et al
Efficient and durable gene marking of hematopoietic progenitor cells in nonhuman primates after nonablative conditioning.
Blood.
94
1999
2271
2286
35
Miller
AD
Buttimore
C
Redesign of retrovirus packaging cell lines to avoid recombination leading to helper virus production.
Mol Cell Biol.
6
1986
2895
2902
36
Cassel
A
Cottler-Fox
M
Doren
S
Dunbar
CE
Retroviral-mediated gene transfer into CD34enriched human peripheral blood stem cells.
Exp Hematol.
21
1993
585
591
37
Shi
PA
Pomper
GJ
Metzger
ME
Donahue
RE
Leitman
SF
Dunbar
CE
Assessment of rapid remobilization intervals with G-CSF and SCF in murine and rhesus macaque models.
Transfusion.
41
2001
1438
1444
38
Tisdale
JF
Hanazono
Y
Sellers
SE
et al
Ex vivo expansion of genetically marked rhesus peripheral blood progenitor cells results in diminished long-term repopulating ability.
Blood.
92
1998
1131
1141
39
Takatoku
M
Sellers
S
Agricola
BA
et al
Avoidance of stimulation improves engraftment of cultured and retrovirally transduced hematopoietic cells in primates.
J Clin Invest.
108
2001
447
455
40
Kluge
KA
Bonifacino
AC
Sellers
S
Agricola
BA
Donahue
RE
Dunbar
CE
Retroviral transduction and engraftment ability of primate hematopoietic progenitor and stem cells transduced under serum-free versus serum-containing conditions.
Mol Ther.
5
2002
316
322
41
Weaver
A
Ryder
D
Crowther
D
Dexter
TM
Testa
NG
Increased numbers of long-term culture-initiating cells in the apheresis product of patients randomized to receive increasing doses of stem cell factor administered in combination with chemotherapy and a standard dose of granulocyte colony-stimulating factor.
Blood.
88
1996
3323
3328
42
Hess
DA
Levac
KD
Karanu
FN
et al
Functional analysis of human hematopoietic repopulating cells mobilized with granulocyte colony-stimulating factor alone versus granulocyte colony-stimulating factor in combination with stem cell factor.
Blood.
100
2002
869
878
43
Miller
DG
Adam
MA
Miller
AD
Gene transfer by retrovirus vectors occurs only in cells that are actively replicating at the time of infection.
Mol Cell Biol.
10
1990
4239
4242
44
Roe
T-Y
Reynolds
TC
Yu
G
Brown
PO
Integration of murine leukemia virus DNA depends on mitosis.
EMBO J.
12
1993
2099
2108
45
Hajihosseini
M
Iavachev
L
Price
J
Evidence that retroviruses integrate into post-replication host DNA.
EMBO J.
12
1993
4969
4974
46
Gothot
A
van der Loo
JCM
Clapp
DW
Srour
EF
Cell cycle-related changes in repopulating capacity of human mobilized peripheral blood CD34+ cells in non-obese diabetic/severe combined immune-deficient mice.
Blood.
92
1998
2641
2649
47
Kittler
EL
Peters
SO
Crittenden
RB
et al
Cytokine-facilitated transduction leads to low-level engraftment in nonablated hosts.
Blood.
90
1997
865
872
48
Dorrell
C
Gan
OI
Pereira
DS
Hawlet
RG
Dick
JE
Expansion of human cord blood CD34+CD38- cells in ex vivo culture during retroviral transduction without a corresponding increase in SCID repopulating cell (SRC) frequency: dissociation of SRC phenotype and function.
Blood.
95
2000
102
110
49
Dao
MA
Hannum
CH
Kohn
DB
Nolta
JA
FLT3 ligand preserves the ability of human CD34+ progenitors to sustain long-term hematopoiesis in immune-deficient mice after ex vivo retroviral-mediated transduction.
Blood.
89
1997
446
456
50
Duarte
RF
Frank
DA
SCF and G-CSF lead to the synergistic induction of proliferation and gene expression through complementary signaling pathways.
Blood.
96
2000
3422
3430
51
Szilvassy
SJ
Meyerrose
TE
Ragland
PL
Grimes
B
Differential homing and engraftment properties of hematopoietic progenitor cells from murine bone marrow, mobilized peripheral blood, and fetal liver.
Blood.
98
2001
2108
2115
52
Peters
SO
Kittler
ELW
Ramshaw
HS
Quesenberry
PJ
Murine marrow cells expanded in culture with IL-3, IL-6, IL-11, and SCF acquire an engraftment defect in normal hosts.
Exp Hematol.
23
1995
461
466
53
Traycoff
CM
Cornetta
K
Yoder
MC
Davidson
A
Srour
EF
Ex vivo expansion of murine hematopoietic progenitor cells generates classes of expanded cells processing different levels of bone marrow repopulating potential.
Exp Hematol.
24
1996
299
306
54
Habibian
HK
Peters
SO
Hsieh
CC
et al
The fluctuating phenotype of the lymphohematopoietic stem cell with cell cycle transit.
J Exp Med.
188
1998
393
398
55
Orlic
D
Girard
LJ
Jordan
CT
Anderson
SM
Cline
AP
Bodine
DM
The level of mRNA encoding the amphotropic retrovirus receptor in mouse and human hematopoietic stem cells is low and correlates with the efficiency of retrovirus transduction.
Proc Natl Acad Sci U S A.
93
1996
11097
11102
56
Orlic
D
Girard
LJ
Anderson
SM
et al
Identification of human and mouse hematopoietic stem cell populations expressing high levels of mRNA encoding retrovirus receptors.
Blood.
91
1998
3247
3254

Author notes

Cynthia E. Dunbar, Hematology Branch, NHLBI, NIH, Building 10, Rm 7C103, 9000 Rockville Pike, Bethesda, MD 20892; e-mail: dunbarc@nhlbi.nih.gov.

Sign in via your Institution