Administration of hematopoietic growth factors is being used increasingly to obtain populations of blood progenitor/stem cells (PBPC) for clinical transplantation. Here we examined the effect of combining stem cell factor (SCF ) and granulocyte colony-stimulating factor (G-CSF ) versus G-CSF alone in a randomized clinical study involving 62 women with early-stage breast cancer. In the first patient cohorts, escalating doses of SCF were administered for 7 days with concurrent G-CSF administration. At baseline, levels of progenitor cells in the bone marrow or blood were comparable in the different patient groups. As with administration of G-CSF alone, the combination of SCF plus G-CSF did not alter the wide variation in levels of PBPC observed between individuals and did not alter the selective nature of PBPC release, with preferential release of day-14 granulocyte-macrophage colony-stimulating factor (GM-CFC) versus day-7 GM-CFC. However, SCF acted to sustain the levels of PBPC after cessation of growth factor treatment; levels of PBPC were elevated 100-fold at later timepoints compared with G-CSF alone. In addition, the maximum levels of PBPC observed were increased approximately fivefold at day 5 of growth-factor administration. The increased levels of PBPC resulted in significantly increased levels of PBPC obtained by leukapheresis. In a subsequent patient cohort, 3-days pretreatment with SCF was introduced and followed by 7 days concurrent SCF plus G-CSF. The 3-days pretreatment with SCF resulted in an earlier wave of PBPC release in response to commencement of G-CSF. In addition, maximum PBPC levels in blood and PBPC yield in leukapheresis products were further increased. Unexpectedly however, SCF pretreatment resulted in progenitor cells with enhanced self-generation potential. Recloning assays documented the ability of approximately 30% of primary granulocyte-macrophage (GM) colonies from control cell populations to generate secondary GM colonies (n = 1,106 primary colonies examined). In contrast approximately 90% of GM colonies from PBPC after SCF pretreatment generated secondary clones and 65% generated secondary colonies. The action of SCF was not explicable in terms of altered SCF, GM-CSF, or G-CSF responsiveness, but SCF pretreatment was associated with maximum serum SCF levels at the time G-CSF was commenced. These results show that PBPC populations mobilized by different growth factor regimens can differ in their functional properties and caution against solely considering number of harvested progenitor cells without regard to their function.

THE ABILITY OF hematopoietic growth factors to mobilize progenitor cells and stem cells into the blood has substantially impacted on clinical bone marrow transplantation and high-dose chemotherapy procedures.1-7 This action of growth factors is seen in humans, for example with granulocyte colony-stimulating factor (G-CSF ),8-11 granulocyte-macrophage colony-stimulating factor (GM-CSF ),12-14 interleukin-3 (IL-3),15,16 and more recently with the mpl-ligand, megakaryocyte growth and development factor (MGDF ).17,18 Although the mechanism of this effect remains unknown, it is probable that it involves cell adhesion components of the hematopoietic microenvironment such as fibronectin and molecules (ie, VLA-4 and VLA-5).19-22 The widespread clinical application of peripheral blood progenitor/stem cell (PBPC) support after high-dose chemotherapy has been limited by both the frequent requirement for multiple collection procedures and the wide variation in response to growth factors observed with different individuals.

The hematopoietic regulator stem cell factor (SCF, also known as kit-ligand or Steel Factor)23,24 has been shown to enhance colony formation by primitive cell populations and displays synergistic activity on myeloid, erythroid, and megakaryocyte progenitors.25-29 In addition to its action to enhance the survival of stem cell populations in vitro, SCF displays multilineage actions in vivo that include expansion of the stem cell compartment.30-32 The action of SCF to mobilize PBPC has also been examined in preclinical models. For example, administration of SCF to monkeys resulted in an up to 300-fold increase in levels of PBPC.30,32 In mice, dogs, and baboons there is evidence suggesting that the combination of SCF plus G-CSF is synergistic, resulting in an increase in the levels of blood progenitor cells.33-36 However, some of these studies lacked a control group that received G-CSF alone.37 

Information on the action of SCF in humans is scant.38 Reports of two early studies have been published in abstract form.39-42 Both studies documented transient and reversible dermatological reactions of the site of injection. One study reported increased levels of progenitor cells per mL of bone marrow, but no changes in blood progenitor cells.43 The action of SCF in combination with G-CSF has also been described in abstract form.44-46 

The study described here was performed to examine the action of SCF plus G-CSF on levels of PBPC in humans. Given the activity of SCF in preclinical studies, we reasoned that the combination with G-CSF might serve to increase the levels of blood progenitor cells and to reduce some of the wide variation seen in individual responses to G-CSF alone.

Clinical details. Patients had early-stage (stage II or III) breast cancer with poor prognostic features, were chemotherapy naive, and all gave informed consent according to a protocol approved by the institutional ethics committees. In the prechemotherapy phase, patients were randomly assigned in the ratio of 2:1 to receive either SCF plus G-CSF or G-CSF alone. The dose of SCF (recombinant methionyl human SCF [r-metHuSCF ]) was escalated in individual patient cohorts (5, 10, or 15 μg/kg/d for 7 days given by single subcutaneous injection), whereas G-CSF was maintained at 12 μg/kg/d and administered as a continuous subcutaneous infusion for 6 days as previously described.5,7 Hereafter the 7 days concurrent administration schedule of SCF plus G-CSF will be referred to as “SCF plus G-CSF.” The number of individuals in each cohort was: G-CSF alone, n = 18 (Cohort A); SCF 5 μg/kg/d plus G-CSF, n = 5 (Cohort B); SCF 10 μg/kg/d plus G-CSF, n = 11 (Cohort C); SCF 15 μg/kg/d plus G-CSF, n = 10 (Cohort D). Subsequently, patients were randomized to receive G-CSF alone versus G-CSF plus 10 days of SCF 10 μg/kg/d of which 3 days of SCF was given before commencement of G-CSF (Cohort E, n = 18). Hereafter this treatment schedule will be referred to as “SCF pretreatment.” For the final 8 patients enrolled in the study, the randomization was between Cohorts C and E and not to Cohort A. For all cohorts, leukapheresis was performed on 3 consecutive days (days 5, 6, 7 relative to commencement of G-CSF ). Patients then went on to receive high-dose chemotherapy with hematopoietic cell support. Patient cohorts were matched in terms of age and stage of disease, although more patients receiving 15 μg/kg/d SCF plus G-CSF had stage III disease (90% versus 50% of G-CSF–alone patients). Specific patient details, safety, and clinical outcomes have been previously reported in abstract form47-49 and will be described in detail elsewhere.

Progenitor cell assays. Bone marrow samples were taken at baseline. Blood samples were taken at baseline, daily during treatment with growth factor and daily after cessation of growth factors.

Blood and bone marrow samples were separated using Ficoll-Paque. Mononuclear cells were washed resuspended and cell counts performed with eosin and crystal violet as previously described.50 Mononuclear cells were examined in triplicate cultures using 104 and 105 viable cells per mL. Cultures were stimulated with G-CSF (500 U/mL), GM-CSF (100 ng/mL), SCF (100 ng/mL), and granulocyte-macrophage (GM) colonies enumerated 14 days later (d14 GM-CFC). For the purpose of this study all colonies grown under these conditions are refered to as d14 GM-CFC and include, for example, Eosinophil-CFC. Cultures stimulated with G-CSF (500 U/mL), and GM-CSF (100 ng/mL) were examined after 7 days (d7 GM-CFC). These culture conditions were deliberately chosen to allow direct comparison of d7 GM-CFC with results from earlier studies.51-54 Cultures stimulated with GM-CSF (100 ng/mL), IL-3 (100 ng/mL), IL-6 (100 ng/mL), SCF (100 ng/mL), and erythropoietin (Epo, 5 U/mL) were examined after 14 days for erythroid colony formation. All cultures were examined by a single observer (GB), blinded to the treatment schedule, using a dissection microscope and with the scoring procedure validated as previously described.50 Results are presented only for those cultures that generated fewer than 250 colonies per plate (typically less than 100 colonies per plate). On some occasions, cell samples were prepared and stored overnight at room temperature; this procedure was shown not to substantially alter colony numbers.50 

In some experiments, serial dilutions of growth factors (G-CSF, GM-CSF, or SCF ) in the absence of any other stimulus were used to stimulate colony formation. All experiments were performed in replicate cultures. All cultures were performed in agar as previously described.51 52 Briefly, a mixture of 0.3% agar, Iscove's Modified Dulbecco's Medium (IMDM), and 25% preselected newborn bovine calf serum was prepared. The mononuclear cells were added and 1 mL dispensed into 35-mm petri dishes that contained the mixture of purified, recombinant growth factors (all provided by Amgen, Thousand Oaks, CA). The agar was allowed to gel and cultures placed in a humidified atmosphere of 5% CO2 in air at 37°C.

Recloning experiments. Sequential individual colonies were removed using a Pasteur pipette as previously described.55 A minimum of 50 colonies was examined in each experiment. Day-14 GM colonies were isolated both from cultures stimulated with G-CSF, GM-CSF, SCF, and from cultures stimulated with GM-CSF, IL-3, IL-6, SCF, and Epo. Under the former culture conditions, colonies of all morphologies were isolated; under the latter culture conditions only GM colonies were isolated (colonies with any erythroid characteristics were ignored). Isolated colonies were deposited into 0.2 mL IMDM and a single cell suspension generated by vigorous pipetting. The cell suspension was then recultured in agar cultures stimulated with SCF (100 ng/mL), GM-CSF (100 ng/mL), and G-CSF (500 U/mL). After a further 14 days of incubation, the total number of clones (>3 cells) and the total number of colonies (>40 cells) was enumerated.

Pharmacokinetic assays. SCF levels were determined on serum samples from all patients taken immediately before administration of SCF (trough levels of SCF ). ELISA kits were used according to the manufacturers instructions (R & D Systems, Minneapolis, MN). All data were corrected for endogenous SCF by subtraction of the individual's zero time concentration. All serum concentrations of SCF remained above baseline until at least day 8.

Baseline levels of progenitor cells. All patients had normal baseline hematological parameters and a normal bone marrow biopsy at commencement of the study. Figure 1A shows the baseline values for bone marrow progenitor cells. There was no difference in levels of bone marrow progenitor cells for patients assigned to receive SCF plus G-CSF versus G-CSF alone. The frequency of mature (day 7, d7) and immature (day 14, d14) granulocyte-macrophage colony-forming cells (GM-CFC) and erythroid progenitor cells (BFU-E) is shown for all patients. There was a 100-fold variation in the frequency of progenitor cells for these individuals with normal bone marrow function. However, the frequency of d7 GM-CFC in the bone marrow was comparable to the frequency of d14 GM-CFC and BFU-E. The baseline level of PBPC was also similar for patients assigned to treatment with or without SCF (Fig 1B). Again there was an approximately 100-fold variation between these individuals. However, in contrast to the results for the bone marrow, the number of mature d7 GM-CFC in blood was approximately 10-fold less than the number of d14 GM-CFC or BFU-E.

Fig. 1.

Baseline values for number of progenitor cells in (A) bone marrow (results expressed per 105 mononuclear cells) and (B) blood (results expressed per mL blood). Results are mean of triplicate cultures for 62 individuals. Patients were randomized to receive G-CSF alone (•, n = 18) or G-CSF plus SCF (○, n = 44). Median results are indicated by horizontal line.

Fig. 1.

Baseline values for number of progenitor cells in (A) bone marrow (results expressed per 105 mononuclear cells) and (B) blood (results expressed per mL blood). Results are mean of triplicate cultures for 62 individuals. Patients were randomized to receive G-CSF alone (•, n = 18) or G-CSF plus SCF (○, n = 44). Median results are indicated by horizontal line.

Close modal

SCF causes sustained elevation in blood progenitor cell levels. Levels of blood progenitor cells were quantitated during and after the administration of growth factors. The wide interindividual variation noted at baseline was also evident following treatment with G-CSF (Fig 2, closed circles). The changes in PBPC occurred in parallel for each cell type with maximum levels at days 5 and 6 of G-CSF treatment. There was an approximately 200-fold increase in d7 GM-CFC, d14 GM-CFC, and BFU-E (median d14 GM-CFC levels at baseline, 100 per mL; median levels at day 6, 20,575 per mL). The level of d7 GM-CFC was consistently lower than d14 GM-CFC or BFU-E despite comparable levels at baseline in the bone marrow (Fig 1A). This difference was approximately 50-fold at the times of peak progenitor cell levels. The level of PBPC following G-CSF treatment had returned to baseline values by day 12 for d7 GM-CFC and day 14 for BFU-E and d14 GM-CFC.

Fig. 2.

Levels of blood progenitor cells for patients treated with G-CSF alone (•, n = 18) or G-CSF plus SCF 10 μg/kd/d (n = 11) for 7 days (○), showing d7 GM-CFC (A), d14 GM-CFC (B), and BFU-E (C). Results are means of triplicate cultures. Median results are indicated for each cohort by horizontal line. Baseline values (day 1) were taken before administration of growth factors. Note the elevated levels of progenitor cells in the G-CSF plus SCF cohort despite similar levels at baseline.

Fig. 2.

Levels of blood progenitor cells for patients treated with G-CSF alone (•, n = 18) or G-CSF plus SCF 10 μg/kd/d (n = 11) for 7 days (○), showing d7 GM-CFC (A), d14 GM-CFC (B), and BFU-E (C). Results are means of triplicate cultures. Median results are indicated for each cohort by horizontal line. Baseline values (day 1) were taken before administration of growth factors. Note the elevated levels of progenitor cells in the G-CSF plus SCF cohort despite similar levels at baseline.

Close modal

Figure 2 also shows the results for all patients who received SCF 10 μg/kg/d plus G-CSF (n = 11). Similar results were seen with all SCF plus G-CSF cohorts (see below), including the dissociation between d14 GM-CFC and d7 GM-CFC. At day 5 there was an approximately 200-fold increase in levels of d7 GM-CFC and 400-fold increase in d14 GM-CFC and BFU-E. Furthermore, the addition of SCF did not alter the 100-fold variation in PBPC levels observed with different individuals. However, unlike levels of PBPC with G-CSF alone, the number of PBPC remained elevated following cessation of growth factors. Thus, at days 10 through 14, the number of PBPC was elevated 10- to 100-fold (Fig 2) compared with levels of PBPC observed with G-CSF. A similar sustained elevation of PBPC was observed with all doses of SCF plus G-CSF (Fig 3), and PBPC levels did not return to baseline levels until after day 14. This difference was highly significant (P < .001 for G-CSF alone v all SCF cohorts at day 14, Wilcoxon Rank-Sum test). The sustained elevation of PBPC levels at later time points clearly established a biological effect of SCF above the action of G-CSF alone.

Fig. 3.

Mean levels of blood white cell counts and progenitor cells for patient cohorts treated with G-CSF alone (•, n = 18), SCF 5 μg/kg/d plus G-CSF (○, n = 5), SCF 10 μg/kg/d plus G-CSF (▪, n = 11), or SCF 15 μg/kg/d plus G-CSF (□, n = 10). Results are also shown for the patient cohort that received SCF pretreatment for 3 days and 7 days concurrent treatment with G-CSF (▴, n = 18). Time is shown relative to the commencement of G-CSF. Results shown are white cell count (A), d7 GM-CFC (B), d14 GM-CFC (C), and BFU-E (D).

Fig. 3.

Mean levels of blood white cell counts and progenitor cells for patient cohorts treated with G-CSF alone (•, n = 18), SCF 5 μg/kg/d plus G-CSF (○, n = 5), SCF 10 μg/kg/d plus G-CSF (▪, n = 11), or SCF 15 μg/kg/d plus G-CSF (□, n = 10). Results are also shown for the patient cohort that received SCF pretreatment for 3 days and 7 days concurrent treatment with G-CSF (▴, n = 18). Time is shown relative to the commencement of G-CSF. Results shown are white cell count (A), d7 GM-CFC (B), d14 GM-CFC (C), and BFU-E (D).

Close modal

The mean results for GM-CFC and BFU-E are compared with changes in the white cell count in Fig 3. An increase in white cell count was clearly evident for all cohorts within 24 hours of commencing G-CSF and reached a maximum approximately 10-fold above baseline. The peak white cell count was approximately 1.5-fold greater with SCF 10 μg/kg/d plus G-CSF compared with G-CSF alone and with the suggestion of an SCF dose response. In all patient cohorts the white cell count fell quickly following cessation of growth factors and had returned to normal by day 14. The kinetics of PBPC mobilization were quite different. The earliest detectable increase was evident at day 3 and the kinetics of PBPC release were remarkably consistent for each progenitor cell type and all patient cohorts. However, with all SCF plus G-CSF cohorts, the discordance between the white cell count and levels of PBPC at later timepoints was notable: the frequency of PBPC amongst nucleated cells at day 12 was increased 10-fold compared with PBPC levels at day 5.

The maximum levels of PBPC observed were greater in the SCF 10 μg/kg/d plus G-CSF and SCF 15 μg/kg/d plus G-CSF cohorts when compared with G-CSF alone. With concurrent 7-day administration of SCF 10 μg/kg/d plus G-CSF, there was an approximately fivefold increase in the maximum levels of d7 GM-CFC, d14 GM-CFC, and BFU-E at days 5 and 6 (P < .001 for d14 GM-CFC at day 5 v levels with G-CSF alone, Wilcoxon Rank-Sum test). Results were similar with SCF 15 μg/kg/d plus G-CSF. Based on these results, the addition of SCF to G-CSF acted to increase the maximum number of PBPC approximately fivefold and sustained the level of these cells in the blood, despite falling white cell counts.

Pretreatment with SCF further enhanced progenitor cell mobilization. SCF acted to generate a sustained elevation of PBPC when combined with G-CSF. We therefore reasoned that manipulation of the administration schedule might allow the peak levels of PBPC seen with G-CSF alone (day 5 to 6) to become coincident with the later mobilization of PBPC seen with SCF plus G-CSF cohorts. A 3-day pretreatment schedule with SCF 10 μg/kg/d was therefore introduced. These patients then continued to receive SCF 10 μg/kg/d plus G-CSF for a further 7 days (SCF pretreatment cohort).

This SCF-pretreatment schedule resulted in an earlier wave of PBPC release following commencement of G-CSF. The rise in blood PBPC was evident at least 1 day earlier for patients who received SCF pretreatment (Fig 3). Thus, the earliest detectable increase in PBPC was at day 2 relative to the commencement of G-CSF. This suggested that SCF pretreatment served to expand a population of cells available for G-CSF–induced mobilization. In addition, the peak level of PBPC was significantly greater than with G-CSF alone and superior to all other SCF plus G-CSF cohorts. The action of SCF to maintain sustained levels of PBPC was also evident with this cohort of patients; at day 12 the level of progenitor cells of each type was approximately fivefold greater than for the equivalent patient group treated concurrently with SCF 10 μg/kg/d plus G-CSF.

Enhanced yields of progenitor cells with SCF plus G-CSF. In all patients leukapheresis was performed on days 5, 6, and 7 as defined relative to the administration of G-CSF. In general, the level of progenitor cells in the blood correlated well with the number of progenitor cells collected (for d7 GM-CFC r = .78, P < .0001; for d14 GM-CFC r = .63, P < .0001; for BFU-E r = .59, P < .0001).

Results of analysis of leukapheresis collections according to patient cohort are shown for all patients in Fig 4. There was no difference in the number of d7 GM-CFC, d14 GM-CFC, BFU-E, or CD34+ cells collected from patients who received G-CSF alone versus SCF 5 μg/kg/d plus G-CSF. However there was a significant increase in the number of d7 GM-CFC, d14 GM-CFC, BFU-E, and CD34+ cells when G-CSF alone was compared with concurrent SCF 10 μg/kg/d plus G-CSF (eg, P < .013 for CD34+ cells; P < .07 for d14 GM-CFC, Wilcoxon Rank-Sum test). There was a further, approximately twofold, increase in number of progenitor cells of all types obtained from patients who received SCF pretreatment when compared with SCF 10 μg/kg/d plus G-CSF. This was significantly different when compared with patients who received G-CSF alone (P < .0001 for d14 GM-CFC; P < .0005 for CD34+ cells, Wilcoxon Rank-Sum test). In addition, SCF pretreatment was superior to concurrent administration of SCF 10 μg/kg/d plus G-CSF in terms of progenitor cells collected (P < .03 for d14 GM-CFC; P = .70 for CD34+ cells, Wilcoxon Rank-Sum test).

Fig. 4.

Leukapheresis collections from days 5 plus 6 plus 7 (relative to G-CSF administration) for each patient cohort: G-CSF alone (G-CSF, ▪); SCF 5 μg/kg/d plus G-CSF (SCF 5, ); SCF 10 μg/kg/d plus G-CSF (SCF 10, ▨); SCF 15 μg/kg/d plus G-CSF (SCF 15, ); and SCF pretreatment (SCF pre, □). Results are mean ± SE for d7 GM-CFC (A), d14 GM-CFC (B), BFU-E (C), and CD34+ cells (D). The apparent decrease in GM-CFC collected in Cohort D was interpreted as reflecting the smaller number of patients in this cohort, the wide interindividual variation, and the increased number of patients with stage III disease in this group.

Fig. 4.

Leukapheresis collections from days 5 plus 6 plus 7 (relative to G-CSF administration) for each patient cohort: G-CSF alone (G-CSF, ▪); SCF 5 μg/kg/d plus G-CSF (SCF 5, ); SCF 10 μg/kg/d plus G-CSF (SCF 10, ▨); SCF 15 μg/kg/d plus G-CSF (SCF 15, ); and SCF pretreatment (SCF pre, □). Results are mean ± SE for d7 GM-CFC (A), d14 GM-CFC (B), BFU-E (C), and CD34+ cells (D). The apparent decrease in GM-CFC collected in Cohort D was interpreted as reflecting the smaller number of patients in this cohort, the wide interindividual variation, and the increased number of patients with stage III disease in this group.

Close modal

Enhanced clonogenic capacity of cells mobilized by SCF pretreatment. Given both the action of SCF on stem cells and the increased numbers of PBPC generated in response to SCF, experiments were performed to document the self-renewal/self-generation characteristics of these cells. Colonies derived from PBPC were isolated, suspended, and recultured to assess recloning potential as a measure of self-generation. The number of secondary clones and secondary colonies derived from each primary colony was documented. Table 1 shows the number of primary GM colonies that contained cells capable of giving rise to secondary clones. An unexpectedly high percentage of GM colonies derived from bone marrow cells generated secondary clones. This was between 49% and 73% (mean 59%) with different bone marrow samples. Moreover, approximately 30% of primary GM colonies gave rise to secondary colonies (range 22% to 47%). Similar results were observed with GM colonies derived from blood: after 5 days of treatment with G-CSF or treatment with SCF 10 μg/kg/d plus G-CSF, approximately 30% of primary GM colonies generated secondary colonies. Although there was some variation between experiments, the results were not significantly different for bone marrow cells versus blood cells from patients who received G-CSF alone or SCF plus G-CSF. The recloning potential was independent of the growth stimulus in the primary culture (G-CSF, GM-CSF, and SCF v GM-CSF, SCF, IL-3, IL-6, and Epo; data not shown). Thus, based on an analysis of over 380 primary GM colonies from G-CSF alone and SCF plus G-CSF mobilized PBPC, the percentage of primary colonies that generated secondary colonies was 29% for both groups. Similar results were obtained when blood PBPC were taken 12 days after commencing growth factor (number secondary clones 36%, secondary colonies 16% for G-CSF alone [n = 385 primary colonies examined]; number secondary clones 35%, secondary colonies 19% for SCF 10 μg/kg/d plus G-CSF [n = 281 primary colonies analyzed]).

Table 1.

Clonogenicity of Primary GM Colonies

Cell SourcePercent Primary Colonies ContainingNo. Clones Generated per Primary Colony.
Clonogenic CellsMedian (range)
Secondary ClonesSecondary ColoniesSecondary ClonesSecondary Colonies
 
BM, baseline 49 27 17 (2-≥300) 4 (1-89) 
(n = 338 primary colonies) 56 28 30 (2-≥300) 5 (1-76) 
 58 22 3 (2-210) 2 (1-45) 
 73 47 5 (2-86) 3 (1-37) 
Mean 59 31   
G-CSF, d5 41 19 8 (2-83) 2 (1-27) 
(n = 383 primary colonies) 48 31 4 (2-49) 3 (1-32) 
 63 34 24 (2-≥300) 4 (1-49) 
 68 31 37 (2-169) 3 (1-23) 
Mean 55 29 
SCF + G-CSF, d5 34 14 5 (2-96) 2 (1-11) 
(n = 465 primary colonies) 38 28 9 (2-68) 2 (1-18) 
 72 58 53 (2-≥300) 9 (1-143) 
 75 33 15 (2-165) 3 (1-130) 
Mean 55 33 
SCF pretreatment, d5 87 44 63 (2-≥300) 5 (1-103) 
(n = 336 primary colonies) 87 69 26 (2-≥300) 5 (1-179) 
 94 69 85 (2-≥300) 4 (1-130) 
 94 86 105 (2-≥300) 23 (1-223) 
Mean 91 67 
Cell SourcePercent Primary Colonies ContainingNo. Clones Generated per Primary Colony.
Clonogenic CellsMedian (range)
Secondary ClonesSecondary ColoniesSecondary ClonesSecondary Colonies
 
BM, baseline 49 27 17 (2-≥300) 4 (1-89) 
(n = 338 primary colonies) 56 28 30 (2-≥300) 5 (1-76) 
 58 22 3 (2-210) 2 (1-45) 
 73 47 5 (2-86) 3 (1-37) 
Mean 59 31   
G-CSF, d5 41 19 8 (2-83) 2 (1-27) 
(n = 383 primary colonies) 48 31 4 (2-49) 3 (1-32) 
 63 34 24 (2-≥300) 4 (1-49) 
 68 31 37 (2-169) 3 (1-23) 
Mean 55 29 
SCF + G-CSF, d5 34 14 5 (2-96) 2 (1-11) 
(n = 465 primary colonies) 38 28 9 (2-68) 2 (1-18) 
 72 58 53 (2-≥300) 9 (1-143) 
 75 33 15 (2-165) 3 (1-130) 
Mean 55 33 
SCF pretreatment, d5 87 44 63 (2-≥300) 5 (1-103) 
(n = 336 primary colonies) 87 69 26 (2-≥300) 5 (1-179) 
 94 69 85 (2-≥300) 4 (1-130) 
 94 86 105 (2-≥300) 23 (1-223) 
Mean 91 67 

GM colonies were grown for 14 days in primary agar cultures, single colonies isolated and recultured for a further 14 days. The number of secondary clones (and colonies) was enumerated and is shown as a percentage. The cell source was bone marrow (BM) taken at baseline or blood taken at day 5 relative to G-CSF administration. Blood was taken after patients received either G-CSF alone, SCF 10 μg/kg/d plus G-CSF for 5 days, or SCF pretreatment. To determine the number of clonogenic cells a mean of 89 colonies was recloned on each occasion (with a mean of 57 colonies generating secondary clones). The number of clones (and colonies) generated per primary colony is also shown. Results were obtained from 4 different patients for each cohort.

The recloning potential of primary GM colonies from patients who received SCF pretreatment was quite different (Table 1). Approximately 90% of primary GM colonies gave rise to secondary clones. This was significantly different to the behavior of primary GM colonies from the other sources (eg, P < .002 for SCF pretreatment v G-CSF alone, unpaired t-test). In addition 65% of primary GM colonies generated secondary colonies (eg, P < .006 for SCF pretreatment v G-CSF alone, unpaired t-test). Moreover, the number of secondary clones generated per primary colony was also significantly increased (Table 1) with numerous primary GM colonies generating over 300 secondary clones (eg, P < .03 for SCF pretreatment v G-CSF alone, unpaired t-test). The number of secondary colonies generated per primary GM colony was also increased, with a maximum of 223 secondary colonies from one primary colony (v a maximum of, eg, 49 colonies from G-CSF PBPC). These secondary colonies were also larger in size (average 1,000 cells per colony, n = 70 colonies).

The high recloning potential of bone marrow and blood GM progenitor cells was unexpected and suggests that these cells have higher self-generation capacity than previously suspected. It seemed most likely that this unexpected result reflected the presence of SCF in the culture dish. In one experiment using bone marrow–derived GM colonies, this idea was supported (with G-CSF and GM-CSF in primary cultures, 11 of 49 colonies gave secondary clones and 5 of 49 gave secondary colonies v G-CSF, GM-CSF, and SCF in primary cultures, in which 30 of 48 gave secondary clones and 18 of 48 gave secondary colonies).

One possible explanation for the enhanced PBPC response with SCF was altered G-CSF responsiveness following SCF administration. The in vitro dose response of PBPC was therefore examined (Fig 5). Response to G-CSF was unchanged when compared with responsiveness of bone marrow cells before growth factor treatment. This was also true for responsiveness to GM-CSF (data not shown) and SCF (Fig 5) regardless of the growth factor combination used to elicit PBPC. Thus, although heightened G-CSF responsiveness has been documented for some subsets of bone marrow progenitor cells,52 this was not observed for populations of growth factor–mobilized PBPC.

Fig. 5.

Growth factor responsiveness was unchanged following SCF administration. Results are shown for G-CSF (•) and SCF (○) titration curves examining d14 GM-CFC in (A) bone marrow at baseline and (B) after 5 days of growth factor treatment (SCF 10 μg/kg/d plus G-CSF ). Similar results were obtained for 4 individuals and for cells analyzed at day 12 of study (data not shown).

Fig. 5.

Growth factor responsiveness was unchanged following SCF administration. Results are shown for G-CSF (•) and SCF (○) titration curves examining d14 GM-CFC in (A) bone marrow at baseline and (B) after 5 days of growth factor treatment (SCF 10 μg/kg/d plus G-CSF ). Similar results were obtained for 4 individuals and for cells analyzed at day 12 of study (data not shown).

Close modal

Serum SCF levels. Given the action of SCF pretreatment, studies were also performed to examine the levels of SCF following different schedules of administration. Results are shown in Fig 6. The serum levels of SCF attained were similar with 7 doses of SCF at either 10 μg/kg/d or 15 μg/kg/d. Maximum levels were achieved after 3 doses and did not continue to rise thereafter. As with other growth factors, the levels then began to fall as the white cell count began to rise. Levels returned to baseline 2 days after cessation of administration (day 9 of study).

Fig. 6.

Serum levels of SCF. Blood was taken before administration of each dose of SCF. Results are mean ± SE for SCF 5 μg/kg/d plus G-CSF (Cohort B, n = 5, ▿), SCF 10 μg/kg/d plus G-CSF (Cohort C, n = 11, •), SCF 15 μg/kg/d plus G-CSF (Cohort D, n = 10, □), and SCF pretreatment (Cohort E, 10 μg/kg, n = 18, ▴). Time is shown relative to administration of G-CSF. Serum concentration is corrected for baseline values.

Fig. 6.

Serum levels of SCF. Blood was taken before administration of each dose of SCF. Results are mean ± SE for SCF 5 μg/kg/d plus G-CSF (Cohort B, n = 5, ▿), SCF 10 μg/kg/d plus G-CSF (Cohort C, n = 11, •), SCF 15 μg/kg/d plus G-CSF (Cohort D, n = 10, □), and SCF pretreatment (Cohort E, 10 μg/kg, n = 18, ▴). Time is shown relative to administration of G-CSF. Serum concentration is corrected for baseline values.

Close modal

With SCF pretreatment, maximum serum levels were achieved after 3 doses and before administration of G-CSF was commenced (day 0 of study). Following commencement of G-CSF, levels fell as the white cell count began to rise. SCF levels returned to their baseline value 2 days after cessation of growth factor. Thus, SCF pretreatment for 3 days effectively attained maximal levels of SCF before administration of G-CSF.

In this report we have shown that coadministration of SCF plus G-CSF resulted in a significant increase in blood levels of d7 GM-CFC, d14 GM-CFC, and BFU-E. The action of SCF was most evident at later time points in which levels of PBPC remained elevated after 12 days. In contrast, by this time the levels of PBPC had returned to baseline for patients who received G-CSF alone. SCF in combination with G-CSF also acted to increase the maximum levels of PBPC. This effect was even more evident when individuals were pretreated with SCF for 3 days; the maximum levels of PBPC were greater and the rising levels were detected earlier. The heightened levels of PBPC observed with SCF plus G-CSF were associated with increased levels of progenitor cells, including CD34+ cells, collected by leukapheresis; the significant advantage of concurrent SCF 10 μg/kg/d plus G-CSF over G-CSF alone was even more evident when 3 days of pretreatment with SCF was performed. Unexpectedly, SCF pretreatment generated GM-CFC that displayed enhanced self-renewal potential. There was no difference in the in vitro growth factor responsiveness to G-CSF, GM-CSF, or SCF of progenitor cells following administration of SCF. However, following SCF pretreatment, elevated SCF levels were maximal before G-CSF administration.

This study clearly documented the benefit of combination treatment compared with G-CSF treatment alone. Despite the evidence suggesting SCF alone can increase levels of PBPC in animal models,32-36 this action is modest or undetectable in humans.41-43 In contrast the action of G-CSF to enhance levels of PBPC is well documented.5,8-11 The combination of both factors increased blood levels of d7 GM-CFC, d14 GM-CFC, and BFU-E and resulted in increased leukapheresis yields. This advantage of SCF plus G-CSF over G-CSF alone was highly statistically significant. This increase in PBPC was evident despite the broad interindividual variation that we have consistently observed.5,9,56,57 Although this wide interindividual variation may be further enhanced as a consequence of prior chemotherapy,38,58 this was not a confounding influence in this study; the greater than 100-fold variation reported here was seen in individuals who all had normal bone marrow function and all had no prior treatment. This highlights the problem of attempting to interpret results from nonrandomized studies examining only small numbers of individuals in a given treatment cohort. This, combined with a modest difference between treatment groups, makes it difficult to evaluate the studies of combinations of growth factors reported to date.59-63 

The mechanism of PBPC mobilization remains unclear. However, it is interesting to note that this phenomenon displays selectivity. Despite comparable numbers of d7 GM-CFC and d14 GM-CFC in the bone marrow, there was consistently a 10- to 50-fold difference in the levels of these cells in the blood. This selectively remains a consistent finding8,9,18 and has also been observed in the mouse.64 This suggests that the mobilization seen with G-CSF cannot simply represent nonselective release. Moreover, it is likely that different mechanisms of mobilization will apply to different growth factors. The kinetics of PBPC release seen here as a result of administration of SCF plus G-CSF differed greatly from those seen with G-CSF alone. Similarly, MGDF stimulates release of PBPC, but with kinetics surprisingly different to G-CSF.18 However, like SCF, MGDF also appears to have an enhanced action when combined with G-CSF.17 

An intriguing observation was the earlier mobilization seen as a result of SCF pretreatment. This suggested that the pool of G-CSF–responsive cells was expanded. In addition, SCF pretreatment increased the peak level of PBPC and the yield of PBPC obtained by leukapheresis. Given the similarities between SCF and flk-2/flt-3 ligand,52 65-67 it is possible that the combination of flk-2/flt-3 ligand plus G-CSF might act in an analogous manner, and that pretreatment might also be advantageous.

Somewhat surprisingly, there was no obvious difference in the in vitro behavior of blood progenitor cells generated in response to concurrent coadministration of both factors (the SCF plus G-CSF cohorts).38 With both factors given together for 7 days there were no differences in in vitro growth factor responsiveness, self-generation characteristics, colony size, or colony morphology. However, an increase in long-term culture–initiating cells has been noted in the apheresis products from patients who received SCF plus G-CSF following cyclophosphamide.38 Similarly, although mouse models suggested that the combination of SCF plus G-CSF might produce a population of human cells ideally suited for gene transfer,37 this was not the case in this study.68 There was no evidence that cells generated by the growth factor combination were more susceptible to gene transfer. However, the increased levels of PBPC did ensure an increased number of target cells.68 

The demonstration that approximately 50% of unselected, day-14 GM colonies from bone marrow– or peripheral blood–contained cells were capable of generating secondary clones was unexpected. This estimate was based on an extensive analysis of GM colonies from control cell samples (n = 1,106 colonies examined). Earlier studies have documented the inability of such colonies to give rise to secondary colonies, thus the depiction of progenitor cells without a “self-renewing” arrow on diagrams of the hematopoietic hierarchy.69 Although the generation of secondary clones of less than 40 cells in size probably reflects the presence of promyelocytes and myelocytes within the colony,51 approximately 30% of primary GM colonies generated secondary colonies. This unexpected recloning potential was probably caused by SCF in the culture dish. SCF might allow cells that would otherwise not express their proliferative potential to read-out in a colony assay. In addition, despite its action to increase colony size, SCF may preserve the self-generation capacity of progenitor cells that still retain some more primitive, “stem-cell” characteristics. The SCF pretreatment generated GM-CFC with apparently greater self-generation potential; approximately 90% of primary GM colonies generated secondary clones and 65% generated secondary colonies. This observation was consistent both with the known action of SCF within the stem-cell compartment and with the elevated SCF serum levels observed before G-CSF administration. This property of GM-CFC generated with SCF pretreatment might suggest that these cells could prove superior in clinical transplantation settings.38 Moreover, it cautions against assuming that firstly, all GM-CFC or CD34+ cell populations generated by in vivo administration of growth factors are equivalent in their functional properties and secondly, that GM-CFC assays and CD34 assays are interchangeable. Furthermore, these results imply that PBPC mobilized by different growth factor treatments cannot necessarily be equated in terms of their clinical utility.

Leiomyosarcoma. A 56-year-old woman presented with abdominal pain and a 30-lb weight loss; a retroperitoneal mass was found. By electron microscopy, all features diagnostic for a metastatic leiomyosarcoma were present. These include caveolae, basal lamina, attachment plaques (all best seen at higher magnification in B), and bundles of microfilaments with dense bodies enclosed (closed arrowheads in A). Additional features of these spindle-shaped tumor cells of smooth muscle cell origin include large electron-dense masses of contracted actin filaments (open arrowheads in A), blunt, club-shaped surface projections (seen at higher magnification in B) and electron-lucent cytoplasmic pools representing admixed glycogen and lipid (P). Note that strands of rough endoplasmic reticulum are not dilated and are inconspicuous in these leiomyosarcoma cells displaying primarily a contractile phenotype. The light microscopic immunoperoxidase profile was positive for vimentin, muscle-specific actin and desmin, and negative for S-100, neurofilament, keratin, and α-1-antitrypsin for this tumor. (A) Original magnification × 8,000; (B) Original magnification × 24,000. (Courtesy of Ann M. Dvorak, MD, Department of Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215.)

Leiomyosarcoma. A 56-year-old woman presented with abdominal pain and a 30-lb weight loss; a retroperitoneal mass was found. By electron microscopy, all features diagnostic for a metastatic leiomyosarcoma were present. These include caveolae, basal lamina, attachment plaques (all best seen at higher magnification in B), and bundles of microfilaments with dense bodies enclosed (closed arrowheads in A). Additional features of these spindle-shaped tumor cells of smooth muscle cell origin include large electron-dense masses of contracted actin filaments (open arrowheads in A), blunt, club-shaped surface projections (seen at higher magnification in B) and electron-lucent cytoplasmic pools representing admixed glycogen and lipid (P). Note that strands of rough endoplasmic reticulum are not dilated and are inconspicuous in these leiomyosarcoma cells displaying primarily a contractile phenotype. The light microscopic immunoperoxidase profile was positive for vimentin, muscle-specific actin and desmin, and negative for S-100, neurofilament, keratin, and α-1-antitrypsin for this tumor. (A) Original magnification × 8,000; (B) Original magnification × 24,000. (Courtesy of Ann M. Dvorak, MD, Department of Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215.)

Close modal

We thank the many staff involved in these studies: Geraint Duggan, Daryl Maher, Chris Juttner, Kerrie Clarke, Dora McPhee, Josephine Iaria, Rosemary Rogers, Don Metcalf, Jan Boyd, Liz O'Flaherty, Elizabetta DeLuca, Ian Russell, John Collins, Jeff Szer, Andrew Grigg, Jenny Marty, and Dora Menchaca.

Supported in part by grants from the National Health and Medical Research Council, Canberra, Australia; The Anti-Cancer Council of Victoria, Australia; the Cooperative Research Centre for Cellular Growth Factors; and Amgen, Thousand Oaks, CA.

Address reprint requests to C.G. Begley, MB, BS, PhD, The Walter and Eliza Hall Institute of Medical Research, PO Royal Melbourne Hospital, Victoria, Australia 3050.

1
Siena
S
Bregni
M
Brando
B
Ravagnani
F
Bonadonna
G
Gianni
AM
Circulation of CD34+ hematopoietic stem cells in the peripheral blood of high-dose cyclophosphamide-treated patients: Enhancement by intravenous recombinant human granulocyte-macrophage colony-stimulating factor.
Blood
74
1989
1905
2
Haas
R
Ho
AD
Bredthauer
U
Cayeux
S
Egerer
G
Knauf
W
Hunstein
W
Successful autologous transplantation of blood stem cells mobilized with recombinant human granulocyte-macrophage colony-stimulating factor.
Exp Hematol
18
1990
94
3
Elias
AD
Ayash
L
Anderson
KC
Hunt
M
Wheeler
C
Schwartz
G
Tepler
I
Mazanet
R
Lynch
C
Pap
S
Pelaez
J
Reich
E
Critchlow
J
Demetri
G
Bibbo
J
Schnipper
L
Griffin
JD
Frei
E
Antman
KH
Mobilization of peripheral blood progenitor cells by chemotherapy and granulocyte-macrophage colony-stimulating factor for hematologic support after high-dose intensification for breast cancer.
Blood
79
1992
3036
4
Huan
SD
Hester
J
Spitzer
G
Yau
JC
Dunphy
FR
Wallerstein
RO
Dicke
K
Spencer
V
LeMaistre
CF
Andersson
BS
Deisseroth
AB
Ventura
GJ
Influence of mobilized peripheral blood cells on the hematopoietic recovery by autologous marrow and recombinant human granulocyte-macrophage colony-stimulating factor after high-dose cyclophosphamide, etoposide, and cisplatin.
Blood
79
1992
3388
5
Sheridan
WP
Begley
CG
Juttner
CA
Szer
J
To
LB
Maher
D
McGrath
KM
Morstyn
G
Fox
RM
Effect of peripheral-blood progenitor cells mobilised by filgrastim (G-CSF ) on platelet recovery after high-dose chemotherapy.
Lancet
339
1992
640
6
Shea
TC
Mason
JR
Breslin
M
Bissent
E
Mullen
M
Taetle
R
Reinfusion and serial measurements of carboplatin-mobilized peripheral-blood progenitor cells in patients receiving multiple cycles of high-dose chemotherapy.
J Clin Oncol
12
1994
1012
7
Sheridan
WP
Begley
CG
To
LB
Grigg
A
Szer
J
Maher
D
Green
MD
Rowlings
PA
McGrath
KM
Cebon
J
Dyson
P
Watson
D
Bayly
J
DeLuca
E
Tomita
D
Hoffman
E
Morstyn
G
Juttner
CA
Fox
RM
Phase II study of autologous filgrastim (G-CSF )-mobilized peripheral blood progenitor cells to restore hemopoiesis after high-dose chemotherapy for lymphoid malignancies.
Bone Marrow Transplant
14
1994
105
8
Dührsen
U
Villeval
JL
Boyd
J
Kannourakis
G
Morstyn
G
Metcalf
D
Effects of recombinant human granulocyte colony-stimulating factor on hematopoietic progenitor cells in cancer patients.
Blood
72
1988
2074
9
DeLuca
E
Sheridan
WP
Watson
D
Szer
J
Begley
CG
Prior chemotherapy does not prevent effective mobilisation by G-CSF of peripheral blood progenitor cells.
Br J Cancer
66
1992
893
10
Baumann I, Testa NG, Lange C, de Wynter E, Luft T, Dexter TM, van Hoef ME, Howell A: Haemopoietic cells mobilised into the circulation by lenograstim as alternative to bone marrow for allogeneic transplants. Lancet 341:369, 1993 (letter)
11
Grigg
AP
Roberts
AW
Raunow
H
Houghton
S
Layton
JE
Boyd
AW
McGrath
KM
Maher
D
Optimizing dose and scheduling of filgrastim (granulocyte colony-stimulating factor) for mobilization and collection of peripheral blood progenitor cells in normal volunteers.
Blood
86
1995
4437
12
Socinski
MA
Cannistra
SA
Elias
A
Antman
KH
Schnipper
L
Griffin
JD
Granulocyte-macrophage colony stimulating factor expands the circulating haemopoietic progenitor cell compartment in man.
Lancet
1
1988
1194
13
Gianni
AM
Siena
S
Bregni
M
Tarella
C
Stern
AC
Pileri
A
Bonadonna
G
Granulocyte-macrophage colony-stimulating factor to harvest circulating haemopoietic stem cells for autotransplantation.
Lancet
2
1989
580
14
Villeval
JL
Duhrsen
U
Morstyn
G
Metcalf
D
Effect of recombinant human granulocyte-macrophage colony stimulating factor on progenitor cells in patients with advanced malignancies.
Br J Haematol
74
1990
36
15
Ganser
A
Lindemann
A
Seipelt
G
Ottmann
OG
Eder
M
Falk
S
Herrmann
F
Kaltwasser
JP
Meusers
P
Klausmann
M
Frisch
J
Schulz
G
Mertelsmann
R
Hoelzer
D
Effects of recombinant human interleukin-3 in aplastic anemia.
Blood
76
1990
1287
16
Ottmann
OG
Ganser
A
Seipelt
G
Eder
M
Schulz
G
Hoelzer
D
Effects of recombinant human interleukin-3 on human hematopoietic progenitor and precursor cells in vivo.
Blood
76
1990
1494
17
Basser
RL
Rasko
JEJ
Clarke
K
Cebon
J
Green
MD
Grigg
AP
Zalcberg
J
Cohen
B
O'Byrne
J
Menchaca
DM
Sheridan
WP
Fox
RM
Begley
CG
Randomized, blinded, placebo-controlled phase I trial of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF ) with filgrastim after dose-intensive chemotherapy in patients with advanced cancer.
Blood
89
1997
3118
18
Rasko
JEJ
Basser
RL
Boyd
J
Mansfield
R
O'Malley
CJ
Hussein
S
Berndt
MC
Clarke
K
O'Byrne
J
Sheridan
WP
Grigg
AP
Begley
CG
Multilineage mobilisation of peripheral blood progenitor cells in humans following administration of PEG-rHuMGDF.
Br J Haematol
97
1997
871
19
Cashman
J
Eaves
AC
Eaves
CJ
Regulated proliferation of primitive hematopoietic progenitor cells in long-term human marrow cultures.
Blood
66
1985
1002
20
Miyake
K
Weissman
IL
Greenberger
JS
Kincade
PW
Evidence for a role of the integrin VLA-4 in lympho-hemopoiesis.
J Exp Med
173
1991
599
21
Hurley
RW
McCarthy
JB
Verfaillie
CM
Direct adhesion to bone marrow stroma via fibronectin receptors inhibits hematopoietic progenitor proliferation.
J Clin Invest
96
1995
511
22
Levesque
JP
Leavesley
DI
Niutta
S
Vadas
M
Simmons
PJ
Cytokines increase human hemopoietic cell adhesiveness by activation of very late antigen (VLA)-4 and VLA-5 integrins.
J Exp Med
181
1995
1805
23
Galli
SJ
Zsebo
KM
Geissler
EN
The kit ligand, stem cell factor.
Adv Immunol
55
1994
1
24
Sheridan WP, McNiece IK: Stem cell factor, in Armitage JO, Antman KH (eds): High Dose Chemotherapy. Baltimore, MD, Williams and Wilkins, 1996, p 429
25
Bernstein
ID
Andrews
RG
Zsebo
KM
Recombinant human stem cell factor enhances the formation of colonies by CD34+ and CD34+lin− cells, and the generation of colony-forming cell progeny from CD34+lin− cells cultured with interleukin-3, granulocyte colony-stimulating factor, or granulocyte-macrophage colony-stimulating factor.
Blood
77
1991
2316
26
Broxmeyer
HE
Cooper
S
Lu
L
Hangoc
G
Anderson
D
Cosman
D
Lyman
SD
Williams
DE
Effect of murine mast cell growth factor (c-kit proto-oncogene ligand) on colony formation by human marrow hematopoietic progenitor cells.
Blood
77
1991
2142
27
de Vries
P
Brasel
KA
Eisenman
JR
Alpert
AR
Williams
DE
The effect of recombinant mast cell growth factor on purified murine hematopoietic stem cells.
J Exp Med
173
1991
1205
28
McNiece
IK
Langley
KE
Zsebo
KM
Recombinant human stem cell factor synergises with GM-CSF, G-CSF, IL-3 and epo to stimulate human progenitor cells of the myeloid and erythroid lineages.
Exp Hematol
19
1991
226
29
Cicuttini
FM
Begley
CG
Boyd
AW
The effect of recombinant stem cell factor (SCF ) on purified CD34-positive human umbilical cord blood progenitor cells.
Growth Factors
6
1992
31
30
Andrews
RG
Knitter
GH
Bartelmez
SH
Langley
KE
Farrar
D
Hendren
RW
Appelbaum
FR
Bernstein
ID
Zsebo
KM
Recombinant human stem cell factor, a c-kit ligand, stimulates hematopoiesis in primates.
Blood
78
1991
1975
31
Bodine
DM
Seidel
NE
Zsebo
KM
Orlic
D
In vivo administration of stem cell factor to mice increases the absolute number of pluripotent hematopoietic stem cells.
Blood
82
1993
445
32
Andrews
RG
Bensinger
WI
Knitter
GH
Bartelmez
SH
Longin
K
Bernstein
ID
Appelbaum
FR
Zsebo
KM
The ligand for c-kit, stem cell factor, stimulates the circulation of cells that engraft lethally irradiated baboons.
Blood
80
1992
2715
33
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
34
Andrews
RG
Briddell
RA
Knitter
GH
Opie
T
Bronsden
M
Myerson
D
Appelbaum
FR
McNiece
IK
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
35
de Revel
T
Appelbaum
FR
Storb
R
Schuening
F
Nash
R
Deeg
J
McNiece
I
Andrews
R
Graham
T
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
36
Drize
N
Chertkov
J
Zander
A
Hematopoietic progenitor cell mobilization into the peripheral blood of mice using a combination of recombinant rat stem cell factor (rrSCF ) and recombinant human granulocyte colony-stimulating factor (rhG-CSF ).
Exp Hematol
23
1995
1180
37
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
38
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
39
Crawford J, Lau D, Erwin R, Rich W, McGuire B, Meyers F: A phase I trial of recombinant human stem cell factor (SCF ) in patients with advanced non-small cell carcinoma. Proc Am Soc Clin Oncol 12:338a, 1993 (abstr)
40
Demetri G, Costa J, Hayes D, Sledge G, Galli S, Hoffman R, Merica E, Rich W, Harkins B, McGuire B, Gordon ML: A phase I trial of recombinant methionyl human stem cell factor (SCF ) in patients with advanced carcinoma pre- and post- chemotherapy (chemo) with cyclophosphamide (C) and doxorubicin (A). Proc Am Soc Clin Oncol 12:367a, 1993 (abstr)
41
Kurtzberg J, Meyers F, McGuire B, Crawford J: Mobilization of peripheral blood progenitor cells in patients given recombinant methionyl human stem cell factor. Proc AACR 34:211a, 1993 (abstr)
42
Demetri GD, Gordon M, Hoffman R, Hayes DF, Sledge G, Sullivan S, Edwards R, Merica E, Baltiato L, Griffin JD: Effects of recombinant methionyl human stem cell factor on hematopoietic progenitor cells in vivo: Preliminary results from a phase I trial. Proc AACR 34:217a, 1993 (abstr)
43
Tong
J
Gordon
MS
Srour
EF
Cooper
RJ
Orazi
A
McNiece
I
Hoffman
R
In vivo administration of recombinant methionyl human stem cell factor expands the number of human marrow hematopoietic stem cells.
Blood
82
1993
784
44
Glaspy J, McNiece IK, LeMaistre F, Menchaca D, Briddell R, Lill M, Jones R, Tami J, Morstyn G, Brown S, Shpall EJ: Effects of stem cell factor (rhSCF ) and filgrastim on the mobilization of peripheral blood progenitor cells (PBPC) and hematological recovery post transplant: Preliminary phaseI/II study results. Br J Haematol 87:156a, 1994 (abstr)
45
Moskowitz C, Stiff P, Gordon M, Gabrilove J, Bayer R, Broun R, Nichols C, Ho AD, Wyres M, Nimer S, McNiece I: The influence of extensive prior chemotherapy on the mobilization of peripheral blood progenitor cells (PBPC) using stem cell factor (rhSCF ) and filgrastim (r-metHuG-CSF ) and on hematological recovery post cyclophosphamide, BCNU, and VP-16 (CBV) in patients (pts) with relapsed non-Hodgkin's lymphoma (NHL): An interim analysis. Blood 84:107a, 1994 (abstr, suppl 1)
46
Tricot G, Jagannath S, Desikan KR, Siegel D, Munshi N, Olson E, Wyres M, Parker W, Barlogie B: Superior moblization of peripheral blood progenitor cells (PBPC) with r-metHuSCF (SCF ) and r-metHUG-CSF (filgrastim) in heavily pretreated multiple myeloma (MM) patients. Blood 88:388a, 1996 (abstr, suppl 1)
47
Basser R, Begley CG, Maher D, To B, Juttner CA, Fox RM, Cebon J, Grigg A, Szer J, Marty J, Sheridan W, Collins J, Russell I, Green MD: The use of peripheral blood progenitor cells (PBPC) mobilized by stem cell factor (SCF ) and filgrastim (G-CSF ) to support multiple cycles of high-dose chemotherapy in untreated women with poor prognosis breast cancer. Br J Haematol 87:91a, 1994 (abstr)
48
Basser R, Begley CG, Mansfield R, To B, Juttner C, Maher D, Fox R, Cebon J, Szer J, Grigg A, Clark K, Marty J, Menchaca D, Thompson B, Russell I, Collins J, Green M: Mobilization of PBPC by priming with stem cell factor (SCF ) before filgrastim compared to concurrent administration. Blood 86:687a, 1995 (abstr, suppl 1)
49
Begley CG, Basser RL, Mansfield R, Maher D, To B, Juttner CA, Fox RM, Cebon J, Grigg A, Szer J, McGrath K, Thomson B, Sheridan W, Menchaca D, Collins J, Russell I, Green MD: Randomized prospective study demonstrating a prolonged effect of SCF with G-CSF (filgrastim) on PBPC in untreated patients: Early results. Blood 84:90a, 1994 (abstr, suppl 1)
50
Begley CG: Human progenitor cell assays, in Morstyn G, Sheridan W (eds): Cell Therapy. New York, NY, Cambridge, 1996
51
Begley
CG
Lopez
AF
Vadas
MA
Metcalf
D
The clonal proliferation in vitro of enriched populations of human promyelocytes and myelocytes.
Blood
65
1985
951
52
Begley
CG
Metcalf
D
Lopez
AF
Nicola
NA
Fractionated populations of normal human marrow cells respond to both human colony-stimulating factors with granulocyte-macrophage activity.
Exp Hematol
13
1985
956
53
Metcalf
D
Begley
CG
Johnson
GR
Nicola
NA
Vadas
MA
Lopez
AF
Williamson
DJ
Wong
GG
Clark
SC
Wang
EA
Biologic properties in vitro of a recombinant human granulocyte-macrophage colony-stimulating factor.
Blood
67
1986
37
54
Burgess
AW
Begley
CG
Johnson
GR
Lopez
AF
Williamson
DJ
Mermod
JJ
Simpson
RJ
Schmitz
A
DeLamarter
JF
Purification and properties of bacterially synthesized human granulocyte-macrophage colony stimulating factor.
Blood
69
1987
43
55
Begley
CG
Rasko
JEJ
Curtis
D
Takagi
K
Metcalf
D
Hilton
D
Roberts
B
Nicola
NA
Rossner
M
Murine flt3 ligand protects M1 leukemic cells from LIF-induced differentiation and suppression of self-renewal.
Exp Hematol
24
1996
1247
56
Basser
RL
To
B
Begley
CG
Juttner
CA
Maher
DW
Szer
J
Cebon
J
Collins
JP
Russell
I
Olver
I
Gill
PG
Fox
RM
Sheridan
WP
Green
MD
Adjuvant treatment of high-risk breast cancer using multicycle high-dose chemotherapy and filgrastim-mobilized peripheral blood progenitor cells.
Clin Cancer Res
1
1995
715
57
Roberts
AW
DeLuca
E
Begley
CG
Basser
R
Grigg
AP
Metcalf
D
Broad inter-individual variations in circulating progenitor cell numbers induced by granulocyte colony-stimulating factor therapy.
Stem Cells
13
1995
512
58
Moskowitz
CH
Stiff
P
Gordon
MS
McNiece
I
Ho
AD
Costa
JJ
Broun
ER
Bayer
RA
Wyres
M
Hill
J
Jelaca-Maxwell
K
Nichols
CR
Brown
SL
Nimer
SD
Gabrilove
J
Recombinant methionyl human stem cell factor and filgrastim for peripheral blood progenitor cell mobilization and transplantation in Non-Hodgkins lymphoma patients: Results of a phase I/II trial.
Blood
89
1997
9
59
Höcker
P
Geissler
K
Kurz
M
Wagner
A
Gerhartl
K
Potentiation of GM-CSF or G-CSF induced mobilization of circulating progenitor cells by pretreatment with IL-3 and harvest by apheresis.
Int J Artificial Organs
5
1993
25
60
D'Hondt
V
Guillaume
T
Humblet
Y
Doyen
C
Chatelain
B
Feyens
AM
Staquet
P
Osselaer
JC
Mull
B
Symann
M
Tolerance of sequential or simultaneous administration of IL-3 and G-CSF in improving peripheral blood stem cells harvesting following multi-agent chemotherapy: A pilot study.
Bone Marrow Transplant
13
1994
261
61
Tong
J
Hoffman
R
Siena
S
Srour
EF
Bregni
M
Gianni
AM
Characterization and quantitation of primitive hematopoietic progenitor cells present in peripheral blood autografts.
Exp Hematol
22
1994
1016
62
Mullen
M
Mealiffe
M
Terstappen
LW
Hardwick
A
Moubayed
M
Oldham
F
Corringham
RET
Ho
AD
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
85
1995
275
63
Huhn
RD
Yurkow
EJ
Tushinski
R
Clarke
L
Sturgill
MG
Hoffman
R
Sheay
W
Cody
R
Philipp
C
Resta
D
George
M
Recombinant human interleukin-3 (rhIL-3) enhances the mobilization of peripheral blood progenitor cells by recombinant human granulocyte colony-stimulating factor (rhG-CSF ) in normal volunteers.
Exp Hematol
24
1996
839
64
Roberts
AW
Metcalf
D
Granulocyte colony-stimulating factor induces selective elevations of progenitor cells in the peripheral blood of mice.
Exp Hematol
22
1994
1156
65
Lyman
SD
James
L
Vanden Bos
T
de Vries
P
Brasel
K
Gliniak
B
Hollingsworth
LT
Picha
KS
McKenna
HJ
Splett
RR
Fletcher
FA
Maraskovsky
E
Farrah
J
Foxworth
ED
Williams
DR
Beckmann
P
Molecular cloning of a ligand for the flt3/flk-2 tyrosine kinase receptor: A proliferative factor for primitive hematopoietic cells.
Cell
75
1993
1157
66
Hannum
C
Culpepper
J
Campbell
D
McClanahan
T
Zurawski
S
Bazan
JF
Kastelein
R
Hudak
S
Wagner
J
Mattson
J
Luh
J
Dudd
G
Martina
N
Peterson
D
Menon
S
Shanafelt
A
Muench
M
Kelner
G
Namikawa
R
Rennick
D
Roncarolo
M-G
Zlotnik
A
Rosnet
O
Dubreuil
P
Birnbaum
D
Lee
F
Ligand for FLT3/FLK2 receptor tyrosine kinase regulates growth of haematopoietic stem cells and is encoded by variant RNAs.
Nature
368
1994
643
67
Rasko
JEJ
Metcalf
D
Rossner
MT
Begley
CG
Nicola
NA
The flt3/flk2 ligand: Receptor distribution and action on murine haemopoietic cell survival and proliferation.
Leukemia
9
1995
2058
68
Elwood
NJ
Zogos
H
Willson
T
Begley
CG
Retroviral transduction of human progenitor cells: Use of G-CSF plus SCF to mobilize progenitor cells in vivo and stimulation by flt3/flk-2 ligand in vitro.
Blood
88
1996
4452
69
Metcalf D: The hemopoietic colony stimulating factors. Amsterdam, The Netherlands, Elsevier, 1984
Sign in via your Institution