This report examines the effects on hematopoietic regeneration of pegylated recombinant human megakaryocyte growth and development factor (PEG-rHuMGDF ) (2.5 μg/kg/d) alone and in combination with recombinant human granulocyte colony stimulating factor (rHu-GCSF ) (10 μg/kg/d) for 21 days in rhesus macaques receiving intense marrow suppression produced by single bolus injections of hepsulfam (1.5 g/m2). In six hepsulfam-only control animals thrombocytopenia (platelet count <100 × 109/L) was observed between days 12 and 25 (nadir 39 ± 20 × 109/L on day 17), and neutropenia (absolute neutrophil count <1 × 109/L) occurred between days 8 and 30 (nadir 0.167 ± 0.120 × 109/L on day 15). PEG-rHuMGDF (2.5 μg/kg/d) injected subcutaneously into four animals from day 1 to day 22 following hepsulfam administration produced trough serum concentrations of 1.9 ± 0.2 ng/mL and increased the platelet count twofold over basal prechemotherapy levels (856 ± 594 × 109/L v baseline of 416 ± 88 × 109/L; P = .01). PEG-rHuMGDF alone also shortened the period of posthepsulfam neutropenia from 22 days to 12 days (P = .01), although the neutropenic nadir was not significantly altered (neutrophil count 0.224 ± 0.112 × 109/L v 0.167 ± 0.120 × 109/L; P < .3). rHu-GCSF (10 μg/kg/d) injected subcutaneously into four animals from day 1 to day 22 following hepsulfam administration produced trough serum concentrations of 1.4 ± 1.1 ng/mL, and reduced the time for the postchemotherapy neutrophil count to attain 1 × 109/L from 22 days to 4 days (P = .005). The postchemotherapy neutropenic nadir was 0.554 ± 0.490 × 109neutrophils/L (P = .3 v hepsulfam-only control of 0.167 ± 0.120 × 109/L). However, thrombocytopenia of <100 × 109 platelets/L was not shortened (persisted from day 12 to day 25), or less severe (nadir of 56 ± 32 × 109 platelets/L on day 14; P = .7 compared with untreated hepsulfam animals). The concurrent administration of rHu-GCSF (10 μg/kg/d) and PEG-rHuMGDF (2.5 μg/kg/d) in four animals resulted in postchemotherapy peripheral platelet counts of 127 ± 85 × 109/L (P = .03 compared with 39 ± 20 × 109/L for untreated hepsulfam alone, and P = .02 compared with 856 ± 594 × 109/L for PEG-rHuMGDF alone), and shortened the period of neutropenia <1 × 109/L from 22 days to 4 days (P = .8 compared with rHu-GCSF alone). Increasing PEG-rHuMGDF to 10 μg/kg/d and maintaining the 21-day schedule of coadministration with rHu-GCSF (10 μg/kg/d) in another four animals produced postchemotherapy platelet counts of 509 ± 459 × 109/L (P < 10−4compared with untreated hepsulfam alone, and P = .04 compared with 2.5 μg/kg/d PEG-rHuMGDF alone), and 4 days of neutropenia. Coadministration of rHu-GCSF and PEG-rHuMGDF did not significantly alter the pharmacokinetics of either agent. The administration of PEG-rHuMGDF (2.5 μg/kg/d) from day 1 through day 22 and rHu-GCSF (10 μg/kg/d) from day 8 through day 22 in six animals produced peak postchemotherapy platelet counts of 747 ± 317 × 109/L (P < 10−4 compared with untreated hepsulfam alone, and P = .7 compared with PEG-rHuMGDF alone), and maintained the neutrophil count < 3.5 × 109/L (P = .008 v rHu-GCSF therapy alone). Thus, both thrombocytopenia and neutropenia are eliminated by initiating daily PEG-rHuMGDF therapy on day 1 and subsequently adding daily rHu-GCSF after 1 week in the rhesus model of hepsulfam marrow suppression. This improvement in platelet and neutrophil responses by delaying the addition of rHu-GCSF to PEG-rHuMGDF therapy demonstrates the importance of optimizing the dose and schedule of cytokine combinations after severe myelosuppressive chemotherapy.

NEUTROPENIC FEVER and thrombocytopenic bleeding complicate high-dose myelosuppressive cancer chemotherapy. Antibiotics and the granulocyte lineage-dominant hematopoietic growth factor, rHu-GCSF or granulocyte-macrophage colony-stimulating factor (GM-CSF ), significantly improve outcomes for patients who are at risk of developing neutropenic fever.1-3 While platelet transfusions protect these patients from thrombocytopenic bleeding, platelet transfusional therapy becomes complicated by limited availability, transmission of infectious diseases, sensitization and possible refractoriness to repeated platelet transfusions.4-6 The availability of recombinant megakaryocyte lineage-dominant hematopoietic growth factors (Mpl-ligands),7-12 permits an assessment of alternatively substituting amplified platelet production for prophylactic platelet transfusions in cancer patients receiving myeloablative chemotherapy. Before evaluating this strategy in patients, we examined the effects of combining granulocyte-dominant rHu-GCSF with megakaryocyte-dominant Mpl-ligands on hematopoietic recovery in a clinically relevant nonhuman primate model of severe chemotherapeutic myelosuppression.

Mpl-ligands induce hematopoietic stem cells to undergo megakaryocytic differentiation, proliferation, and endoreduplication,13-16 thereby expanding the formation of megakaryocyte cytoplasmic substrate destined for fragmentation and release as functional circulating platelets.7-10,16-19 The regulatory role of native Mpl-ligand, thrombopoietin, in the process of platelet production is evident from the capacity of excess soluble Mpl-receptor to neutralize both the megakaryocyte colony-stimulating activity and platelet-elevating activity in thrombocytopenic plasma.7,8,10,15 Mice engineered for deletion of the Mpl-receptor20 or endogenous thrombopoietin21 exhibit platelet counts about one sixth of normal without affecting other blood cell counts. In addition, circulating levels of Mpl-ligand in patients with postchemotherapy thrombocytopenia are elevated, and decrease after the peripheral platelet concentrations return toward normal by platelet transfusions or marrow recovery.10,15 In this study, the effects of PEG-rHuMGDF alone and in combination with rHu-GCSF, are investigated in a model of chemotherapy-induced intense myelosuppression in rhesus macaques.

Animals studied.A total of 32 rhesus monkeys (26 males and 6 females) weighing 5 to 9 kg were used for this study. All procedures were approved by the Institutional Animal Care and Use Committee and conducted in accordance with Federal guidelines (Guide for the Care and Use of Laboratory Animals. National Institutes of Health, Bethesda, MD, NIH Publ. No. 86-23).

Study design.The effects of injecting PEG-rHuMGDF (2.5 μg/kg/d) subcutaneously each day for 21 days was assessed in four normal control animals without associated hepsulfam administration. Hepsulfam, a myelosuppressive agent,22-24 was given to the remaining 28 animals by intravenous bolus injections. The hepsulfam animals were allocated to six different treatment groups. These animals (except for group 6) received daily subcutaneous injections from day 1 through day 22 consisting of: (1) saline controls (n = 6); (2) PEG-rHuMGDF (n = 4); (3) rHu-GCSF (n = 4); (4) concurrent PEG-rHuMGDF (2.5 μg/kg/d) plus rHu-GCSF (10 μg/kg/d) (n = 4); (5) concurrent PEG-rHuMGDF (10 μg/kg/d) plus rHu-GCSF (10 μg/kg/d) (n = 4); and (6) PEG-rHuMGDF (2.5 μg/kg/d) on day 1 through day 22, and rHu-GCSF(10 μg/kg/d) beginning on day 8 and continuing through day 22 (n = 6).

Enrofloxacin (2.5 mg/kg as Batril; Mobay Corp, Shawnee, KS) was administered by intramuscular injection twice daily beginning on day 1 and continuing through at least day 21 until the neutrophil count was >500/μL for two successive determinations. Fluconazol (5 mg/kg as Diflucan; Pfizer, Brooklyn, NY) was given orally once daily. Blood was obtained for complete blood counts by femoral vein puncture before hepsulfam administration and three times weekly thereafter (Monday, Wednesday, and Friday) for 6 weeks. Blood for serum chemistry determinations was drawn at baseline and at the end of cytokine administration. Bone marrow aspirates were obtained before hepsulfam administration and 3, 7, 14, and 21 days later.

Reagents.Hepsulfam (sulfamic acid diester, NSC 329680)23 was a gift from the Division of Cancer Treatment, National Cancer Institute, Bethesda, MD. The lyophilized drug was solubilized in diluent containing 10% (vol/vol) ethanol and 40% (vol/vol) propylene glycol in 0.05 mol/L phosphate buffer (pH 7.4), and then diluted in 150 mL of normal saline. Hepsulfam (1.5 g/m2) was administered over 30 minutes by single bolus intravenous infusion.

PEG-rHuMGDF is a truncated molecule including the N-terminal of human c-Mpl ligand, in this case having 163 amino acids, which is expressed in Escherichia coli and derivatized with polyethylene glycol on the N-terminus by reductive alkylation.10,25 PEG-rHuMGDF was formulated in an aqueous buffer, sterilized by filtration, and provided by Amgen Inc, Thousand Oaks, CA. Dosing was based on protein weight. rHu-GCSF was provided as filgrastim by Amgen Inc.

Laboratory procedures.Peripheral platelet counts, mean platelet volumes, red blood cell counts, total white blood cell counts, and neutrophil counts were determined in whole blood collected every other day in Na2EDTA (2 mg/mL) using Serono/Baker model 9000 whole blood analyzer (Allentown, PA).26,27 The neutrophil counts were determined from electronic leukocyte counts and manual 500-cell leukocyte differential counts on Wright-Giemsa-stained peripheral blood films. The baseline mean blood cell counts were 416 ± 88 × 109/L for platelets, 5.69 ± 0.49 × 1012/L for erythrocytes, 10.2 ± 3.4 × 109/L for leukocytes, and 4.5 ± 2.1 × 109/L for neutrophils.

Serum concentrations of PEG-rHuMGDF and thrombopoietin (TPO) were determined using an enzyme-linked immunosorbent assay (ELISA) involving an initial polyclonal antibody capture procedure followed by enzyme product formation and determination.16,28 Serum levels of exogenous and endogenous G-CSF were also determined using ELISA involving initial antibody capture procedure and subsequent enzyme product formation and determination.1,29 

Megakaryocyte number, size, and ploidy were measured by flow cytometry using a previously reported method for multiparameter correlative marrow analysis with a single-argon-ion-laser FACScan analyzer (Becton Dickinson, San Jose, CA).28,30-32 Cell DNA in aspirated marrow was stained with propidium iodide, and surface membrane receptors were analyzed with antibodies labeled with fluorescein and phycoerythrin. Megakaryocytes expressing platelet glycoprotein (GP) IIb/IIIa were enumerated in relation to the nucleated erythroid precursors expressing glycophorin A.6 Measurements of megakaryocyte diameters were based on the time-of-flight principle, ie, time required for a cell in suspension to pass through a focused light beam.30,33 Megakaryocytes were selected on the basis of their distinct immunofluorescence at levels above that of control cells labeled with an unrelated monoclonal antibody (MoAb). In each sample, 2,000 to 3,000 megakaryocytes were analyzed. Flow cytometry was performed using FACScan (Becton Dickinson). Bone marrow aspirates were obtained baseline and after 3, 7, 14, and 21 days of treatment.

Estimates of marrow megakaryocyte mass were used to represent the marrow substrate giving rise to circulating platelets, and was calculated as the product of megakaryocyte numbers and mean megakaryocyte volumes.6,16,18,28 Normal rhesus marrow values (n = 16) averaged: megakaryocyte diameter of 39 μm (range, 21 μm for 2N to 56 μm for 64N cells), volume of 30.1 ± 3.60 × 103 f L, and megakaryocyte number of 6.36 ± 1.41 × 106 megas/kg, giving a total megakaryocyte mass of 19.5 ± 6.62 × 1010 f L/kg. The normal modal ploidy was 16N.

Steady-state platelet mass turnover (platelet concentration multiplied by mean platelet volume and divided by platelet life span and by platelet recovery) was used to estimate the rate at which viable platelet mass was delivered to the peripheral blood.16,18,28 To measure platelet survival time, autologous platelets were labeled with 111In-oxine using the method described previously34; the labeled platelets functioned normally.35 Platelet survival time, ie, the average time in circulation, was then calculated using computer least-squares fitting of the raw data to a gamma-function modeling program.34 Baseline rhesus platelet lifespan (n = 10) was 5.5 ± 0.6 days. The recovery of labeled platelets in the circulation at equilibrium was estimated by extrapolating the survival curve to time zero and estimating the blood volume (70 mL/kg) using the formula: recovery in the circulation = total circulating platelet radioactivity/total platelet radioactivity injected × 100 (%). Basal platelet recovery in rhesus was 85% ± 4%. Platelet mass turnover, a measure of steady state platelet production and destruction, was calculated by the formula: platelet mass turnover rate (f L plats/μL/d) = platelet count/μL multiplied by mean platelet volume (f L) and divided by platelet survival time (days) and proportion of 111In-platelets recovered immediately after intravenous infusion. Platelet mass turnover in normal rhesus (n = 10) was 5.17 ± 1.27 × 105 f L plats/μL/d.

Marrow biopsies were obtained from the proximal femur before hepsulfam treatment, and after 3 weeks of cytokine therapy. The biopsies were processed for paraffin sectioning and stained for morphologic evaluation by light microscopy.

Data analysis.Data were analyzed using SIGMA STAT (Jandel Scientific Software, San Rafael CA). Comparisons between two groups were performed using the two-tailed Student's t-test, unless the data were not distributed randomly, in which case nonparametric analyses were performed. Analysis of variance (ANOVA) was used to compare values for a particular group at various time points.36 Unless otherwise stated, variance about the mean is given as ± 1 standard deviation (SD).

Effects of PEG-rHuMGDF on platelet production in normal rhesus controls.Daily subcutaneous injections of PEG-rHuMGDF (2.5 μg/kg/d) produced serum trough levels of 1.60 ± 0.19 ng/mL (P < 10−4 compared with endogenous Mpl-ligand levels of <100 pg/mL). The corresponding peripheral platelet counts increased fivefold, attaining peak levels at 2 weeks of 2,197 ± 249 × 109/L (Fig 1, left and Table 1; P = .001 v baseline of 416 ± 88 × 109/L). During the period of induced thrombocytosis, the mean platelet volume (MPV) decreased from the baseline value of 8.3 ± 1.0 f L to 7.2 ± 0.9 f L (P = .01). Platelet ultrastructure was normal (data not shown), and platelet lifespan averaged 132 ± 7 hours (P = .68 compared with the mean baseline value of 130 ± 14 hours). PEG-rHuMGDF (2.5 μg/kg/d) increased platelet mass turnover sixfold, from 5.2 ± 1.3 to 32.7 ± 1.5 × 105 f L plats/μL/d (P < 10−4). After discontinuing therapy, platelet counts fell to baseline values and MPVs normalized within 10 days (Fig 1). The peripheral neutrophil counts were not significantly altered from baseline values (Fig 1, right; P > .2), and peripheral erythrocyte counts and volumes were not changed from baseline (P > .5).

Fig. 1.

Effects of PEG-rHuMGDF on hematopoiesis in untreated controls. PEG-rHuMGDF (2.5 μg/kg/d) increased the concentration of circulating platelets sixfold by 2 weeks. The platelet count normalized within 10 days after stopping therapy (left). Peripheral leukocyte, neutrophil, or erythrocyte counts did not change significantly during PEG-rHuMGDF administration (right).

Fig. 1.

Effects of PEG-rHuMGDF on hematopoiesis in untreated controls. PEG-rHuMGDF (2.5 μg/kg/d) increased the concentration of circulating platelets sixfold by 2 weeks. The platelet count normalized within 10 days after stopping therapy (left). Peripheral leukocyte, neutrophil, or erythrocyte counts did not change significantly during PEG-rHuMGDF administration (right).

Close modal
Table 1.

Effects of PEG-rHuMGDF on Marrow Megakaryocytes and Platelet Production in Normal Rhesus Macaques

Duration PEG-rHu MGDF Therapy (2.5 μg/kg/d)MegakaryocytesPlatelets
Ratio to NE (×103)*No.Volume (×103 fL)MassIncrease (×N)Concentration (×109/L)Volume (fL)Mass turnoverIncrease (×N)
(×106 megas/kg)(×1010 fL/kg)(×105 fL plats/μL/d)
Baseline 1.20 ± 0.26 6.36 ± 1.41 30.1 ± 3.60 19.5 ± 6.62 1.0  416 ± 88 8.3 ± 1.0 5.17 ± 1.27 1.0 
3 d 2.21 ± 0.84 11.7 ± 4.45 57.2 ± 3.39 67.4 ± 29.5 3.4  474 ± 108 8.0 ± 0.5 7.95 ± 1.64 1.5 
7 d 6.25 ± 2.73 33.2 ± 14.6 48.6 ± 1.44 160 ± 68.6 8.2 1,084 ± 153 7.6 ± 0.9 17.2 ± 2.49 3.2 
14 d 8.42 ± 3.52 55.5 ± 11.7 40.1 ± 8.71 231 ± 7.20 11.8 2,197 ± 249 7.2 ± 0.9 32.7 ± 1.52 6.2 
Duration PEG-rHu MGDF Therapy (2.5 μg/kg/d)MegakaryocytesPlatelets
Ratio to NE (×103)*No.Volume (×103 fL)MassIncrease (×N)Concentration (×109/L)Volume (fL)Mass turnoverIncrease (×N)
(×106 megas/kg)(×1010 fL/kg)(×105 fL plats/μL/d)
Baseline 1.20 ± 0.26 6.36 ± 1.41 30.1 ± 3.60 19.5 ± 6.62 1.0  416 ± 88 8.3 ± 1.0 5.17 ± 1.27 1.0 
3 d 2.21 ± 0.84 11.7 ± 4.45 57.2 ± 3.39 67.4 ± 29.5 3.4  474 ± 108 8.0 ± 0.5 7.95 ± 1.64 1.5 
7 d 6.25 ± 2.73 33.2 ± 14.6 48.6 ± 1.44 160 ± 68.6 8.2 1,084 ± 153 7.6 ± 0.9 17.2 ± 2.49 3.2 
14 d 8.42 ± 3.52 55.5 ± 11.7 40.1 ± 8.71 231 ± 7.20 11.8 2,197 ± 249 7.2 ± 0.9 32.7 ± 1.52 6.2 
*

Ratio of marrow megakaryocytes to nucleated erythroid cells determined by comparing nucleated GPIIb/IIIa-positive cells to nucleated glycophorin A-positive cells using flow cytometry (see Materials and Methods).

PEG-rHuMGDF (2.5 μg/kg/d) expanded megakaryocyte volume to 57.2 ± 3.39 ×103 f L by the third day of injections (P < 10−4v the baseline of 30.1 ± 3.60 × 103 f L), and increased megakaryocyte number to 55.5 ± 11.7 × 106/kg by day 14 (P < 10−4v baseline of 6.36 ± 1.41 × 106/kg). Megakaryocyte modal ploidy was increased from 16 to 32, with increases in 64N and the appearance of a 128N cohort (P = .01). Megakaryocytes mass, the product of the total number of megakaryocytes and their mean volume,6,18 increased greater than 10-fold to 231 ± 7.20 × 1010 f L/kg by day 14 (P < .0001 compared with basal values).

Effects of hepsulfam on hematopoiesis in untreated controls.The hematologic effects of hepsulfam (1.5 g/m2 intravenous bolus) was determined in six control rhesus macaques that received daily subcutaneous injections of saline for 21 days (Fig 2). Thrombocytopenia of less than 100 × 109 platelets/L was observed between day 12 and day 25 following hepsulfam treatment, with a nadir of 39 ± 20 × 109 platelets/L on day 17 (Fig 2, left). Thereafter, peripheral platelet counts slowly returned to normal values, ie, platelet counts remained significantly reduced at 3 months, as previously reported.22 

Fig. 2.

Effects of hepsulfam on hematopoiesis. Hepsulfam (1.5 g/m2 intravenous bolus) produced thrombocytopenia (<100 × 109platelets/L) between day 12 and 25, with a nadir of 39 ± 20 × 109 platelets/L on day 17 (left). Neutropenia of less than 1 × 109 neutrophils/L occurred between day 8 and 30 following hepsulfam, with a nadir of 0.167 ± 0.120 × 109 neutrophils/L on day 15 (right).

Fig. 2.

Effects of hepsulfam on hematopoiesis. Hepsulfam (1.5 g/m2 intravenous bolus) produced thrombocytopenia (<100 × 109platelets/L) between day 12 and 25, with a nadir of 39 ± 20 × 109 platelets/L on day 17 (left). Neutropenia of less than 1 × 109 neutrophils/L occurred between day 8 and 30 following hepsulfam, with a nadir of 0.167 ± 0.120 × 109 neutrophils/L on day 15 (right).

Close modal

Neutropenia of less than 1 × 109 neutrophils/L occurred between day 8 and day 30 following hepsulfam administration, with a nadir of 0.167 ± 0.120 × 109 neutrophils/L on day 15 (Fig 2, right). After hepsulfam treatment and in association with repeated blood sampling, anemia developed progressively, reaching a nadir hemaglobin of 8 g/dL on day 24, with gradual normalization over the subsequent month. Reticulocytes disappeared transiently from the circulation between days 5 and 12 following hepsulfam dosing.

Effects of PEG-rHuMGDF on posthepsulfam hematopoietic regeneration.Daily subcutaneous injections of PEG-rHuMGDF (2.5 μg/kg/d) beginning 1 day following hepsulfam injections and continuing for 21 days produced 7-day serum trough levels of 1.49 ± 0.29 ng/mL and 14-day trough levels of 1.91 ± 0.16 ng/mL (P > .2). The peripheral platelet counts exhibited a biphasic response, increasing progressively over prechemotherapy values for 9 days, then declining transiently to near baseline on day 15 (P = .01 comparing peak at 9 days and valley at 15 days using paired analysis), and subsequently climbing twofold over baseline by day 24 (P = .05 comparing valley at day 15 and peak at day 24 using paired analysis; peak platelet count of 856 ± 594 × 109/L v basal level of 416 ± 88 × 109/L; P = .01), thereafter falling to control levels over 10 days (Fig 3, left). PEG-rHuMGDF increased the proportion of megakaryocytes attaining the 64N and 128N ploidy classes (P < .005 in both cases compared with normal controls), with a reciprocal reduction in the 16N ploidy class (P < .005 compared with normal controls).

Fig. 3.

Effects of PEG-rHuMGDF on posthepsulfam hematopoietic regeneration. Daily subcutaneous injections of PEG-rHuMGDF (2.5 μg/kg/d), beginning 1 day after hepsulfam administration and continuing for 21 days, produced a biphasic increase in the peripheral platelet counts at or above prechemotherapy values (left). PEG-rHuMGDF dosing after hepsulfam chemotherapy shortened the duration of neutropenia from 22 to 12 days (P = .01), although the neutropenic nadir was not significantly changed (right). The open symbols depict the counts for untreated hepsulfam controls, and the closed symbols designate counts in PEG-rHuMGDF-treated hepsulfam animals.

Fig. 3.

Effects of PEG-rHuMGDF on posthepsulfam hematopoietic regeneration. Daily subcutaneous injections of PEG-rHuMGDF (2.5 μg/kg/d), beginning 1 day after hepsulfam administration and continuing for 21 days, produced a biphasic increase in the peripheral platelet counts at or above prechemotherapy values (left). PEG-rHuMGDF dosing after hepsulfam chemotherapy shortened the duration of neutropenia from 22 to 12 days (P = .01), although the neutropenic nadir was not significantly changed (right). The open symbols depict the counts for untreated hepsulfam controls, and the closed symbols designate counts in PEG-rHuMGDF-treated hepsulfam animals.

Close modal

PEG-rHuMGDF dosing had no significant effect on the nadir of neutropenia produced by hepsulfam chemotherapy, ie, 0.224 ± 0.112 × 109 neutrophils/L, compared with 0.167 ± 0.120 × 109/L for hepsulfam alone (P > .3). However, the duration of neutropenia was significantly shortened from 22 days to 12 days (P < .01), and reached basal values after 30 days (Fig 3, right). Endogenous levels of G-CSF were not detectably increased. PEG-rHuMGDF did not affect the suppressive effect of hepsulfam on the reticulocyte count (P > .4; data not shown).

Effects of rHu-GCSF on posthepsulfam hematopoietic regeneration.Daily subcutaneous injections of rHu-GCSF (10 μg/kg/d) beginning 1 day after hepsulfam treatment and continuing for 21 days produced 7-day serum trough levels of 0.43 ± 0.27 ng/mL and 14-day trough levels of 1.42 ± 1.09 ng/mL (P > .3). rHu-GCSF alone had no effect on the nadir or duration of postchemotherapy thrombocytopenia, ie, the platelet count remained <100 × 109 platelets/L between day 12 and day 25, with a nadir of 56 ± 32 × 109/L on day 14 (Fig 4, left; P = .7). The endogenous Mpl-ligand levels increased to 0.24 ± 0.07 ng/mL at 7 days and 0.36 ± 0.08 ng/mL at 14 days (P > .5). Although hepsulfam markedly reduced hematopoietic progenitors, identifiable megakaryocytes shifted ploidy to higher classes compared with normal controls, similar to the effects produced by elevated levels of exogenous Mpl-ligand.

Fig. 4.

Effects of rHu-GCSF on posthepsulfam hematopoietic regeneration. rHu-GCSF (10 μg/kg/d) beginning 1 day after hepsulfam treatment and continuing for 21 days did not significantly improve the nadir or duration of postchemotherapy thrombocytopenia (left). Twentyone days of rHu-GCSF (10 μg/kg/d) injections shortened the period of neutropenia (<1 × 109 neutrophils/L) from 22 to 4 days (right); the intensity of the neutropenia was not significantly affected (right). The open symbols depict the counts for untreated hepsulfam controls, and the closed symbols designate counts in rHu-GCSF–treated hepsulfam animals.

Fig. 4.

Effects of rHu-GCSF on posthepsulfam hematopoietic regeneration. rHu-GCSF (10 μg/kg/d) beginning 1 day after hepsulfam treatment and continuing for 21 days did not significantly improve the nadir or duration of postchemotherapy thrombocytopenia (left). Twentyone days of rHu-GCSF (10 μg/kg/d) injections shortened the period of neutropenia (<1 × 109 neutrophils/L) from 22 to 4 days (right); the intensity of the neutropenia was not significantly affected (right). The open symbols depict the counts for untreated hepsulfam controls, and the closed symbols designate counts in rHu-GCSF–treated hepsulfam animals.

Close modal

Posthepsulfam injections of rHu-GCSF (10 μg/kg/d) shortened the period of neutropenia (<1 × 109 neutrophils/L) from 22 days to 4 days (Fig 4, right; P = .005), although the nadir was not significantly affected (0.554 ± 0.490 × 109 neutrophils/L v hepsulfam control of 0.167 ± 0.120 × 109 neutrophils/L; P = .3). rHu-GCSF did not affect reticulocyte counts significantly (P = .5).

Effects of combining PEG-rHuMGDF and rHu-GCSF on hematopoietic regeneration.Administering daily subcutaneous injections of PEG-rHuMGDF (2.5 μg/kg/d) concurrently with rHu-GCSF (10 μg/kg/d) produced platelet counts of 127 ± 85 × 109/L (Fig 5, left), compared with 39 ± 20 × 109/L for hepsulfam alone controls (P = .03), and 856 ± 594 × 109/L for posthepsulfam PEG-rHuMGDF alone (P = .02). This combination of PEG-rHuMGDF and rHu-GCSF produced a neutropenic nadir of 1.18 ± 0.99 × 109/L (P = .01 compared with hepsulfam only controls). The 7-day serum trough levels of PEG-rHuMGDF averaged 1.06 ± 0.11 ng/mL, and the 14-day mean trough level was 2.40 ± 0.92 (P > .1). The rHu-GCSF 14-day mean trough level was 2.02 ± 1.92 ng/mL (P = 0.6 v 1.42 ± 1.09 ng/mL for rHu-GCSF alone).

Fig. 5.

Effects of combining PEG-rHuMGDF and rHu-GCSF on hematopoietic regeneration. Administering PEG-rHuMGDF (2.5 μg/kg/d, or 10 μg/kg/d) concurrently with rHu-GCSF (10 μg/kg/d) increased platelet counts in a dose-dependent fashion (left) compared with hepsulfam alone controls (P < .03 in both cases), but significantly less than the peak platelet count of 856 ± 594 × 109/L for posthepsulfam PEG-rHuMGDF (2.5 μg/kg/d) alone (P < .02 in both cases). Combination PEG-rHuMGDF and rHu-GCSF shortened the period of neutropenia (<1 × 109 neutrophils/L) to 4 days (right); (P = .001 v hepsulfam alone controls) without significantly increasing the nadir (P < .2 for hepsulfam only controls). The (○) depict the counts for untreated hepsulfam controls. The (▴) represent counts in hepsulfam animals concurrently receiving 2.5 μg/kg PEG-rHuMGDF and 10 μg/kg rHu-GCSF. The (•) designate counts in hepsulfam animals concurrently receiving 10 μg/kg PEG-rHuMGDF and 10 μg/kg rHu-GCSF.

Fig. 5.

Effects of combining PEG-rHuMGDF and rHu-GCSF on hematopoietic regeneration. Administering PEG-rHuMGDF (2.5 μg/kg/d, or 10 μg/kg/d) concurrently with rHu-GCSF (10 μg/kg/d) increased platelet counts in a dose-dependent fashion (left) compared with hepsulfam alone controls (P < .03 in both cases), but significantly less than the peak platelet count of 856 ± 594 × 109/L for posthepsulfam PEG-rHuMGDF (2.5 μg/kg/d) alone (P < .02 in both cases). Combination PEG-rHuMGDF and rHu-GCSF shortened the period of neutropenia (<1 × 109 neutrophils/L) to 4 days (right); (P = .001 v hepsulfam alone controls) without significantly increasing the nadir (P < .2 for hepsulfam only controls). The (○) depict the counts for untreated hepsulfam controls. The (▴) represent counts in hepsulfam animals concurrently receiving 2.5 μg/kg PEG-rHuMGDF and 10 μg/kg rHu-GCSF. The (•) designate counts in hepsulfam animals concurrently receiving 10 μg/kg PEG-rHuMGDF and 10 μg/kg rHu-GCSF.

Close modal

Increasing PEG-rHuMGDF to 10 μg/kg/d in combination with rHu-GCSF (10 μg/kg/d) produced an exaggerated biphasic platelet response with a transient thrombocytopenic nadir on day 15 and subsequent peak platelet response of 509 ± 459 × 109 platelets/L (P < 10−4v hepsulfam only controls, and P = .01 v 127 ± 85 × 109 platelets/L for 2.5 μg/kg/d PEG-rHuMGDF combination dosing, and P = .03 v 856 ± 594 × 109 platelets/L for 2.5 μg/kg/d PEG-rHuMGDF only dosing; Fig 5).

Posthepsulfam injections of PEG-rHuMGDF (2.5 μg/kg/d) on days 1 to 22, with the addition of rHu-GCSF (10 μg/kg/d) from days 8 to 22, reproduced the biphasic increase in posthepsulfam platelet counts, peaking at 747 ± 317 × 109/L (P = .7 v 856 ± 594 × 109/L produced by 2.5 μg/kg/d PEG-rHuMGDF alone, P < 10−4v 56 ± 32 posthepsulfam rHu-GCSF alone, and P = .001 v prehepsulfam pretreatment baseline of 416 ± 88 × 109/L; Fig 6, left). Neutropenia was completely abolished by this dosing regimen (Fig 6, right), ie, neutrophil count >3.5 × 109/L throughout the postchemotherapy interval (P = .01 v the nadir neutrophil count for rHu-GCSF therapy alone). The serum trough concentrations of PEG-rHuMGDF for 7 days (1.34 ± 0.54 ng/mL) and 14 days (1.71 ± 0.45 ng/mL) were similar (P = .3). The 14-day trough levels of rHu-GCSF were 1.34 ± 1.16 ng/mL, similar to the 1.79 ± 1.25 ng/mL in the cohort receiving concurrent combined dosing (P = .5).

Fig. 6.

Prevention of both thrombocytopenia and neutropenia. PEG-rHuMGDF (2.5 μg/kg/d) singly for 7 days after hepsulfam, and adding daily rHu-GCSF (10 μg/kg/d) subsequently for 2 weeks restored the posthepsulfam platelet response produced by 2.5 μg/kg/d PEG-rHuMGDF alone (left) and abolished the period of neutropenia (right). The (○) depict the counts for untreated hepsulfam controls. The (•) represent counts in hepsulfam animals receiving 2.5 μg/kg PEG-rHuMGDF from day 1 through day 22, and 10 μg/kg rHu-GCSF from day 8 through day 22.

Fig. 6.

Prevention of both thrombocytopenia and neutropenia. PEG-rHuMGDF (2.5 μg/kg/d) singly for 7 days after hepsulfam, and adding daily rHu-GCSF (10 μg/kg/d) subsequently for 2 weeks restored the posthepsulfam platelet response produced by 2.5 μg/kg/d PEG-rHuMGDF alone (left) and abolished the period of neutropenia (right). The (○) depict the counts for untreated hepsulfam controls. The (•) represent counts in hepsulfam animals receiving 2.5 μg/kg PEG-rHuMGDF from day 1 through day 22, and 10 μg/kg rHu-GCSF from day 8 through day 22.

Close modal

Coadministration of rHu-GCSF did not appear to significantly affect the levels of PEG-rHuMGDF, ie, 14-day levels of PEG-rHuMGDF were similar at 1.91 ± 0.16, 2.40 ± 0.92, and 1.71 ± 0.45 ng/mL for PEG-rHuMGDF (2.5 μg/kg/d only), PEG-rHuMGDF (2.5 μg/kg/d) and rHu-GCSF (10 μg/kg/d), and PEG-rHuMGDF (2.5 μg/kg/d) followed by rHu-GCSF (10 μg/kg/d), respectively (P = .219 by ANOVA). The 14-day serum trough concentrations of rHu-GCSF were similar for dosing rHu-GCSF (10 μg/kg/d) alone, concurrent administration with PEG-rHuMGDF (2.5 μg/kg/d), and delayed co-administration beginning 1 week after initiating PEG-rHuMGDF (2.5 μg/kg/d), ie, 1.42 ± 1.09, 2.02 ± 1.92, and 1.61 ± 1.34 ng/mL (P = .48 by ANOVA).

This study demonstrates in a rhesus model of hepsulfam-induced marrow suppression that stimulating megakaryocytopoiesis with PEG-rHuMGDF (2.5 μg/kg/d) abolishes postchemotherapy thrombocytopenia and attenuates neutropenia, while stimulating neutrophil production by rHu-GCSF (10 μg/kg/d) markedly truncates the period of postchemotherapy neutropenia without increasing platelet production, and coadministration of PEG-rHuMGDF and rHu-GCSF eliminates both thrombocytopenia and neutropenia when rHu-GCSF therapy is added after initiating postchemotherapy PEG-rHuMGDF treatment.

The present rhesus hepsulfam myelosuppressive model is useful for the preclinical evaluation of hematopoietic cytokines because it reproduces chemotherapeutic myelosuppression in patients, ie, predictable and severe thrombocytopenia lasting 13 days and neutropenia lasting 22 days, with gradual return to baseline values.22 Hepsulfam is an alkylating agent structurally related to busulfan (1,4 butanediol dimethanesulfonate), that is toxic to early hematopoietic progenitor cells,23,24,37 with little associated damage to non-hematologic tissues. The rhesus hepsulfam model has been used previously to characterize the effects of other recombinant human cytokines (GM-CSF, interleukin-3 [IL-3], IL-6, IL-11) on early hematopoietic progenitor cells and hematopoietic recovery.22,38 

PEG-rHuMGDF stimulates megakaryocyte-lineage growth and development from hematopoietic progenitors (Fig 1 and Table 1),16,28,39,40 clears slowly from circulating blood as a result of pegylation,16 and is well tolerated in preclinical16,28,41,42 and initial clinical studies.43 Thus, PEG-rHuMGDF is a potent megakaryocyte lineage-dominant growth factor, analogous to the other late-acting hematopoietic cytokines, recombinant human erythropoietin (rHu-EPO) and rHu-GCSF. Under physiologic conditions the peripheral concentrations of erythrocytes, neutrophils, and platelets are maintained constant by independently regulating the production of each specific hematopoietic lineage via the corresponding hematopoietic cytokine to compensate for changing peripheral demands.2,13,14,16,18,44-48 Mpl ligands, similar to other cytokines regulating hematopoiesis, trigger intracellular signaling pathways in hematopoietic progenitors that mediate megakaryocyte development, proliferation, and differentiation, including tyrosine phosphorylation of membrane-associated kinases and adaptor molecules, ie, the Shc-Ras-raf1-MEK-ERK pathway, the Jak2-STAT3 and 5 pathway, PLC-γ, and Mpl itself.14,49-54 The differential between the increase in megakaryocyte mass and platelet turnover probably represents an unsteady state, although ineffective production cannot be ruled out.

After initiating PEG-rHu-MGDF therapy following hepsulfam-induced myelosuppression, there is a 3-day delay before the peripheral concentration of platelets rises (Fig 3), demonstrating that the processes of final megakaryocyte cytoplasmic maturation and platelet delivery are largely independent of PEG-rHuMGDF, consistent with studies of platelet release from megakaryocytes in vitro.17 Thereafter, the peripheral platelet counts increase progressively over prechemotherapy values for 9 days, then decline transiently to near baseline values on day 15, subsequently climb twofold over baseline by day 24, and fall to baseline levels over the ensuing 10 days (Fig 3, left). Because hepsulfam has destroyed the majority of hematopoietic progenitor cells, the initial platelet peak represents PEG-rHuMGDF stimulation of marrow megakaryocytes that survived hepsulfam toxicity, and responded to PEG-rHuMGDF-induced maturation mitosis (endoreduplication) and delivery of the corresponding expanded cytoplasm as additional circulating platelets (Table 1). The approximate doubling of the platelet count over baseline in the initial peak supports the interpretation that PEG-rHuMGDF induces on average one additional endomitotic event, thereby doubling substrate cytoplasm dedicated to platelet production. Furthermore, this result implies that most morphologically identifiable marrow megakaryocytes resist the lethal effects of hepsulfam. The subsequent valley in the platelet counts is attributable to hepsulfam's lethal contraction of the early hematopoietic progenitor cell reservoir that has not yet been replaced by PEG-rHuMGDF–induced proliferation. The second platelet peak reflects compensatory proliferative amplification of megakaryocyte progenitors and their subsequent maturation mediated by PEG-rHuMGDF after myelosuppressive chemotherapy. The steady fall in the concentration of peripheral platelets after discontinuing PEG-rHuMGDF therapy (Figs 1 and 4) reflects the 5 to 6 day survival time of circulating platelets together with continuing platelet production from already mature marrow megakaryocytes. As the concentration of circulating platelets increases over baseline in response to PEG-rHuMGDF postchemotherapy, the mean platelet volume decreases by approximately 15%, returning to normal only after PEG-rHuMGDF injections are discontinued, similar to the reduction in platelet volume observed without associated hepsulfam chemotherapy.16,28 

The effects of PEG-rHuMGDF on postchemotherapy platelet production in the present study are in accord with reports that recombinant Mpl-ligands accelerate platelet recovery in several different murine chemotherapy models, including myelosuppressive chemotherapy,55,56 combination of carboplatin and irradiation,41,57,58 and bone marrow59-61 or peripheral blood stem cell transplantation.62 PEG-rHuMGDF specifically produces log-linear dose-dependent responses in platelet recovery and prevents thrombocytopenia in mice receiving carboplatin chemotherapy,55 and significantly reduces the severity and duration of thrombocytopenia, and improves mortality in the more severe model using irradiation plus carboplatin.41 Transplantation of PEG-rHuMGDF-mobilized peripheral blood progenitor cells results in accelerated platelet recovery, especially if PEG-rHuMGDF is administered following transplantation.62 In addition, bone marrow transplants from donor mice pretreated with rHu-TPO exhibit accelerated platelet recovery.60 In nonhuman primates, rHu-MGDF or PEG-rHuMGDF accelerates platelet recovery following sublethal total body irradiation (TBI)63; control animals exhibit more than 12 days of severe thrombocytopenia (<20,000 μL), while animals receiving either rHu-MGDF or PEG-rHuMGDF for 18 days average ≤0.5 days of severe thrombocytopenia. In this model, the concurrent administration of PEG-rHuMGDF and rHu-GCSF, both beginning 1 day after TBI, accelerated multilineage hematopoietic recovery without evidence of lineage competition.63 Additional studies in this model indicate that PEG-rHuMGDF is effective even when administered three times per week, but that there is significant decrease in efficacy if therapy is initiated on day 5, as opposed to day 1.42 Recently, rHu-TPO has been reported not to improve platelet recovery following myeloablative TBI and infusion of small numbers of highly purified CD34 positive DR dull cells.19 

The present study shows that PEG-rHuMGDF alone facilitates postchemotherapy neutrophil recovery, as evidenced by its capacity to shorten the period of neutropenia following severe myelosuppressive chemotherapy from 22 to 12 days (Fig 3, right; <0.01 compared with untreated hepsulfam controls). Although the mechanism(s) whereby PEG-rHuMGDF improve neutrophil recovery are not presently understood, recent evidence suggests that megakaryocytes may produce cytokines exhibiting hematopoietic activities, including IL-1, IL-3, IL-6, and GM-CSF.64-66 Alternatively, there is considerable cytokine pleiotropy evident for lineage-dominant cytokines in vitro.13,49,67-70 The possibility that PEG-rHuMGDF might increase the production of endogenous G-CSF sufficiently to mediate the improved neutrophil response is not excluded, since the present assay lacks the sensitivity to rule out a biologically significant increase in endogenous G-CSF levels, and mRNA levels for G-CSF in megakaryocytes were not measured. The present observations are in agreement with the reported improvement in neutrophil counts produced by rHu-MGDF in nonhuman primates following radiation-induced myelosuppression.42 Myelosuppressed mice receiving PEG-rHuMGDF alone, also show improved leukocyte and red blood cell recoveries, and markedly reduced mortality (94 % v 15% with PEG-rHuMGDF ). However, rHu-GCSF therapy alone does not improve platelet recovery (Fig 4, left), despite measurable increases in serum endogenous Mpl-ligand.

In the present study rHu-GCSF (10 μg/kg/d) alone reduces the time for postchemotherapy neutropenia from 22 days to 4 days, although the intensity of the neutropenic nadir is not significantly attenuated (Fig 4, right), and neither the duration nor intensity of thrombocytopenia is improved (Fig 4, left). The present data showing that rHu-GCSF does not induce megakaryocyte growth and development, has been observed by others previously.14,71 Presumably, the explanation for the inability of rHu-GCSF to prevent transient neutropenia, as opposed to the elimination of thrombocytopenia by high-dose PEG-rHuMGDF, is related in part to the removal of neutrophils from the peripheral blood within hours after neutrophils enter the blood stream, contrasted to the platelets' 5- to 6-day lifespan after entry into the circulation.

Coadministration of rHu-GCSF with PEG-rHuMGDF reproduces the postchemotherapy neutrophil responses induced by rHu-GCSF alone, and produces dose-dependent increases in postchemotherapy peripheral platelet counts compared with untreated hepsulfam controls, ie, 39 ± 20 × 109/L for untreated hepsulfam, 127 ± 85 × 109/L and 509 ± 459 × 109/L for 2.5 μg/kg/d and 10 μg/kg/d PEG-rHuMGDF, respectively. However, the platelet responses following concurrent therapy with rHu-GCSF and PEG-rHuMGDF are significantly less than the posthepsulfam response produced by PEG-rHuMGDF treatment alone, ie, 856 ± 594 × 109/L (P < .02 in both cases). The hypothetical mechanism(s) whereby rHu-GCSF may blunt PEG-rHuMGDF–induced platelet responses include lineage competition for early pluripotent progenitors, relative ligand-receptor specificities, binding affinities, signal transduction, receptor redundancy/pleiotropy, and altered pharmacokinetics, impacting on a markedly depleted reservoir of early hematopoietic progenitors surviving hepsulfam chemotherapy. The present study does not confirm or exclude any of these possibilities. However, changes in pharmacokinetics do not appear to be the explanation, as the pharmacokinetics of rHu-GCSF and PEG-rHuMGDF in the present study are not significantly affected by either of these cytokines. It should also be noted that coadministration of rHu-GCSF and PEG-rHuMGDF in a rhesus model of TBI-myeloablation, did not show diminution in megakarycoyte responsiveness.63 

Because PEG-rHuMGDF induces both platelet and neutrophil responses after marrow suppressive chemotherapy (Fig 4), we studied the effects on hematopoietic recovery of adding rHu-GCSF after instituting PEG-rHuMGDF therapy. By administering PEG-rHuMGDF for 1 week before adding rHu-GCSF for 2 subsequent weeks, both the platelet and neutrophil counts are maintained at or above baseline values, which was not achievable by concurrently administering the two cytokines.

Thus, in this rhesus model of hepsulfam marrow suppression, initiating PEG-rHuMGDF therapy for 7 days and adding rHu-GCSF thereafter completely eliminates postchemotherapy thrombocytopenia and neutropenia. The combination of lineage-dominant PEG-rHuMGDF and rHu-GCSF following potentially curative myelosuppressive chemotherapy may be a useful strategy for minimizing hematopoietic toxicity in patients receiving high-dose marrow-lethal chemotherapy with marrow or peripheral blood progenitor cell transplantation. The present study demonstrates the importance of optimizing the dose and schedule of cytokine combinations following severe myelosuppressive chemotherapy.

We thank Deborah White and Evan Dessasau for expert technical assistance. We also thank Dr William P. Sheridan for useful suggestions during this study and for reviewing the manuscript.

Supported by National Institutes of Health Grants No. HL31950 and RR00165 and by a research grant from Amgen, Inc.

Address reprint requests to Laurence A. Harker, MD, Division of Hematology and Oncology, Emory University School of Medicine, PO Drawer AR, Atlanta, GA 30322.

1
Sheridan
WP
Morstyn
G
Wolf
M
Dodds
A
Lusk
J
Maher
D
Layton
SE
Green
MD
Souza
L
Fox
RM
Granulocyte colony-stimulating factor and neutrophil recovery after high-dose chemotherapy and autologous bone marrow transplant.
Lancet
2
1989
891
2
Groopman
JE
Molina
JM
Scadden
DT
Hematopoietic growth factors. Biology and clinical application.
N Engl J Med
321
1989
1449
3
Crawford
J
Ozer
H
Stoller
R
Reduction in the incidence of chemotherapy-induced febrile neutropenia in patients with small cell lung cancer by granulocyte colony-stimulating factor (r-metHuG-CSF ).
N Engl J Med
315
1991
164
4
Slichter SJ: Principles of platelet transfusion therapy, in Hoffman R, Benz EJ, Jr, Shattil SJ, Furie B, Cohen HJ (eds): Hematology: Basic Principles and Practice. New York, NY, Churchill Livingstone, 1991, p 1610
5
Mollison PL, Engelfriet CP, Contreras M: The transfusion of platelets, leucocytes and plasma components, in Mollison PL, Engelfriet CP, Contreras M (eds): Blood Transfusion in Clinical Medicine. London, UK, Blackwell Scientific, 1993, p 638
6
Slichter
SJ
Harker
LA
Thrombocytopenia mechanisms and management of defects in platelet production.
Clin Haematol
7
1978
523
7
Wendling
F
Maraskovsky
E
Debili
N
Florindo
C
Teepe
M
Titeux
M
Methia
N
Breton-Gorius
J
Cosman
D
Vainchenker
W
c-Mpl ligand is a humoral regulator of megakaryocytopoiesis.
Nature
369
1994
571
8
Kaushansky
K
Lok
S
Holly
RD
Broudy
VC
Lin
N
Bailey
MC
Forstrom
JW
Buddle
MM
Oort
PJ
Hagen
FS
Roth
GJ
Papayannopoulou
T
Foster
DC
Promotion of megakaryocyte progenitor expansion and differentiation by the c-Mpl ligand thrombopoietin.
Nature
369
1994
568
9
deSauvage
FJ
Hass
PE
Spencer
SD
Malloy
BE
Gurney
AL
Spencer
SA
Darbonne
WC
Henzel
WJ
Wong
SC
Kuang
W-J
Oles
KJ
Hultgren
B
Solberg
LA Jr
Goeddel
DV
Eaton
DL
Stimulation of megakaryocytopoiesis and thrombopoiesis by the c-Mpl ligand.
Nature
369
1994
533
10
Bartley
TD
Bogenberger
J
Hunt
P
Li
Y-S
Lu
HS
Martin
F
Chang
M-S
Samal
B
Nichol
JL
Swift
S
Johnson
MJ
Hsu
R-Y
Parker
VP
Suggs
S
Skrine
JD
Merewether
LA
Clogston
C
Hsu
E
Hokom
MM
Hornkohl
A
Choi
E
Pangelinan
M
Sun
Y
Mar
V
McNinch
J
Simonet
L
Jacobsen
F
Xie
C
Shutter
J
Chute
H
Basu
R
Selander
L
Trollinger
D
Sieu
L
Padilla
D
Trail
G
Elliott
G
Izumi
R
Covey
T
Crouse
J
Garcia
A
Xu
W
Del Castillo
J
Biron
J
Cole
S
Hu
MC-T
Pacifici
R
Ponting
I
Saris
C
Wen
D
Yung
YP
Lin
H
Bosselman
RA
Identification and cloning of a megakaryocyte growth and development factor that is a ligand for the cytokine receptor Mpl.
Cell
77
1994
1117
11
Sohma
Y
Akahori
H
Seki
N
Hori
T
Ogami
K
Kato
T
Shimada
Y
Kawamura
K
Miyazaki
H
Molecular cloning and chromosomal localization of the human thrombopoietin gene.
FEBS Lett
353
1994
57
12
Kuter
DJ
Beeler
DL
Rosenberg
RD
The purification of megapoietin: A physiological regulator of megakaryocyte growth and platelet production.
Proc Natl Acad Sci USA
91
1994
11104
13
Hoffman
R
Regulation of megakaryocytopoiesis.
Blood
74
1989
1196
14
Kaushansky
K
Thrombopoietin: The primary regulator of platelet production.
Blood
86
1995
419
15
Nichol
JL
Hokom
MM
Hornkohl
A
Sheridan
WP
Ohashi
H
Kato
T
Li
YS
Bartley
TD
Choi
E
Bogenberger
J
Skrine
JD
Knudten
A
Chen
J
Trail
G
Sleeman
L
Cole
S
Grampp
G
Hunt
P
Megakaryocyte growth and development factor. Analyses of in vitro effects on human megakaryopoiesis and endogenous serum levels during chemotherapy-induced thrombocytopenia.
J Clin Invest
95
1995
2973
16
Harker
LA
Hunt
P
Marzec
UM
Kelly
AB
Tomer
A
Hanson
SR
Stead
RB
Regulation of platelet production and function by megakaryocyte growth and development factor (MGDF ) in nonhuman primates.
Blood
87
1996
1833
17
Choi
ES
Hokom
M
Bartley
T
Li
Y-S
Ohashi
H
Kato
T
Nichol
JL
Skrine
J
Knudten
A
Chen
J
Hornkohl
A
Grampp
G
Sleeman
L
Cole
S
Trail
G
Hunt
P
Recombinant human megakaryocyte growth and development factor (rHuMGDF ), a ligand for c-Mpl, produces functional human platelets in vitro.
Stem Cells
13
1995
317
18
Harker
LA
Finch
CA
Thrombokinetics in man.
J Clin Invest
48
1969
963
19
Neelis KJ, Wognum AW, Eaton DL, Thomas R, Wagemaker G: Preclinical evaluation of thrombopoietin in rhesus monkeys. Blood 86:256a, 1995 (abstr, suppl 1)
20
Gurney
AL
Carver-Moore
K
de Sauvage
FJ
Moore
MW
Thrombocytopenia in c-mpl-deficient mice.
Science
265
1994
1445
21
de Sauvage EJ, Luoh S-M, Carver-Moore K, Ryan A, Dowd M, Eaton DL, Moore MW: Deficiencies in early and late stages of megakaryocytopoiesis in TPO-KO mice. Blood 86:255a, 1995 (abstr, suppl 1)
22
Winton
EF
Srinivasiah
J
Kim
BK
Hillyer
CD
Strobert
EA
Orkin
JL
Swenson
RB
McClure
HM
Myers
LA
Saral
R
Effects of recombinant human interleukin-6 (rhIL-6) and rhIL-3 on hematopoietic regeneration as demonstrated in a nonhuman primate chemotherapy model.
Blood
84
1994
65
23
Hendricks
CB
Grochow
LB
Rowinsky
EK
Forastiere
AA
McGuire
WP
Ettinger
DS
Sartorius
S
Lubejko
B
Donehower
RC
Phase I and pharmacokinetic study of hepsulfam (NSC 329680).
Cancer Res
51
1991
5781
24
Ravdin
PM
Havlin
KA
Marshall
MV
Brown
TD
Koeller
JM
Kuhn
JG
Rodriguez
G
Von Hoff
DD
A phase I clinical and pharmacokinetic trial of hepsulfam Center, San Antonio 78284-7884.
Cancer Res
51
1991
6268
25
Chang
MS
McNinch
J
Basu
R
Shutter
J
Hsu
RY
Perkins
C
Mar
V
Suggs
S
Welcher
A
Li
L
et al
Cloning and characterization of the human megakaryocyte growth and development factor (MGDF ) gene.
J Biol Chem
270
1995
511
26
Hanson
SR
Pareti
FI
Ruggeri
ZM
Kunicki
TJ
Montgomery
RR
Zimmerman
TS
Harker
LA
Effects of monoclonal antibodies against the platelet glycoprotein IIb/IIIa complex on thrombosis and hemostasis in the baboon.
J Clin Invest
81
1988
149
27
Cadroy
Y
Hanson
SR
Kelly
AB
Marzec
UM
Evatt
BL
Kunicki
TJ
Montgomery
RR
Harker
LA
Relative antithrombotic effects of monoclonal antibodies targeting different platelet glycoprotein-adhesive molecule interactions in non-human primates.
Blood
83
1994
3218
28
Harker
LA
Marzec
UM
Hunt
P
Kelly
AB
Tomer
A
Cheung
E
Hanson
SR
Stead
RB
Dose-response effects of pegylated human megakaryocyte growth and development factor (PEG-rHuMGDF ) on platelet production and function in nonhuman primates.
Blood
88
1996
511
29
Maher
DW
Lieschke
GJ
Green
M
Bishop
J
Stuart-Harris
R
Wolf
M
Sheridan
WP
Kefford
JF
Cebon
J
Olver
I
McKendrick
J
Toner
G
Bradstock
K
Lieschke
M
Cruickshank
S
Tomita
DK
Hoffman
EW
Fox
RM
Morstyn
G
Filgrastim in patients with chemotherapy-induced febrile neutropenia: A double-blind, placebo-controlled trial.
Ann Intern Med
121
1994
492
30
Shapiro HM: Practical Flow Cytometry. New York, NY, Wiley-Liss, 1995
31
Tomer
A
Friese
P
Conklin
R
Bales
W
Archer
L
Harker
LA
Burstein
SA
Flow cytometric analysis of megakaryocytes from patients with abnormal platelet counts.
Blood
74
1989
594
32
Grosset
DG
Georgiadis
D
Stirling
S
Cowburn
P
Kelman
AW
Faichney
A
Lees
KR
Subclinical embolism in patients undergoing cardiac valve implantation and coronary artery bypass surgery.
Eur J Cardiothorac Surg
8
1994
63
33
Tomer
A
Harker
LA
Burstein
SA
Flow cytometric analysis of normal human megakaryocytes.
Blood
71
1988
1244
34
Tomer
A
Hanson
SR
Harker
LA
Autologous platelet kinetics in patients with severe thrombocytopenia: Discrimination between disorders of production and destruction.
J Lab Clin Med
118
1991
546
35
Savage
B
McFadden
PR
Hanson
SR
Harker
LA
The relation of platelet density to platelet age: Survival of low and high-density 111Indium-labeled platelets in baboons.
Blood
68
1986
386
36
Bailar
JC
Mosteller
F
Guidelines for statistical reporting in articles for medical journals.
Ann Intern Med
108
1988
266
37
Ravdin
PM
Havlin
KA
Marshall
MV
Brown
TD
Koeller
JM
Kuhn
JG
Rodriguez
G
Von Hoff
DD
A phase I clinical and pharmacokinetic trial of hepsulfam.
Cancer Res
51
1991
6268
38
Winton
EF
Jacobs
PC
Rozmiarek
SK
Stahl
CP
Myers
LA
Liehl
E
Stoll
RE
Anderson
DC
McClure
HM
Studies of an improved rhesus hematopoietic progenitor cell assay.
Exp Hematol
20
1992
401
39
Odell
TT
McDonald
TP
Detwiler
TC
Stimulation of platelet production by serum of platelet-depleted rats.
Proc Soc Exp Biol Med
108
1961
428
40
Harker
LA
Kinetics of thrombopoiesis.
J Clin Invest
47
1968
458
41
Hokom
MM
Lacey
D
Kinstler
OB
Choi
E
Kaufman
S
Faust
J
Rowan
C
Dwyer
E
Nichol
JL
Grasel
T
Wilson
J
Steinbrink
R
Hecht
R
Winters
D
Boone
T
Hunt
P
Pegylated megakaryocyte growth and development factor abrogates the lethal thrombocytopenia associated with carboplatin and irradiation in mice.
Blood
86
1995
4486
42
Farese AM, Hunt P, Grab LB, MacVittie TJ: Evaluation of administration protocols of pegylated-megakaryocyte growth and development factor on platelet recovery in a primate model of radiation-induced bone marrow aplasia. Blood 86:497a, 1995 (abstr)
43
Basser R, Clarke K, Fox R, Green M, Cebon J, Marty J, Menchaca D, Tomita D, Bealey G: Randomized, double-blind, placebo-controlled phase I trial of pegylated megakaryocyte growth and development factor (PEG-rHuMGDF ) administered to patients with advanced cancer before and after chemotherapy-early results. Blood 86:257a, 1995 (abstr, suppl 1)
44
Metcalf D: Implications of the polyfunctionality of hemopoietic regulators. Stem Cells 12:259, 1994 (suppl 1)
45
Jakubowski AA, Golde DW: Therapeutic use of cytokines, in Beutler E, Lichtman MA, Coller BS, Kipps TJ (eds): Hematology. New York, NY, McGraw-Hill, 1995, p 155
46
Souza
LM
Boone
TC
Gabrilove
JL
Recombinant human granulocyte colony stimulating factor: Effects on normal and leukemic myeloid cells.
Science
232
1986
61
47
Zsebo
KM
Cohen
AM
Murdock
DC
Recombinant human granulocyte colony stimulating factor: Molecular and biological characterization.
Immunology
172
1986
175
48
Yuo
A
Kitagawa
S
Ohsaka
A
Recombinant human granulocyte colony-stimulating factor as an activator of human granulocytes: Potentiation of responses triggered by receptor-mediated agonists and stimulation of C3bi receptor expression and adherence.
Blood
74
1989
2144
49
Morella
KK
Bruno
E
Kumaki
S
Lai
C-F
Fu
J
Wang
H-M
Murray
L
Hoffman
R
Tumour
M
Benit
L
Gisselbrecht
S
Zhuang
H
Wojchowski
DM
Baumann
H
Gearing
DP
Signal transduction by the receptors for thrombopoietin (c-mpl) and interleukin-3 in hematopoietic and nonhematopoietic cells.
Blood
86
1995
557
50
Drachman
JG
Griffin
JD
Kaushansky
K
The c-mpl ligand (thrombopoietin) stimulates tyrosine phosphorylation of Jak2, Shc and c-mpl.
J Biol Chem
270
1995
4979
51
Ritchie A, Vadhan-Raj S, Broxmeyer HE: Thrombopoietin suppresses growth factor withdrawal induced apoptosis and behaves as a survival factor for the human growth factor-dependent cell line M07e. Blood 86:21a, 1995 (abstr, suppl 1)
52
Katagiri T, Miyazawa K, Tauchi T, Nishimaki J, Gotoh A, Broxmeyer HE, Toyama K: Comparative analysis of signal transduction pathways between thrombopoietin and erythropoietin involves RAS- and JAK-stat-mediated signalings. Blood 86:151a, 1995 (abstr, suppl 1)
53
Hill RJ, Zozulya S, Lu J, Hollenbach P, Bogenberger J, Gishizky M: Stimulation of the c-MPL receptor with megakaryocyte growth and development factor (MGDF ) induces SHC-dependent differentiation phenotype in hematopoietic cells. Blood 86:696a, 1995 (abstr, suppl 1)
54
Ohashi H, Morita H, Misaizu T, Tsunakawa H, Tahara T, Tsumura H, Miyazaki H, Kato T: Receptor activation mechanisms and signal transduction pathways of truncated or chimeric c-mpl. Blood 86:905a, 1995 (abstr, suppl 1)
55
Ulich
TR
Castillo
J
Yin
S
Swift
S
Padilla
D
Senaldi
G
Bennett
L
Shutter
J
Bogenberger
J
Sun
D
Samal
B
Shimamoto
G
Lee
R
Steinbrink
R
Boone
T
Sheridan
WT
Hunt
P
Megakaryocyte growth and development factor ameliorates carboplatin-induced thrombocytopenia in mice.
Blood
86
1995
971
56
Akahori H, Kabaya K, Shibuya K, Obuchi M, Kato T: Effects of recombinant human thrombopoietin (rhTPO) and granulocyte colony-stimulating factor (rHuG-CSF ) on pancytopenia induced by chemotherapy. Blood 86:892a, 1995 (abstr, suppl 1)
57
Thibodeaux H, Mathias J, Eaton DL, Thomas GR: Evaluation of thrombopoietin (TPO) in murine models of thrombocytopenia induced by whole body irradiation and cancer chemotherapeutic agents. Blood 86:497a, 1995 (abstr, suppl 1)
58
Grossmann A, Lenox JS, Humes J, Ren HP, Kaushansky K, Sprugel KH: Effects of the combined administration of TPO and G-CSF on recovery from myelosuppression in mice. Blood 86:371a, 1995 (abstr, suppl 1)
59
Molineaux G, Hartley CA, McElroy P, McCrea C, McNiece LK: Effects of treatment with MGDF and G-CSF on the acceleration of stimultaneous neutrophil and platelet recovery after bone marrow transplant in mice. Blood 86:227a, 1995 (abstr, suppl 1)
60
Fibbe
WE
Heemskerk
DPM
Laterveer
L
Pruijt
JFM
Foster
D
Kaushansky
K
Willemze
R
Accelerated reconstitution of platelets and erythrocytes after syngeneic transplantation of bone marrow cells derived from thrombopoietin pretreated donor mice.
Blood
86
1995
3308
61
Kabaya K, Akahori H, Shibuya K, Nitta Y, Kusaka M, Kato T, Miyazaki H: Improvement of thrombocytopenia after bone marrow-transplantation by recombinant human thrombopoietin (rhTPO) in mice. Blood 86:114a, 1995 (abstr, suppl 1)
62
Molineux G, Hartley CA, McElroy P, McCrea C, McNiece IK: Megakaryocyte growth and development factor (MGDF ) accelerates platelet recovery in peripheral blood progenitor cell (PBPC) transplant recipients. Blood 86:461a, 1995 (abstr, suppl 1)
63
Farese
AM
Hunt
P
Grab
LB
MacVittle
TJ
Combined administration of recombinant human megakaryocyte growth and development factor and granulocyte colony-stimulating factor enhances multilineage hematopoietic reconstitution in nonhuman primates after radiation-induced marrow aplasia.
J Clin Invest
97
1996
1
64
Jiang
S
Levine
JD
Fu
Y
Deng
B
London
R
Groopman
JE
Avraham
H
Cytokine production by primary bone marrow megakaryocytes.
Blood
84
1994
4151
65
Wickenhauser
C
Lorenzen
J
Thiele
J
Hillienhof
A
Jungheim
B
Hansmann
ML
Fischer
R
Secretion of cytokines (interleukins-1a,-3, and -6 and granulocyte macrophage colony-stimulating factor) by normal human bone marrow megakaryocytes.
Blood
85
1995
685
66
Navarro
S
Debili
N
LeCouedic
J-P
Klein
B
Breton-Gorius
J
Doly
J
Vainchecker
W
Interleukin-6 and its receptor are expressed by human megakaryocytes: In vitro effects on proliferation and endoreplication.
Blood
77
1991
461
67
Kaushansky
K
Karplus
PA
Hematopoietic growth factors: Understanding functional diversity in structural terms.
Blood
82
1993
3229
68
McNiece
IK
McGrath
HE
Quesenberry
PJ
Granulocyte colony-stimulating factor augments in vitro megakaryocyte colony formation by interleukin-3.
Exp Hematol
16
1988
807
69
Ku L, Kaushansky K, Ogawa M: Thrombopoietin, the ligand for the Mpl receptor, synergises with steel factor and other early-acting cytokines in supporting proliferation of primitive hematopoietic progenitors of mice. Blood 86:256a, 1995 (abstr, suppl 1)
70
Sitnicka E, Lin N, Fox N, Priestley GV, Broudy V, Wolf NS, Kaushansky K: The effect of thrombopoietin on the proliferation and differentiation of murine hematopoietic stem cells. Blood 86:419a, 1995 (abstr, suppl 1)
71
Hunt
P
Li
Y-S
Nichol
JL
Hokom
MM
Bogenberger
JM
Swift
SE
Skrine
JD
Hornkohl
AC
Lu
H
Clogston
C
Merewether
LA
Johnson
MJ
Parker
V
Knudten
A
Farese
A
Hsu
RY
Garcia
A
Stead
R
Bosselman
RA
Bartley
TD
Purification and biologic characterization of plasma-derived megakaryocyte growth and development factor.
Blood
86
1995
540
Sign in via your Institution