Myelopoietins (MPOs) constitute a family of engineered, chimeric molecules that bind and activate the IL-3 and G-CSF receptors on hematopoietic cells. This study investigated the in vivo hematopoietic response of rhesus monkeys administered MPO after radiation-induced myelosuppression. Animals were total body irradiated (TBI) in 2 series, with biologically equivalent doses consisting of either a 700 cGy dose of Cobalt-60 (60Co) γ-radiation or 600 cGy, 250 kVp x-irradiation. First series: On day 1 after 700 cGy irradiation, cohorts of animals were subcutaneously (SC) administered MPO at 200 μg/kg/d (n = 4), or 50 μg/kg/d (n = 2), twice daily, or human serum albumin (HSA) (n = 10). Second series: The 600 cGy x-irradiated cohorts of animals were administered either MPO at 200 μg/kg/d, in a daily schedule (n = 4) or 0.1% autologous serum (AS) , daily, SC (n = 11) for 23 days. MPO regardless of administration schedule (twice a day or every day) significantly reduced the mean durations of neutropenia (absolute neutrophil count [ANC] < 500/μL) and thrombocytopenia (platelet < 20 000/μL) versus respective control-treated cohorts. Mean neutrophil and platelet nadirs were significantly improved and time to recovery for neutrophils (ANC to < 500/μL) and platelets (PLT < 20 000/μL) were significantly enhanced in the MPO-treated cohorts versus controls. Red cell recovery was further improved relative to control-treated cohorts that received whole blood transfusions. Significant increases in bone marrow-derived clonogenic activity was observed by day 14 after TBI in MPO-treated cohorts versus respective time-matched controls. Thus, MPO, administered daily was as effective as a twice daily schedule for multilineage recovery in nonhuman primates after high-dose, radiation-induced myelosuppression.

The major dose limiting sequelae, consequent to radiation and/or chemotherapy are neutropenia and thrombocytopenia. Protocols involving dose intensification and/or schedule compression will most likely exacerbate the duration and degree of myelosuppression associated with standard dose therapy. Although G-CSF or GM-CSF are used clinically to reduce the obligate neutropenic consequences of cytotoxic therapy, the effective treatment of thrombocytopenia has remained elusive.1-7 Recently, cytokines such as IL-11 and the cognate ligand (L) of the c-mpl receptor (Mpl-L) offer renewed potential for increasing platelet production after cytotoxic therapy.8-16 The Mpl-L, the full length, glycosylated, recombinant human (rHu) thrombopoietin (Tpo), and the nonpegylated or pegylated (PEG) truncated form (encompassing the Epo-like domain) of the Mpl-L, designated as megakaryocyte growth and development factor (MGDF), significantly induced recovery of thrombopoiesis in myelosuppressed nonhuman primates.8,11,12,17 The extension of these models to combination studies showed that the Mpl-L in combination with G-CSF augmented the G-CSF–induced recovery of neutrophils without evidence of lineage competition after radiation-induced myelosuppression.11,17 Furthermore, the combination of daniplestim, an engineered IL-3 receptor agonist with a truncated Mpl-L (Epo-like domain), enhanced hemopoietic regeneration and reduced clinical support requirements noted with respective daniplestim or truncated Mpl-L monotherapy in the irradiated nonhuman primate.18 

Another approach to inducing multilineage hematopoietic recovery, subsequent to myelosuppressive therapy, has been to engineer new molecules such as chimeric growth factor (GF) receptor agonists that possess greater biologic activity than either GF agonist alone. Furthermore, the ability to engineer each GF component of the chimeric molecule offers the potential for enhancing the efficacy of GFs with narrow therapeutic windows when used as monotherapy, such as IL-3. These recently described families of chimeric proteins are composed of combined GF agonists for either IL-3 and G-CSF receptors (myelopoietin), IL-3 and Mpl-L receptors (promegapoietins), or Flt-3L and G-CSF receptors (progenipoietins).19-24 

The combination of engineered IL-3 and G-CSF receptor agonists to form myelopoietin (MPO) was based on a consistent amount of in vitro and in vivo evidence, suggesting the combination would enhance both neutrophil and platelet regeneration within a favorable toxicity profile.25-31 In fact, the chimeric IL-3/G-CSF receptor agonist, MPO, had a significantly higher potency for multilineage CFU stimulatory activity on human CD34+ bone marrow cells compared with mixtures of the IL-3 and G-CSF receptor agonists.20 The in vivo priming effect of IL-3 for the action of other cytokines on early hematopoietic progenitors suggested that maximum stimulation would be achieved by the combination of IL-3 and G-CSF.32 In addition, the combined therapeutic administration of daniplestim (IL-3 receptor agonist) and G-CSF to high-dose, sublethally irradiated nonhuman primates further decreased the cytopenic durations and nadirs for both platelets and neutrophils relative to the respective cytokine monotherapy.33 Also, preliminary pharmacokinetic studies of the MPO molecule in normal rhesus monkeys demonstrated an increased biologic half-life and suggested that a reduced administration schedule would be biologically equivalent to the normally required twice daily schedule used with native, or the engineered IL-3, daniplestim.31,33,34 This study evaluated the preclinical efficacy of myelopoietin, using twice daily or daily administration schedules on multilineage hematopoietic recovery in a nonhuman primate model of high-dose, radiation-induced myelosuppression.

Conduct of experiments

Experimental protocols evaluating the therapeutic efficacy of MPO were conducted using 2 models of radiation-induced myelosuppression. The same team of investigators conducted “Series 1” at the Armed Forces Radiobiology Research Institute (AFRRI), Bethesda, MD, and “Series 2” at the Greenebaum Cancer Center (GCC) at the University of Maryland, Baltimore, MD.

Animals

Male rhesus monkeys, Macaca mulatta, mean weight 3.5 ± 0.2 kg, were respectively housed in individual stainless steel cages in conventional holding rooms at the AFRRI or the Veterinary Resources Department at GCC in animal facilities accredited by the American Association for Accreditation of Laboratory Animal Care. Research was conducted according to the principles enunciated in the Guide for the Care and Use of Laboratory Animals,35 prepared by the Institute of Laboratory Animal Resources, National Research Council and research protocols approved by respective Institutional Animal Care and Use Committees.

Irradiation

Monkeys, after a prehabituation period, were irradiated in plexiglass restraining chairs for both protocols.First series. In the AFRRI protocol, monkeys received bilateral, total body irradiation (TBI) with Cobalt-60 (60Co) γ-radiation to a total midline tissue dose of 700 cGy at a dose rate of 40 cGy/min. Second series. In the GCC protocol, monkeys received unilateral TBI with 250 kVp x-radiation at 13 cGy/min in the posterior-anterior position, rotated 180° at the mid-dose (300 cGy) to the anterior-posterior position for completion of the total 600 cGy midline tissue exposure. The 700 cGy 60Co γ-radiation exposure is bioequivalent to the 600 cGy x-irradiation relative to hematopoietic myelosuppression (see Results, Table1). Dosimetry was performed using paired 0.5 cm3 ionization chambers, with calibration factors traceable to the National Institute of Standards and Technology.

Table 1.

Comparison of radiation-induced hematologic effects: 700 cGy 60Co γ-irradiation versus 600 cGy 250 kVp x-irradiation

700 cGy 60Co γ-Irradiation 600 cGy 250 kVp x-Irradiation
Neutropenia duration (days) 14.8 ± 0.7  16.2 ± 0.4  
ANC nadir (μL−1)  0 ± 0  8 ± 4  
ANC recovery (days)  19.7 ± 0.5  21.7 ± 0.4  
Thrombocytopenia duration (days)  11.9 ± 1.6  10.4 ± 0.8  
PLT nadir (μL−1)  5 000 ± 600  3 000 ± 1 000 
PLT recovery (days)  20.7 ± 1.4  20.7 ± 0.9 
700 cGy 60Co γ-Irradiation 600 cGy 250 kVp x-Irradiation
Neutropenia duration (days) 14.8 ± 0.7  16.2 ± 0.4  
ANC nadir (μL−1)  0 ± 0  8 ± 4  
ANC recovery (days)  19.7 ± 0.5  21.7 ± 0.4  
Thrombocytopenia duration (days)  11.9 ± 1.6  10.4 ± 0.8  
PLT nadir (μL−1)  5 000 ± 600  3 000 ± 1 000 
PLT recovery (days)  20.7 ± 1.4  20.7 ± 0.9 

Monkeys were total body irradiated to 700 cGy with60Co γ-irradiation (n = 10) or 600 cGy with 250 kVp x-irradiation (n = 11) as midline tissue doses and treated with control protein (HSA or autologous serum) according to protocol (Methods). Neutropenia is defined as an absolute neutrophil count (ANC) <500/μL and thrombocytopenia is defined as a platelet count (PLT) <20 000/μL. ANC and PLT recovery are the days required to reach sustained levels of ANC ≥500/μL and PLT ≥20 000/μL. Data represents mean values ± SEM.

Recombinant cytokines

Myelopoietin (SC-68420) (MPO), an engineered, chimeric hematopoietic growth factor that binds and activates the IL-3 and G-CSF receptors was produced in Escherichia coli through the use of a plasmid-based expression vector. It was expressed in insoluble inclusion bodies within the E coli cells. Washed inclusion bodies were solubilized in urea buffer and disulfide bonds formed through air oxidation after lowering the urea concentration. MPO was purified by ion exhange chromatography and filtration. Endotoxin levels were undetectable (Limulus amebocyte assay [Associates of Cape Cod, Inc, Woods Hole, MA]). Purified protein was stored as a frozen solution in 10 mmol/L Tris buffer, pH 8.0. In the previous study daniplestin (SC-55494) was provided by Searle R&D (St Louis, MO). G-CSF was purchased as Neupogen (Filgrastin) (Amgen Inc, Thousand Oaks, CA).

Study design

In each experimental series, animals were irradiated at day 0 and randomly assigned to a treatment protocol. First series, 700 cGy γ-irradiation protocols. On day 1 after 700 cGy γ-irradiation, animals received MPO at either 200 μg/kg/d (n = 4) or 50 μg/kg/d (n = 2), or HSA (Plasbumin-5, Miles Inc, Cutter Biological, Elkhart, IN). MPO or control protein, 15 μg/kg/d, n = 10, was injected twice daily (BID) as a divided dose (10-hour interval) as 1 mL subcutaneous (SC) injection bolus for 18 to 23 days. Administration was stopped when the white blood cell count reached 1 × 105/μL. Of the 4 animals injected with 200 μg/kg of MPO, 1 animal each was stopped after 18, 20, 22, and 23 days of administration. Both animals administered MPO at 50 μg/kg/d received the full 23-day schedule.Second series, 600 cGy x-irradiation protocols. On day 1 after 600 cGy x-irradiation, animals received MPO 200 μg/kg/d, n = 5 or 0.1% autologous serum (AS, n = 11) as control protein in 1 mL bolus SC injections once a day (QD) for 18 days.

Rationale for cytokine dose and schedule

The dose (50 μg/kg and 200 μg/kg) and administration schedule (BID vs QD) for MPO were based on several observations: (1) Native rhuIL-3 or engineered the IL-3 receptor agonist, daniplestim, have 20- to 50-fold less binding affinity for rhesus IL-3 receptor than the homologous, rhesus IL-3.36,37 (2) We previously demonstrated the enhanced therapeutic efficacy of daniplestim administered BID at 100 μg/kg/d relative to 25 μg/kg/d.31 (3) The MPO chimeric is constructed at approximately equimolar ratios of engineered IL-3 and G-CSF receptor agonists. Therefore, the 200 μg/kg dose of MPO provides approximately 100 μg/kg of engineered IL-3. (4) Monotherapy protocols that use native rhuIL-3 or daniplestim, required BID administration for optimal biologic response34 (Farese et al, unpublished observations, 1998). (5) The plasma pharmacokinetic parameters of daniplestim in irradiated monkeys31 and MPO in normal monkeys (Table 2, herein) suggested that an abbreviated schedule (QD vs BID) of MPO would induce an equivalent biologic response to the full BID schedule.

Table 2.

Plasma pharmacokinetic parameters for MPO after intravenous or subcutaneous administration to normal rhesus monkeys

Pharmacokinetic Parameters Rhesus
Intravenous* 
 Dose (μg/kg)  10    
Half-lives (h)  
 First 0.533 ± 0.051  
 Second  25.3 ± 14.0  
 Clearance (mL/min/kg)  0.415 ± 0.070  
 VC (L/kg) 0.0220 ± 0.0026  
 VSS (L/kg) 0.0706 ± 0.0481  
Subcutaneous* 
 Dose (μg/kg) 30    
 CMAX (ng/mL)  97.4 ± 29.1 
 TMAX (h)  3.3 ± 1.3  
 AUC ([ng/mL]h) 806 ± 368  
 Bioavailability (%)  62.5 
Pharmacokinetic Parameters Rhesus
Intravenous* 
 Dose (μg/kg)  10    
Half-lives (h)  
 First 0.533 ± 0.051  
 Second  25.3 ± 14.0  
 Clearance (mL/min/kg)  0.415 ± 0.070  
 VC (L/kg) 0.0220 ± 0.0026  
 VSS (L/kg) 0.0706 ± 0.0481  
Subcutaneous* 
 Dose (μg/kg) 30    
 CMAX (ng/mL)  97.4 ± 29.1 
 TMAX (h)  3.3 ± 1.3  
 AUC ([ng/mL]h) 806 ± 368  
 Bioavailability (%)  62.5 
*

Values are the mean (±SEM) for 3 monkeys.

VC and VSS are the apparent central compartment and steady state volumes of distribution, respectively.

Values are calculated from 0 to 24 hours for monkeys.

Clinical support

Clinical support consisting of an antimicrobial regimen and whole blood transfusions were provided to all animals as required. An antibiotic regimen and transfusions of fresh, irradiated (1500 cGy,60Co γ-irradiation) whole blood were administered as previously described.11,33 

Hematologic evaluations

Peripheral blood.

Peripheral blood was obtained from the saphenous vein to assay complete blood (Baker, System 9000, Serono-Baker, Allentown, PA or Sysmex K-4500, Long Grove, IL) and differential counts (Wright-Giemsa Stain, Ames Automated Slide Stainer, Elkhart, IN). Assessment of hematologic evaluations has been previously described.31 

Bone marrow.

Approximately 2 mL of heparinized-bone marrow (BM) was aspirated from the humerus and/or iliac crest of anesthetized primates (Ketaset, Fort Dodge, IA, 10 mg/kg intramuscular). Low density (< 1.077 g/cm3) mononuclear cells (MNC) were separated and cultured as previously described.31 GM-CFC and E-BFU derived colonies (> 50 cells) were expressed as the number of CFC/105 MNC. MK-CFC (10-50 cells/colony) are not distinguished from MK-BFC in this study, and are termed MK-CFC.

Pharmacokinetic analysis

The pharmacokinetics of MPO were determined in female rhesus monkeys (n = 3) after intravenous (IV) bolus (10 μg/kg) and SC (30 μg/kg) administration. MPO concentrations in plasma were determined by sandwich ELISA (enzyme-linked immunosorbent assay) procedures. The ELISA uses a murine monoclonal anti–G-CSF antibody, bound in microtiter plate wells, to bind MPO present in plasma samples. After removal of unbound plasma proteins by washing with buffer, MPO was quantified by the addition of goat polyclonal antidaniplestim antibody that is conjugated to horseradish peroxidase. The color produced by incubation with peroxidase substrate is read at 650 nm, and MPO concentrations are determined by reference to a standard curve prepared with MPO in plasma with a sensitivity limit of 0.150 ng/mL.

Statistical analysis

The Normal Scores Test was used to make pairwise comparisons of the durations of neutropenia and thrombocytopenia and evaluate the statistical significance between nadirs, the time to recovery and transfusion and antibiotic requirements. The tests were carried out using the software package StatXact (Cytel Software Corp, Cambridge, MA). The exact P values were obtained for all analysis.

Comparison of the MPO-treated cohorts with the combined daniplestim/G-CSF cohort in the published data set33 used a 1-way analysis of variance (ANOVA) across all groups, followed by linear contrasts representing each type comparison. Appropriate transforms were used to obtain homogeneous variance.

Bone marrow-derived clonogenic activities were analyzed by 2 methods. A repeated measures analysis was performed, followed by a 2-sided Dunnett's test to compare treatment days after TBI with baseline values. The comparisons of treated groups versus the time-matched, HSA- or AS-treated controls were made with a 1-way ANOVA, followed by a 1-sided Dunnett's test.

Relative hematologic effects of total body exposure to 700 cGy γ-irradiation versus 600 cGy 250 kVp x-irradiation

The acute hematologic suppression consequent to a midline tissue dose of 700 cGy γ-irradiation is not significantly different from that observed consequent to 600 cGy of 250 kVp x-irradiation (Table 1). The key parameters, indicative of the relative degree of myelosuppression, namely, the duration of neutropenia, absolute neutrophil count (ANC) < 500/μL, 14.8 and 16.2 days,P = .065, and thrombocytopenia, platelet count (PLT) < 20 000/μL, 11.9 and 10.4 days, P = .207, absolute neutrophil (0 and 8/μL, P = .139), and platelet nadirs, 5 000 and 3 000/μL, P = .197, are not statistically different after exposure to 700 cGy 60Co γ-irradiation or 600 cGy 250 kVp x-irradiation, respectively (Table 1). Inspection of the BM-derived clonogenic activity noted in Figures 2 and 4 for the respective control-treated cohorts also substantiates the equivocal hematopoietic myelosuppressive effects of these different quality radiation exposures.

Pharmacokinetics of MPO in normal animals

Elimination of MPO from rhesus monkey plasma occurred in 2 phases after bolus IV administration of 10 μg/kg. The mean values of the first and second phase half-lives were 0.533 and 25.3 hours, respectively (Table 2). The total body clearance of MPO was 0.415 mL/min/kg. The central compartment (Vc) and steady state (VSS) volumes of distribution were 0.0220 and 0.0706 L/kg, respectively. These volume of distribution values are similar to the plasma volume and whole blood volume, respectively, and indicate little, if any, distribution from blood into other tissues.

The maximal plasma concentration (Cmax) after a 30 μg/kg SC dose was 97.4 ng/mL at 3.3 hours (TMAX). MPO was measurable in plasma up to 24 to 48 hours after SC administration. Mean area under the plasma concentration-time curves (AUC) was 806 (ng/mL) hour. The apparent SC bioavailability (based on separate groups of animals for IV and SC doses) was 62.5%. Human whole body clearance of rhIL-3 and G-CSF have been reported to be approximately 4 to 5 mL/min and 0.5-0.7 mL/min/kg, respectively. The whole body clearance of MPO that would be expected in man was estimated by using an allometric scaling procedure. Linear extrapolation predicted that human clearance for MPO would approximate 0.2 mL/min/kg.

First series, 700 cGy γ-irradiation, MPO, BID administration protocol

Neutrophil recovery.

Neutrophil parameters were significantly improved by administration of MPO at a total daily dose of 200 μg/kg/d in a BID protocol after 700 cGy total body γ-irradiation. The duration of neutropenia (ANC < 500/μL) was decreased from 14.8 days in control-treated animals to 7.3 days (P = .008) and the neutrophil nadir was improved from absolute neutropenia in the control group to 390/μL (P = .005) (Table 3, Figure1A) in the MPO-treated cohort. The time to recovery of ANC to ≥ 500/μL was significantly improved from 19.7 days in the control-treated group to 12.3 days (P = .007) in the 200 μg/kg/d, BID, MPO-treated group. Consequently, the days on antibiotics were significantly reduced from 17.4 days for controls to 7.5 days (P = .005) for MPO-treated animals. Treatment with a lower dose of MPO (50 μg/kg/d), although less effective than the 200 μg/kg dose, improved the neutropenic duration (12.0 days), neutrophil nadir (36/μL), recovery time to ANC ≥ 500/μL (15.5 days), and days on antibiotics (12.5 days) relative to control-treated animals (Table 3).

Table 3.

Neutrophil and platelet-related parameters in sublethally γ-irradiated (700 cGy) or x-irradiated (600 cGy) rhesus monkeys treated with myelopoietin: duration, nadir, time to recovery, and clinical support

Neutrophil-Related Parameters
TreatmentDuration (days) Neutropenia ANC Nadir (μL−1) Time to Recovery (days)Antibiotics Required (days)
Series 1 (700 cGy) 
 HSA  14.8 ± 0.7   0  19.7 ± 0.5 17.4 ± 1.1  
 MPO, BID (200 μg/kg)  7.3 ± 3.13-150,3-151 390 ± 553-151,3-152 12.3 ± 3.13-151 7.5 ± 3.33-151 
 MPO, BID (50 μg/kg)3-153 12.0  36 15.5  12.5  
Previous data set (700 cGy)  
 HSA 14.8 ± 0.7   0  19.7 ± 0.5 17.4 ± 1.1  
 Daniplestim alone, BID  14.1 ± 1.8 92 ± 3.03-151,3-155 19.8 ± 0.93-155 13.4 ± 1.83-150 
 G-CSF alone, QD  12.4 ± 0.53-150 5 ± 4 16.2 ± 0.63-151 13.8 ± 0.63-151 
 Daniplestim, BID/G-CSF, QD  10.6 ± 0.73-151 124 ± 473-151,3-155 15.0 ± 0.63-151,3-155 9.2 ± 1.73-151,3-155 
Series 2 (600 cGy)  
 0.1% AS-control  16.2 ± 0.4  8 ± 4  21.7 ± 0.4 19.3 ± 0.5  
 MPO, QD  7.2 ± 2.03-151 377 ± 423-151,3-152 9.2 ± 3.83-151,3-152 10.0 ± 1.93-151 
Platelet-related parameters
 
Treatment  Duration (d) Thrombocytopenia  PLT Nadir (μL−1)  Time to Recovery (d) Transfusions  
Series 1 (700 cGy)  
 HSA 11.9 ± 1.6  5,000 ± 600  20.7 ± 1.4 1.7 ± 0.5  
 MPO (200 μg/kg)  2.0 ± 1.23-151 35,000 ± 10,0003-151 7.5 ± 3.33-151 1.0 ± 0.7 
 MPO (50 μg/kg)3-153 6.0  10,000 15.5  2.0  
Previous data set (700 cGy)  
 HSA 11.9 ± 1.6  5,000 ± 600  20.7 ± 1.4 1.7 ± 0.5  
 Daniplestim alone, BID  3.5 ± 1.23-151,3-155 14,400 ± 3,0003-151,3-155 13.2 ± 3.33-150 0.4 ± 0.23-150 
 G-CSF alone, QD  7.2 ± 0.93-150 7,400 ± 2,0003-150 16.4 ± 0.73-150 1.4 ± 0.8  
 Daniplestim, BID/G-CSF, QD 1.9 ± 0.83-151,3-155 22,200 ± 5,3003-151,3-155 8.6 ± 3.53-151,3-155 0 ± 03-151 
Series 2 (600 cGy) 
 0.1% AS-control  10.4 ± 0.8  3,000 ± 1,000 20.7 ± 0.9  2.6 ± 0.4  
 MPO, QD  3.2 ± 1.03-151 28,000 ± 10,4003-151 11.8 ± 2.23-151 0 ± 03-151 
Neutrophil-Related Parameters
TreatmentDuration (days) Neutropenia ANC Nadir (μL−1) Time to Recovery (days)Antibiotics Required (days)
Series 1 (700 cGy) 
 HSA  14.8 ± 0.7   0  19.7 ± 0.5 17.4 ± 1.1  
 MPO, BID (200 μg/kg)  7.3 ± 3.13-150,3-151 390 ± 553-151,3-152 12.3 ± 3.13-151 7.5 ± 3.33-151 
 MPO, BID (50 μg/kg)3-153 12.0  36 15.5  12.5  
Previous data set (700 cGy)  
 HSA 14.8 ± 0.7   0  19.7 ± 0.5 17.4 ± 1.1  
 Daniplestim alone, BID  14.1 ± 1.8 92 ± 3.03-151,3-155 19.8 ± 0.93-155 13.4 ± 1.83-150 
 G-CSF alone, QD  12.4 ± 0.53-150 5 ± 4 16.2 ± 0.63-151 13.8 ± 0.63-151 
 Daniplestim, BID/G-CSF, QD  10.6 ± 0.73-151 124 ± 473-151,3-155 15.0 ± 0.63-151,3-155 9.2 ± 1.73-151,3-155 
Series 2 (600 cGy)  
 0.1% AS-control  16.2 ± 0.4  8 ± 4  21.7 ± 0.4 19.3 ± 0.5  
 MPO, QD  7.2 ± 2.03-151 377 ± 423-151,3-152 9.2 ± 3.83-151,3-152 10.0 ± 1.93-151 
Platelet-related parameters
 
Treatment  Duration (d) Thrombocytopenia  PLT Nadir (μL−1)  Time to Recovery (d) Transfusions  
Series 1 (700 cGy)  
 HSA 11.9 ± 1.6  5,000 ± 600  20.7 ± 1.4 1.7 ± 0.5  
 MPO (200 μg/kg)  2.0 ± 1.23-151 35,000 ± 10,0003-151 7.5 ± 3.33-151 1.0 ± 0.7 
 MPO (50 μg/kg)3-153 6.0  10,000 15.5  2.0  
Previous data set (700 cGy)  
 HSA 11.9 ± 1.6  5,000 ± 600  20.7 ± 1.4 1.7 ± 0.5  
 Daniplestim alone, BID  3.5 ± 1.23-151,3-155 14,400 ± 3,0003-151,3-155 13.2 ± 3.33-150 0.4 ± 0.23-150 
 G-CSF alone, QD  7.2 ± 0.93-150 7,400 ± 2,0003-150 16.4 ± 0.73-150 1.4 ± 0.8  
 Daniplestim, BID/G-CSF, QD 1.9 ± 0.83-151,3-155 22,200 ± 5,3003-151,3-155 8.6 ± 3.53-151,3-155 0 ± 03-151 
Series 2 (600 cGy) 
 0.1% AS-control  10.4 ± 0.8  3,000 ± 1,000 20.7 ± 0.9  2.6 ± 0.4  
 MPO, QD  3.2 ± 1.03-151 28,000 ± 10,4003-151 11.8 ± 2.23-151 0 ± 03-151 

Series 1. Monkeys were total body irradiated (TBI) to 700 cGy with60Co-γ-radiation and treated with control protein, HSA (n = 10) or MPO at 200 μg/kg/d (n = 4) or 50 μg/kg/d (n = 2), BID, according to protocol (Methods). Data represents mean values ± SEM.

Series 2. Monkeys were total body irradiated (TBI) to 600 cGy with x-radiation and treated with control protein, autologous serum (AS) (n = 11) or MPO at 200 μg/kg/d, QD, (n = 5) according to protocol (Methods). Data represents mean values ± SEM.

Previous Data Set. Published study showing effects of coadministered daniplestim (BID) plus G-CSF (QD) relative to daniplestim and G-CSF monotherapy in the same 700 cGy 60Co-γ-radiation animal model performed in the same approximate time frame as Series 1.33 The control cohort (n = 10) is the same as series 1. The duration of neutropenia is defined as days of ANC <500/μL and thrombocytopenia is defined as days of platelets <20,000/μL. Time to recovery is the number of days required for the ANC to reach ≥500/μL and platelets to reach ≥20,000/μL.

F3-150

Statistically different from HSA/AS-treated controls (P ≤ .05).

F3-151

Statistically different from HSA/AS-treated controls (P ≤ .01).

F3-152

Statistically different from daniplestim(BID)/G-CSF(QD) treated animals (P ≤ .02).

F3-153

Values for MPO treated with 50 μg/kg/d were not evaluated statistically at n = 2.

F3-155

Statistically different from G-CSF-treated animals (P ≤ .05).

Fig. 1.

Effect of Myelopoietin (MPO) administration on peripheral blood (A) neutrophils (ANC), (B) platelets (PLT), (C) red blood cells (RBC), and (D) nucleated red blood cells (NRBC) in 700 cGy total body60Co-γ-irradiated animals.

Animals were administered MPO (BID, 200 μg/kg/d total dose, n = 4) or HSA (n = 10) as described in Methods. Average whole blood transfusions per animal: Control = 1.7, MPO = 1.0. Data represents mean values ± SEM.

Fig. 1.

Effect of Myelopoietin (MPO) administration on peripheral blood (A) neutrophils (ANC), (B) platelets (PLT), (C) red blood cells (RBC), and (D) nucleated red blood cells (NRBC) in 700 cGy total body60Co-γ-irradiated animals.

Animals were administered MPO (BID, 200 μg/kg/d total dose, n = 4) or HSA (n = 10) as described in Methods. Average whole blood transfusions per animal: Control = 1.7, MPO = 1.0. Data represents mean values ± SEM.

Close modal

Platelet recovery.

The BID administration of MPO at a total daily dose of 200 μg/kg/d significantly enhanced the recovery of platelets after 700 cGy total body γ-irradiation. The 11.9-day duration of thrombocytopenia (PLT < 20 000/μL) noted for control-treated animals was significantly reduced to 2.0 days (P = .001) in the MPO-treated group (200 μg/kg/d) (Table 3, Figure 1B). The mean platelet nadir in MPO-treated animals was also significantly improved (35 000/μL) relative to the control-treated cohort (5 000/μL)(P = .005). Recovery to platelet levels of ≥ 20 000/μL after 700 cGy TBI occurred earlier in the MPO-treated cohort, 7.5 days versus 20.7 days (P = .005) for the control-treated animals. Administration of the lower dose of MPO (50 μg/kg/d), although less effective than the 200 μg/kg dose, improved the duration of thrombocytopenia (6.0 days), platelet nadir (10 000 μL), and recovery of PLT to ≥ 20 000/μL (15.5 days) relative to the control-treated cohorts (Table 3).

Production of red blood cells (RBC), nucleated red blood cells (NRBC) and transfusion requirements.

The recovery of RBC in the MPO-treated cohort (200 μg/kg/d, BID) appears equivalent to the control cohort (Figure 1C). However, the appearance of NRBCs in the peripheral blood occurred earlier after MPO administration (17 to 23 days) than the HSA-treated controls (24 days) (Figure 1D). In addition, the HSA-treated cohort received an average of 1.7 transfusions per animal, whereas the MPO-treated cohort required 1.0 transfusion per animal (Table 3).

Bone marrow-derived clonogenic activity.

Bone marrow-derived GM-CFC, E-BFU, MK-CFC, and GEMM-CFC activity was evaluated at baseline and day 7, 14, 21, 48, and 100 after 700 cGy TBI. In the HSA-treated controls, the concentration of MK-CFC was significantly reduced through 21-day after exposure, whereas GM-CFC and E-BFU were significantly decreased through 48-day after TBI (Figure2). The number of recognizable GEMM-CFC were very low in baseline BM samples. An increase in GEMM-CFC per 105 BM cells was only observed at 48-day after TBI in the control animals (Figure 2). The recovery of clonogenic activity measured in the MPO-treated cohort (200 μg/kg/d, BID) was significantly enhanced relative to that noted for time-matched controls or respective baseline values (Figure 2A through D). All clonogenic activity noted for GM-CFC, MK-CFC, E-BFU, and GEMM-CFC in MPO-treated animals had returned to baseline values within 14 days after TBI.

Fig. 2.

Effect of Myelopoietin (MPO) administration (200 μg/kg/d) on the bone marrow-derived concentration of GM-CFC, MK-CFC, E-BFU, and GEMM-CFC in 700 cGy total body60Co-γ-irradiated animals.

Clonogenic activity was observed before (baseline [BL]) and on days 7, 14, 21, 48, and 100 after TBI and administration of MPO (n = 4, 200 μg/kg/d, BID) or a control protein (n = 10, HSA or AS) as described in Methods. All animals were assayed at each time point. Clonogenic concentrations (per 105 MNC) are reported as mean values ± SEM. Baseline GEMM-CFC are detectable in low concentration. There is no significant differences between the baseline values of MPO and control-treated animals. Asterisk (*) denotes significantly different from BL (P < .05); Hdenotes significantly greater than control protein, time-matched controls (P < .05); ↓ denotes zero values; ND denotes not done.

Fig. 2.

Effect of Myelopoietin (MPO) administration (200 μg/kg/d) on the bone marrow-derived concentration of GM-CFC, MK-CFC, E-BFU, and GEMM-CFC in 700 cGy total body60Co-γ-irradiated animals.

Clonogenic activity was observed before (baseline [BL]) and on days 7, 14, 21, 48, and 100 after TBI and administration of MPO (n = 4, 200 μg/kg/d, BID) or a control protein (n = 10, HSA or AS) as described in Methods. All animals were assayed at each time point. Clonogenic concentrations (per 105 MNC) are reported as mean values ± SEM. Baseline GEMM-CFC are detectable in low concentration. There is no significant differences between the baseline values of MPO and control-treated animals. Asterisk (*) denotes significantly different from BL (P < .05); Hdenotes significantly greater than control protein, time-matched controls (P < .05); ↓ denotes zero values; ND denotes not done.

Close modal

Second series, 600 cGy x-irradiation, MPO QD administration protocol

Neutrophil recovery.

Neutrophil regeneration was profoundly affected by the administration of MPO at 200 μg/kg/d in a QD schedule after TBI. Significant reduction in the duration of neutropenia and improvement in neutrophil nadir were evident in the MPO-treated animals compared with AS-treated controls 7.2 days versus 16.2 days and 377/μL versus 8/μL,P < .001 respectively (Table 3, Figure3A). The recovery of ANC to ≥ 500/μL was also significantly enhanced in MPO-treated animals relative to the controls, 9.2 days versus 21.7 days, P < .001. Consequently, the antibiotic requirements were reduced from 19.3 days for the controls to 10.0 days (P < .001) for MPO-treated animals (Table 3).

Fig. 3.

Effect of Myelopoietin (MPO) administration on recovery of peripheral blood (A) neutrophils (ANC), (B) platelets (PLT), (C) red blood cells (RBC), and (D) nucleated red blood cells (NRBC) in 600 cGy total body x-irradiated (250 kVp) animals.

Animals were administered MPO (QD, 200 μg/kg/d, n = 5) or control protein (n = 11) as described in Methods. Average whole blood transfusions per animal: Control = 2.6, MPO = 0. Data represents mean values ± SEM.

Fig. 3.

Effect of Myelopoietin (MPO) administration on recovery of peripheral blood (A) neutrophils (ANC), (B) platelets (PLT), (C) red blood cells (RBC), and (D) nucleated red blood cells (NRBC) in 600 cGy total body x-irradiated (250 kVp) animals.

Animals were administered MPO (QD, 200 μg/kg/d, n = 5) or control protein (n = 11) as described in Methods. Average whole blood transfusions per animal: Control = 2.6, MPO = 0. Data represents mean values ± SEM.

Close modal

Platelet recovery.

Administration of MPO at 200 μg/kg/d, QD for 18 consecutive days after TBI significantly reduced the duration of thrombocytopenia relative to AS-treated controls 3.2 days versus 10.4 days, respectively, P < .001 (Table 3, Figure 3B). The depth of platelet nadir was also significantly improved by MPO administration from the control value of 3 000/μL to 28 000/μL (Table 3, Figure3B). Recovery times of platelet count to ≥ 20 000/μL were also significantly decreased by QD administration with MPO (11.8 days) versus the control-treated cohort (20.7 days) (P < .001).

Production of red blood cells (RBC), nucleated red blood cells (NRBC) and transfusion requirements.

The MPO-induced recovery of RBCs was modestly improved relative to the control cohort (Figure 3C). However, the RBC recovery observed in the AS-treated animals reflects the effect of whole blood transfusions. The control cohort necessitated an average 2.6 whole blood transfusions per animal, whereas the MPO-treated animals were transfusion independent (P < .001) (Table 3). MPO administration after 600 cGy TBI induced an earlier appearance of NRBC in a fashion similar to that noted for the 700 cGy TBI protocol (Figure 3D).

Bone marrow-derived clonogenic activity.

Bone marrow-derived GM-CFC, MK-CFC, and E-BFU activity (CFC/105 MNCs) were evaluated before (baseline) and at days 7, 14, 21, and 46 after 600 cGy TBI. In the control-treated animals, all clonogenic activity was significantly decreased at least through 21 days after x-irradiation. Administration of MPO-stimulated recovery of GM-CFC and MK-CFC to within preirradiation values by 21 days after irradiation (Figure 4). Interestingly, E-BFU recovery was also enhanced by day 14 after QD administration of MPO.

Fig. 4.

Effect of Myelopoietin (MPO) administration on the bone marrow-derived concentration of GM-CFC, MK-CFC, and E-BFU in 600 cGy total body x-irradiated (250 kVp) animals.

Clonogenic activity was observed before (baseline [BL]) and on days 7, 14, 21, and 46 after TBI and administration of MPO (n = 5) or a control protein (n = 11, HSA or AS) as described in Methods. Clonogenic concentrations (per 105 MNC) are reported as mean values ± SEM. Asterisk (*) denotes significantly different from BL (P < .05); H denotes significantly greater than control protein, time-matched controls (P < .05); ↓ denotes zero values; ND denotes not done.

Fig. 4.

Effect of Myelopoietin (MPO) administration on the bone marrow-derived concentration of GM-CFC, MK-CFC, and E-BFU in 600 cGy total body x-irradiated (250 kVp) animals.

Clonogenic activity was observed before (baseline [BL]) and on days 7, 14, 21, and 46 after TBI and administration of MPO (n = 5) or a control protein (n = 11, HSA or AS) as described in Methods. Clonogenic concentrations (per 105 MNC) are reported as mean values ± SEM. Asterisk (*) denotes significantly different from BL (P < .05); H denotes significantly greater than control protein, time-matched controls (P < .05); ↓ denotes zero values; ND denotes not done.

Close modal

Comparison of MPO-induced hematopoietic recovery to that of coadministered daniplestim plus G-CSF.

We previously investigated the relative efficacy of coadministered daniplestim (BID) plus G-CSF (QD) to monotherapy with daniplestim (BID) or G-CSF (QD) in the equivalent 700 cGy γ-irradiated rhesus monkeys.33 We have added the key parameters in Table 3 for ready comparison to those generated herein. In that study, the coadministration of daniplestim/G-CSF significantly enhanced recovery of all neutrophil and platelet-related parameters relative to the control-treated cohort (Table 3). Furthermore, the coadministration of daniplestim/G-CSF improved all neutrophil and platelet-related parameters relative to the efficacy of daniplestim or G-CSF monotherapy.

Herein, we show that MPO, whether administered in a BID or QD schedule improved all neutrophil-related parameters noted with the coadministered daniplestim/G-CSF (Table 3). Duration of neutropenia, neutrophil nadir, recovery to an ANC ≥ 500/μL and number of days on antibitoics were all improved. Furthermore, significant improvements were noted in the ANC nadir for both BID (P < .001) and QD (P < .01) protocols and in the ANC time to recovery for the MPO, QD (P = .02) protocol relative to the daniplestim/G-CSF cohort. With regard to platelet and RBC-relative parameters, the effect of MPO (BID or QD) was equivalent to that of combined daniplestim/G-CSF (Table 3). In this regard, the QD administration schedule of MPO was as effective in enhancing hematopoietic recovery as the BID schedule.

Myelopoietin, a novel, chimeric molecule that binds and activates the IL-3 and G-CSF receptors had marked neutrophil and platelet restorative activity in a nonhuman primate model of radiation-induced myelosuppression. Furthermore, the favorable pharmacodynamic profile exhibited by MPO conferred an equivalent therapeutic efficacy between BID or QD administration protocols. MPO administration in either protocol significantly improved hematopoietic parameters indicative of myelopoiesis, thrombocytopoiesis, and erythropoiesis subsequent to high-dose sublethal radiation exposure. The neutrophil and platelet nadirs were significantly improved, neutropenic and thrombocytopenic durations were significantly reduced, and respective neutrophil and platelet recovery time to ≥ 500/μL and ≥ 20 000/μL were significantly enhanced. RBC recovery was improved subsequent to increased erythroid clonogenic activity. Increased hematopoietic recovery resulted in fewer transfusions and significantly reduced number of days on antibiotics for all MPO-treated animals.

The therapeutic benefit of the chimeric MPO, whether administered BID or QD, compares favorably with the coadministration of daniplestim (BID) and G-CSF(QD) after radiation-induced myelosuppression (Table3).33 MPO administration improved all neutrophil-related parameters relative to the cohort coadministered daniplestim/G-CSF: The duration of neutropenia, recovery time of ANC to ≥ 500/μL and neutrophil nadir were all improved. In fact, significant improvements were noted in neutrophil nadirs and time to recovery. Platelet- and RBC-related parameters, in the MPO-treated groups (BID or QD) were equivalent to that of the combined daniplestim/G-CSF cohort. Low-level immunoreactivity to MPO was only detected at greater than 21 d after initiation of treatment and had no effect on myeloid reconstitution.

The utility of the IL-3 molecule in a combined cytokine protocol has been suggested by a number of in vitro and in vivo studies.25-31,38,39 Specifically, the addition of IL-3 and G-CSF increased in vitro bone marrow-derived myeloid and megakaryocyte colony formation25-28 and in vivo use of IL-3 was noted to prime hematopoietic progenitor cells for subsequent ex vivo stimulation by lineage-dominant cytokines such as G-CSF or GM-CSF.32,38IL-3-induced stimulation of the erythroid lineage has also been noted in normal and myelosuppressed rhesus monkeys.31,40Furthermore, the noted trophic and survival-promoting aspects of IL-3 and G-CSF offer additional support for their use in combination cytokine protocols.41-47 Brandt et al44demonstrated that the presence of IL-3 delayed the induction of apoptosis in highly enriched human hematopoietic progenitor cells. The potential to reduce radiation-induced apoptosis is yet another aspect of the ability of IL-3 to act as a priming agent for the subsequent proliferative and differentiative stimuli of endogenous and/or exogenous cytokines secreted within or added to the postirradiation hematopoietic milieu.41,45,48 

Several previous studies evaluated the coadministration of multilineage and lineage dominant cytokines in nonhuman primate models of radiation or drug-induced myelosuppression in an attempt to stimulate multilineage regeneration. Those reported include IL-3/GM-CSF, IL-3/IL-6, IL-6/G-CSF, IL-6/GM-CSF, the fusion protein PIXY321, Tpo/G-CSF, and MGDF/G-CSF.11,12,17,34,39,49-54 Of these protocols, the most efficacious combination for production of both cell lineages, ie, neutrophil and platelet, within their respective models, were MGDF/G-CSF, Tpo/G-CSF, daniplestim (Synthokine)/G-CSF, and IL-3/GM-CSF.11,12,17,33,49 Each of the other combinations showed efficacy in either 1 lineage (IL-3/IL-6) or, if both lineages were enhanced (IL-6/G-CSF, IL-6/GM-CSF), the responses were not significantly greater than the respective cytokines used alone.34,50,51,53 

Of particular interest is the combination of the Mpl-L (PEG-rHuMGDF) and G-CSF, which significantly enhanced multilineage, hematopoietic recovery with no evidence of lineage competition in the same 700 cGy γ-radiation-induced myelosuppression model used herein.11The PEG-rHuMGDF plus G-CSF protocol significantly diminished thrombocytopenia, and the severity of platelet nadir relative to controls.11 Neutrophil recovery was also augmented and the combined PEG-rHuMGDF plus G-CSF protocol further decreased the neutropenic duration compared with G-CSF monotherapy. After radiation-induced myelosuppression, efficacy of MPO (BID or QD) compared favorably with regard to all parameters of platelet recovery, and further improved the duration of neutropenia and ANC nadir noted in our previous study with the combined PEG-MGDF/G-CSF protocol.

These results are concordant with the in vitro data suggesting that MPO is a multifunctional agonist of the human IL-3 and G-CSF receptor (R) complexes. Monahan et al55,56 have documented an affinity conversion event when comparing MPO binding to single IL-3R or G-CSFR positive cells versus that observed with double (IL-3 plus G-CSF) R positive cells. Essentially, MPO binds to both IL-3R and G-CSFRs on dual, IL-3R/G-CSFR cells with an affinity closely matching that of IL-3 and G-CSF for their Rs. In contrast, MPO binds with significantly lower affinity to single IL-3R positive cells, whereas MPO binds to G-CSFR positive cells with the same affinity as G-CSF. It is likely that by virtue of its IL-3 component, MPO recruits primitive multilineage HPCs and early myeloid cells into proliferation. IL-3Rs have been detected on unfractionated bone marrow and on purified CD34+ cells.36,57 Further analysis of IL-3R expression on CD34+ subsets purified from normal rhesus bone marrow indicated that Rs are expressed on both CD34bright RhLA-DRdull and CD34bright RhLA-DRbright cells.58Additional studies in the murine system have shown that cells within both primitive and mature hematopoietic stem cells express receptors for both IL-3 and G-CSF.59 

In addition to its capacity to stimulate primitive multilineage and early myeloid lineage dominant HPCs, MPO may also augment the hematopoietic milieu in the postirradiation microenvironment. The enhanced production of neutrophils after MPO-administration may be complemented by enhanced megakaryocytopoiesis and thrombopoiesis because of the combined autocrine and paracrine action of endogenously produced IL-1, IL-3, IL-6, IL-11, GM-CSF, c-kitL, and/or thrombopoietin. Marrow-derived megakaryocytes are capable of spontaneously secreting IL-1, IL-3, IL-6, and GM-CSF.60-62IL-3 can also enhance the secretion of IL-3, IL-6, and GM-CSF from marrow-derived megakaryocytes. Thus, the benefit derived from MPO may exceed the direct influence of the chimeric protein on the IL-3 and G-CSF receptor positive target cells.

These results will be of significance in the design considerations for clinical trials particularly because native IL-3 has required BID administration for optimal biologic effect, whether in monotherapy or combined cytokine therapeutic protocols. The equivalent therapeutic efficacy of MPO administered QD relative to that of the BID schedule would also forecast enhanced clinical utility. Based on the animal pharmacokinetic and pharmacodynamic data, effective plasma concentrations of MPO should be easily achieved and sustained for at least 12 to 24 hours after subcutaneous administration to man. The ability to use the most abbreviated and optimal schedules will aid in the design of the most favorable clinical protocols.

The demonstrated efficacy of MPO (SC 68420) in significantly improving the hematopoietic recovery after high-dose, sublethal irradiation in the nonhuman primate forecasts clinical efficacy for this engineered chimeric IL-3 and G-CSF receptor agonist for the treatment of both granulocytopenia and thrombocytopenia in the myelosuppressed patient.

We wish to thank Michael Flynn, Nelson Fleming, Lisa B. Lind, Lorelei Dacquel Smith, Daniel Casey, and Heather Webster for their superb technical assistance and William Jackson for assistance with the statistical analysis.

Reprints:Thomas J. MacVittie, Greenebaum Cancer Center, S9D11, University of Maryland, 22 S Greene St, Baltimore, MD 21201.

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

1
Morstyn
G
Campbell
L
Souza
LM
et al
Effect of granulocyte colony stimulating factor on neutropenia induced by cytotoxic chemotherapy.
Lancet.
1
1988
668
2
Sheridan
WP
Morstyn
G
Wolf
M
et al
Granulocyte colony-stimulating factor and neutrophil recovery after high-dose chemotherapy and autologous bone marrow transplantation.
Lancet.
2
1989
891
3
Antman
KS
Griffin
JD
Elias
A
et al
Effect of recombinant human granulocyte-macrophage colony-stimulating factor on chemotherapy-induced myelosuppression.
N Engl J Med.
319
1988
593
4
Vadhan-Raj
S
Keating
M
LeMaistre
PL
et al
Effects of recombinant human granulocyte-macrophage colony-stimulating factor in patients with myelodysplastic syndrome.
N Engl J Med.
317
1987
1545
5
Moore
JO
Dodge
RK
Amrein
PC
et al
Granulocyte-colony stimulating factor (Filgrastim) accelerates granulocyte recovery after intensive postremission chemotherapy for acute myeloid leukemia with aziridinyl benzoquinone and mitoxantrone: Cancer and Leukemia Group B Study 9022.
Blood.
89
1997
780
6
Crawford
J
Ozer
H
Stoller
R
et al
Reduction by granulocyte-colony stimulating factor of fever and neutropenia induced by chemotherapy in patients with small-cell lung cancer.
N Engl J Med.
325
1991
164
7
Rowe
JM
Anderson
JW
Mazza
JJ
et al
A randomized placebo-controlled phase III study of granulocyte-macrophage colony-stimulating factor in adult patients (>55 to 70 years of age) with acute myelogenous leukemia: a study of the Eastern Cooperative Oncology Group (E1490).
Blood.
86
1995
457
8
Akahori
H
Shibuya
A
Obuchi
M
et al
Effect of recombinant human thrombopoietin in nonhuman primates with chemotherapy-induced thrombocytopenia.
Br J Haematol.
94
1996
722
9
Grossman
A
Lenox
J
Ren
HP
et al
Thrombopoietin accelerates platelet, red blood cell, and neutrophilic recovery in myelosuppressed mice.
Exp Hematol.
24
1996
1238
10
Shibuya
K
Akahori
H
Takahashi
K
Tahara
E
Kato
T
Miyazaki
H
Multilineage hematopoietic recovery by a single injection of pegylated recombinant human megakaryocyte growth and development factor in myelosuppressed mice.
Blood.
91
1998
37
11
Farese
AM
Hunt
P
Grab
LB
MacVittie
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
2145
12
Harker
LA
Marzec
UM
Kelly
AB
et al
Prevention of thrombocytopenia and neutropenia in a nonhuman primate model of marrow suppressive chemotherapy by combining pegylated recombinant human megakaryocyte growth and development factor and recombinant human granulocyte colony-stimulating factor.
Blood.
89
1997
155
13
Du
XX
Keller
D
Goldman
S
Williams
DA
Functional effects of interleukin-11 treatment in vivo following bone marrow transplantation (BMT) and combined modality therapy in mice [abstract].
Exp Hematol.
20
1992
768a
14
Du
XX
Neben
T
Goldman
S
Williams
DA
Effects of recombinant interleukin-11 on hematopoietic reconstitution in transplant mice: acceleration of recovery of peripheral blood neutrophils and platelets.
Blood.
81
1993
27
15
Leonard
JP
Quinto
CM
Goldman
SJ
Kozitza
MK
Neben
TY
Recombinant human interleukin-11(rhIL-11) multilineage hematopoietic recovery in mice after a myelosuppressive regimen of sublethal irradiation and carboplatin.
Blood.
83
1994
1499
16
Isaacs
C
Robert
NJ
Bailey
FA
et al
Randomized placebo-controlled study of recombinant human interleukin-11 to prevent chemotherapy-induced thrombocytopenia in patients with breast cancer receiving dose-intensive cyclophosphamide and doxorubicin.
J Clin Oncol.
15
1997
3368
17
Neelis
KJ
Dubbelman
YD
Qingliang
L
Thomas
GR
Eaton
DL
Wagemaker
G
Simultaneous administration of TPO and G-CSF after cytoreductive treatment of rhesus monkeys prevents thrombocytopenia, accelerates platelet and red cell reconstitution, alleviates neutropenia, and promotes the recovery of immature bone marrow cells.
Exp Hematol.
25
1997
1084
18
Farese
A
MacVittie
TJ
Lind
LB
Smith
WG
McKearn
JP
The combined administration of daniplestim and MpL ligand augments the hematopoietic reconstitution observed with single cytokine administration in a nonhuman primate model of myelosuppression.
Thrombopoietin: From Molecule to Medicine.
Murphy
MJ
Kutner
DJ
1998
143
AlphaMed Press
Miamisburg
19
Monahan
JB
Hood
WF
Joy
WD
et al
Functional characterization of SC 68420—a multifunctional agonist which activates both IL3 and G-CSF receptors.
Blood.
86
1995
154a
(abstract).
20
McKearn
JP
Hood
WF
Monahan
JB
et al
Myelopoietin—a multifunctional agonist of human IL-3 and G-CSF receptors.
Blood.
86
1995
259a
(abstract).
21
Kahn
LE
Staten
NR
Wood
DC
et al
Development of engineered hematopoietic growth factors for multilineage expansion and maturation of CD34+ progenitors.
Exp Hematol.
24
1996
1072
(abstract).
22
Kahn
LE
Blystone
LW
Villani-Price
D
et al
In vitro and in vivo biology of an engineered novel factor for expansion and development of platelet producing cells.
Blood.
88
1996
351a
(abstract).
23
Giri
JG
Kahn
LE
Staten
NR
et al
An engineered novel factor for expansion and development of platelet producing cells.
Exp Hematol.
24
1996
1072
(abstract).
24
Giri
JG
Kahn
LE
Doshi
PD
et al
Promegapoietin, an engineered chimeric growth factor for platelet producing cells.
Blood.
88
1996
351a
(abstract).
25
McNiece
IK
McGrath
HE
Granulocyte colony-stimulating factor augments in vitro megakaryocyte colony formation by interleukin-3.
Exp Hematol.
16
1988
807
26
Paquette
RL
Zhou
JY
Yang
YC
Clark
SC
Koeffler
HP
Recombinant gibbon interleukin-3 acts synergistically with recombinant human G-CSF and GM-CSF in vitro.
Blood.
71
1988
1596
27
McNiece
IK
Andrews
R
Stewart
M
Clark
S
Boone
T
Quesenberry
P
Action of interleukin-3, G-CSF, and GM-CSF on highly enriched human hematopoietic progenitor cells: Synergistic interaction of GM-CSF plus G-CSF.
Blood.
74
1989
110
28
Ikebuchi
K
Wong
GG
Clark
SC
Ihle
JN
Hirai
T
Ogawa
M
Interleukin-6 enhancement of interleukin 3-dependent proliferation of multipotential hemopoietic progenitors.
Proc Natl Acad Sci USA.
84
1987
9035
29
Krumwieh
D
Weinmann
E
Seiler
FR
Different effects of interleukin-3 (IL-3) on the hematopoiesis of subhuman primates due to various combinations with granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte colony stimulating factor (G-CSF).
Int J Cell Cloning.
8
1990
229
30
Ulich
TR
Del Castillo
J
McNiece
IK
et al
Acute and subacute hematologic effects of multi-colony stimulating factor in combination with granulocyte colony-stimulating factor in vivo.
Blood.
75
1990
48
31
Farese
AM
Herodin
F
Grab
LB
et al
Acceleration of hematopoietic reconstitution with a synthetic cytokine (SC-55494) after radiation-induced marrow aplasia.
Blood.
87
1996
581
32
Aglietta
M
Sanavio
F
Stacchini
A
et al
Interleukin-3 in vivo: kinetic of response of target cells.
Blood.
82
1993
2054
33
MacVittie
TJ
Farese
AM
Herodin
F
Grab
LB
Baum
CM
McKearn
JP
Combination therapy of radiation-induced bone marrow aplasia in nonhuman primates using synthokine SC-55494 and recombinant human granulocyte colony-stimulating factor.
Blood.
87
1996
4129
34
Winton
EF
Srinivasiah
J
Kim
BK
et al
Effect of recombinant human interleukin-6 and rhIL-3 on hematopoietic regeneration as demonstrated in a nonhuman primate chemotherapy model.
Blood.
84
1994
65
35
The Institute of Laboratory Animal Resources, National Research Council
Guide for the Care and Use of Laboratory Animals.
1996
86
23
National Institutes of Health
Washington, DC
36
van Gils
F
Budel
L
Burger
H
van Leen
R
Lowenberg
B
Wagemaker
G
Interleukin-3 (IL-3) receptors on rhesus monkey cells: species specificity of human IL-3, binding characteristics, and lack of competition with GM-CSF.
Exp Hematol.
22
1994
248
37
Klein
B
Thomas
J
Klover
J
et al
Comparison of daniplestim and human interleukin-3: impact of structure on functional activity.
Blood.
92
1998
174b
(abstract).
38
Donahue
RE
Seehra
J
Metzger
M
et al
Human IL-3 and GM-CSF act synergistically in stimulating hematopoiesis in primates.
Science.
241
1988
1820
39
Winton
EF
Hillyer
CD
Srinivasiah
J
et al
A nonhuman primate model for the study of effects of cytokines on hematopoietic reconstitution following chemotherapy-induced marrow damage.
Advances in the Treatment of Radiation Injuries: Advances in the Biosciences.
MacVittie
TJ
Weiss
JF
Browne
D
1994
75
Pergamon
Great Britain
40
Wagemaker
G
van Gils
FCJM
Burger
H
et al
Highly increased production of bone marrow derived blood cells by administration of homologous interleukin-3 to rhesus monkeys.
Blood.
76
1990
2235
41
Collins
MK
Marvel
J
Malde
P
Lopez-Rivas
A
Interleukin-3 protects murine bone marrow cells from apoptosis induced by DNA damaging agents.
J Exp Med.
176
1992
1043
42
Collins
MKL
Perkins
GR
Rodriguez-Tarduchy
G
Nieto
MA
Lopez-Rivas
A
Growth factors as survival factors: regulation of apoptosis.
Bioessays.
16
1994
133
43
Bodine
D
Crosier
P
Clark
SC
Effects of hematopoietic growth factors on the survival of primitive stem cells in liquid suspension culture.
Blood.
78
1991
914
44
Brandt
JE
Bhalla
K
Hoffman
R
Effects of interleukin-3 and c-kit ligand on the survival of various classes of human hematopoietic progenitor cells.
Blood.
83
1994
1507
45
Yee
NS
Paek
I
Besmer
P
Role of kit-ligand in proliferation and suppression of apoptosis in mast cells: basis for radiosensitivity of white spotting and steel mutant mice.
J Exp Med.
179
1994
1777
46
Williams
GT
Smith
CA
Spooncer
E
Dexter
TM
Taylor
DR
Haemopoietic colony stimulating factors promote cell survival by suppressing apoptosis.
Nature.
343
1990
76
47
Lin
Y
Benchimol
S
Cytokines inhibit p53-mediated apoptosis but not G1 arrest.
Mol Cell Biol.
15
1995
6045
48
Silva
A
Wyllie
A
Collins
MKL
p53 is not required for regulation of apoptosis or radioprotection by interleukin-3.
Blood.
89
1997
2717
49
Farese
AM
Williams
DE
Seiler
FR
MacVittie
TJ
Combination protocols of cytokine therapy with interleukin-3 and granulocyte-macrophage colony-stimulating factor in a primate model of radiation-induced marrow aplasia.
Blood.
82
1993
3012
50
MacVittie
TJ
Farese
AM
Patchen
ML
Myers
LA
Therapeutic efficacy of recombinant interleukin-6 alone and combined with recombinant human IL-3 in a nonhuman primate model of high-dose, sublethal radiation-induced marrow aplasia.
Blood.
84
1994
2515
51
Gonter
PW
Hillyer
CD
Strobert
EA
et al
Enhanced post-chemotherapy platelet and neutrophil recovery using IL-6 and rhGM-CSF in a nonhuman primate model.
Blood.
82
1993
365a
(abstract).
52
Gonter
PW
Hillyer
CD
Strobert
EA
et al
The effect of varying ratios of administered rhIL-3 and rhGM-CSF on post-chemotherapy marrow regeneration in a nonhuman primate model.
Blood.
82
1993
365a
(abstract).
53
Farese
AM
Myers
LA
MacVittie
TJ
Therapeutic efficacy of the combined administration of either recombinant human interleukin-6 and rh-granulocyte colony stimulating factor or rh-granulocyte-macrophage colony stimulating factor in a primate model of radiation-induced marrow aplasia.
Exp Hematol.
22
1994
684a
(abstract).
54
Williams
DE
Dunn
JT
Park
LS
et al
A GM-CSF/ IL-3 fusion protein promotes neutrophil and platelet recovery in sublethally irradiated rhesus monkeys.
Biotechnol Ther.
4
1993
17
55
Monahan
JB
Hood
WF
Joy
WD
et al
Receptor binding and signalling characteristics of MPO-1 (SC-68420)—a multifunctional IL-3 and G-CSF receptor agonist.
Exp Hematol.
24
1996
1154
(abstract).
56
McKearn
J
Monahan
J
Hood
W
et al
Myelopoietin—a multifunctional chimeric protein family.
Exp Hematol.
24
1996
1154
(abstract).
57
Sato
N
Caux
C
Kitamura
T
et al
Expression and factor-dependent modulation of interleukin-3 receptor subunits on human hematopoietic cells.
Blood.
82
1993
752
58
Wognum
AW
Visser
TP
de Jong
MO
Egeland
T
Wagemaker
G
Differential expression of receptors for interleukin-3 on subsets of CD34-expressing hematopoietic cells for rhesus monkeys.
Blood.
86
1995
581
59
McKinstry
WJ
Li
CL
Rasko
JEJ
Nicola
NA
Johnson
GR
Metcalf
D
Cytokine receptor expression on hematopoietic stem and progenitor cells.
Blood.
89
1997
65
60
Jiang
S
Levine
JD
Fu
Y
et al
Cytokine production by primary bone marrow megakaryocytes.
Blood.
84
1994
4151
61
Wickenhauser
C
Lorenzen
J
Thiele
J
et al
Secretion of cytokines (Interleukins-1 , -3, and -6 and granulocyte macrophage colony-stimulating factor) by normal human bone marrow megakaryocytes.
Blood.
85
1995
685
62
Navarro
S
Debili
N
Le Couedic
J-P
et al
Interleukin-6 and its receptor are expressed by human megakaryocytes: In vitro effects on proliferation and endoreplication.
Blood.
77
1991
461
Sign in via your Institution