Abstract

Leridistim is from the myelopoietin family of proteins, which are dual receptor agonists of the human interleukin-3 and G-CSF receptor complexes. This study investigated the effect of dosage, administration route, and schedule of leridistim to stimulate multilineage hematopoietic recovery in total body irradiated rhesus monkeys. Animals were x-irradiated on day 0 (600 cGy, 250 kVp) and then received, on day 1, leridistim s.c. in an abbreviated, every-other-day schedule at 200 μg/kg, or daily at 50 μg/kg, or i.v. daily or every-other-day schedules at 200 μg/kg dose. Other cohorts received G-CSF (Neupogen® [Filgrastim]) in an every-other-day schedule at 100 μg/kg/day, or autologous serum (0.1%) s.c. daily. Hematopoietic recovery was assessed by bone marrow clonogenic activity, peripheral blood cell nadirs, duration of cytopenias, time to recovery to cellular thresholds, and requirements for clinical support. Leridistim, administered s.c. every other day, or i.v. daily, significantly improved neutrophil, platelet, and lymphocyte nadirs, shortened the respective durations of cytopenia, hastened trilineage hematopoietic recovery, and reduced antibiotic and transfusion requirements. A lower dose of leridistim administered daily s.c. enhanced recovery of neutrophil and platelet parameters but did not affect lymphocyte recovery relative to controls. Leridistim, a novel engineered hematopoietic growth factor administered at the appropriate dose, route and schedule, stimulates multilineage hematopoietic reconstitution in radiation-myelosuppressed nonhuman primates.

Introduction

Acute marrow myelosuppression and delayed immune reconstitution remain as major hematopoietic sequelae consequent to many chemotherapeutic regimens. Realizing the full potential of cytotoxic therapy depends upon controlling the associated cytopenia and clinical support requirements. G-CSF, GM-CSF, and interleukin-11 (IL-11) provide a measure of therapeutic support for neutropenia and thrombocytopenia respectively [1–, 6]. The therapeutic efficacy of the Mpl-Ls, which were remarkably efficient in preclinical models of myelosuppression [7–, 10], have yet to demonstrate clinically meaningful benefit in two reported trials [11,, 12].

Continued efforts to enhance neutrophil regeneration have resulted in the development of a modified, pegylated form of G-CSF and the family of chimeric growth factors (GF) called myelopoietins [13–, 17]. SC-68420, a myelopoietin, (family of engineered IL-3 and G-CSF GF receptor agonists), recently has been shown to effectively stimulate neutrophil recovery in a nonhuman primate model of radiation-induced myelosuppression [18]. Stimulation of hematopoiesis with myelopoietin s.c. was as effective in a daily (qd) administration schedule as a twice daily (bid) schedule.

In addition to the myelosuppressive effects of chemotherapy, the immune system is also significantly compromised. This includes both the cellular components that constitute lymphoid progenitors, as well as effector and memory cells of both innate and acquired immunity. Subsequent to irradiation and/or chemotherapy, T cells repopulate, but there is a marked, prolonged CD4+ cell depletion, CD4+ and CD8+ subset alterations, and limited T-cell receptor repertoire diversity [19,, 20].

We report herein that leridistim, another molecule from the myelopoietin family but structurally unique from the myelopoietin, SC-68420, evaluated previously [21], retains equivalent efficacy for stimulating hematopoietic multilineage recovery, including an early stimulation of lymphoid recovery, when administered i.v. in a daily schedule or s.c. in an alternate-day schedule. The lower dose of leridistim, in a daily schedule, significantly enhanced hematopoiesis but not lymphocyte recovery.

Materials and Methods

Animals

Male rhesus monkeys, Macaca mulatta, mean weight 3.5 ± 0.2 kg, were housed in individual stainless steel cages in conventional holding rooms at the Veterinary Resources Department at the University of Maryland, Greenebaum Cancer Center, in animal facilities accredited by the American Association for Accreditation of Laboratory Animal Care (http://www.aaalac.org). Monkeys were provided 10 air changes/hour of 100% fresh air, conditioned to 72° ± 2°F with a relative humidity of 50% ± 20% and maintained on a 12-hour light/dark full spectrum light cycle, with no twilight. Monkeys were provided with commercial primate chow, supplemented with fresh fruit and tap water ad libitum. Research was conducted according to the principles enunciated in the Guide for the Care and Use of Laboratory Animals [22], prepared by the Institute of Laboratory Animal Resources, National Research Council (http://www.nas.edu/nrc) and research protocol approved by the Institutional Animal Care and Use Committee (http://www.umresearch.umd.edu/IACUC).

Irradiation

Monkeys, following a prehabituation period, were total body irradiated (TBI) in Lucite® restraining chairs. The 250 kVp x-irradiation was unilateral at 13 cGy/minute 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. Dosimetry was performed using paired 0.5 cm3 ionization chambers, with calibration factors traceable to the National Institute of Standards and Technology (http://www.nist.gov).

Recombinant Cytokines

Leridistim is from the myelopoietin family of engineered, chimeric hematopoietic growth factors that bind and activate the IL-3 and G-CSF receptors [23]. It was produced in E. coli through the use of a plasmid-based expression vector and 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. Leridistim was purified by ion exchange chromatography and filtration. Purified protein was stored as a frozen solution in 10 mM Tris buffer, ph 8.0. Neupogen® (Filgrastim), recombinant human (rHu) G-CSF, was produced in E. coli with a specific activity of 1.0 ± 0.6 × 108 U/mg (as measured by cell mitogenesis assay).

Study Design

Animals were irradiated on day 0 to 600 cGy x-irradiation and randomly assigned to treatment groups. On day 1 animals received leridistim s.c. at 200 μg/kg every other day (qod), n = 8 or 50 μg/kg every day (qd), n = 7; leridistim i.v. at 200 μg/kg/qd or qod, n = 3 each; or G-CSF, s.c., at 100 μg/kg/qod, n = 5. The controls received 0.1% autologous serum (AS), n = 9, in 1 ml bolus s.c. injections for 18 days. Leridistim was administered until an ANC ≥ 3,000/μl was attained (range 13-18 days).

Rationale for Cytokine Dose and Schedule

The primary focus of the leridistim schedule was to enhance neutrophil recovery; platelet, red cell, and lymphocyte responses were secondary objectives and, therefore, optimum schedules have not been investigated. The dose (50 μg/kg or 200 μg/kg), administration schedule (qd versus qod), and route (i.v. versus s.c.) were based on several observations: A) native rHuIL-3 or an engineered IL-3 receptor agonist, daniplestim, has 20- to 50-fold less binding affinity for rhesus IL-3 receptors than homologous, rhesus IL-3 [24,, 25]; B) we previously demonstrated enhanced therapeutic efficacy of daniplestim administered s.c., bid at 100 μg/kg/day relative to 25 μg/kg/day [26], and the myelopoietin, SC-68420, s.c. at bid or qd schedules (200 μg/kg/day) [18]; C) the myelopoietins are constructed to contain approximately equimolar ratios of IL-3 and G-CSF receptor agonists, and therefore, the 200 μg/kg dose of leridistim provides approximately 100 μg/kg each of engineered IL-3 and G-CSF proteins. Since our primary focus was on leridistim in an abbreviated, qod schedule, administered s.c. at 200 μg/kg, we included a cohort administered G-CSF, qod, s.c. at 100 μg/kg; D) monotherapy protocols using native rHuIL-3 or daniplestim, required bid administration for optimal biological response [27] (Farese et al., unpublished observations), and E) the plasma pharmacokinetic parameters of daniplestim in irradiated monkeys [26] and myelopoietin (SC-68420) in normal monkeys suggested that an abbreviated s.c. schedule (qd or qod) and qd i.v. administration of similarly engineered myelopoietins would induce an equivalent biological response to the full bid schedule [18].

Clinical Support

Clinical support consisting of an antimicrobial regimen and whole blood transfusions was provided to all animals as required.

Antimicrobials

An antibiotic regimen was initiated prophylactically when the WBC was <1,000/μl. Gentamicin (Elkin Sinn, an AH Robbins subsidiary; Cherry Hill, NJ) 10 mg/day, i.m., qd was administered during the first 7 days of treatment and Baytril® (Bayer Corporation; Shawnee Mission, KS; http://www.bayerus.com) 10 mg/day i.m., qd was administered for the entire period of antimicrobial treatment. The administration of antibiotics continued until the animal maintained a WBC ≥1,000/μl for 3 consecutive days and had attained an ANC ≥500/μl [7,, 26].

Transfusions

Fresh, irradiated (1,500 cGy, Cobalt-60 γ-irradiation) whole blood (approximately 30 ml/transfusion) from a random donor pool (monkeys >10 kg body weight), was administered when the platelet (PLT) count was <20,000/μl and the hematocrit (HCT) was <18%. Alternatively, fresh, irradiated (1,500 cGy, Cobalt-60 γ-irradiation) packed red blood cells (approximately 12.5 ml/transfusion) were administered when the PLT count was within normal limits and the HCT was <15%.

Hematological 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; http://www.sysmex.com) and differential counts (Wright-Giemsa Stain, Ames Automated Slide Stainer; Elkhart, IN). Assessment of hematologic evaluations has been previously described [26].

Flow Cytometric Studies

Cell Preparation

For each test, a 100 μl sample of EDTA-anticoagulated whole blood (WBC ∼10-15 × 103/μl) combined with mouse serum (1% final concentration) (Sigma; St. Louis, MO; http://www.sigma-aldrich.com) was incubated for 20 minutes at 4°C to block for nonspecific binding of monoclonal antibodies. For the lymphocyte panel, cells were then stained with either CD3 fluorescein isothiocyanate (FITC) (PharMingen; San Diego, CA; http://www.pharmingen.com), CD4 allophycocyanin (APC), CD8 peridinin chlorophyll protein (PerCP), and CD20 phycoerythrin (PE) (Becton Dickinson; San Jose, CA; http://www.bd.com) or their appropriate isotype controls according to the manufacturer's recommendation. For the memory and naïve lymphocyte panel, cells were stained with CD45RA FITC, CD62L PE, CD4 APC, CD20 FITC (Becton Dickinson), CD8 APC, CD16 PE (PharMingen), or CD3 Biotin-Streptavidin PerCP. Samples with CD3 Biotin (a generous gift from Dr. Michael Rosenzweig) were incubated for 30 minutes, washed, and then stained with Streptavidin-PerCP (Becton Dickinson). All fluorochromes were incubated in the dark for 20 minutes at 4°C. After incubation, cells were lysed for 10 minutes at room temperature in the dark using 1.5 ml FACS Lysing Solution (Becton Dickinson). Cells were then washed twice at 4°C with 2 ml of 1% bovine serum albumin (Sigma). Finally, cells were reconstituted with 300 μl of phosphate buffered saline if the cells were acquired on the same day or with 300 μl of 1% paraformaldehyde (Electron Microscopy Sciences; Ft. Washington, PA) for cells that were fixed and acquired the next day. Cells were vortexed prior to acquisition.

Cell Acquisition

Cells were acquired using a dual laser (488 nm blue argon laser and a 635 nm red diode laser) Becton Dickinson FACSCalibur flow cytometer. Instrument calibration and sensitivity were performed initially using CaliBRITE beads and FACSComp software (Becton Dickinson). Prior to acquisition, single fluorochrome positive compensation tubes were used to ensure correct compensation of individual fluorochromes. All events (25,000) were acquired and saved. A lymphocyte gate was used to display positive events.

Analysis

Gating strategies incorporated a primary gate (R1) that excluded dead cells and debris from the analysis. For analysis of lymphocyte surface markers (CD3, CD4, CD8, CD20) and memory and naïve surface markers (CD45RA, CD62L), a lymphocyte gate was drawn (R2). CD3+CD45RA+CD62L+ events were designated as the “naïve” phenotype, and CD3+CD45RACD62L events were designated as the “memory” phenotype. Isotype controls were used to set the quadrant markers for plots with respective fluorochromes. Region and quadrant statistics were displayed as both % gated and % total. The absolute lymphocyte count (WBC times the percent lymphocytes) multiplied by the desired percent positive events enabled the reporting of cells per microliter for the lymphocyte markers.

Bone Marrow

Approximately 2 ml of heparinized-bone marrow (BM) were aspirated from the humerus and/or iliac crest of anesthetized primates (Ketaset; Fort Dodge, IA, 10 mg/kg and Buprenex® injectable, Richmond and Cole Pharmaceuticals; Richmond, VA, 10 μg/kg, i.m.). The clonogenicity of BM progenitor cells were assayed in a short-term culture assay. Culture medium contained 0.9% methylcellulose (MethoCult H4230, Stem Cell Technologies; Vancouver, BC; http://www.stemcell.com) in Iscove's modified Dulbecco's medium. In addition, a combination of recombinant human cytokines, G-CSF (5 ng/ml), stem cell factor (SCF) (50 ng/ml), erythropoietin (Epo) (2 U/ml), megakaryocyte growth and development factor (MGDF) (20 ng/ml) (Amgen; Thousand Oaks, CA), IL-3 (20 ng/ml), GM-CSF (5 ng/ml), and IL-6 (40 ng/ml), (Sandoz Pharmaceuticals; East Hanover, NJ; http://www.pharma.novartis.com) were added to each culture dish. Mononuclear cells (MNC) were cultured at a plating density of 3 to 5 × 104 cells/ml (days 0, 21, 48 post-TBI) or 1 × 105 cells/ml (day 7, 14 post-TBI). Cells were incubated for 10 days at 37°C with 5% CO2 in air in a fully humidified incubator. GM-colony-forming cells (GM-CFC) and BFU-E-derived colonies (>50 cells) were expressed as the number of colony-forming cells/105 MNC. Megakaryocyte (MK)-CFC are not distinguished from MK-BFU in this study. All MK colonies ranged from 10 to 50 cells.

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; http://www.cytel.com). The exact p values were obtained for all analyses.

BM-derived clonogenic activities were analyzed for each endpoint at each day across the control and leridistim treatment groups using the Kruskal-Wallis Test. Post hoc tests were made if the p value for the Kruskal-Wallis Test was ≤0.05 and were made using a Bonferroni corrected Mann-Whitney Test.

Results

Neutrophil Recovery: s.c. Leridistim Administration

Neutrophil parameters were significantly improved by s.c. administration of leridistim in a qod schedule at 200 μg/kg dose after 600 cGy TBI (Fig. 1A, Table 1). The duration of neutropenia (ANC <500/μl) was 19.1 days versus 1.0 day (p < 0.001), the neutrophil nadir was 16/μl versus 601/μl (p < 0.001), and the time to recovery of ANC ≥500/μl was 24.5 days to 4.1 days (p < 0.001) for control and leridistim-treated groups, respectively. As a result, the days on antibiotics were significantly lower at 5.8 for the leridistim-treated cohort compared with 19.6 for the controls (p < 0.001).

Table 1.

Neutrophil, platelet, and lymphocyte-related parameters in sublethally x-irradiated rhesus monkeys treated with leridistim: duration, nadir, time to recovery, and clinical support

Neutrophil-related parameters
Treatment (dose, route, schedule) Duration (d) neutropenia ANC nadir (μl–1) Time to recovery (d) Antibiotic support (d)
0.1% AS, s.c., qd 19.1 ± 2.3 16 ± 6 24.5 ± 2.2 19.6 ± 1.7
Leridistim
    50 μg/kg, s.c., qd 3.3 ± 1.6* 400 ± 116* 11.0 ± 2.4* 8.0 ± 2.2*
    200 μg/kg, s.c., qod 1.0 ± 0.6* 601 ± 123* 4.1 ± 2.2* 5.8 ± 1.0*
    200 μg/kg, i.v., qd 2.7 ± 1.8* 442 ± 227* 7.0 ± 4.4* 4.3 ± 3.0*
    200 μg/kg, i.v., qod 9.0 ± 1.7 63 ± 30 16.3 ± 1.2 14.7 ± 0.3
G-CSF
    100 μg/kg, s.c., qod 14.2 ± 1.9 10 ± 4 19.4 ± 1.7 17.8 ± 1.8
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 7.2 ± 2.0* 205 ± 33* 9.2 ± 3.8* 10.0 ± 1.9*
Platelet-related parameters
Treatment (dose, route, schedule) Duration (d) thrombocytopenia PLT nadir (μl–1) Time to recovery (d) Transfusions
0.1% AS, s.c, qd 6.9 ± 0.5 4,250 ± 1,634 18.1 ± 0.4 1.1 ± 0.4
Leridistim
    50 μg/kg, s.c., qd 3.4 ± 1.2 18,429 ± 6,070 11.0 ± 2.9* 0
    200 μg/kg, s.c., qod 1.6 ± 1.0* 21,900 ± 3,900* 7.8 ± 3.0* 0
    200 μg/kg, i.v., qd 2.7 ± 1.8 24,667 ± 14,518 10.3 ± 5.2* 0
    200 μg/kg, i.v., qod 3.7 ± 1.5 10,333 ± 5,457 15.0 ± 1.5 0.3 ± 0.3
G-CSF
    100 μg/kg, s.c., qod 8.2 ± 1.8 5,000 ± 1,304 18.4 ± 1.6 0.8 ± 0.6
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 3.2 ± 1.0* 14,800 ± 7,870* 11.8 ± 2.2* 0
Lymphocyte-related parameters
Treatment (dose, route, schedule) Duration (d) lymphocytopenia ALC nadir (μl–1) Time to recovery (d)
AS-control 24.1 ± 1.3 130 ± 10 25.1 ± 1.3
Leridistim
    50 μg/kg, s.c., qd 22.0 ± 2.2 91 ± 22 23.0 ± 2.2
    200 μg/kg, s.c., qod 17.1 ± 1.7* 198 ± 27 18.6 ± 1.7*
    200 μg/kg, i.v., qd 16.3 ± 0.7* 89 ± 27 17.3 ± 0.7*
    200 μg/kg, i.v., qod 24.7 ± 2.9 117 ± 9 26.0 ± 3.2
G-CSF
    100 μg/kg, s.c., qod 28.2 ± 3.7 116 ± 10 29.2 ± 3.7
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 16.0 ± 1.2* 181 ± 26 17.0 ± 1.2*
Neutrophil-related parameters
Treatment (dose, route, schedule) Duration (d) neutropenia ANC nadir (μl–1) Time to recovery (d) Antibiotic support (d)
0.1% AS, s.c., qd 19.1 ± 2.3 16 ± 6 24.5 ± 2.2 19.6 ± 1.7
Leridistim
    50 μg/kg, s.c., qd 3.3 ± 1.6* 400 ± 116* 11.0 ± 2.4* 8.0 ± 2.2*
    200 μg/kg, s.c., qod 1.0 ± 0.6* 601 ± 123* 4.1 ± 2.2* 5.8 ± 1.0*
    200 μg/kg, i.v., qd 2.7 ± 1.8* 442 ± 227* 7.0 ± 4.4* 4.3 ± 3.0*
    200 μg/kg, i.v., qod 9.0 ± 1.7 63 ± 30 16.3 ± 1.2 14.7 ± 0.3
G-CSF
    100 μg/kg, s.c., qod 14.2 ± 1.9 10 ± 4 19.4 ± 1.7 17.8 ± 1.8
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 7.2 ± 2.0* 205 ± 33* 9.2 ± 3.8* 10.0 ± 1.9*
Platelet-related parameters
Treatment (dose, route, schedule) Duration (d) thrombocytopenia PLT nadir (μl–1) Time to recovery (d) Transfusions
0.1% AS, s.c, qd 6.9 ± 0.5 4,250 ± 1,634 18.1 ± 0.4 1.1 ± 0.4
Leridistim
    50 μg/kg, s.c., qd 3.4 ± 1.2 18,429 ± 6,070 11.0 ± 2.9* 0
    200 μg/kg, s.c., qod 1.6 ± 1.0* 21,900 ± 3,900* 7.8 ± 3.0* 0
    200 μg/kg, i.v., qd 2.7 ± 1.8 24,667 ± 14,518 10.3 ± 5.2* 0
    200 μg/kg, i.v., qod 3.7 ± 1.5 10,333 ± 5,457 15.0 ± 1.5 0.3 ± 0.3
G-CSF
    100 μg/kg, s.c., qod 8.2 ± 1.8 5,000 ± 1,304 18.4 ± 1.6 0.8 ± 0.6
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 3.2 ± 1.0* 14,800 ± 7,870* 11.8 ± 2.2* 0
Lymphocyte-related parameters
Treatment (dose, route, schedule) Duration (d) lymphocytopenia ALC nadir (μl–1) Time to recovery (d)
AS-control 24.1 ± 1.3 130 ± 10 25.1 ± 1.3
Leridistim
    50 μg/kg, s.c., qd 22.0 ± 2.2 91 ± 22 23.0 ± 2.2
    200 μg/kg, s.c., qod 17.1 ± 1.7* 198 ± 27 18.6 ± 1.7*
    200 μg/kg, i.v., qd 16.3 ± 0.7* 89 ± 27 17.3 ± 0.7*
    200 μg/kg, i.v., qod 24.7 ± 2.9 117 ± 9 26.0 ± 3.2
G-CSF
    100 μg/kg, s.c., qod 28.2 ± 3.7 116 ± 10 29.2 ± 3.7
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 16.0 ± 1.2* 181 ± 26 17.0 ± 1.2*

Monkeys were total body irradiated (250 kVp) to 600 cGy with x-irradiation and treated s.c. with control protein, autologous serum (AS) (n = 9) or leridistim at 200 μg/kg/d, qod (n = 8), or 50 μg/kg/d, qd (n = 7) or i.v. with leridistim at 200 μg/kg/d either qd or qod (n = 3) per cohort according to protocol (Materials and Methods). 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. Data represent mean values ± SE.

*Statistically different from AS-treated controls (p ≤ 0.01).

Statistically different from AS-treated controls (p ≤ 0.05).

‡ Previous data set. Published study showing effects of SC-68420, myelopoietin, qd, s.c. at 200 μg/kg/d in the same 600 cGy x-irradiation model [18]. This study showed s.c., qd, and bid administration were equivalent in bioeffect. Published cell nadirs were calculated using the nadir of the cohort. Cell nadirs reported herein are calculated as the mean of the cell nadir of the individual animals in each cohort.

Table 1.

Neutrophil, platelet, and lymphocyte-related parameters in sublethally x-irradiated rhesus monkeys treated with leridistim: duration, nadir, time to recovery, and clinical support

Neutrophil-related parameters
Treatment (dose, route, schedule) Duration (d) neutropenia ANC nadir (μl–1) Time to recovery (d) Antibiotic support (d)
0.1% AS, s.c., qd 19.1 ± 2.3 16 ± 6 24.5 ± 2.2 19.6 ± 1.7
Leridistim
    50 μg/kg, s.c., qd 3.3 ± 1.6* 400 ± 116* 11.0 ± 2.4* 8.0 ± 2.2*
    200 μg/kg, s.c., qod 1.0 ± 0.6* 601 ± 123* 4.1 ± 2.2* 5.8 ± 1.0*
    200 μg/kg, i.v., qd 2.7 ± 1.8* 442 ± 227* 7.0 ± 4.4* 4.3 ± 3.0*
    200 μg/kg, i.v., qod 9.0 ± 1.7 63 ± 30 16.3 ± 1.2 14.7 ± 0.3
G-CSF
    100 μg/kg, s.c., qod 14.2 ± 1.9 10 ± 4 19.4 ± 1.7 17.8 ± 1.8
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 7.2 ± 2.0* 205 ± 33* 9.2 ± 3.8* 10.0 ± 1.9*
Platelet-related parameters
Treatment (dose, route, schedule) Duration (d) thrombocytopenia PLT nadir (μl–1) Time to recovery (d) Transfusions
0.1% AS, s.c, qd 6.9 ± 0.5 4,250 ± 1,634 18.1 ± 0.4 1.1 ± 0.4
Leridistim
    50 μg/kg, s.c., qd 3.4 ± 1.2 18,429 ± 6,070 11.0 ± 2.9* 0
    200 μg/kg, s.c., qod 1.6 ± 1.0* 21,900 ± 3,900* 7.8 ± 3.0* 0
    200 μg/kg, i.v., qd 2.7 ± 1.8 24,667 ± 14,518 10.3 ± 5.2* 0
    200 μg/kg, i.v., qod 3.7 ± 1.5 10,333 ± 5,457 15.0 ± 1.5 0.3 ± 0.3
G-CSF
    100 μg/kg, s.c., qod 8.2 ± 1.8 5,000 ± 1,304 18.4 ± 1.6 0.8 ± 0.6
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 3.2 ± 1.0* 14,800 ± 7,870* 11.8 ± 2.2* 0
Lymphocyte-related parameters
Treatment (dose, route, schedule) Duration (d) lymphocytopenia ALC nadir (μl–1) Time to recovery (d)
AS-control 24.1 ± 1.3 130 ± 10 25.1 ± 1.3
Leridistim
    50 μg/kg, s.c., qd 22.0 ± 2.2 91 ± 22 23.0 ± 2.2
    200 μg/kg, s.c., qod 17.1 ± 1.7* 198 ± 27 18.6 ± 1.7*
    200 μg/kg, i.v., qd 16.3 ± 0.7* 89 ± 27 17.3 ± 0.7*
    200 μg/kg, i.v., qod 24.7 ± 2.9 117 ± 9 26.0 ± 3.2
G-CSF
    100 μg/kg, s.c., qod 28.2 ± 3.7 116 ± 10 29.2 ± 3.7
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 16.0 ± 1.2* 181 ± 26 17.0 ± 1.2*
Neutrophil-related parameters
Treatment (dose, route, schedule) Duration (d) neutropenia ANC nadir (μl–1) Time to recovery (d) Antibiotic support (d)
0.1% AS, s.c., qd 19.1 ± 2.3 16 ± 6 24.5 ± 2.2 19.6 ± 1.7
Leridistim
    50 μg/kg, s.c., qd 3.3 ± 1.6* 400 ± 116* 11.0 ± 2.4* 8.0 ± 2.2*
    200 μg/kg, s.c., qod 1.0 ± 0.6* 601 ± 123* 4.1 ± 2.2* 5.8 ± 1.0*
    200 μg/kg, i.v., qd 2.7 ± 1.8* 442 ± 227* 7.0 ± 4.4* 4.3 ± 3.0*
    200 μg/kg, i.v., qod 9.0 ± 1.7 63 ± 30 16.3 ± 1.2 14.7 ± 0.3
G-CSF
    100 μg/kg, s.c., qod 14.2 ± 1.9 10 ± 4 19.4 ± 1.7 17.8 ± 1.8
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 7.2 ± 2.0* 205 ± 33* 9.2 ± 3.8* 10.0 ± 1.9*
Platelet-related parameters
Treatment (dose, route, schedule) Duration (d) thrombocytopenia PLT nadir (μl–1) Time to recovery (d) Transfusions
0.1% AS, s.c, qd 6.9 ± 0.5 4,250 ± 1,634 18.1 ± 0.4 1.1 ± 0.4
Leridistim
    50 μg/kg, s.c., qd 3.4 ± 1.2 18,429 ± 6,070 11.0 ± 2.9* 0
    200 μg/kg, s.c., qod 1.6 ± 1.0* 21,900 ± 3,900* 7.8 ± 3.0* 0
    200 μg/kg, i.v., qd 2.7 ± 1.8 24,667 ± 14,518 10.3 ± 5.2* 0
    200 μg/kg, i.v., qod 3.7 ± 1.5 10,333 ± 5,457 15.0 ± 1.5 0.3 ± 0.3
G-CSF
    100 μg/kg, s.c., qod 8.2 ± 1.8 5,000 ± 1,304 18.4 ± 1.6 0.8 ± 0.6
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 3.2 ± 1.0* 14,800 ± 7,870* 11.8 ± 2.2* 0
Lymphocyte-related parameters
Treatment (dose, route, schedule) Duration (d) lymphocytopenia ALC nadir (μl–1) Time to recovery (d)
AS-control 24.1 ± 1.3 130 ± 10 25.1 ± 1.3
Leridistim
    50 μg/kg, s.c., qd 22.0 ± 2.2 91 ± 22 23.0 ± 2.2
    200 μg/kg, s.c., qod 17.1 ± 1.7* 198 ± 27 18.6 ± 1.7*
    200 μg/kg, i.v., qd 16.3 ± 0.7* 89 ± 27 17.3 ± 0.7*
    200 μg/kg, i.v., qod 24.7 ± 2.9 117 ± 9 26.0 ± 3.2
G-CSF
    100 μg/kg, s.c., qod 28.2 ± 3.7 116 ± 10 29.2 ± 3.7
SC-64820, myelopoietin
    200 μg/kg, s.c., qd 16.0 ± 1.2* 181 ± 26 17.0 ± 1.2*

Monkeys were total body irradiated (250 kVp) to 600 cGy with x-irradiation and treated s.c. with control protein, autologous serum (AS) (n = 9) or leridistim at 200 μg/kg/d, qod (n = 8), or 50 μg/kg/d, qd (n = 7) or i.v. with leridistim at 200 μg/kg/d either qd or qod (n = 3) per cohort according to protocol (Materials and Methods). 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. Data represent mean values ± SE.

*Statistically different from AS-treated controls (p ≤ 0.01).

Statistically different from AS-treated controls (p ≤ 0.05).

‡ Previous data set. Published study showing effects of SC-68420, myelopoietin, qd, s.c. at 200 μg/kg/d in the same 600 cGy x-irradiation model [18]. This study showed s.c., qd, and bid administration were equivalent in bioeffect. Published cell nadirs were calculated using the nadir of the cohort. Cell nadirs reported herein are calculated as the mean of the cell nadir of the individual animals in each cohort.

Figure 1.

Effect of leridistim administration on peripheral blood (A) neutrophils (ANC), (B) platelets (PLT), (C) red blood cells (RBC), and (D) lymphocytes (ALC) in 600 cGy x-irradiated rhesus monkeys.  Animals were s.c. administered leridistim in either 200 μg/kg in a qod (n = 8) or 50 μg/kg/d in a qd (n = 7) schedule or autologous serum (AS) (0.1%) as a control protein (n = 9) as described in  Materials and Methods. Data represents mean values ± standard error (SE).

Leridistim administered s.c. at a lower dose of 50 μg/kg/d in a qd schedule also significantly (p < 0.001) improved all neutrophil-related parameters relative to the control-treated cohort (Fig. 1, Table 1). However, the 200 μg/kg, s.c., qod, leridistim administration was significantly (p = 0.032) better than the 50 μg/kg, s.c., qd schedule at improving neutrophil recovery (Table 1).

Neutrophil Recovery: G-CSF Administration

G-CSF administered s.c. in a qod schedule at 100 μg/kg, induced a modest, although not statistically significant (p > 0.05) improvement in duration of neutropenia, 14.2 days versus 19.1 days for the controls, and time to recovery of ANC to >500/μl, 19.4 days versus 24.5 days for controls (Table 1). The neutrophil nadir and antibiotic support were unaffected by administration of G-CSF.

Neutrophil Recovery: i.v. Leridistim Administration

Intravenous administration of leridistim via qd or qod schedules significantly decreased respective durations of neutropenia (2.7 days and 9.0 days versus 19.1 days) (p ≤ 0.012), neutrophil nadirs (442/μl and 63/μl versus 16/μl) (p ≤ 0.03), and improved the time to recovery of ANC ≥ 500/μl (7.0 days and 16.3 days versus 24.7 days) (p ≤ 0.018) relative to controls (Table 1). The qd schedule of leridistim administration was markedly, although not significantly (p ≥ 0.1) different from the qod schedule in improving all neutrophil-related parameters. Antibiotic support was significantly lower with the qd administration schedule (4.3 days, p = 0.006) but not with the qod schedule (14.7 days, p = 0.061) compared with the controls at 19.6 days.

Platelet Recovery

Leridistim administration independent of schedule, dose, or route was equally effective in improving all platelet-related parameters (Table 1, Fig. 1B). Leridistim administered s.c., either qod (200 μg/kg) or qd (50 μg/kg) resulted in a significantly shorter duration of thrombocytopenia (platelet count <20,000/μl), 1.6 days and 3.4 days, respectively, compared with 6.9 days for controls (p ≤ 0.014). The mean platelet nadir was significantly greater, 21,900/μl and 18,429/μl versus 4,250/μl for controls (p ≤ 0.013), while time to recovery (day ≥20,000/μl) was significantly shorter, 7.8 days and 11.0 days, versus the control 18.1 days following qod or qd leridistim administration, respectively. The leridistim, s.c.-treated cohorts were transfusion independent while the controls required on average 1.1 transfusions/animal. G-CSF administered s.c., in the qod schedule at 100 μg/kg did not improve upon the platelet-related parameters noted for the control-treated animals (Table 1).

Production of RBC

Leridistim administered s.c. in the 200 μg/kg qod or the 50 μg/kg qd schedule improved maintenance of RBC levels over the second to third week post x-irradiation (Fig. 1C). This effect was most likely mediated through the enhanced platelet recovery and lack of inopportune hemorrhage. The control cohort, however, received on average one transfusion/animal during this time frame, which was necessitated by a combined PLT count <20,000/μl and hematocrit <18%. The recovery curve noted for the control cohort reflects the effects of the whole blood transfusions.

Lymphocyte Recovery

Leridistim administered s.c. at 200 μg/kg in the qod schedule significantly enhanced an early period of absolute lymphocyte (ALC) recovery (ALC >1,000/μl), whereas the recovery pattern noted for the lower dose, 50 μg/kg/d qd cohort was not significantly different from the control-treated cohort (Table 1, Fig. 1D). The duration of lymphopenia (ALC <1,000/μl) was shorter, 17.1 versus 24.1 days for controls (p = 0.003), and the time for ALC recovery was also shorter, 18.6 versus 25.1 days in the controls (p = 0.005) in the leridistim-treated cohort. This enhanced ALC recovery phase, however, could not be sustained following cessation of leridistim treatment. The ALC recovery slowed and converged with that of the control cohort for the remainder of the study.

With respect to the i.v. administration of leridistim, only the qd schedule (200 μg/kg/d) resulted in a significantly shorter duration of lymphopenia (p = 0.006) and improved recovery of ALC >1,000/μl (p = 0.006). G-CSF administered s.c. in the qod schedule at 200 μg/kg did not improve upon the lymphocyte-related parameters noted for the control-treated animals (Table 1).

Flow Cytometric Analysis of Lymphocyte Subsets Following Leridistim Administration (200 μg/kg, qod, sc)

All T cell subsets (CD3+, CD4+, CD3+/CD8+, natural killer [NK] cells [CD3CD8+, CD16+]) and B cells (CD20+) were significantly decreased within several days of 600 cGy TBI. Further analysis at weekly intervals demonstrated that the early but transient lymphocyte recovery was CD3+CD8+ T cell dominant. Modest but nonsignificant improvement was noted in recovery of CD4+ cells to >500/μl (40.6 days) relative to the controls (54.1 days) (p = 0.101), whereas B cell recovery was unaffected by leridistim treatment (data not shown). The time to recovery of CD3+CD8+ cells to ≥500/μl was 28.1 days for leridistim-treated animals and 47.3 days for the controls, respectively (p = 0.019) (Table 2). Thereafter, the CD3+CD8+ cells from both leridistim- and control-treated cohorts recovered at equivalent rates returning to preirradiation values (1,101/μl and 1,220/μl) by 56 days post-irradiation (923/μl and 1,111/μl, respectively) (Table 2). The CD4+ T cells in both the control- and leridistim-treated cohorts recovered at a much slower rate than the CD3+CD8+ T cells. Preirradiation CD4+ cells were 2,061/μl for control and 1,602/μl for the leridistim-treated cohort. CD4+ cells at 56 days post-irradiation were 584/μl and 504/μl for control- and leridistim-treated cohorts, respectively, representing approximately 30% of their preirradiation values (Table 2). These differential recovery rates in CD4+ and C3+CD8+ cells resulted in inversion of the CD4/CD8 ratio. Preirradiation CD4/CD8 ratios for control (1.69), and leridistim cohorts (1.46) were reduced to 0.53 and 0.55, respectively, at 56 days post-irradiation. Further analysis revealed that regardless of T cell subset, CD4+ or CD3+CD8+, the predominant cell subsets that recovered were memory (CD45RA/CD62L) rather than naïve (CD45RA+/CD62L+) phenotype (data not shown).

Table 2.

Recovery of T lymphocytes in sublethally x-irradiated rhesus monkeys treated with leridistim

Table 2.

Recovery of T lymphocytes in sublethally x-irradiated rhesus monkeys treated with leridistim

BM-Derived Clonogenic Activity

Bone marrow-derived GM-CFC, BFU-E, and MK-CFC activity (CFC/105 MNCs) were evaluated pre-irradiation and at 7, 14, 21, and 46 days after 600 cGy x-irradiation in control and leridistim-treated cohorts (Fig. 2). Leridistim administered at 200 μg/kg qod stimulated earlier recovery of GM-CFC, BFU-E, and MK-CFC than noted with the control-treated animals. Significant recovery was noted for the MK-CFC within 7 days post-x-irradiation (p = 0.018) and GM-CFC (p < 0.002) by day 14 post-x-irradiation, with continued recovery in concentration of clonogenic cells to within preirradiation levels by 21 days post-exposure. Recovery of lineage-specific CFC in the control-treated cohort was variable: GM-CFC were at 58% of baseline at day 46, whereas the concentration of BFU-E and MK-CFC had recovered to preirradiation values.

Figure 2.

Effect of leridistim administration on the bone marrow-derived concentration of GM-CFC, MK-CFC, and BFU-E 600 cGy x-irradiated rhesus monkeys.  Clonogenic activity (CFCs/105 MNCs) was observed preirradiation (baseline) and on days 7, 14, 21, and 46 post-exposure. Animals were administered s.c. leridistim in either 200 μg/kg in a qod (n = 8) or 50 μg/kg/d in a qd (n = 7) schedule or autologous serum (AS) (0.1%) as a control protein (n = 9) as described in Materials and Methods. All animals were assayed at each time point indicated. Data are reported as mean percent of baseline values ± SE. *denotes significant difference from time matched control values (p ≤ 0.05); ‡ denotes significant difference from baseline values (p < 0.05);denotes zero values.

Discussion

Leridistim administered s.c. in an alternate day schedule significantly enhanced multilineage, e.g., neutrophil, platelet, and erythroid recovery, and stimulated an early but transient recovery of CD3+CD8+ T lymphocytes, in high-dose, sublethally irradiated rhesus monkeys. The abbreviated leridistim schedule administered via the s.c. route was as effective as the conventional qd or bid myelopoietin (SC-68420) schedules [18]. The multilineage efficacy of leridistim administered via s.c. or i.v. routes in the qod schedule is of interest considering the ineffectiveness of its component growth factors, e.g., native or engineered IL-3 (daniplestim) or G-CSF, when administered less frequently than bid or qd schedules, respectively [27]. G-CSF administered in an s.c., qod schedule did not significantly enhance neutrophil-related parameters. Whereas, G-CSF administered in a conventional qd schedule either alone or in combination with daniplestim effectively enhanced neutrophil recovery in two similar models of radiation-induced myelosuppression [7,, 8,, 28]. The s.c., qod administration of leridistim significantly improved the neutrophil and platelet nadirs, neutropenic, thrombocytopenic, and lymphopenic durations were significantly shorter, and respective neutrophil (≥500/μl), platelet (≥20,000/μl), and lymphocyte (≥1,000/μl) recovery times were significantly enhanced. RBC recovery was improved relative to the control cohort. Additionally, the increased recovery of peripheral blood hematopoietic cells was supported by the accelerated recovery of marrow-derived lineage-specific clonogenic progenitors. The increased hematopoietic recovery in leridistim-treated groups resulted in fewer whole blood transfusions and significantly reduced the number of days of antibiotic therapy support. The reduced dose of leridistim stimulated significant recovery in all neutrophil- and platelet-related parameters relative to the control cohort. The leridistim schedule of 200 μg/kg, s.c., qod improved upon all neutrophil and platelet parameters (except transfusions) noted for the 50 μg/kg qd schedule. Also, the 200 μg/kg dose of leridistim was required to stimulate the early increase in lymphocyte recovery, whereas the 50 μg/kg, s.c., qd dose was ineffective in modulating lymphocyte parameters.

The therapeutic efficacy of leridistim administered s.c. in either qod (200 μg/kg) or qd (50 μg/kg) schedules or via an i.v. route at the 200 μg/kg dose in a daily schedule compared favorably with the previously published effects of the structurally distinct, myelopoietin (SC-68420), which stimulated hematopoiesis when administered s.c., in qd or bid schedules in the same model presented herein [18]. The probable mechanisms responsible for the stimulation observed with the abbreviated schedule remain in effect: the IL-3 component may prime hematopoietic progenitor cells for subsequent stimulation by growth factors released within the radiation-damaged marrow microenvironment [29–, 31], IL-3 and G-CSF can exert trophic and survival-promoting effects on responsive progenitors within the post-irradiation microenvironment [32–, 37], IL-3 receptors are present on CD34+ cell subsets purified from human and rhesus marrow [24,, 38,, 39], and combined IL-3 and G-CSF are noted for their synergistic effects on myelopoiesis and megakaryocytopoiesis [26,, 30,, 31,, 40–, 42]. Leridistim binds with high affinity to both the IL-3 and G-CSF receptor complexes. Using TF-1 cells that express the IL-3 receptor but not the G-CSF receptor, leridistim binds with an IC50 of 14.2 ± 2.0 nM. This is fourfold less potent than rHuIL-3 (IC50 of 3.5 ± 0.5 nM) and twofold less potent than the cognate IL-3 receptor agonist present in leridistim (IL-315-125, 50D, IC50 = 6.9 ± 1.7 nM). However, using AML-193 cells that express both IL-3 and G-CSF receptors, leridistim binds to the IL-3 receptor with about sixfold higher affinity than its cognate IL-3 receptor agonist (IC50 = 2.0 ± 0.7 versus 11.6 ± 3.5 nM, respectively). In contrast to the affinity shift observed for the IL-3 receptor in AML-193 cells, the G-CSF receptor binding by leridistim was equivalent to that of G-CSF. Leridistim was also shown to activate signaling pathways downstream of both IL-3 and G-CSF receptor complexes (J.B. Monahan, personal communication).

The efficacy of leridistim is not easily explained by pharmacokinetic data. Plasma concentrations following i.v. administration of myelopoietin (SC-68420) to normal rhesus macaques indicated an elimination half-life of 3.3 hours and a volume of distribution similar to whole blood volume indicating little if any distribution from blood into other tissues [18]. s.c. administration of myelopoietin (SC-64820) [18] or leridistim [23] to normal rhesus macaques resulted in detectable plasma levels for at least 24-48 hours post-dose. In fact, the leridistim plasma concentration at 24 hours was approximately 0.5 nM, which is a concentration that results in maximal stimulation of CFU activity using human CD34+ cells. The effect of myelosuppression and circulating neutrophil count on the pharmacokinetics of leridistim is under investigation.

An interesting finding emerging from this study is that leridistim stimulated an earlier recovery of CD3+CD8+ memory phenotype T lymphocytes than noted with the controls B lymphocyte recovery was unaffected. The CD3+CD8+ T lymphocyte recovery was observed in the leridistim-treated cohort in the third week post-irradiation coincident with the last few days of leridistim administration. Thereafter, both control and leridistim-treated CD3+CD8+ T cells continued to recover at the same rate, whereas CD4+ cells in both cohorts recovered much slower and remained at levels approximately 30% of preirradiation values at 56 days post-exposure. The relatively rapid recovery of CD3+CD8+ cells compared with that of CD4+ cells is well documented in patients receiving multiple cycle chemotherapy [20,, 43].

The biology associated with administration of IL-3 and G-CSF would not predict a demonstrable effect on lymphopoiesis. IL-3 deficient mice have normal lymphopoiesis [44,, 45] and in vitro exposure of murine hematopoietic cells to IL-3 has been shown to suppress T and B cell generation [46,, 47]. IL-3 administered to normal rhesus macaques failed to stimulate a lymphocyte response [48]. Other studies suggest a positive influence of IL-3 on lymphoid development [49–, 51]. We have not assayed for the presence of marrow-derived lymphohematopoietic stem or progenitor cells (LHSC) in the control or leridistim-treated rhesus monkey. Therefore, we cannot determine if leridistim stimulated an increase in lymphoid progenitors. However, we will hypothesize that in addition to the previously noted favorable effects of IL-3 on pluripotent stem/progenitor cells, leridistim may interact within the marrow and thymic microenvironments with GFs such as IL-7, IL-11, c-kitL, flt-3L, and/or thymic stromal-derived lymphopoietin [52–, 61] to increase survival and/or generation of LHSC, T cell progenitors, and immature thymic precursors during the period immediate post-irradiation. As stated previously, our experiments were not focused on stimulating lymphoid regeneration, however the observations reported herein suggest that leridistim, within the post-irradiation microenvironment, initiated an early period of CD3+CD8+ T cell expansion. Administration of leridistim for a longer period, at the appropriate dose, may continue to influence T cell regeneration.

Several previous studies evaluated the co-administration of multilineage and lineage-dominant cytokines in nonhuman primate models of radiation-induced myelosuppression. Of particular interest are the combinations of the Mpl-Ls (Peg-rHuMGDF or thrombopoietin) and G-CSF [7,, 8,, 62]. Combinations of either Mpl-L with G-CSF further improved the neutrophil recovery noted with G-CSF monotherapy. Leridistim, employed via the schedule herein further improved, although not significantly so, the recovery of neutrophils observed in the comparable radiation model [7].

The differential recovery kinetics observed between the i.v. and s.c. routes and that noted with the abbreviated schedule and dose should be important in designing appropriate clinical trials for leridistim. The data reported herein suggest that if appropriate dose and schedule can be achieved in clinical trials, leridistim should provide multilineage reconstitution following radiation- or drug-induced myelosuppression.

Acknowledgements

The authors wish to thank Michael Flynn, Carrie Redinger, Tamara Chalk, Jaime Fenimore, and Scott Perry for their superb technical assistance and William Jackson for assistance with the statistical analyses.

Research supported by contract provided by Pharmacia Corporation. The views presented herein are those of the authors.

References

1

Morstyn
 
G
,
Campbell
 
L
,
Souza
 
LM
, et al.
Effect of granulocyte colony stimulating factor on neutropenia induced by cytotoxic chemotherapy
.
Lancet
 
1988
;
1
:
668
672
.

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
 
1989
;
2
:
891
895
.

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
 
1988
;
319
:
593
598
.

4

Vadhan-Raj
 
S
,
Keating
 
M
,
LeMaistre
 
A
, et al.
Effects of recombinant human granulocyte-macrophage colony-stimulating factor in patients with myelodysplastic syndrome
.
N Engl J Med
 
1987
;
317
:
1545
1552
.

5

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
 
1991
;
325
:
164
170
.

6

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
 
1997
;
15
:
3368
3377
.

7

Farese
 
AM
,
Hunt
 
P
,
Grab
 
LB
, et al.
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
 
1996
;
97
:
2145
2151
.

8

Neelis
 
KJ
,
Dubbelman
 
YD
,
Qingliang
 
L
, et al.
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
 
1997
;
25
:
1084
1093
.

9

Grossman
 
A
,
Lenox
 
J
,
Ren
 
HP
, et al.
Thrombopoietin accelerates platelet, red blood cell, and neutrophilic recovery in myelosuppressed mice
.
Exp Hematol
 
1996
;
24
:
1238
1246
.

10

Shibuya
 
K
,
Akahori
 
H
,
Takahashi
 
K
, et al.
Multilineage hematopoietic recovery by a single injection of pegylated recombinant human megakaryocyte growth and development factor in myelosuppressed mice
.
Blood
 
1998
;
91
:
37
45
.

11

Archimbaud
 
E
,
Ottmann
 
OG
,
Yin
 
JA
, et al.
A randomized, double-blinded, placebo-controlled study with pegylated recombinant human megakaryote growth and development factor (PEG-rHuMGDF) as an adjunct to chemotherapy for adults with de novo acute myeloid leukemia
.
Blood
 
1999
;
94
:
3694
3701
.

12

Schiffer
 
CA
,
Miller
 
K
,
Larson
 
RA
, et al.
A double-blind, placebo-controlled trial of pegylated recombinant human megakaryocyte growth and development factor as an adjunct to induction and consolidation therapy for patients with acute myeloid leukemia
.
Blood
 
2000
;
95
:
2530
2534
.

13

McKearn
 
J
,
Monahan
 
J
,
Hood
 
W
, et al.
Myelopoietin—a multifunctional chimeric protein family
.
Exp Hematol
 
1996
;
24
:
1154a

14

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
 
1995
;
86
:
154a

15

Farese
 
AM
,
Roskos
 
L
,
Cheung
 
E
, et al.
A single administration of r-metHuG-SD/01 (SD01) significantly improves neutrophil recovery following autologous bone marrow transplantation
.
Blood
 
1998
;
92
:
112a

16

Farese
 
AM
,
Roskos
 
L
,
Stead
 
RB
, et al.
r-metHuG-CSF-SD/01 (SD/01) significantly improves neutrophil recovery in myelosuppressed non human primates
.
Blood
 
1999
;
94
:
49a

17

Molineux
 
G
,
Kinstler
 
O
,
Briddell
 
B
, et al.
A new form of Filgrastim with sustained duration in vivo and enhanced ability to mobilize PBPC in both mice and humans
.
Exp Hematol
 
1999
;
27
:
1724
1734
.

18

MacVittie
 
TA
,
Farese
 
AM
,
Smith
 
WG
, et al.
Myelopoietin, an engineered chimeric IL-3 and G-CSF receptor agonist, stimulates multilineage hematopoietic recovery in a nonhuman primate model of radiation-induced myelosuppression
.
Blood
 
2000
;
95
:
837
845
.

19

Mackall
 
CL
,
Fleisher
 
TA
,
Brown
 
MR
, et al.
Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy
.
N Engl J Med
 
1995
;
332
:
143
149
.

20

Mackall
 
CL
,
Fleisher
 
TA
,
Brown
 
MR
, et al.
Distinctions between CD8+ and CD4+ T-cell regenerative pathways result in prolonged T-cell subset imbalance after intensive chemotherapy
.
Blood
 
1997
;
89
:
3700
3707
.

21

Broudy
 
VC
,
Lin
 
NL
,
Kaushansky
 
K
.
Thrombopoietin (c-Mpl ligand) acts synergistically with erythropoietin, stem cell factor, and interleukin-11 to enhance murine megakaryocyte colony growth and increases megakaryocyte ploidy in vitro
.
Blood
 
1995
;
85
:
1719
1726
.

22

The Institute of Laboratory Animal Resources, National Research Council
:
Guide for the Care and Use of Laboratory Animals
.
Washington
:
National Institutes of Health
,
1996
:
86
.

23

Smith
 
WG
,
Giri
 
JG
,
Abegg
 
A
, et al.
Preclinical pharmacology of the myelopoietin leridistim, a multifunctional agonist of IL-3 and G-CSF receptors
.
Blood
 
1998
;
92
:
380a

24

van Gils
 
F
,
Budel
 
L
,
Burger
 
H
, et al.
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
 
1994
;
22
:
248
255
.

25

Klein
 
B
,
Thomas
 
J
,
Klover
 
J
, et al.
Comparison of daniplestim and human interleukin-3: impact of structure on functional activity
.
Blood
 
1998
;
92
:
174a

26

Farese
 
AM
,
Herodin
 
F
,
McKearn
 
JP
, et al.
Acceleration of hematopoietic reconstitution with a synthetic cytokine (SC-55494) after radiation-induced marrow aplasia
.
Blood
 
1996
;
87
:
581
591
.

27

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
 
1994
;
84
:
65
73
.

28

MacVittie
 
TJ
,
Farese
 
AM
,
Herodin
 
F
, et al.
Combination therapy for radiation-induced bone marrow aplasia in nonhuman primates using synthokine SC-55494 and recombinant human granulocyte colony-stimulating factor
.
Blood
 
1996
;
87
:
4129
4135
.

29

Wickenhauser
 
C
,
Lorenzen
 
J
,
Thiele
 
J
, et al.
Secretion of cytokines (interleukins-1 alpha, −3, and −6 and granulocyte macrophage colony-stimulating factor) by normal human bone marrow megakaryocytes
.
Blood
 
1995
;
85
:
685
691
.

30

Aglietta
 
M
,
Sanavio
 
F
,
Stacchini
 
A
, et al.
Interleukin-3 in vivo: kinetic of response of target cells
.
Blood
 
1993
;
82
:
2054
2061
.

31

Donahue
 
RE
,
Seehra
 
J
,
Metzger
 
M
, et al.
Human IL-3 and GM-CSF act synergistically in stimulating hematopoiesis in primates
.
Science
 
1988
;
241
:
1820
1823
.

32

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
 
1994
;
83
:
1507
1514
.

33

Collins
 
MK
,
Marvel
 
J
,
Malde
 
P
, et al.
Interleukin-3 protects murine bone marrow cells from apoptosis induced by DNA damaging agents
.
J Exp Med
 
1992
;
176
:
1043
1051
.

34

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
 
1994
;
179
:
1777
1787
.

35

Silva
 
A
,
Wyllie
 
A
,
Collins
 
MKL
.
p53 is not required for regulation of apoptosis or radioprotection by interleukin-3
.
Blood
 
1997
;
89
:
2717
2722
.

36

Collins
 
MKL
,
Perkins
 
GR
,
Rodriguez-Tarduchy
 
G
, et al.
Growth factors as survival factors: regulation of apoptosis
.
Bioessays
 
1994
;
16
:
133
138
.

37

Lin
 
Y
,
Benchimol
 
S
.
Cytokines inhibit p53-mediated apoptosis but not p53-mediated G1 arrest
.
Mol Cell Biol
 
1995
;
15
:
6045
6054
.

38

Sato
 
N
,
Caux
 
C
,
Kitamura
 
T
, et al.
Expression and factor-dependent modulation of interleukin-3 receptor subunits on human hematopoietic cells
.
Blood
 
1993
;
82
:
752
761
.

39

Wognum
 
AW
,
Visser
 
TP
,
de Jong
 
MO
, et al.
Differential expression of receptors for interleukin-3 on subsets of CD34-expressing hematopoietic cells for rhesus monkeys
.
Blood
 
1995
;
86
:
581
591
.

40

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. In:
MacVittie
 
TJ
,
Weiss
 
JF
,
Browne
 
D
, eds.
Advances in the Treatment of Radiation Injuries: Advances in the Biosciences
.
Great Britain
:
Pergamon
,
1994
:
75
81

41

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
 
1996
;
88
:
351a

42

Paquette
 
RL
,
Zhou
 
JY
,
Yang
 
YC
, et al.
Recombinant gibbon interleukin-3 acts synergistically with recombinant human G-CSF and GM-CSF in vitro
.
Blood
 
1988
;
71
:
1596
1600
.

43

Mackall
 
CL
,
Fleisher
 
TA
,
Brown
 
MR
, et al.
Lymphocyte depletion during treatment with intensive chemotherapy for cancer
.
Blood
 
1994
;
84
:
2221
2228
.

44

Chen
 
Q
,
Solar
 
G
,
Eaton
 
DL
, et al.
IL-3 does not contribute to platelet production in C-Mpl-deficient mice
.
Stem cells
 
1998
;
16
(
suppl 2
):
31
36
.

45

Nishinakamura
 
R
,
Nakayama
 
N
,
Irabayashi
 
Y
, et al.
Mice deficient for the IL-3/GM-CSF/IL-5 βc receptor exhibit lung pathology and impaired immune response, while β IL-3 receptor-deficient mice are normal
.
Immunity
 
1995
;
2
:
211
222
.

46

Hirayama
 
F
,
Ogawa
 
M
.
Negative regulation of early T lymphopoiesis by interleukin-3 and interleukin-1α
.
Blood
 
1995
;
86
:
4527
4531
.

47

Hirayama
 
F
,
Clark
 
SC
,
Ogawa
 
M
.
Negative regulation of early B lymphopoiesis by interleukin 3 and interleukin 1α
.
Proc Natl Acad Sci USA
 
1994
;
91
:
469
473
.

48

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
 
1990
;
76
:
2235
2241
.

49

Miller
 
JS
,
McCullar
 
V
,
Punzel
 
M
, et al.
Single adult human CD34+/Lin/CD38 progenitors give rise to natural killer cells, B-lineage cells, dendritic cells, and myeloid cells
.
Blood
 
1999
;
93
:
96
106
.

50

Castelli
 
MP
,
Black
 
PL
,
Schneider
 
M
, et al.
Protective, restorative and therapeutic properties of recombinant human IL-1 in rodent models
.
J Immunol
 
1988
;
140
:
3830
3837
.

51

Brown
 
MP
,
Nosaka
 
T
,
Tripp
 
RA
, et al.
Reconstitution of early lymphoid proliferation and immune function in Jak3-deficient mice by interleukin-3
.
Blood
 
1999
;
94
:
1906
1914
.

52

Mackarehtschian
 
K
,
Hardin
 
JD
,
Moore
 
KA
, et al.
Targeted disruption of the flk2/flt3 gene leads to deficiencies in primitive hematopoietic progenitors
.
Immunity
 
1995
;
3
:
147
161
.

53

Hirayama
 
F
,
Aiba
 
Y
,
Ikebuchi
 
K
, et al.
Differentiation in culture of murine primitive lymphohematopoietic progenitors toward T-cell lineage
.
Blood
 
1999
;
93
:
4187
4195
.

54

Carding
 
SR
,
Hayday
 
AC
,
Bottomly
 
K
.
Cytokines in T-cell development
.
Immunol Today
 
1991
;
12
:
239
245
.

55

Rodewald
 
HR
,
Kretzschmar
 
K
,
Swat
 
W
, et al.
Intrathymically expressed c-kit ligand (stem cell factor) is a major factor driving expansion of very immature thymocytes in vivo
.
Immunity
 
1995
;
3
:
313
319
.

56

von Freeden-Jeffry
 
U
,
Vieira
 
P
,
Lucian
 
LA
, et al.
Lymphopenia in interleukin (IL)-7 gene-deleted mice identifies IL-7 as a nonredundant cytokine
.
J Exp Med
 
1995
;
181
:
1519
1526
.

57

Peschon
 
JJ
,
Morrissey
 
PJ
,
Grabstein
 
KH
, et al.
Early lymphocyte expansion is severely impaired in interleukin-7 receptor deficient mice
.
J Exp Med
 
1994
;
180
:
1955
1960
.

58

Borge
 
OJ
,
Adolfsson
 
J
,
Jacobsen
 
AM
, et al.
Lymphoid-restricted development from multipotent candidate murine stem cells: distinct and complimentary functions of the c-kit and flt3-ligands
.
Blood
 
1999
;
94
:
3781
3790
.

59

Morrissey
 
PJ
,
Conlon
 
P
,
Braddy
 
S
, et al.
Administration of IL-7 to mice with cyclophosphamide-induced lymphopenia accelerates lymphocyte repopulation
.
J Immunol
 
1991
;
146
:
1547
1552
.

60

Faltynek
 
CR
,
Wang
 
S
,
Miller
 
D
, et al.
Administration of human recombinant IL-7 to normal and irradiated mice increases the numbers of lymphocytes and some immature cells of myeloid lineage
.
J Immunol
 
1992
;
149
:
1276
1282
.

61

Friend
 
SL
,
Hosier
 
S
,
Nelson
 
A
, et al.
A thymic stromal cell line supports in vitro development of surface IgM+ B cells and produces a novel growth factor affecting B and T lineage cells
.
Exp Hematol
 
1994
;
22
:
321
328
.

62

Neelis
 
KJ
,
Hartong
 
SCC
,
Egeland
 
T
, et al.
The efficacy of single-dose administration of thrombopoietin with coadministration of either granulocyte/macrophage or granulocyte colony-stimulating factor in myelosuppressed rhesus monkeys
.
Blood
 
1997
;
90
:
2565
2573
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)