Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Nov 11.
Published in final edited form as: Am J Hematol. 2018 May 11:10.1002/ajh.25136. doi: 10.1002/ajh.25136

rBPI21 (Opebacan) Promotes Rapid Trilineage Hematopoietic Recovery in a Murine Model of High-Dose Total Body Irradiation

Kenneth J Janec 1, Huaiping Yuan 2, James E Norton Jr 1, Rowan H Kelner 1, Christian K Hirt 2, Rebecca A Betensky 3, Eva C Guinan 1,4,5,*
PMCID: PMC6230507  NIHMSID: NIHMS972570  PMID: 29752735

Abstract

The complexity of providing adequate care after radiation exposure has drawn increasing attention. While most therapeutic development has focused on improving survival at lethal radiation doses, acute hematopoietic syndrome (AHS) occurs at substantially lower exposures. Thus, it is likely that a large proportion of such a radiation-exposed population will manifest AHS of variable degree and that the medical and socioeconomic costs of AHS will accrue. Here, we examined the potential of rBPI21 (opebacan), used without supportive care, to accelerate hematopoietic recovery after radiation where expected survival was substantial (42–75%) at 30 days). rBPI21 administration was associated with accelerated recovery of hematopoietic precursors and normal marrow cellularity, with increases in megakaryocyte numbers particularly marked. This translated into attaining normal trilineage peripheral blood counts 2–3 weeks earlier than controls. Elevations of hematopoietic growth factors observed in plasma and the marrow microenvironment suggest the mechanism is likely multifactorial and not confined to known endotoxin-neutralizing and cytokine down-modulating activities of rBPI21. These observations deserve further exploration in radiation models and other settings where inadequate hematopoiesis is a prominent feature. These experiments also model the potential of therapeutics to limit the allocation of scarce resources after catastrophic exposures as an endpoint independent of lethality mitigation.

Keywords: hematopoiesis, total body irradiation, endotoxin, bactericidal/permeability-increasing protein (BPI), radiation disaster

Introduction

Perhaps more than Hiroshima, the accidents at Chernobyl and Three Mile Island, natural disaster at Fukushima, and omnipresent threat of nuclear detonation highlight that unexpected radiation exposure is a reality that could affect considerable numbers of individuals. As the scope of the resource and managerial challenges after such an event has been increasingly well-delineated, we understand that radiation mitigation strategies must meet an array of efficacy, toxicity, and implementation criteria to be of general value.111 A number of promising strategies that emphasize improving survival of very severely affected individuals have recently emerged, often demanding the use of extensive personnel and other resource intensive medical management.12 and rev. in13,14

A wide range of partial or total body irradiation (TBI) doses can induce profound hematopoietic suppression without causing lethal damage to other organs. It is therefore unsurprising that most proposed radiation mitigation strategies, including hematopoietic cell transplantation, improve some aspect of radiation-induced acute hematopoietic syndrome (AHS). Indeed, in 2015, filgrastim (recombinant human granulocyte colony stimulating factor, rhG-CSF) and pegfilgrastim (PEGylated rhG-CSF), hematopoietic growth factors used to improve recovery from myelosuppressive cancer therapy including transplantation, became the first agents approved for the indication of improving survival in patients exposed to TBI doses associated with hematopoietic suppression.13

Even lower TBI doses associated with AHS commonly cause some transient damage to gastrointestinal mucosa.1517 As a result, bacteria (including Gram-negative bacteria bearing endotoxin, also known as lipopolysaccharide or LPS, in their plasma membrane) leak into the systemic circulation.1519 In humans, who are particularly endotoxin-sensitive, and in animal model systems, both bacterial sepsis and endotoxemia have marked effects on bone marrow (BM) and peripheral blood (PB) cells, and endotoxin itself can induce myelosuppression and hematopoietic stem cell (HSC) exhaustion.2022 Endotoxin sensitivity is regulated by interactive proteins that enhance or downregulate responsiveness. Amongst the most potent is bactericidal/permeability-increasing protein (BPI) whose N-terminus binds endotoxin avidly.23 This interaction both precludes endotoxin from binding to TLR4, its pro-inflammatory, pro-apoptotic receptor, and potentiates endotoxin clearance. As BPI is abundant in the cytoplasmic granules of polymorphonuclear neutrophils (PMN) and monocytes,2326 TBI-induced hematopoietic suppression results in loss of BPI and its endotoxin neutralization capacity.

Opebacan (rBPI21, a 21kD recombinant N-terminal fragment of human BPI) has many advantages as a potential ameliorator of hematopoietic damage caused by radiation. It can be administered subcutaneously (SC), has a long shelf-life and an excellent safety profile in approximately 1200 people of all ages in Phase I-III trials of sepsis and one myeloablative chemoradiotherapy study.2732 rBPI21 has considerable anti-infective properties25,26,29,32,33 and synergizes with antibiotics against multiply-resistant bacteria,34 potentially of value in radiation disaster settings where concomitant trauma contributes to morbidity and mortality.35,36 We previously explored the potential of rBPI21 to mitigate radiation-induced lethality in a murine model.37 Thirty-day (D30) mortality after 7 Gy TBI was ≥95%, and neither SC rBPI21 nor oral (PO) enrofloxacin (ENR, a fluoroquinolone, used to model suggested supportive care post-radiation catastrophe) alone was sufficient to improve survival. However, combinatorial rBPI21 and ENR treatment, starting 24 hours after TBI, improved D30 survival from ≤5% to ≥65–90% and markedly mitigated AHS.

At the time of our prior study, there was no known role for rBPI21 in hematopoietic regulation or radiation mitigation. rBPI21 and ENR co-administration limited our ability to understand their individual contributions to hematopoietic recovery. Fluoroquinolones had demonstrated some radiation protection but inadequate mitigation in murine models.38,39 Moreover, FDA warnings about fluoroquinolone toxicities40 and the recent association with gastrointestinal perforation41 have been concerning. We therefore became interested in studying the role of single-agent rBPI21 in ameliorating AHS and exploring the hypothesis that rBPI21 might participate in stress hematopoietic regulation. An easily deployable therapeutic, with an established safety record, that can ameliorate trilineage AHS independent of effects on lethality should improve recovery from, and decrease the resources necessary to support individuals after, significant (but not uniformly lethal) radiation. Herein, we report results of using SC rBPI21 as sole treatment in this setting.

Materials and Methods

Animal Model and rBPI21 Treatment Regimen

Studies were conducted in accordance with Dana-Farber Cancer Institute (DFCI) Animal Care and Use Committee (OLAW Assurance # A3023-01) approved protocol 13-001. At 12 weeks of age, mice were placed in a Rad Disk rodent isolation irradiation cage (Braintree Scientific) and administered a single dose of either 6.25 Gy or 6.5 Gy TBI at 103 rad/min by a Gammacell 40 Exactor (Best Theratronics, Ontario) cesium source irradiator. Littermates were saved as unirradiated, untreated age-matched controls. Irradiated mice were randomly assigned to 3 groups: 1) no treatment (TBI alone) or, starting 24 hours after TBI, 14 days of twice daily (separated by 6–8 hours) SC injection of either 2) rBPI21 at 40mg/kg/day [2.0mg/ml] (Margaux Biologicals, Inc.) in formulation buffer, composed of citrate buffer saline (5mM citrate, 150mM sodium chloride, pH 5.0) with 0.2% poloxamer 188 and 0.002% polysorbate 80, filtered through a sterile 0.22μm PES vacuum filter, or 3) equal volume of the identical formulation buffer (vehicle control, VEH, from Catalent). Mice were monitored and euthanized as previously reported.37 PB and BM studies performed on D3 (to establish the degree of BM ablation), D15 and D19 (to assess recovery) and D30 (recovery and survival endpoint). In some cases, D10 studies were performed.

Processing and Analysis of Blood

Terminal collection of PB was carried out by cardiac puncture and held in EDTA coated microtainer tubes (Becton-Dickinson). CBCs were performed on a Hemavet 950 FS analyzer (Drew Scientific) by DFCI core technicians. Reticulocyte staining was performed using the Retic-Count kit (Becton-Dickinson) per manufacturer’s instructions. Plasma was collected as previously described.37

Processing and Evaluation of Bones, BM Supernatants and BM Cells

One femur/mouse was fixed in 10% neutral buffered formalin for 24–48 hours at 4°C, placed in Kristensen’s decalcification solution for 48 hours at 4°C and paraffin-embedded. 5 μm sections were stained with H&E. Two observers independently determined BM cellularity/femur and megakaryocyte count/high power field (HPF) using an Olympus BX43 with Olympus SC100 for image capture. BM cellularity was assessed at 40× as the percentage of the BM cavity occupied by nucleated hematopoietic cells and not microenvironmental or adipose constituents. Megakaryocyte counts were calculated as the median value from 10 random HPF/femur at 200× magnification with a 0.40 numerical aperture. All images were taken with CellSens Standard software at 400× magnification with a 0.65 numerical aperture of a 40× objective lens. Contralateral bones were held in complete medium (RPMI 1640 supplemented with 1% HEPES, 1% Pen/Strep, 1% L-glutamine, and 10% heat-inactivated FBS) before being processed. To acquire BM supernatant, the epiphyses of both bones in one hind limb were cut off after dissection and each diaphyseal shaft was flushed with 250 μl of complete medium. Flushed BM cells were pelleted by centrifugation at 300×g for 5 minutes at 4°C. Diluted BM supernatant was stored at −80°C. BM diaphyses were then flushed with an additional total 10 ml of ice-cold complete media to ensure removal of remaining BM cells. The eluate was combined with the first flush pellet and centrifuged at 300×g for 5 minutes at 4°C. The pellet was resuspended in RBC lysis buffer (#R7757 Sigma) per manufacturer’s instructions. Viable BM cell counts/limb were enumerated on a hemocytometer using trypan-blue exclusion.

Quantification of Mediators in Plasma and BM Supernatant

Mediator levels were analyzed by multi-analyte bead-based kit (#MCYTOMAG-70K-PMX, EMD Millipore) per manufacturer’s instructions and read on a Bio-Plex 200 System (Bio-Plex 200 System, Bio-Rad). TPO levels were analyzed by ELISA (Elabscience, #E-EL-M0640) per manufacturers’ directions and read at 450 nm with a microplate reader (SpectraMax M3, Molecular Devices, LLC). To estimate the dilution of BM supernatant in complete medium, we measured the diameter and length of 13 femurs using an Olympus BX43 to calculate the volume of the cavity/femur (median 6.6 mm3, range 2.8–10.8 mm3). We estimated that 50% of the cavity was fluid volume, yielding an estimated volume of 3.3 μL/femur. This estimate was used to correct for the dilution of the BM supernatant protein levels. Reported BM supernatant data were calculated using these assumptions.

BM Cell Flow Cytometry Analysis

Non-specific binding was blocked by incubating in stain buffer (Becton-Dickinson) with 2% normal rat serum at 4°C for 20 minutes. Thereafter, antibody staining was performed by the addition of a pre-mixed FITC-conjugated lineage-specific (Lin) cocktail of antibodies (145-2C11, RB6-8C5, M1/70, RA3-6B2, Ter-119; Biolegend), APC-conjugated c-Kit (2B8; Biolegend), and PE-Cy7-conjugated Sca-1 (D7; Becton-Dickinson) diluted [1:20] in FACS staining buffer (Becton-Dickinson) at 4°C for 25 minutes protected from light. Fluorescent-minus-one samples were used as gating controls. 7-AAD [1:100] was used for viability discrimination. Data was acquired on a Miltenyi MACSQuant and analyzed in FlowJo v.10.0.8 (Treestar). Cells negative for lineage-cocktail markers (Lin) were further designated as Lin/Sca-1/c-Kit+ (LK) or Lin/Sca-1+/c-Kit+ (LSK). Population frequencies were determined on a per limb basis and the absolute number determined using BM cell counts/limb. Because D3 cellularity was very low, BM cells were pooled between animals within respective groups prior to preparation for flow cytometry.

Statistical Analysis

Mantel-Cox log-rank was used to compare survival curves (GraphPad Prism 6). PB parameters and BM cell counts were analyzed using an unpaired parametric t-test with Welch’s correction (GraphPad Prism 6). For CBC values, outliers were removed prior to analysis using a modified Z-score (Excel); equal variance was not assumed. For HSC analyses, we fit separate regression models for the log transformed LSK and LK data. We fit linear models in day, as well as models that treated day as a repeated measure. We included interaction terms to enable us to consider differences between TBI alone and VEH and between TBI doses. We selected our final models for the LSK and LK analyses using the Bayesian Information Criterion (BIC). We confirmed approximate normality of the residuals from the models through histogram plots and applied a Bonferroni correction (separately within the LSK analyses and the LK analyses) for the multiple testing across days. The alpha (α= 0.05) for statistical significance was prospectively identified and not varied for any data analysis. For analyses of histopathology results and cytokine parameters, ordinary one-way ANOVAs with Dunnett’s multiple comparison test were used to compare multiple groups. Where appropriate, n sizes are shown in the figure legends. Where indicated in figures, * P < 0.05, ** P < 0.01, *** P < 0.001, **** P < 0.0001.

Results

6.25 Gy and 6.5 Gy produce a range of intermediate survivals after single fraction TBI

In our studies examining effects of rBPI21 and ENR co-administration on radiation lethality, a single 7 Gy fraction of TBI produced rapid and profound hematologic suppression and resulted in 95% lethality by D30 in 12 week-old male BALB/c mice.37 To explore whether rBPI21 as a single therapeutic could ameliorate severe AHS in a model of single fraction TBI where survival was possible but not universal, we examined the lethality of TBI at 4 different TBI doses: 6 Gy, 6.25 Gy, 6.5 Gy and 7 Gy. Survival determined by Mantel-Cox log rank was significantly different across the 4 dose groups examined (P < 0.0001, Supplementary Fig. S1). D30 survival of the 6.25 Gy cohorts ranged from 60–75% (mean 68%, total n = 19 from 2 independent experiments). D30 survival of the 6.5 Gy cohorts ranged from 42–58% (mean 50%, total n = 24 from 2 independent experiments). As the survival at these doses was both in the desired intermediate range and sufficient to permit comparisons between untreated and treated cohorts, subsequent experiments to examine hematopoietic effects were performed at both 6.25 and 6.5 Gy.

rBPI21 accelerated trilineage PB count recovery after AHS

White blood cell (WBC) and PMN recovery

Both 6.25 and 6.5 Gy TBI were associated with sustained PB pancytopenia, including >15 days of leukopenia and neutropenia in irradiated controls (Fig. 1). In contrast, by D15 after 6.25 Gy, rBPI21-treated mice had a trend to more (2- to 3-fold) WBC and PMN than either control (Fig. 1A,B). After 6.5 Gy (Fig. 1E,F), that difference was significant for WBC (rBPI21 vs TBI alone P = 0.01 or VEH P = 0.009) and PMN (rBPI21 vs TBI alone P = 0.009 or VEH P = 0.01). While median WBC and PMN counts remained low and virtually unchanged through D19 in both irradiated controls, rBPI21 treatment was associated with higher counts after 6.25 Gy (WBC: rBPI21 vs TBI alone or VEH P =0.0003; PMN: rBPI21 vs TBI alone or VEH P = 0.0001) and 6.5 Gy (WBC: rBPI21 vs TBI alone P = 0.004 or VEH P = 0.003; PMN: rBPI21 vs TBI alone or VEH P = 0.002). Only rBPI21-treated mice had D19 median WBC (6.25 Gy) and PMN (both doses) above the lower limit of normal (LLN). Similar recovery was not observed in irradiated controls until D30, when virtually all groups had median values above the LLN. rBPI21-treated mice also experienced accelerated monocyte recovery (Supplementary Fig. S2).

Figure 1.

Figure 1

Platelet recovery

PLT counts recovered with a similar trajectory (Fig. 1C,G). By D15, rBPI21-treated mice had greater median PLT than irradiated controls (6.25 Gy, P = 0.005 vs either irradiated control; 6.5 Gy, P = 0.04 vs either irradiated control). Between D15-19, rBPI21-treated mice continued to recover PLT counts rapidly, by 3-fold (6.25 Gy) and 5-fold (6.5 Gy), nearing or exceeding the LLN. During this period, counts in irradiated controls rose < 2-fold, remaining well below the LLN. The median D19 PLT count of rBPI21-treated mice was significantly greater than TBI alone or VEH counts (P < 0.0001 for each) at both TBI doses. By D30, virtually all groups had median counts at or exceeding the LLN.

Red cell recovery

Median hematocrit (HCT) universally lay below the LLN by D15 (Supplementary Fig. S2). VEH and rBPI21-treated groups had lower D15 HCT than TBI alone after 6.5 Gy (VEH vs TBI alone P = 0.0006 and rBPI21 vs TBI alone P = 0.004) but not 6.25 Gy, raising the possibility that injection related bleeding may have contributed to falls in HCT in the more compromised 6.5 Gy mice. Bleeding at rBPI21 or VEH injection sites (total 28 injections over 14 days) was observed either immediately or as SC hemorrhage at necropsy and was more frequently noted after 6.5 Gy. At D19, HCT had fallen further in irradiated controls, but had risen in rBPI21-treated mice, producing significant differences (6.25 Gy: rBPI21 vs TBI and vs VEH P < .0001; 6.5 Gy: rBPI21 vs TBI P = 0.0001 and rBPI21 vs VEH P = 0.0015). By D30, all groups had median HCT above or nearing the LLN. Changes in hemoglobin (Hgb) and red blood cell count (RBC) were similar (Supplementary Fig. S2). There was a trend to elevated reticulocyte frequency in rBPI21-treated mice by D15 (Fig. 1D,H). By D19, reticulocytes comprised 18% of circulating RBC in rBPI21-treated mice in comparison to low, unchanged values (< 4%) in irradiated controls (6.25 Gy: rBPI21 vs either TBI alone or VEH P = 0.01; 6.5 Gy: rBPI21 vs TBI alone P = 0.0004 or VEH P = 0.0002).

rBPI21 accelerated the rate of recovery of BM hematopoietic populations

rBPI21 accelerated recovery of BM cell counts

On D3, BM cell counts/hindlimb were profoundly reduced (Fig. 2A,D). There was a trend to greater ablation at 6.5 Gy (P = 0.09). Neither VEH nor rBPI21 impacted the degree of hematopoietic suppression on D3 (6.25 Gy: TBI alone vs VEH P = 0.4 or rBPI21 P = 0.6; 6.5 Gy: TBI alone vs VEH P = 0.4 or rBPI21 P = 0.8). We evaluated the time to normal count recovery by comparing each irradiated treatment group to unirradiated controls. In rBPI21-treated mice, median BM counts increased by 7.3-fold (6.25 Gy) and 1.7-fold (6.5 Gy) between D3-15 and reached or exceeded the LLN by D19 (Fig. 2A,D). In contrast, counts remained ≤ 5% of the LLN between D3-19 in irradiated controls, regardless of TBI dose. While all D30 group medians fell above the LLN, rBPI21-treated group values exceeded the normal median whereas irradiated controls remained 33–53% below.

Figure 2.

Figure 2

We also compared counts between irradiated treatment groups (Fig. 2A,D). After 6.25 Gy, counts were greater in the rBPI21-treated group by D15 (rBPI21 vs TBI alone P = 0.0012 or VEH P = 0.0006), D19 (rBPI21 vs TBI alone P = 0.0002 or VEH P = 0.0003) and D30 (rBPI21 vs TBI alone or VEH P = 0.03). Similarly, counts were greater in the rBPI21-treated group after 6.5 Gy by D15 (rBPI21 vs TBI alone P = 0.017 or VEH P = 0.21), D19 (rBPI21 vs TBI alone P = 0.001 or VEH P = 0.004) and D30 (rBPI21 vs TBI alone P = 0.033 or VEH P = 0.008).

rBPI21 accelerated recovery of LK and LSK counts

We first compared values in each irradiated treatment group to values in normal age-matched mice and characterized the fold increase in LK and LSK populations to illustrate the trajectory each group followed during recovery. Median LK counts in rBPI21-treated mice rose early, increasing 253-fold (6.25 Gy) and 97-fold (6.5 Gy) between D3-15. By D19, approximately 50% of rBPI21-treated mice had exceeded (6.25 Gy) or neared the LLN (6.5 Gy, Fig. 2B,E). By D30 all rBPI21-treated mice in both TBI dose groups had LK counts within the normal range. In contrast, D3-15 fold-increases were much lower (range, none to 26-fold) in irradiated controls. By D19, median values in both irradiated control groups at both TBI doses remained at 4–13% of the LLN. Capacity for delayed recovery of LK in irradiated controls was evident in the 10- to 30-fold increases observed from D19-30. By Day 30, the majority of irradiated control mice had LK counts in the normal range.

LSK recovery was comparable. In rBPI21-treated mice, median LSK counts increased by 4.6-fold (6.25 Gy) and 3.3-fold (6.5 Gy) between D3-15 and the majority (6.25 Gy) or some (6.5 Gy) had LSK counts that exceeded the LLN of by D19 (Fig. 2C,F). By D30, the fraction above the LLN had increased for both TBI doses. In contrast, median LSK counts remained unchanged between D3-15 in both irradiated controls regardless of TBI dose, and by D19 their LSK counts were still 6–18% of the LLN. Despite greater fold increases (ranging 3- to 13-fold) from D19-30, less than 50% of the irradiated controls had LSK counts that neared or exceeded the LLN at D30 after 6.25 Gy and virtually none did so after 6.5 Gy.

We also compared irradiated treatment groups to one another (Table 1). For all modelling approaches, we found non-significant interactions representing differences between TBI alone and VEH and therefore we combined these two groups into a single category referred to as irradiated controls for the subsequent analyses. The linear model for LK estimated a significantly higher slope for rBPI21 versus irradiated controls (difference in slopes of 0.036, SE = 0.006, P < 0.0001). The linear model for LSK also obtained a significantly higher slope for rBPI21 than irradiated controls (difference in slopes of 0.024, SE = 0.005, P < 0.0001). The selected model for LK retained the interactions between day and rBPI21 vs irradiated controls and no terms involving dose. The selected model for LSK retained the interactions between day and rBPI21 vs irradiated controls as well as the interactions between day and dose. The repeated measures models for both LSK and LK obtained significantly higher values for rBPI21 versus irradiated control results (after Bonferroni correction) for D10, 15 and 19. This is also true at D30, though is not significant at the corrected p-value (Table 1). Global P-values (5 degrees of freedom) for each analysis for the comparison between rBPI21 and irradiated controls are both < 0.0001.

Table 1.

rBPI21 accelerated recovery of progenitor and stem cell populations

Day Difference in slope of rBPI21 vs combined TBI controls SE p-value
LK 3 0.2 0.23 0.383

10 0.99 0.38 0.011

15 1.34 0.22 <0.0001

19 1.28 0.21 <0.0001

30 0.39 0.19 0.04

LSK 3 0.007 0.19 0.972

10 1.07 0.31 0.0008

15 0.65 0.18 0.003

19 0.97 0.17 <0.0001

30 0.35 0.15 0.022

Data aggregated from 2–5 independent experiments at each TBI dose, n = 3–9 per group. SE = standard error

rBPI21 accelerated recovery of in situ BM cellularity and megakaryocyte counts

The loss of BM hematopoietic elements at D3 was profound (Fig. 2A,D) at both TBI doses, which precluded meaningful histologic comparisons. Thereafter, rBPI21-associated recovery of BM cellularity was more complete than either irradiated control at each time point and TBI dose (Fig. 3A–D). In contrast, we observed no change in the cellularity of either irradiated control through D19 regardless of TBI dose, and D30 median cellularity remained ≤ 50% of normal. These histopathologic results were consistent with the BM cell counts shown in Fig. 2A,D. rBPI21-treated mice had earlier, more robust megakaryocyte recovery than irradiated controls on each day evaluated at 6.25 Gy, and D19-30 at 6.5 Gy (Fig. 3A–C,E).

Figure 3.

Figure 3

rBPI21 was associated with increased cytokine and hematopoietic growth factor levels

The rapid hematopoietic recovery in rBPI21-treated mice led us to examine early levels (D3) of hematopoietic and inflammatory mediators that could have contributed to these later outcomes. For the majority of mediators assessed, D3 plasma levels in rBPI21-treated mice were significantly elevated in comparison to both unirradiated and irradiated controls (Fig. 4A–H). In some cases, these effects were only observed at 6.5 Gy or in more singular patterns (Supplementary Fig. S3).

Figure 4.

Figure 4

We observed fewer effects in BM supernatant, although the high-volume flush required for sample collection introduced substantial dilution of the extracellular fluid from the cavity which may have impacted assay sensitivity. As described in Methods, we corrected for this dilution to provide some reasonable comparability of concentrations of mediators in the dilute supernatants with that in undiluted plasma. Significant rBPI21-associated elevation of G-CSF, KC, and CCL-2 were observed on D3 (Fig 4I–K). For KC and CCL-2, median BM supernatant levels were higher than in plasma whereas G-CSF levels were comparable.

Because thrombopoietin (TPO) participates in regulating both HSC and megakaryocyte production,42,43 we determined D3 TPO levels in plasma and BM supernatant (Supplementary Table S1). Levels were not different between the TBI alone and VEH-treated cohorts (at 6.25 Gy P = 0.24, at 6.5 Gy P = 0.94), thus they were combined as irradiated controls for TPO analyses. In comparison to unirradiated controls, plasma levels in irradiated controls rose by 1.6- (6.25 Gy) to 1.5- (6.5 Gy) fold while BM supernatant levels rose 4.5-fold (6.25 Gy) and 9.8-fold (6.5 Gy). The addition of rBPI21-treatment led to further increases in both plasma and supernatant levels, which were significant in most cases.

rBPI21 improved survival of mice irradiated at 6.5 Gy

Although these studies were designed primarily to examine effects of rBPI21 on AHS, a limited number of small survival cohorts were also followed. At 6.25 Gy, rBPI21 did not improve D30 survival (rBPI21 vs TBI P = 0.2 and rBPI21 vs VEH, P = 0.93) However, at 6.5 Gy, which was associated with greater D30 mortality, rBPI21 administration resulted in a near doubling of survival (mean 78%, range 67–92) in comparison to VEH (mean 42%, range 33–50, P = 0.0065 by Mantel-Cox; Supplementary Fig. S4).

Discussion

The numerous, interactive challenges to providing adequate medical care following substantial radiation exposure are well-delineated in manuscripts describing the intensive resource and operational requirements of responding to a catastrophic event.111 Optimal management of individuals with unintended radiation exposure will likely depend upon factors including exposure (dose, type of radiation and body surface involved), extent and type of concomitant trauma, and facilities and resources available for triage and treatment.111,35 As AHS occurs at relatively lower radiation doses than other acute radiation toxicities10,4446 it is likely that many individuals within a radiation exposed population will manifest hematologic injury of variable degree and duration. Most candidate, and the approved (Neupogen and Neulasta), radiation mitigators improve post-TBI hematopoietic recovery as well as survival although the effects are not equal for all hematopoietic lineages.13,14,47,48 In order to obtain FDA designation as a radiation mitigator, the path to approval specifies studies demonstrating a survival advantage in lethal/near-lethal TBI model systems. However, the larger number of individuals will experience more variable partial or TBI exposures less likely to lead to death but highly likely to result in clinically relevant AHS.1,10,45,49 Streamlined approaches that improve the likelihood and rapidity of trilineage hematopoietic recovery in such populations will decrease the frequency of pancytopenia-related complications and reduce demand on scare resources.

In this report, at TBI doses where D30 survival was 42–75% without intervention, we nonetheless observed profound persistent pancytopenia lasting several weeks. Treatment with single agent SC rBPI21 without supportive care substantially decreased the duration and degree of trilineage hematopoietic impairment in both BM and PB and accelerated return to normal values, often by 2–3 weeks. The rapid rise in short-lived PB cells such as PMN, PLT and reticulocytes as well as the recovery of BM megakaryocytes suggests this was largely due to improved production. Notably, a different N-terminal BPI construct was recently shown to promote HSC, progenitor and myeloid cell recovery post-irradiation in a perfused murine BM-on-a-chip model of AHS (RBC, megakaryocyte and PLT production were not quantitated).50 In addition, in a small phase I/II study we conducted in patients receiving myeloablative chemoradiotherapy conditioning for allogeneic hematopoietic cell transplantation, a 72-hour rBPI21 infusion was well-tolerated and associated with considerably more rapid PLT recovery and substantially fewer infections and Grade 3–5 toxicities in the first 35 days post-transplant than that observed in protocol-eligible cotemporaneous controls.51 In aggregate, these data support the hypothesis that rBPI21 could impact transfusion requirements, pancytopenia-related complications including infections, and associated material, personnel and hospitalization utilization post-irradiation.

Hematopoietic cell transplant patients become endotoxemic and BPI deficient after chemoradiotherapy-based myeloablation.37 Moreover, endotoxin ligation of TLR4 and downstream signaling play a major role in aberrant myeloid differentiation, HSC exhaustion and death during sepsis or experimental endotoxemia.21,22,5254 Together, these observations led us to investigate a potential role for rBPI21-mediated endotoxin neutralization in AHS mitigation. As rBPI21 effects had been attributed almost solely to its ability to neutralize endotoxin and ameliorate inflammatory cytokine release during endotoxemia,2326,30,31,34 we anticipated that rBPI21 administration would be associated with decreased HSC death or damage and decreased levels of inflammatory or hematopoietic mediators that had previously been described as elevated after radiation or during sepsis-related emergency hematopoiesis.22,5558 Instead, rBPI21 administration markedly augmented levels of these mediators in plasma and/or BM supernatant. Data from two unrelated systems suggest that rBPI21 may produce this effect at least in part by an endotoxin-independent mechanism. In retinopathy models, rBPI21 was found to act directly on retinal epithelial, pericyte and endothelial cells,59,60 influencing growth and apoptotic regulation and inhibition of VEGF-induced angiogenesis. In an organ-on-a-chip model of AHS, an N-terminal rBPI construct used to perfuse sterile BM stroma and HSC substantially increased hematopoietic cell proliferation.50 These observations support the notion that endotoxin-independent, direct interactions with cells, potentially including endothelium and pericytes in the hematopoietic microenvironment, led to some of the rBPI21 effects observed.

Independent of mechanism, elevation of hematopoietic regulators, has been previously reported in association with AHS mitigators. Notably, the survival benefit associated with CBLB502 (flagellin)-mediated TBI mitigation was correlated with elevation of plasma G-CSF and IL-6 levels.55 As blocking either molecule decreased the efficacy of CBLB502, the authors suggested this combination might be a biomarker of an effective AHS mitigation treatment. Furthermore, co-administration of IL-6 and G-CSF has been shown to provide more effective AHS mitigation than either alone.61 In rBPI21-treated mice, levels of these two mediators were 50–100-fold greater than in VEH-treated mice. Moreover, some mediators that rose in association with rBPI21 administration, including G-CSF, TPO, and IL-6, have been evaluated as possible AHS mitigators in multiple species where they have accelerated single- or multi-lineage recovery.1214,47,48,58,62,63 Other mediators that rose in association with rBPI21 treatment are also known to effect hematopoietic cells. For example, the myeloid growth factor GM-CSF is included as a radiation countermeasure in the Strategic National Stockpile.13 IL-9 can potentiate megakaryocytopoiesis in conjunction with other mediators, including TPO,64 and has a critical role in resolving inflammation.65 CCL2 can reduce HSC cycling, potentially supporting a longer period for repair after TBI and increased HSC survival.66 Thus, it is possible that the quantitative and temporal relationship of mediators observed in response to rBPI21 treatment, in conjunction with amelioration of endotoxin-related toxicity, support both hematopoietic recovery and survival through a variety of independent and interactive mechanisms.

Comparisons between therapeutics are always colored by differences in model selection and experimental design and regimens. With these caveats, the effects of rBPI21 on the time to onset, rate and completeness of hematopoietic recovery after AHS reported here exceed or equal those observed with other agents, including rhG-CSF administration for a similar duration in murine models of sub-lethal TBI.61 Most reports detail greatest effects on PMN recovery, while effects of equivalent magnitude on erythropoiesis and/or thrombopoiesis are less commonly reported.13,14 While PMN are essential to decreasing infectious risk and maintaining tissue integrity, PLT also play a critical role in wound repair and vascular integrity. PLT reconstitution is also an important endpoint as collection of platelets for transfusion is costly, time intensive, yields a product with limited shelf-life, and administration is associated with high risk of reactions and infection.67 However, the goal of the experiments reported here was not to establish the relative merits of rBPI21, but rather to determine the capacity of rBPI21 to rapidly resolve TBI-induced pancytopenia when given as a single agent in the absence of supportive care. This study suggests that there are likely multiple mechanisms, some novel, that contribute to the effects on hematopoiesis observed here and that these deserve further exploration in regard to normal hematopoietic regulation and other stress conditions such as myelosuppressive therapy, sepsis and acquired or congenital marrow failure. Finally, this report emphasizes the need to identify meaningful endpoints other than improved survival to foster development of approaches that limit the allocation of scarce resources to the large number of individuals expected to experience consequential, but not necessarily irreversible, syndromes such as AHS.

Supplementary Material

Supp info

Acknowledgments

The content of this publication does not necessarily reflect the views or policies of the Department of Defense or the US Government.

References

  • 1.Dainiak N, Ricks RC. The evolving role of haematopoietic cell transplantation in radiation injury: potentials and limitations. BJR Suppl. 2005;27:169–174. [Google Scholar]
  • 2.Flood AB, Nicolalde RJ, Demidenko E, et al. A Framework for Comparative Evaluation of Dosimetric Methods to Triage a Large Population Following a Radiological Event. Radiat Meas. 2011;46(9):916–922. doi: 10.1016/j.radmeas.2011.02.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hick JL, Weinstock DM, Coleman CN, et al. Health care system planning for and response to a nuclear detonation. Disaster Med Public Health Prep. 2011;5(Suppl 1):S73–88. doi: 10.1001/dmp.2011.28. [DOI] [PubMed] [Google Scholar]
  • 4.Knebel AR, Coleman CN, Cliffer KD, et al. Allocation of scarce resources after a nuclear detonation: setting the context. Disaster Med Public Health Prep. 2011;5(Suppl 1):S20–31. doi: 10.1001/dmp.2011.25. [DOI] [PubMed] [Google Scholar]
  • 5.Shimura T, Yamaguchi I, Terada H, Robert Svendsen E, Kunugita N. Public health activities for mitigation of radiation exposures and risk communication challenges after the Fukushima nuclear accident. J Radiat Res. 2015;56(3):422–429. doi: 10.1093/jrr/rrv013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Weisdorf D, Chao N, Waselenko JK, et al. Acute radiation injury: contingency planning for triage, supportive care, and transplantation. Biol Blood Marrow Transplant. 2006;12(6):672–682. doi: 10.1016/j.bbmt.2006.02.006. [DOI] [PubMed] [Google Scholar]
  • 7.Coleman CN, Weinstock DM, Casagrande R, et al. Triage and treatment tools for use in a scarce resources-crisis standards of care setting after a nuclear detonation. Disaster Med Public Health Prep. 2011;5(Suppl 1):S111–121. doi: 10.1001/dmp.2011.22. [DOI] [PubMed] [Google Scholar]
  • 8.Bouville A, Chumak VV, Inskip PD, Kryuchkov V, Luckyanov N. The chornobyl accident: estimation of radiation doses received by the Baltic and Ukrainian cleanup workers. Radiat Res. 2006;166(1 Pt 2):158–167. doi: 10.1667/RR3370.1. [DOI] [PubMed] [Google Scholar]
  • 9.Brenner DJ, Chao NJ, Greenberger JS, et al. Are We Ready for a Radiological Terrorist Attack Yet? Report From the Centers for Medical Countermeasures Against Radiation Network. Int J Radiat Oncol Biol Phys. 2015;92(3):504–505. doi: 10.1016/j.ijrobp.2015.02.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.DiCarlo AL, Maher C, Hick JL, et al. Radiation injury after a nuclear detonation: medical consequences and the need for scarce resources allocation. Disaster Med Public Health Prep. 2011;5(Suppl 1):S32–44. doi: 10.1001/dmp.2011.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Williams JP, McBride WH. After the bomb drops: a new look at radiation-induced multiple organ dysfunction syndrome (MODS) Int J Radiat Biol. 2011;87(8):851–868. doi: 10.3109/09553002.2011.560996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.MacVittie TJ, Farese AM, Jackson W., 3rd Defining the full therapeutic potential of recombinant growth factors in the post radiation-accident environment: the effect of supportive care plus administration of G-CSF. Health Phys. 2005;89(5):546–555. doi: 10.1097/01.hp.0000173143.69659.5b. [DOI] [PubMed] [Google Scholar]
  • 13.Singh VK, Garcia M, Seed TM. A review of radiation countermeasures focusing on injury-specific medicinals and regulatory approval status: part II. Countermeasures for limited indications, internalized radionuclides, emesis, late effects, and agents demonstrating efficacy in large animals with or without FDA IND status. Int J Radiat Biol. 2017;93(9):870–884. doi: 10.1080/09553002.2017.1338782. [DOI] [PubMed] [Google Scholar]
  • 14.Singh VK, Romaine PL, Newman VL, Seed TM. Medical countermeasures for unwanted CBRN exposures: part II radiological and nuclear threats with review of recent countermeasure patents. Expert Opin Ther Pat. 2016;26(12):1399–1408. doi: 10.1080/13543776.2016.1231805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Booth C, Tudor G, Tudor J, Katz BP, MacVittie TJ. Acute gastrointestinal syndrome in high-dose irradiated mice. Health Phys. 2012;103(4):383–399. doi: 10.1097/hp.0b013e318266ee13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.MacVittie TJ, Farese AM, Bennett A, et al. The acute gastrointestinal subsyndrome of the acute radiation syndrome: a rhesus macaque model. Health Phys. 2012;103(4):411–426. doi: 10.1097/HP.0b013e31826525f0. [DOI] [PubMed] [Google Scholar]
  • 17.Zhang L, Sun W, Wang J, et al. Mitigation effect of an FGF-2 peptide on acute gastrointestinal syndrome after high-dose ionizing radiation. Int J Radiat Oncol Biol Phys. 2010;77(1):261–268. doi: 10.1016/j.ijrobp.2009.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Packey CD, Ciorba MA. Microbial influences on the small intestinal response to radiation injury. Curr Opin Gastroenterol. 2010;26(2):88–94. doi: 10.1097/MOG.0b013e3283361927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Walker RI, Ledney GD, Galley CB. Aseptic endotoxemia in radiation injury and graft-vs-host disease. Radiat Res. 1975;62(2):242–249. [PubMed] [Google Scholar]
  • 20.Chua HL, Plett PA, Sampson CH, et al. Long-term hematopoietic stem cell damage in a murine model of the hematopoietic syndrome of the acute radiation syndrome. Health Phys. 2012;103(4):356–366. doi: 10.1097/HP.0b013e3182666d6f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rodriguez S, Chora A, Goumnerov B, et al. Dysfunctional expansion of hematopoietic stem cells and block of myeloid differentiation in lethal sepsis. Blood. 2009;114(19):4064–4076. doi: 10.1182/blood-2009-04-214916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhang H, Rodriguez S, Wang L, et al. Sepsis Induces Hematopoietic Stem Cell Exhaustion and Myelosuppression through Distinct Contributions of TRIF and MYD88. Stem Cell Reports. 2016;6(6):940–956. doi: 10.1016/j.stemcr.2016.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Gioannini TL, Weiss JP. Regulation of interactions of Gram-negative bacterial endotoxins with mammalian cells. Immunol Res. 2007;39(1–3):249–260. doi: 10.1007/s12026-007-0069-0. [DOI] [PubMed] [Google Scholar]
  • 24.Borregaard N. Bactericidal mechanisms of the human neutrophil. An integrated biochemical and morphological model. Scandinavian Journal of Haematology. 1984;32(3):225–230. doi: 10.1111/j.1600-0609.1984.tb01685.x. [DOI] [PubMed] [Google Scholar]
  • 25.Elsbach P, Weiss J. Bactericidal/permeability increasing protein and host defense against gram-negative bacteria and endotoxin. Current Opinion in Immunology. 1993;5(1):103–107. doi: 10.1016/0952-7915(93)90088-a. [DOI] [PubMed] [Google Scholar]
  • 26.Levy O. A neutrophil-derived anti-infective molecule: bactericidal/permeability-increasing protein. Antimicrobial Agents & Chemotherapy. 2000;44:2925–2931. doi: 10.1128/aac.44.11.2925-2931.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bauer RJ, Wedel N, Havrilla N, White M, Cohen A, Carroll SF. Pharmacokinetics of a recombinant modified amino terminal fragment of bactericidal/permeability-increasing protein (rBPI21) in healthy volunteers. J Clin Pharmacol. 1999;39(11):1169–1176. [PubMed] [Google Scholar]
  • 28.Demetriades D, Smith JS, Jacobson LE, et al. Bactericidal/permeability-increasing protein (rBPI21) in patients with hemorrhage due to trauma: results of a multicenter phase II clinical trial. rBPI21 Acute Hemorrhagic Trauma Study Group. J Trauma. 1999;46(4):667–676. doi: 10.1097/00005373-199904000-00018. discussion 676–667. [DOI] [PubMed] [Google Scholar]
  • 29.Levin M, Quint PA, Goldstein B, et al. Recombinant bactericidal/permeability-increasing protein (rBPI21) as adjunctive treatment for children with severe meningococcal sepsis: a randomised trial. rBPI21 Meningococcal Sepsis Study Group.[see comment] Lancet. 2000;356(9234):961–967. doi: 10.1016/s0140-6736(00)02712-4. [DOI] [PubMed] [Google Scholar]
  • 30.von der Mohlen MA, Kimmings AN, Wedel NI, et al. Inhibition of endotoxin-induced cytokine release and neutrophil activation in humans by use of recombinant bactericidal/permeability-increasing protein. J Infect Dis. 1995;172(1):144–151. doi: 10.1093/infdis/172.1.144. [DOI] [PubMed] [Google Scholar]
  • 31.von der Mohlen MA, van Deventer SJ, Levi M, et al. Inhibition of endotoxin-induced activation of the coagulation and fibrinolytic pathways using a recombinant endotoxin-binding protein (rBPI23) Blood. 1995;85(12):3437–3443. [PubMed] [Google Scholar]
  • 32.Wiezer MJ, Meijer C, Sietses C, et al. Bactericidal/permeability-increasing protein preserves leukocyte functions after major liver resection. Ann Surg. 2000;232(2):208–215. doi: 10.1097/00000658-200008000-00010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Balakrishnan A, Schnare M, Chakravortty D. Of Men Not Mice: Bactericidal/Permeability-Increasing Protein Expressed in Human Macrophages Acts as a Phagocytic Receptor and Modulates Entry and Replication of Gram-Negative Bacteria. Front Immunol. 2016;7:455. doi: 10.3389/fimmu.2016.00455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Weitz A, Spotnitz R, Collins J, Ovadia S, Iovine NM. Log reduction of multidrug-resistant Gram-negative bacteria by the neutrophil-derived recombinant bactericidal/permeability-increasing protein. International journal of antimicrobial agents. 2013;42(6):571–574. doi: 10.1016/j.ijantimicag.2013.07.019. [DOI] [PubMed] [Google Scholar]
  • 35.Baranov A, Gale RP, Guskova A, et al. Bone marrow transplantation after the Chernobyl nuclear accident. N Engl J Med. 1989;321(4):205–212. doi: 10.1056/NEJM198907273210401. [DOI] [PubMed] [Google Scholar]
  • 36.Kiang JG, Jiao W, Cary LH, et al. Wound trauma increases radiation-induced mortality by activation of iNOS pathway and elevation of cytokine concentrations and bacterial infection. Radiat Res. 2010;173(3):319–332. doi: 10.1667/RR1892.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Guinan EC, Barbon CM, Kalish LA, et al. Bactericidal/permeability-increasing protein (rBPI21) and fluoroquinolone mitigate radiation-induced bone marrow aplasia and death. Sci Transl Med. 2011;3(110):110ra118. doi: 10.1126/scitranslmed.3003126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kim K, Pollard JM, Norris AJ, et al. High-throughput screening identifies two classes of antibiotics as radioprotectors: tetracyclines and fluoroquinolones. Clin Cancer Res. 2009;15(23):7238–7245. doi: 10.1158/1078-0432.CCR-09-1964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Plett PA, Sampson CH, Chua HL, et al. Establishing a murine model of the hematopoietic syndrome of the acute radiation syndrome. Health Phys. 2012;103(4):343–355. doi: 10.1097/HP.0b013e3182667309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.FDA Drug Safety Communication. [Accessed 10–13–2017];FDA updates warnings for oral and injectable fluoroquinolone antibiotics due to disabling side effects. https://www.fda.gov/drugs/drugsafety/ucm511530.htm.
  • 41.Hsu SC, Chang SS, Lee MG, et al. Risk of gastrointestinal perforation in patients taking oral fluoroquinolone therapy: An analysis of nationally representative cohort. PLoS One. 2017;12(9):e0183813. doi: 10.1371/journal.pone.0183813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.de Graaf CA, Metcalf D. Thrombopoietin and hematopoietic stem cells. Cell Cycle. 2011;10(10):1582–1589. doi: 10.4161/cc.10.10.15619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kaushansky K, Lok S, Holly RD, et al. Promotion of megakaryocyte progenitor expansion and differentiation by the c-Mpl ligand thrombopoietin. Nature. 1994;369(6481):568–571. doi: 10.1038/369568a0. [DOI] [PubMed] [Google Scholar]
  • 44.Fliedner TM, Graessle D, Meineke V, Dorr H. Pathophysiological principles underlying the blood cell concentration responses used to assess the severity of effect after accidental whole-body radiation exposure: an essential basis for an evidence-based clinical triage. Exp Hematol. 2007;35(4 Suppl 1):8–16. doi: 10.1016/j.exphem.2007.01.006. [DOI] [PubMed] [Google Scholar]
  • 45.Mauch P, Constine L, Greenberger J, et al. Hematopoietic stem cell compartment: Acute and late effects of radiation therapy and chemotherapy. International Journal of Radiation Oncology*Biology*Physics. 1995;31(5):1319–1339. doi: 10.1016/0360-3016(94)00430-S. [DOI] [PubMed] [Google Scholar]
  • 46.Waselenko JK, MacVittie TJ, Blakely WF, et al. Medical management of the acute radiation syndrome: recommendations of the Strategic National Stockpile Radiation Working Group. Ann Intern Med. 2004;140(12):1037–1051. doi: 10.7326/0003-4819-140-12-200406150-00015. [DOI] [PubMed] [Google Scholar]
  • 47.Farese AM, Cohen MV, Katz BP, et al. Filgrastim improves survival in lethally irradiated nonhuman primates. Radiat Res. 2013;179(1):89–100. doi: 10.1667/RR3049.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hankey KG, Farese AM, Blaauw EC, et al. Pegfilgrastim Improves Survival of Lethally Irradiated Nonhuman Primates. Radiat Res. 2015;183(6):643–655. doi: 10.1667/RR13940.1. [DOI] [PubMed] [Google Scholar]
  • 49.Fliedner TM, Powles R, Sirohi B, Niederwieser D European Group for B, Marrow Transplantation Nuclear Accident C. Radiologic and nuclear events: the METREPOL severity of effect grading system. Blood. 2008;111(12):5757–5758. doi: 10.1182/blood-2008-04-150243. author reply 5758–5759. [DOI] [PubMed] [Google Scholar]
  • 50.Torisawa YS, Mammoto T, Jiang E, et al. Modeling Hematopoiesis and Responses to Radiation Countermeasures in a Bone Marrow-on-a-Chip. Tissue Eng Part C Methods. 2016;22(5):509–515. doi: 10.1089/ten.TEC.2015.0507. [DOI] [PubMed] [Google Scholar]
  • 51.Guinan E, Avigan DE, Soiffer RJ, et al. Pilot experience with opebacan/rBPI 21 in myeloablative hematopoietic cell transplantation. F1000Res. 2015;4:1480. doi: 10.12688/f1000research.7558.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Navarini AA, Lang KS, Verschoor A, et al. Innate immune-induced depletion of bone marrow neutrophils aggravates systemic bacterial infections. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(17):7107–7112. doi: 10.1073/pnas.0901162106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Skirecki T, Kawiak J, Machaj E, et al. Early severe impairment of hematopoietic stem and progenitor cells from the bone marrow caused by CLP sepsis and endotoxemia in a humanized mice model. Stem Cell Res Ther. 2015;6:142. doi: 10.1186/s13287-015-0135-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Takizawa H, Fritsch K, Kovtonyuk LV, et al. Pathogen-Induced TLR4-TRIF Innate Immune Signaling in Hematopoietic Stem Cells Promotes Proliferation but Reduces Competitive Fitness. Cell Stem Cell. 2017;21(2):225–240. e225. doi: 10.1016/j.stem.2017.06.013. [DOI] [PubMed] [Google Scholar]
  • 55.Krivokrysenko VI, Shakhov AN, Singh VK, et al. Identification of granulocyte colony-stimulating factor and interleukin-6 as candidate biomarkers of CBLB502 efficacy as a medical radiation countermeasure. J Pharmacol Exp Ther. 2012;343(2):497–508. doi: 10.1124/jpet.112.196071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Vijay-Kumar M, Aitken JD, Sanders CJ, et al. Flagellin treatment protects against chemicals, bacteria, viruses, and radiation. J Immunol. 2008;180(12):8280–8285. doi: 10.4049/jimmunol.180.12.8280. [DOI] [PubMed] [Google Scholar]
  • 57.Zhang P, Quinton LJ, Gamble L, Bagby GJ, Summer WR, Nelson S. The granulopoietic cytokine response and enhancement of granulopoiesis in mice during endotoxemia. Shock. 2005;23(4):344–352. doi: 10.1097/01.shk.0000158960.93832.de. [DOI] [PubMed] [Google Scholar]
  • 58.Van der Meeren A, Mouthon MA, Vandamme M, Squiban C, Aigueperse J. Combinations of cytokines promote survival of mice and limit acute radiation damage in concert with amelioration of vascular damage. Radiat Res. 2004;161(5):549–559. doi: 10.1667/rr3164. [DOI] [PubMed] [Google Scholar]
  • 59.Geraldes P, Yamagata M, Rook SL, et al. Glypican 4, a membrane binding protein for bactericidal/permeability-increasing protein signaling pathways in retinal pigment epithelial cells. Invest Ophthalmol Vis Sci. 2007;48(12):5750–5755. doi: 10.1167/iovs.07-0470. [DOI] [PubMed] [Google Scholar]
  • 60.Yamagata M, Rook SL, Sassa Y, et al. Bactericidal/permeability-increasing protein’s signaling pathways and its retinal trophic and anti-angiogenic effects. Faseb J. 2006;20(12):2058–2067. doi: 10.1096/05-5662com. [DOI] [PubMed] [Google Scholar]
  • 61.Patchen ML, Fischer R, MacVittie TJ. Effects of combined administration of interleukin-6 and granulocyte colony-stimulating factor on recovery from radiation-induced hemopoietic aplasia. Exp Hematol. 1993;21(2):338–344. [PubMed] [Google Scholar]
  • 62.Moroni M, Ngudiankama BF, Christensen C, et al. The Gottingen minipig is a model of the hematopoietic acute radiation syndrome: G-colony stimulating factor stimulates hematopoiesis and enhances survival from lethal total-body gamma-irradiation. Int J Radiat Oncol Biol Phys. 2013;86(5):986–992. doi: 10.1016/j.ijrobp.2013.04.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Patchen ML, MacVittie TJ, Williams JL, Schwartz GN, Souza LM. Administration of interleukin-6 stimulates multilineage hematopoiesis and accelerates recovery from radiation-induced hematopoietic depression. Blood. 1991;77(3):472–480. [PubMed] [Google Scholar]
  • 64.Fujiki H, Kimura T, Minamiguchi H, et al. Role of human interleukin-9 as a megakaryocyte potentiator in culture. Exp Hematol. 2002;30(12):1373–1380. doi: 10.1016/s0301-472x(02)00966-9. [DOI] [PubMed] [Google Scholar]
  • 65.Roediger B, Weninger W. Resolving a chronic inflammation mystery. Nat Med. 2017;23(8):914–916. doi: 10.1038/nm.4384. [DOI] [PubMed] [Google Scholar]
  • 66.Broxmeyer HE, Pelus LM, Kim CH, Hangoc G, Cooper S, Hromas R. Synergistic inhibition in vivo of bone marrow myeloid progenitors by myelosuppressive chemokines and chemokine-accelerated recovery of progenitors after treatment of mice with Ara-C. Exp Hematol. 2006;34(8):1069–1077. doi: 10.1016/j.exphem.2006.04.007. [DOI] [PubMed] [Google Scholar]
  • 67.Garraud O, Cognasse F, Tissot JD, et al. Improving platelet transfusion safety: biomedical and technical considerations. Blood Transfus. 2016;14(2):109–122. doi: 10.2450/2015.0042-15. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp info

RESOURCES