THE SYSTEMIC inflammatory response syndrome (SIRS) constitutes the primary host response to a variety of severe clinical insults, such as trauma, burns, pancreatitis, or major surgical interventions.1 Distinct from SIRS, the term sepsis is applied to the clinical state when bacterial infections cause a systemic hyperinflammatory immune response.1 However, this exaggerated host inflammatory response may not lead to an efficient elimination of the infectious agent; rather, it may contribute to a state of immune deactivation that has been termed compensatory anti-inflammatory response syndrome (CARS).2 Concomitantly, CARS may increase susceptibility to secondary infections. The main pathways of the inflammatory circuit during SIRS, sepsis, and CARS are summarized in Fig 1.

Fig. 1.

The main pathways of the inflammatory circuits during SIRS and sepsis are summarized. Exogenous G-CSF (dashed lines) can interrupt a fulminant proinflammatory and anti-inflammatory response by downmodulating the action of proinflammatory TNF- and IL-1, by increasing IL-1ra (and soluble TNFR) secretion, by decreasing IL-8, by improving IL-6 consumption via IL6R (p80), by upregulating HO-1, by improving LPS clearance, and by modulating the incidence of apoptotic hematopoietic cells.

Fig. 1.

The main pathways of the inflammatory circuits during SIRS and sepsis are summarized. Exogenous G-CSF (dashed lines) can interrupt a fulminant proinflammatory and anti-inflammatory response by downmodulating the action of proinflammatory TNF- and IL-1, by increasing IL-1ra (and soluble TNFR) secretion, by decreasing IL-8, by improving IL-6 consumption via IL6R (p80), by upregulating HO-1, by improving LPS clearance, and by modulating the incidence of apoptotic hematopoietic cells.

Close modal

The overall aim of this review is to discuss the current rationale for administering G-CSF to critically ill patients at risk of or with sepsis/SIRS to improve neutrophil function and to modulate the otherwise predestined release of inflammatory mediators. Thus, by direct and indirect effects, G-CSF may prevent the fatal course of sepsis in critically ill patients and promote recovery.

According to the general view, SIRS and sepsis are caused by trauma- or infection-induced overproduction of tumor necrosis factor-α (TNF-α) and interleukin-1 (IL-1).1 Severe sepsis with organ failure may develop when endotoxin (lipopolysaccharide [LPS]) from gram-negative bacteria and/or soluble products of gram-positive bacteria, mostly superantigens, lead to macrophage activation and production of macrophage and T-cell–derived cytokines, including TNF-α, IL-1, interferon-γ (IFN-γ), and IL-12. In turn, these cytokines may synergize with LPS to stimulate the release of nitric oxide (NO) via NOS-2 in a variety of cell types.3-8 

NO has been proposed as the direct mediator of the cardiovascular failure in sepsis9 and septic shock.10 Although NO is produced during sepsis from cytokine-regulated NOS-2, a strong correlation between NO production, organ failure, and an exaggerated proinflammatory response has only been documented in pediatric patients.11 In the majority of adult patients, a clear correlation is more difficult to prove, which may be due to suppression of TNF and IL-1 by anti-inflammatory cytokines, such as by IL-10, which downregulates NO synthase activity and the synthesis of proinflammatory mediators.12 NO synthase inhibition reduced TNF-stimulated IL-8 production in a human endothelial cell line, whereas exogenously added NO enhanced the release of IL-8.13 In addition, NO synthase inhibition blocked IL-8 and IL-6 production in LPS-stimulated human whole blood.14 Conversely, direct exposure of whole blood to NO caused a dose-dependent stimulation of IL-8 but had no effect on IL-6 release. Moreover, the hydroxyl radical scavenger dimethyl sulfoxide (DMSO) prevented NO induction of IL-8, suggesting the participation of the hydroxyl radical in the NO-induced IL-8 production.14 Thus, NO and reactive oxygen intermediates (ROI) may play an essential role in the cytokine pattern in critically ill patients with SIRS and sepsis.

In contrast, counterregulatory Th2 type cytokines, such as IL-415 and transforming growth factor-β (TGF-β),4,16 have been shown to prevent NOS-2 induction. Importantly, the highly reactive peroxynitrite (ONOO) is formed as a reaction product of NO plus ROI (Fig 1). ROI either result from xanthine oxidase activity17 or from neutrophils activated by bacteria or their soluble products. In addition to a number of other effects, peroxynitrite causes microvascular permeability and edema formation, leading to clinical signs of septic shock.18 As a potent oxidant, peroxynitrite reacts with lipids, thiols, DNA, and proteins and may constitute an inducer of apoptosis, as examplified in a study with vascular smooth muscle cells.7,19 

The fatal course of sepsis may be due to systemic immune dysregulation and immunosuppression. In septic mice, apoptosis has been shown to occur primarily in hematopoietic cells, such as T and B lymphocytes, as well as in parenchymal cells, such as bowel, lung, and, to a lesser extent, skeletal muscles and kidneys, but not in liver, brain, or heart.20 Thus, widespread lymphocyte depletion by apoptosis may contribute to lymphopenia observed in patients with sepsis and to decreased elimination of bacteria and bacterial products in sepsis.

Moreover, it has been shown that endothelial cell apoptosis can occur via an oxygen radical-dependent mechanism.21,22Experimental work by Ayala et al23,24 as well as Hotchkiss et al20 shows that systemic apoptosis of the immune system is the most marked feature in experimental animals dying of multiple organ failure during septic shock induced by massive bacterial infection, as already proposed by Bone.2 

In severely ill patients with sepsis, peripheral blood cell-derived TNF-α and IL-1β cytokine production after stimulation with LPS in vitro was transiently downregulated and restored within 72 hours in survivors, but there was no restoration in nonsurvivors.25Thus, persistent downregulation of proinflammatory cytokine production could be related to increased mortality in critically ill patients with sepsis. This downregulation may not result from the induction of tolerance, but through the selective removal of immune cells capable of producing these cytokines. Recent evidence suggests that, in addition, FAS-ligand (FasL) may mediate apoptosis in selected cell populations. Type 1 (Th1) T helper lymphocytes, activated by bacteria, could be subsequently killed after FAS-ligand induction in the same or in a different T-cell population, a process that has been termed activation-induced cell death (AICD).26 AICD predominantly, but not exclusively, occurs in Th1 cells.26,27Whereas T cells exposing FAS are highly sensitive to AICD, FAS-ligand–expressing cells are resistant.26,27 This may lead to a selective survival of Th2 cells and a preponderance of Th2 cells after proinflammatory response during SIRS and sepsis. In turn, predominant depletion of Th1 cells by apoptosis may contribute to the development of CARS. The term CARS is based on the presence of an inactivating and compensatory response to the original inciting event, the proinflammatory response.2,28 This state of immunosuppression also results in increased susceptibility to infection,2 which may further contribute to an infection-induced fatal course of sepsis.

Agents contributing to the anti-inflammatory response include corticosteroids, prostaglandins, IL-4,15IL-10,29 IL-11, IL13, TGF-β,16 soluble receptors to TNF (sTNF-R), and receptor antagonists to IL-1 (IL-1ra)30-33 (Fig 1). The inhibitory Th2 type cytokines IL-4, IL-10, and IL-13 downregulate monocyte activation states after encountering LPS.29,34,35 

To analyze the relative contribution of SIRS alone or in combination with infection, ie, sepsis, ex vivo cytokine production by neutrophils isolated from patients who had undergone cardiac surgery with cardiopulmonary bypass and from patients with sepsis were studied. These stressful conditions related to inflammation, independent of infection, rapidly dampened the reactivity of circulating neutrophils. The release of IL-8 by neutrophils in both groups of patients was significantly reduced whether activated by LPS or by heat-killed streptococci.36 LPS-induced mediators such as IL-10 may be responsible for the observed anergy in these patients. In sepsis, immune deactivation is also mediated by activation of TGF-β via proteases, such as plasmin.37 

Colony-stimulating factors, such as G-CSF and granulocyte-macrophage colony-stimulating factor (GM-CSF), appear to constitute a separate, unique class of mediators. These mediators not only trigger recruitment of progenitors from stem cells, but also modulate adhesion receptor expression as well as upregulate cell protective molecules. In mesenchymal cells such as fibroblasts and mesothelial cells, the G-CSF gene is clearly regulated in a coordinate manner with other genes encoding cytokines such as GM-CSF and IL-6,38-40 ie, these genes are all induced by the same stimuli with similar kinetics. After exposure of cytokines, such as TNF, induced by infections, G-CSF and GM-CSF are coinduced.38,39 In turn, both, G-CSF and GM-CSF may stimulate common effector pathways,41,42 while resulting effects of both cytokines may also differ significantly, as will be discussed in detail below.

In the clinical setting, three considerations are required for the successful treatment of patients with sepsis: (1) to stabilize blood pressure and maintain regional organ and tissue perfusion, (2) to modulate the hyperinflammatory immune response induced by specific (infections; sepsis) or nonspecific (ROI; SIRS) stimulators, and (3) to counteract systemic nonresponsiveness and anergy. The use of biological response modifiers for the treatment of patients with SIRS and sepsis is limited by the fact that the molecular nature of the inducer(s) is not precisely known and is likely not a single common denominator. In addition, differing clinical states have not yet been defined by immunological parameters, and treatment protocols are up to now not directed at a distinct immunological status of the patient.

These limitations become a challenge when biological response modifiers are considered as interventional therapies. Can any one single therapeutic approach help restore homeostasis in sepsis and SIRS regardless of its timing? Clearly, monotherapies such as inhibitors of TNF-α or IL-1 are limited in this regard. The putative goal for anti-inflammatory monotherapies has been to block or eliminate the inducing agent(s) or to modulate the proinflammatory cytokine cascade. The failure to demonstrate any benefit from such treatments43,44 may be due in part to the erroneous assumption that the pathogenesis of sepsis and SIRS in all patients is essentially one of persistent, uncontrolled inflammation driven by the overproduction of a single inflammatory mediator.2 

An alternative approach to the anti-inflammatory monotherapies has been to consider multipotent molecules, such as hematopoietic growth factors, which can address the various immunologic responses simultaneously in the septic patient. One of the hematopoietic growth factors, ie, G-CSF, plays a central role in proliferation, maturation, and functional activation of neutrophils.42,45-50 G-CSF has recently been shown to stimulate host immunity by increasing leukocyte number and by upregulating neutrophil function in postoperative/posttraumatic patients at risk of sepsis (Fig 2).51 IL-6 and G-CSF provide growth and survival signals to progenitors of the granulocyte lineage.52 G-CSF promoted mobilization and survival of CD34+ stem cells in the bone marrow53 and in the peripheral blood.54 This was due, at least in part, to suppression of apoptosis.53,54 Also, incubation of neutrophils from acquired immunodeficiency syndrome (AIDS) patients with G-CSF in vitro significantly decreased apoptosis.55 In patients with acute respiratory distress syndrome (ARDS), neutrophils from bronchoalveolar lavage fluid (BAL) showed only modest signs of apoptosis, presumably due to high amounts of G-CSF and GM-CSF as well as other cytokines like IL-6 in BAL.56 Thus, G-CSF may represent one of several survival factors for neutrophils to inhibit apoptosis during SIRS and sepsis and to preserve neutrophil function.

Fig. 2.

Comparison of the quantitative follow-up of FMLP-induced (10−4 mol/L) oxygen radical production by isolated neutrophils in the postoperative/posttraumatic period between the study group and the control group. Oxygen radical production was assayed by chemiluminescence measurements. Chemiluminescence response is expressed as a percentage of the baseline value, ie, chemiluminescence response at the first postoperative day (100%). In the study group, the baseline value at the first postoperative day represents the chemiluminescence response before infusion of rhG-CSF. Dosages of rhG-CSF administered in the study group are indicated as bars on the x axis. Each point of the curves is the mean ± SEM of the chemiluminescence response of the 10 patients of the study group and of the control group, respectively, at the various days. *P < .05 equals a statistically significant difference between chemiluminescence response at the first postoperative day, ie, baseline value, and the chemiluminescence response during the following days within the study group and within the control group, respectively. +P < .05 equals a statistically significant difference between the chemiluminescence response of the study group and that of the control group at the various postoperative days. (○) Study group; (×) control group. (Modified and reprinted with permission.51)

Fig. 2.

Comparison of the quantitative follow-up of FMLP-induced (10−4 mol/L) oxygen radical production by isolated neutrophils in the postoperative/posttraumatic period between the study group and the control group. Oxygen radical production was assayed by chemiluminescence measurements. Chemiluminescence response is expressed as a percentage of the baseline value, ie, chemiluminescence response at the first postoperative day (100%). In the study group, the baseline value at the first postoperative day represents the chemiluminescence response before infusion of rhG-CSF. Dosages of rhG-CSF administered in the study group are indicated as bars on the x axis. Each point of the curves is the mean ± SEM of the chemiluminescence response of the 10 patients of the study group and of the control group, respectively, at the various days. *P < .05 equals a statistically significant difference between chemiluminescence response at the first postoperative day, ie, baseline value, and the chemiluminescence response during the following days within the study group and within the control group, respectively. +P < .05 equals a statistically significant difference between the chemiluminescence response of the study group and that of the control group at the various postoperative days. (○) Study group; (×) control group. (Modified and reprinted with permission.51)

Close modal

Besides these direct effects on granulocytes, G-CSF has been shown to exert direct and indirect effects on other immunocompetent cells. Direct effects are mediated by cells that express G-CSF receptors (G-CSFR). G-CSFR were clearly detected by flow cytometry on adult human peripheral granulocytes and monocytes, but not on lymphocytes.57 Administered prophylactically, direct and indirect effects of G-CSF might block the hyperinflammatory cytokine cascade mediated by proinflammatory cytokines before culminating in severe tissue damage. Moreover, these effects might help to restore immune dysfunction and dysbalance in critically ill patients. The addition of G-CSF promoted an anti-inflammatory response pattern in several usually proinflammatory test systems. For example, T cells of G-CSF–treated mice predominantly produced anti-inflammatory type-2 cytokines (IL-4 and IL-10).58 Moreover, G-CSF administration to human volunteers increased Th2 type cytokines in LPS-stimulated ex vivo blood cultures.59Reduced Th1 cytokine and increased Th2 cytokine expression are expected to downmodulate NOS-2 activation, as schematically demonstrated in Fig 1.

As summarized in Fig 1, TNF-α can stimulate IL-1 secretion in tissue macrophages, and this cytokine in turn can activate IL-1ra production to counteract IL-1–mediated inflammatory cascades.60Exogenous G-CSF (dashed lines in Fig 1) rapidly stimulates IL-1ra production.59,61,62 The autocrine cytokine cascade of inflammatory mediators could thus be interrupted.

Moreover, G-CSF promotes IL-6 use via increased expression and shedding of the IL-6 receptor (IL-6R, p80), as shown independently in two patient populations.63,64 Indeed, IL-6 and G-CSF may act similarly by stimulating intracellular pathways, such as phosphorylation of STAT3 transcription factor.65-69 G-CSF and IL-6 cooperate and can even substitute for each other in recruitment of myeloid cells in G-CSFR and IL-6 knock out mouse models.52 G-CSF and IL-6 may act in a dual role to amplify inflammatory responses when the inciting pathogen is present within the host and to downmodulate the response when the pathogen has been eradicated.68 

Bacterial endotoxin stimulates macrophages to produce proinflammatory cytokines, such as TNF-α and IL-1, which in turn stimulate fibroblasts and endothelial cells to produce G-CSF.70 Dunn et al71 identified regulatory elements in the G-CSF promotor activated by NF-IL-6 and NF-κB, and Smith et al72 reported binding for the transcription factors PU.1 and C/EBP-α. Consequently, TNF-α synthesizing monocytes regulate G-CSF production by autocrine effects that are probably different from endothelial cells and fibroblasts. Inducers of G-CSF release may be distinct in different cell types, ie, macrophages, fibroblasts, and endothelial cells.

G-CSF serum concentrations have been reported to be less than 30 pg/mL in most healthy persons73 and rarely exceed concentrations greater than 170 pg/mL70,73-76(Table 1). Early during active infections, such as pneumonia, cholecystitis, or urinary tract infection, and sepsis, serum concentrations of G-CSF may increase rapidly to more than 50,000 pg/mL,70,74,77-80 but rapidly decrease due to clearance through binding and use. After an infectious episode, G-CSF serum concentrations returned to levels less than 200 pg/mL.70,74,79,80 

Table 1.

G-CSF Serum Concentrations During Infection and Sepsis

G-CSF Serum Concentrations (pg/mL)Reference
HealthyInfection/Sepsis
During Upon Resolution
<100  3,199  <100  74  
<170  4,326  <50 70  
<78    75  
 863   77  
 11,478  78  
 >50,000  <200  80  
<163   73  
<83    76 
G-CSF Serum Concentrations (pg/mL)Reference
HealthyInfection/Sepsis
During Upon Resolution
<100  3,199  <100  74  
<170  4,326  <50 70  
<78    75  
 863   77  
 11,478  78  
 >50,000  <200  80  
<163   73  
<83    76 

In recent years, G-CSF has been demonstrated to be effective and safe in reducing the incidence of infection in high-risk patients, ie, after myelosuppressive anticancer chemotherapy,45,81,82 as well as in neutropenic and agranulocytotic patients.82-84 Its effectiveness is due in part to G-CSF’s effects on increasing neutrophil count and functional receptors, such as G-CSFR as well as monomeric Fc receptors. Moreover, G-CSF might be beneficial in nonneutropenic patients with SIRS, infections, and sepsis with impaired neutrophil function. In these nonneutropenic patients, high concentrations of proinflammatory cytokines, such as TNF-α and IL-1, may cause accumulation of ROI in granulocytic effectors, thereby causing increased apoptosis of neutrophils,85 inefficient phagocytic function, and thus a state of immune dysfunction.

Secondary phagocyte defects and reductions in neutrophil numbers and functional impairment have been documented in a variety of clinical states. Patients with impaired neutrophil function are at increased risk to develop sepsis and to progress into septic shock and multiple organ failure.86,87 Preexisting immunosuppression due to the underlying disease has been reported in patients with diabetes,88,89 acute necrotizing pancreatitis,90 or alcoholism.91 Acquired immunosuppression and impairment of neutrophil function may result from previous medication (immunosuppressive and cytostatic drugs), operative trauma,92-94 blood transfusions,95 anesthetics, hypnotics, and sedatives96,97 as well as sympathomimetics.98 Migration to an infectious focus, ingestion, and killing of bacteria by neutrophils from critically ill patients with SIRS and sepsis has been reported to be significantly compromised.99-103 

However, boosting numbers and functions of granulocytes, macrophages, and lymphocytes is a prerequisite for withstanding microorganism invasion. Endogenous and exogenous G-CSF may be crucial for host defense in patients with SIRS and sepsis to avoid progression into severe sepsis with organ failure and septic shock by improving neutrophil functions and inhibiting neutrophil apoptosis.23,24,55 In addition to stimulating the proliferation and maturation of neutrophils,45 G-CSF enhances chemotaxis,46 phagocytic activity,47bactericidal function,47 respiratory burst,42,49 and antibody-dependent cellular cytotoxicity (ADCC)48 of isolated human neutrophils. Administration of G-CSF acts synergistically with antibiotics and improves outcome of severe infections.104-106 

G-CSF administration improved survival in several animal models of sepsis,105-108 even when applied therapeutically after the onset of sepsis105 (Fig 3). These animal data imply that stimulation of neutrophil function and viability represents a major goal for treating patients with ongoing infections.

Fig. 3.

Cumulative mortality in septic rats treated with G-CSF at the induction of sepsis or after 4 hours. *P < .05; logrank test, n = 24 in all groups. In rats, polymycrobial peritonitis was induced by a cecal perforation and 10 μg/kg recombinant human G-CSF was administered intravenously every 12 hours, with the first dose at sepsis induction or 4 hours postinduction. (Modified and reprinted with permission.169)

Fig. 3.

Cumulative mortality in septic rats treated with G-CSF at the induction of sepsis or after 4 hours. *P < .05; logrank test, n = 24 in all groups. In rats, polymycrobial peritonitis was induced by a cecal perforation and 10 μg/kg recombinant human G-CSF was administered intravenously every 12 hours, with the first dose at sepsis induction or 4 hours postinduction. (Modified and reprinted with permission.169)

Close modal

There is now increasing evidence that an inappropriate endogenous G-CSF response may be associated with an adverse outcome to sepsis. For example, low serum G-CSF concentrations (0 to 125 pg/mL) on admission have been shown to be associated with fatal outcome in patients with acute bacterial infections.78 

In patients undergoing a successful response to infections, G-CSF serum concentrations should be high immediately and decrease consecutively, when G-CSF is used and neutrophil counts increase. In patients not responding successfully to infections, serum G-CSF concentrations will remain high, but neutrophil counts would not increase. Similar patterns are expected to occur when patients are treated with exogenous G-CSF.

In patients with bacteremia80 or at the time sepsis was diagnosed,79 endogenous G-CSF concentrations were found to be markedly increased in all patients, both survivors and nonsurvivors. However, concentrations significantly decreased with recovery in the survivors, but remained increased in the nonsurvivors. In the same study, the levels of G-CSF, IL-6, and, to some extent, TNF-α rapidly decreased in nonfatal bacteremic infections within the first 24 hours; however, concentrations remained increased or showed a more gradual decrease in the nonsurvivors within 18 to 96 hours.80 In a different study, application of exogenous rhG-CSF in 19 patients with granulocytopenia complicated by sepsis increased granulocyte counts and improved survival when G-CSF serum concentrations decreased.109 However, in the five patients with persistently high plasma G-CSF levels, leukocyte counts were not increased and the patients died.109 Thus, either early increased endogenous production or enhanced use of administered G-CSF (ie, G-CSF responsiveness) correlated with the patients’ successful outcome.

Thus, G-CSF responsiveness has to be monitored by leukocyte counts, G-CSF serum concentrations, and G-CSF–responsive gene products (see below) to ascertain successful treatment with exogenous G-CSF.

In a phase II study in patients at risk of or with sepsis, we successfully identified patients who could and could not respond to exogenous G-CSF administration. In responding patients, an increase in G-CSF serum concentrations after exogenous application of rhG-CSF was followed by an immediate decrease and a rapid increase of neutrophil counts and CD64 expression.61 In nonresponders, in which G-CSF substitution did not increase leukocyte counts, molecular mechanisms have to be defined. Impaired G-CSFR expression may contribute to nonresponsiveness. Whereas G-CSF induced increased G-CSFR expression,110 GM-CSF, TNF, LPS, f-met-leu-phe (fMLP), phorbol ester (TPA), and C5a were shown to downregulate the neutrophil G-CSFR in vitro.111 Binding assays using 125I-labeled rG-CSF showed that the number of rG-CSF binding sites on the surface of neutrophil progenitor cells is regulated by cytoplasmic cAMP and protein kinase A.112 Rapid internalization of G-CSFRs111 may mask the amount of surface expression of G-CSFRs detected under G-CSF–responsive conditions. These conditions explain results showing reduced G-CSFRs in G-CSF–treated cell lines and neutrophils.113 Differential G-CSF responsiveness may ultimately explain the failure to demonstrate beneficial effects on nosocomial infections in a recently performed study in patients with acute traumatic brain injury or cerebral hemorrhage prophylactically treated with G-CSF.114 

Prophylactic administration with G-CSF appears to be superior to an endogenous immune response to fight infection in critically ill patients. During an infection, both G-CSF and GM-CSF are produced.38,39 Both cytokines induce leukocyte maturation and activation.41,42 In contrast to G-CSF, GM-CSF may cause serious side effects when applied exogenously and thus independently of G-CSF.45,115 

G-CSF inhibited leukotriene E4(LTE4) formation in humans in vivo116 and potently reduced the activity of NOS-2 in peritoneal macrophages.117 In contrast, GM-CSF upregulated NOS-2118-122 and enhanced LTB4 generation of neutrophils.123-125 McDonald et al126 showed that IL-8 may be responsible for the synthesis of LTB4 and platelet-activating factor after GM-CSF treatment. Thereby, the combined presence of IL-8 and of GM-CSF at inflammatory foci may contribute to the amplification of the inflammatory response. GM-CSF stimulated adhesion receptor expression, such as intercellular adhesion molecule-1 (ICAM-1).127 However, G-CSF downregulated ICAM-1 on IL-1–stimulated human endothelial cells.128Yong129 further showed that G-CSF, but not GM-CSF, promotes transendothelial migration of neutrophils. ROI released by adherent neutrophils may cause capillary leakage and lung toxicity, which occurred in patients with sepsis treated with GM-CSF.45,115 Massive IL-8 is considered to indicate the amount of ROI induced in vitro.13,14 

In contrast to GM-CSF, G-CSF is expected to downmodulate the proinflammatory immune response, which otherwise might result in severe tissue damage and organ failure. This rationale favors the administration of G-CSF to critically ill patients at risk of sepsis and before other proinflammatory cytokines are systemically present.

Functional antigens on neutrophils, such as CD64, CD32, CD16, and CD11b, defined by specific clusters of differentiation (CD), can be easily monitored by flow cytometry. rhG-CSF reconstituted and increased functional antigen CD64 (monomeric Fc receptor; FcγRI) expression in postoperative/posttraumatic patients at risk of or with sepsis,61 and the same increase of CD64 expression has been described on immature and mature neutrophils in normals and patients with malignancies130-132 (Table2A). Functionally, exogenous G-CSF enhanced ADCC in patients with advanced squamous cell carcinoma of the oral cavity and pharynx through FcγRI (CD64).130 Also, expression of FcγRII (CD32) increased when G-CSF was administered (Table2B).132,133 Functionally, G-CSF enabled neutrophils to kill hybridoma cell lines through FcγRII (CD32).134 FcγRIII (CD16) expression was upregulated by exogenous G-CSF in healthy volunteers75 and in patients with paroxysmal nocturnal hemoglobinuria,135 but did not change61,133 and even decreased131,132 in others (Table 2C). In human newborn infants with presumed bacterial sepsis, functional activation of neutrophils (C3bi = CD11b expression) was determined 24 hours after the application of 10 μg/kg·d of rhG-CSF.136 

Table 2.

Effects of G-CSF on the Expression of Functional Surface Markers of Neutrophils (Fc Receptors)

A. CDReceptor Effect of G-CSF on Expression of CD64, FcR Type I Subjects, Clinical Background, Comments Reference
CD64  FcγRI  ↑  With chemotherapy + autologous bone marrow transplantation  132  
CD64  FcγRI  ↑  Mature neutrophils of healthy volunteers, G-CSF in vitro  132  
CD64 FcγRI  ↑  With advanced squamous cell carcinoma, ↑ ADCC 130  
CD64  FcγRI  ↑  With cytotoxic chemotherapy + malignancies, healthy volunteers  131  
CD64 FcγRI  ↑  Postoperative/posttraumatic at risk of or with sepsis  61  
B. CD  Receptor Effect of G-CSF on Expression of CD32, FcR Type II Subjects, Clinical Background, Comments  Reference 
CD32  FcγRII  ↑  ↑ killing of hybridoma cell lines, application of G-CSF in vitro  134  
CD32  FcγRII  — Healthy volunteers  137  
CD32  FcγRII  —  With cytotoxic chemotherapy + malignancies, healthy volunteers  131 
CD32  FcγRII  ↑  With chemotherapy + autologous bone marrow transplantation  132  
CD32  FcγRII  ↑  With non-Hodgkin lymphoma  133  
CD32  FcγRII  ↑-↓ Postoperative/posttraumatic at risk of or with sepsis  61 
C. CD  Receptor  Effect of G-CSF on Expression of CD16, FcR Type III  Subjects, Clinical Background, Comments  Reference  
CD16 FcγRIII  ↑  Healthy volunteers  75  
CD16 FcγRIII  ↑  With paroxysmal nocturnal haemoglobinuria 135  
CD16  FcγRIII  ↓  With cytotoxic chemotherapy + malignancies, healthy volunteers  131  
CD16 FcγRIII  ↓  With chemotherapy + autologous bone marrow transplantation  132  
CD16  FcγRIII  —  With non-Hodgkin lymphoma  133  
CD16  FcγRIII  — Postoperative/posttraumatic at risk of or with sepsis 61 
A. CDReceptor Effect of G-CSF on Expression of CD64, FcR Type I Subjects, Clinical Background, Comments Reference
CD64  FcγRI  ↑  With chemotherapy + autologous bone marrow transplantation  132  
CD64  FcγRI  ↑  Mature neutrophils of healthy volunteers, G-CSF in vitro  132  
CD64 FcγRI  ↑  With advanced squamous cell carcinoma, ↑ ADCC 130  
CD64  FcγRI  ↑  With cytotoxic chemotherapy + malignancies, healthy volunteers  131  
CD64 FcγRI  ↑  Postoperative/posttraumatic at risk of or with sepsis  61  
B. CD  Receptor Effect of G-CSF on Expression of CD32, FcR Type II Subjects, Clinical Background, Comments  Reference 
CD32  FcγRII  ↑  ↑ killing of hybridoma cell lines, application of G-CSF in vitro  134  
CD32  FcγRII  — Healthy volunteers  137  
CD32  FcγRII  —  With cytotoxic chemotherapy + malignancies, healthy volunteers  131 
CD32  FcγRII  ↑  With chemotherapy + autologous bone marrow transplantation  132  
CD32  FcγRII  ↑  With non-Hodgkin lymphoma  133  
CD32  FcγRII  ↑-↓ Postoperative/posttraumatic at risk of or with sepsis  61 
C. CD  Receptor  Effect of G-CSF on Expression of CD16, FcR Type III  Subjects, Clinical Background, Comments  Reference  
CD16 FcγRIII  ↑  Healthy volunteers  75  
CD16 FcγRIII  ↑  With paroxysmal nocturnal haemoglobinuria 135  
CD16  FcγRIII  ↓  With cytotoxic chemotherapy + malignancies, healthy volunteers  131  
CD16 FcγRIII  ↓  With chemotherapy + autologous bone marrow transplantation  132  
CD16  FcγRIII  —  With non-Hodgkin lymphoma  133  
CD16  FcγRIII  — Postoperative/posttraumatic at risk of or with sepsis 61 

Abbreviations: ↑, increase; —, no effect; ↓, decrease.

Membrane-bound CD14 (mCD14) is a relevant surface antigen for effective endotoxin clearance. An enhanced expression of CD14 on neutrophils has been reported in healthy individuals after subcutaneous administration of G-CSF.131,137 Improved re-expression of mCD14 has been observed in G-CSF–treated postoperative/posttraumatic patients.64 The effects of G-CSF on mCD14 might explain a better LPS clearance.138 

The events leading to the upregulation of functional antigens by endogenous and exogenous G-CSF are explained in part by STAT1 and STAT3 phosphorylation65 and by transcription factors PU.1 and C/EBP-α72 activated through the G-CSF receptor. Both transcription factors play an important role in differentiation (PU.1) and induction of regulatory genes, induced by increased cytoplasmic cAMP112,139 and protein kinase A activation,112including the G-CSFR itself.112 These molecular events are relevant to understand specific as well as promiscous effects by G-CSF. Clearly, CD64 appears to be a highly valid marker to follow responsiveness to endogenous and exogenous G-CSF.

Effects of exogenous G-CSF on the cytokine response pattern.

There is some concern that G-CSF–induced increases in neutrophil count and function might aggravate inflammatory responses and thus be harmful for the host. This concern is based on the functional properties of G-CSF to stimulate phospholipase A241 and to stimulate oxidative burst formation in granulocytes.42 On the other hand, these properties are most relevant to control infection and to attenuate or even prevent an otherwise progressive immune dysregulation. Direct effects (via G-CSF receptors on granulocytes and monocytes, expression of CD64, increase in neutrophil counts, and priming of neutrophil function) and indirect effects of G-CSF (clearance of endotoxin and bacteria and counterregulation of induction of proinflammatory cytokines) can prevent nonspecific tissue damage, especially by preserving endothelial cell function. Endothelial cell damage is expected to occur through systemically high TNF-α, IL-1, or IL-8 (Fig 1). Clearly, IL-8 is a cytokine released upon oxidative stress by almost every cell type.13,14 G-CSF treatment attenuates circulating IL-8.61 Further support for this concept of achieving less oxidative stress by G-CSF is provided in vivo by animal studies, when exogenous G-CSF improved rather than worsened organ function and survival in models using LPS- or live bacteria-induced proinflammatory response and disease.140Exogenous G-CSF increased microvascular flow and survival rate and prevented neutrophil-mediated tissue damage in fulminant intra-abdominal sepsis in rats, at least in part, by downmodulating the potent vasoconstrictor endothelin-1.105 Hemoconcentration and lactic acidosis were also attenuated, which is consistent with reduced endothelial damage and plasma leakage.105 

Table 3 summarizes the inflammatory cytokine response pattern resulting from application of exogenous G-CSF to animals.105,108,140-144 The relative contribution of direct (via G-CSF receptors on monocytes) and indirect effects (better control of infection, T cells) of G-CSF to this pattern has yet to be clarified. Clearly, G-CSF is a member of the inflammatory cytokine cascade and displays important regulatory properties by attenuating the proinflammatory cytokine response that would otherwise predominate.

Table 3.

Effects of G-CSF on Bacterial Products, Cytokines, and Survival Administered Before Induction of Sepsis in Animals

Animal Sepsis ModelEffects of G-CSF on Reference
Endotoxin Levels Bacterial Counts TNF-αInterleukins Survival
Mouse/rat  LPS-induced septic shock    ↓   ↑  108  
Canine Intraperitoneal E coli clot  ↓  ↓  ↓  ↑  141  
Rat  Fecal inoculum    ↓  ↑  142  
Porcine  Pseudomonas sepsis  ↓  ↓   ↑  143  
Rat  Cecal perforation  ↓ ↓  ↓   ↑  105  
Mouse  Superantigen mediated shock     ↓ IL-2  ↑  140  
Mouse LPS-induced septic shock    —  ↓ IL-1β  ↑ 144  
     ↓ IFN-γ 
     ↓ IL-10 
Animal Sepsis ModelEffects of G-CSF on Reference
Endotoxin Levels Bacterial Counts TNF-αInterleukins Survival
Mouse/rat  LPS-induced septic shock    ↓   ↑  108  
Canine Intraperitoneal E coli clot  ↓  ↓  ↓  ↑  141  
Rat  Fecal inoculum    ↓  ↑  142  
Porcine  Pseudomonas sepsis  ↓  ↓   ↑  143  
Rat  Cecal perforation  ↓ ↓  ↓   ↑  105  
Mouse  Superantigen mediated shock     ↓ IL-2  ↑  140  
Mouse LPS-induced septic shock    —  ↓ IL-1β  ↑ 144  
     ↓ IFN-γ 
     ↓ IL-10 

Abbreviations: ↑, increase; −, no effect; ↓, decrease.

Another secondary feature of exogenous G-CSF administration is its effects on T cells. In mice, G-CSF has been shown to polarize T-cell subpopulations towards an increased anti-inflammatory type-2 cytokine response (characterized by the cytokines IL-4 and IL-10) and a reduced proinflammatory type-1 cytokine production (IL-2 and IFN-γ)58 (Table 4). In a similar pattern, G-CSF administration altered serum concentrations of cytokines in healthy volunteers145 and ex vivo cytokine production of whole blood in response to gram-positive and gram-negative bacterial products,59,145 respectively, in that more anti-inflammatory and less proinflammatory cytokines were released (Table 4). Although, G-CSF administration to healthy volunteers caused a reproducible increase of proinflammatory mediators, such as TNF-α, after ex vivo and in vivo stimulation with LPS, the increases of counterregulatory and anti-inflammatory cytokines, such as the soluble p55 and p75 TNF receptors (sTNF-Rs) and IL-1ra, were significantly higher.62,145 Similarly, Hartung et al59 found more anti-inflammatory cytokines, such as IL-1ra, sTNF-R-type I and -type II, and IL-10, but less proinflammatory cytokines, such as TNF and IFN-γ, in blood cultures stimulated with bacterial products from volunteers treated with 480 μg G-CSF applied subcutaneously.

Table 4.

Comparison of the Effects of G-CSF on Proinflammatory and Anti-Inflammatory Cytokine Patterns

Effect of G-CSF on the ModelReference
Proinflammatory Cytokine PatternAnti-Inflammatory Cytokine Pattern
↓ IL-2, IFN-γ ↑ IL-4  Mice, T-cell cytokine production  58  
↑ TNF-α  ↑ p55 and p75 sTNF-Rs, IL-1ra Healthy volunteers, serum concentrations  145  
— IL-1β  — IL-6  
↑ TNF-α ↑↑ p55 and p75 sTNF-Rs ↑↑ IL-1ra  Ex vivo whole blood cytokine production  145  
↓ TNF, IFN-γ, GM-CSF  ↑ p55 and p75 sTNF-Rs ↑ IL-1ra, IL-10  Healthy volunteers, ex vivo whole blood cytokine production  59  
↑ TNF, IL-8  ↑ sTNF-Rs  Healthy volunteers, LPS IV  62  
 ↑ IL-1ra ↑ IL-6  G-CSF pretreatment IV 2 hours before LPS  
− TNF ↑ sTNF-Rs  G-CSF pretreatment SC 24 hours before LPS  62 
↓ IL-8  — Il-1ra  
 — IL-6 
Effect of G-CSF on the ModelReference
Proinflammatory Cytokine PatternAnti-Inflammatory Cytokine Pattern
↓ IL-2, IFN-γ ↑ IL-4  Mice, T-cell cytokine production  58  
↑ TNF-α  ↑ p55 and p75 sTNF-Rs, IL-1ra Healthy volunteers, serum concentrations  145  
— IL-1β  — IL-6  
↑ TNF-α ↑↑ p55 and p75 sTNF-Rs ↑↑ IL-1ra  Ex vivo whole blood cytokine production  145  
↓ TNF, IFN-γ, GM-CSF  ↑ p55 and p75 sTNF-Rs ↑ IL-1ra, IL-10  Healthy volunteers, ex vivo whole blood cytokine production  59  
↑ TNF, IL-8  ↑ sTNF-Rs  Healthy volunteers, LPS IV  62  
 ↑ IL-1ra ↑ IL-6  G-CSF pretreatment IV 2 hours before LPS  
− TNF ↑ sTNF-Rs  G-CSF pretreatment SC 24 hours before LPS  62 
↓ IL-8  — Il-1ra  
 — IL-6 

Abbreviations: ↑↑, strong increase; ↑, increase; —, no effect; ↓, decrease; IV, intravenously; SC, subcutaneously.

G-CSF exerts differential effects on cytokine release in human endotoxemia when administered either intravenously forn 2 hours or subcutaneously for 24 hours before endotoxin.62 When administered 2 hours before endotoxin, G-CSF actually augmented an LPS-induced inflammatory cytokine response, whereas when administered 24 hours before LPS challenge, G-CSF attenuated the LPS-induced proinflammatory state and caused significant downregulation of inflammation (Table 4).62 A direct explanation implies the involvement of a G-CSF–induced transcription of gene product(s) that functions to regulate the proinflammatory effects induced by endotoxin. If G-CSF causes modulation of LPS-receptor expression, such as CD14, the anti-inflammatory gene product(s) may be either involved in endotoxin removal or in neutralizing oxidative stress.62Further experimental work is necessary to understand the regulatory pathways induced by G-CSF in the absence of a proinflammatory stimulus. Results are fundamental to work out a prophylactic G-CSF treatment schedule applicable to patients at risk of sepsis.

Modulation of adhesion receptors of neutrophils and the endothelium by G-CSF.

Adhesion molecules, such as selectins, regulate the primary binding of neutrophils to the endothelium and β2-integrins are necessary for subsequent transendothelial migration.146-149G-CSF may decrease selectin expression and diminish binding to and enhance transmigration of neutrophils through the endothelium. Therefore, G-CSF may prevent endothelial damage by avoiding adhesion of activated neutrophils to inflamed vascular endothelium.

Downregulation of L-selectin by G-CSF may be beneficial during distinct pathological inflammatory responses, which are associated with the adhesion of hyperactivated neutrophils to the endothelium. L-selectin–deficient mice were resistant to LPS-induced toxic shock and death, because neutrophil, lymphocyte, and monocyte migration to the peritoneum in response to an inflammatory stimulus was significantly inhibited.150 In healthy volunteers, exogenous G-CSF reversibly downregulated surface expression of L-selectin (synonymous [syn.]: leukocyte adhesion molecule-1 [LAM-1]) on human neutrophils151 and a specific surface metalloproteinase, called L-selectin sheddase, may be involved.152 In 11 postoperative/posttraumatic patients at risk of or with sepsis and treated with rhG-CSF, L-selectin expression on granulocytes was downregulated within 24 to 48 hours.61 

Increased plasma concentrations of soluble E-selectin, indicating endothelial damage, were closely associated with multiple organ dysfunction and death in patients with SIRS.153 Patient survival was zero when soluble E-selectin levels were greater than 30 U/mL.153 Exogenous G-CSF may have been protective to the endothelium, because in postoperative/posttraumatic patients at risk of or with sepsis, soluble E-selectin showed a sharp increase when rhG-CSF was reduced from 1 to 0.5 μg/kg·d.154 

The intercellular adhesion molecule (ICAM-1; CD54) is expressed on the surface of inflamed endothelial cells and plays an important role in specific binding for transmigration of activated granulocytes.147-149 An increase in soluble ICAM (sICAM) may also be indicative of endothelial damage. In this context, it has been reported that sICAM-1 serum concentrations were increased in adult patients with sepsis and corresponded with severity of disease, with subsequent organ failure, and possibly also with outcome.155 In postoperative/posttraumatic patients at risk of or with sepsis,61 sICAM concentrations were relatively stable during the infusion of rhG-CSF at 1 μg/kg·d; however, levels increased when rhG-CSF was tapered to 0.5 μg/kg·d and after cessation of rhG-CSF. Nethertheless, extrapolating sICAM concentrations to endothelial damage remains controversial, because ICAM-1 is also expressed and can be shed from monocytes.156 

Despite upregulation of CD11b/18, G-CSF had no effect on neutrophil adhesion but was a powerful stimulator of transmigration.129 In whole blood cultures, a time- and G-CSF concentration-dependent upregulation of the expression of CD11b has been observed.75 In healthy volunteers, the nadir in circulating neutrophils occurred 30 minutes postinjection of 300 μg G-CSF subcutaneously and coincided with significant upregulation of the expression of CD11b on circulating neutrophils.75 This effect was short-lasting in that CD11b was maximally expressed at 2 hours postinjection and returned to baseline levels at 4 hours postinjection.75 

Taken together, diminished but not abolished binding of preactivated neutrophils to endothelial cells and enhanced transmigration, induced by exogenous rhG-CSF, might effectively protect the endothelium from injury by hyperactivated neutrophils.

The encouraging results in nonneutropenic patients have supported expanding the application of G-CSF into nonneutropenic, critically ill patients with secondary functional impairment of neutrophils.

G-CSF should benefit the course of disease in patients with pneumonia and subsequent organ dysfunction (Table 5). The neutrophil has been strongly implicated in the pathogenesis of inflammatory lung injury,50 and there has been theoretical concern that G-CSF and neutrophil activation might exacerbate lung injury. Exogenous G-CSF clearly worsened lung injury and survival when applied during very severe pulmonary infection in rats.50In other studies in which lung injury was exacerbated in G-CSF–treated animals and humans, a beneficial survival effect remained significant (Table 5).50,157,158 In ethanol-treated rats with experimentally induced pneumonia, all control animals, but less than 10 % of the G-CSF–treated rats died.159 Also, exogenous G-CSF improved survival in models in which pneumonia was induced in aminals by various microorganisms, such as Klebsiella pneumoniae,159 Streptococcus pneumoniae,160,161and Pseudomonas aeruginosa.162 In a canine model of peritonitis and septic shock, G-CSF application did not aggravate sepsis-related pulmonary dysfunction despite increases in circulating as well as lung lavage neutrophils.141 In mice, exogenous rhG-CSF has been shown to decrease LPS-induced pulmonary edema and alveolar capillary leakage.140 In a canine model of peritonitis and septic shock, exogenous G-CSF did not aggravate sepsis-related pulmonary dysfunction despite increased circulating as well as lung lavage neutrophils.141 

Table 5.

Comparison of the Beneficial and Harmful Effects of G-CSF on Infectious Lung Injury in Patients

Effect of G-CSF on Infectious Lung Injury Effect of G-CSF on Clinical Parameters Model Reference
↑  +  One neutropenic, immunosuppressed patient with adult respiratory distress syndrome  157  
↑  −  3/9 patients with drug-induced agranulocytosis with pre-existing life-threatening infection  158  
↓  +  One patient with adult respiratory distress syndrome during chemotherapy  163  
↓  +  One patient with alcoholism, leukopenia, and pneumococcal sepsis  91 
∅︀  +  20 postoperative/posttraumatic patients at risk of or with sepsis  165  
∅︀  ∅︀  44 patients with acute traumatic brain injury or cerebral hemorrhage  114  
↓  756 hospitalized patients with community-acquired pneumonia 164 
Effect of G-CSF on Infectious Lung Injury Effect of G-CSF on Clinical Parameters Model Reference
↑  +  One neutropenic, immunosuppressed patient with adult respiratory distress syndrome  157  
↑  −  3/9 patients with drug-induced agranulocytosis with pre-existing life-threatening infection  158  
↓  +  One patient with adult respiratory distress syndrome during chemotherapy  163  
↓  +  One patient with alcoholism, leukopenia, and pneumococcal sepsis  91 
∅︀  +  20 postoperative/posttraumatic patients at risk of or with sepsis  165  
∅︀  ∅︀  44 patients with acute traumatic brain injury or cerebral hemorrhage  114  
↓  756 hospitalized patients with community-acquired pneumonia 164 

Abbreviations: ↑, harmful; ∅︀, no effect; ↓, beneficial; +, improved; −, worsened.

In human volunteers, rhG-CSF pretreatment prevented the accumulation of neutrophils in the lung during the first 2 hours after endotoxin administration.62 This prevention occurred in the presence of increased CD11b and CD18 expression on the neutrophils, ie, under otherwise proadhesive conditions.62 In neutropenic patients, G-CSF application predominantly resulted in an improvement in pulmonary function.91,157,158,163 G-CSF has also been proven to benefit nonneutropenic patients with pneumonia and subsequent organ dysfunction. In a study involving 756 hospitalized patients with community-acquired pneumonia, rhG-CSF accelerated clearance of pulmonary infiltrates and reduced the occurrence of serious complications: empyema, ARDS, disseminated intravascular coagulation, and septic shock.164 The frequency of nosocomial pneumonia was higher in patients with acute traumatic brain injury or cerebral hemorrhage prophylactically receiving 300 μg/d rhG-CSF (4/14) than in the controls (2/17); however, the difference with respect to the control group was not statistically significant.114 For several reasons, this study was underpowered to detect a difference and thus not ideal to evaluate a potential benefit or adverse effect from the prophylactic application of G-CSF in patients with high risk for nosocomial infections. In a different study with a small number of postoperative/posttraumatic patients at risk of or with sepsis, the application of rhG-CSF did not affect pulmonary function.165 Combined with the molecular events discussed above, G-CSF is unlikely to promote nonspecific adhesion of activated neutrophils to inflamed tissues and thus cause tissue damage as long as other proinflammatory mediators such as TNF-α, IL-1, and GM-CSF are low or absent.

The present studies in humans and animals stress the importance of the appropriate timing, dosage, and patient selection for a prophylactic treatment protocol with G-CSF in pneumonia. G-CSF prophylactically administered after hemorrhage in mice improved survival from pneumonia due to Pseudomonas aeruginosa; however, the protective effect was highly dependent on the dosing schedule used.162 In a nonneutropenic infection model of Streptococcus pneumoniaepulmonary infection, rhG-CSF improved lung clearance in both splenectomized and sham-operated mice compared with controls; however, rhG-CSF improved survival in the splenectomized mice but not in the sham-operated mice.160 In ethanol-fed rats, G-CSF was unable to provide protection against fatal Pneumococcal pneumonia despite increasing the numbers of circulating neutrophils.161 

Recently, a dramatic increase in mortality has been reported when mice were pretreated with G-CSF before induction of pneumonia withKlebsiella pneumoniae.166 In vitro, G-CSF increased capsular polysaccharide (a bacterial virulence factor) production ofKlebsiella pneumoniae, resulting in impaired phagocytic uptake and killing by neutrophils.166 Hemorrhagic shock in rats was accompanied by acute lung injury with neutrophil infiltration and an increase of G-CSF mRNA levels in the lung produced mainly by bronchial epithelial cells.167 G-CSF instillation into rat lungs mediated neutrophil recruitment, pulmonary edema, and hypoxia, indicating that local production of G-CSF may be involved in lung damage and ARDS.168 

Thus, depending on certain circumstances, application of G-CSF for prophylaxis and therapy of pneumonia may be beneficial or detrimental. Appropriate dosage, timing, and indication are not yet clearly defined to gain maximal beneficial effects with minimal side effects in patients. In summary, recent studies114,164,166,168indicate the need for additional controlled studies to define the role of G-CSF in community-aquired and nosocomial pneumonia.

At present, exogenous G-CSF is widely used and has been proven to be effective and safe in reducing the incidence of infection and sepsis in immunocompromised patients with nonmyeloid tumors after myelosuppressive anticancer chemotherapy45,81,82 as well as in neutropenic and agranulocytotic patients.82-84 

In human newborn infants with presumed bacterial sepsis, application of G-CSF significantly increased C3bi (CD11b) expression of their neutrophils, indicating functional activation.136 In nonneutropenic, posttraumatic/postoperative patients with a high risk of sepsis, none of the patients treated with rhG-CSF developed sepsis; however, three patients in the control group did.51 In fulminant intra-abdominal sepsis in rats, mortality decreased from 92% to 46% and to 42%, respectively, with the first dose of G-CSF at sepsis induction or 4 hours postinduction, ie, during manifested sepsis, respectively (Fig 3).169 Based on animal studies, G-CSF should be effective not only in prophylaxis, but also in treatment of sepsis.

In future use, G-CSF may be effective in nonneutropenic critically ill patients suffering from yet unrecognized impairment of neutrophil function and tissue injury. Beneficial effects of exogenous G-CSF have been observed on the resolution of infection, especially pneumonia, and subsequent sepsis, multiple organ dysfunction, and septic shock. These effects of exogenous G-CSF result directly from a better control of infection and indirectly from an amelioration of an overwhelming, damaging, proinflammatory response.

Ischemia/reperfusion injury is also a frequent consequence of surgical injury, hemorrhagic shock, and trauma. Improved endotoxin elimination by exogenous G-CSF138 may contribute to the recently described endothelial cell protection by the immediate induction of IL-1ra. This latter property of G-CSF extends the therapeutic potential to ischemia/reperfusion injury. Short, clinically relevant visceral ischemia produced TNF-α– and IL-1–dependent lung injury in mice.170 Recently, in rats with visceral ischemia/reperfusion injury, treatment with rhG-CSF increased survival, reduced serum TNF-α induction, reduced myeloperoxidase activity in the ileum and in the lung, and improved mean arterial blood pressure.117 Such results may be explained by our recent observation that G-CSF stimulated hemoxygenase-1 (HO-1) expression in microvascular endothelial cells in vitro (unpublished observation). If HO-1 induction by G-CSF is a general phenomenon in endothelial cells, then this may represent an important response against oxidative damage.

G-CSF has been demonstrated to alleviate neutropenia in patients with advanced AIDS or AIDS-related complex receiving zidovudine.83 In addition, the prophylactic use of G-CSF may prevent endotoxin-induced shock in AIDS patients. The incidence of infection with gram-negative bacteria is increased in AIDS patients who suffer from accelerated leukocyte apoptosis and defective neutrophil functions.55,171 Accelerated apoptosis contributes to impaired neutrophil function in AIDS patients.55 However, incubation with G-CSF in vitro significantly decreased apoptosis in neutrophils of these patients.55 In addition, repeated pretreatment with rhG-CSF successfully protected mice with retrovirus-induced murine acquired immunodeficiency syndrome (MAIDS) from hypersensitivity to LPS-induced lethal shock, and this protective effect coincided with the suppression of IFN-γ production.144 

Application of rhG-CSF may also find widespread use in critically ill patients, because no serious side effects have been observed in multiple therapeutic trials in immunocompromised, agranulocytotic, neutropenic, and nonneutropenic patients.45,51,61,82,136,164,165 G-CSF is well-tolerated, and the most common side effects, bone pain and occasionally reported skin rash, appear to be dose-dependent.45,82 Transgenic mice that overexpressed G-CSF had a 100-fold increase in G-CSF serum concentrations (1,041 ± 242 pg/mL in sera) and peripheral neutrophils. These G-CSF–overexpressing transgenic mice developed osteoporosis because of increased osteoclastic activity. Thus, high G-CSF serum concentrations could have a negative influence on bone homeostasis in vivo when active over extended periods of time.172 

Animal studies suggest that prolonged therapy with G-CSF may also have anti-inflammatory effects in colitis. Inflammatory bowel disease is associated with mucosal neutrophil recruitment and activation, mediated in part by arachidonic acid metabolites. G-CSF attenuates the immune response to sepsis and ameliorates glycogen storage disease Ib-related colitis. In immune complex colitis in rabbits, treatment with rhG-CSF resulted in a marked decrease of proinflammatory mediators, such as LTB4 and thromboxane B2 (TXB2), but mucosal generation of the protective prostaglandin E2(PGE2) was preserved.173 

In critically ill patients with a secondary functional impairment of host defense, G-CSF improved leukocyte recruitment and, in addition, appears to activate a number of cell protective mechanisms awaiting more detailed investigations.

The present data support the hypothesis that G-CSF treatment may be extended to a group of critically ill patients with secondary impairment of neutrophil function occurring as a result of extensive tissue damage, oxidative stress, and/or ischemia/reperfusion. G-CSF both augments the replication and function of neutrophils. G-CSF also stimulates the elimination of endotoxin and attenuates the hyperinflammatory state by increasing IL-1ra and by intermittently modulating the adhesion receptor L-selectin. These events would counteract the progression of sepsis and, presumably, the incidence and severity of multiple organ dysfunction. Refinement in the use of G-CSF, either prophylactically or in states with acute infection, reperfusion, or severe tissue injury relies on the experimental as well as clinical evidence that G-CSF stimulates neutrophil function and concomitantly attenuates an uncontrolled inflammatory response. However, the successful administration of G-CSF to nonneutropenic patients with impairment of neutrophil function will strongly depend on optimal timing and dosage. The prophylactic administration of G-CSF to surgical patients with low G-CSF serum concentrations will result in appropriate serum concentrations of about 1,000 pg/mL G-CSF in responding patients when administered at about 1 μg/kg·d.51,61,165 These doses are associated with beneficial effects on neutrophil function in the absence of granulocytosis greater than 50,000/μL.51,61,165 

An ongoing inflammation is possibly also targeted by G-CSF with beneficial effects if the G-CSF administration is associated with its efficient use. Evidence of use includes functional activation (CD64 expression, phagocytic activity, and less apoptosis), decreased L-selectin expression on neutrophils, and a concomitant increase of serum IL-1ra as well as a decrease of serum proinflammatory cytokine concentrations (such as IL-8). Thus, the definition of subgroups of patients sensitive to G-CSF treatment, which by now includes cases with pneumonia, diabetes, acute necrotizing pancreatitis, alcoholism, AIDS, and ischemia/reperfusion injury, may further extend our understanding of immunomodulatory effects by this hematopoietic growth factor.

Additional controlled studies with critically ill patient populations at a high risk to encounter severe infections and sepsis are necessary to further define and use the regulatory properties of G-CSF in a window of effective treatment protocols.

Supported by GM-40586-II awarded by the National Institute of General Medical Sciences, US Public Health Services.

1
American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference Committee
American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference: Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis.
Crit Care Med
20
1992
864
2
Bone
R
Sir Isaac Newton, sepsis, SIRS, and CARS.
Crit Care Med
24
1996
1125
3
Stuehr
DJ
Kwon
NS
Gross
SS
Thiel
BA
Levi
R
Nathan
CF
Synthesis of nitrogen oxides from L-arginine by macrophage cytosol: Requirement for inducible and constitutive components.
Biochem Biophys Res Commun
161
1989
420
4
Sparrow
JR
Nathan
C
Vodovotz
Y
Cytokine regulation of nitric oxide synthase in mouse retinal pigment epithelial cells in culture.
Exp Eye Res
59
1994
129
5
Xie
QW
Nathan
C
Promoter of the mouse gene encoding calcium-independent nitric oxide synthase confers inducibility by interferon-gamma and bacterial lipopolysaccharide.
Trans Assoc Am Physicians
106
1993
1
6
Nussler
AK
Billiar
TR
Inflammation, immunoregulation, and inducible nitric oxide synthase.
J Leukoc Biol
54
1993
171
7
O’Connor
M
Salzman
AL
Szabó
C
Role of peroxinitrite in the protein oxidation and apoptotic DNA fragmentation in vascular smooth muscle cells stimulated with bacterial lipopolysaccharide and interferon-γ.
Shock
8
1997
439
8
Christ
EA
Meals
E
English
BK
Streptococcal pyogenic exotoxins A (SpeA) and C (SpeC) stimulate the production of inducible nitric oxide synthase (iNOS) protein in RAW 264.7 macrophages.
Shock
8
1997
450
9
Szabo
C
Thiemermann
C
Invited opinion: Role of nitric oxide in hemorrhagic, traumatic, and anaphylactic shock and thermal injury.
Shock
2
1994
145
10
Kilbourn
RG
Szabo
C
Traber
DL
Beneficial versus detrimental effects of nitric oxide synthase inhibitors in circulatory shock: Lessons learned from experimental and clinical studies.
Shock
7
1997
235
11
Doughty
LA
Kaplan
SS
Carcillo
JA
Inflammatory cytokine and nitric oxide responses in pediatric sepsis and organ failure.
Crit Care Med
24
1996
1137
12
Friedman
G
Jankowski
S
Marchant
A
Goldman
M
Kahn
RJ
Vincent
JL
Blood interleukin 10 levels parallel the severity of septic shock.
J Crit Care
12
1997
183
13
Villarete
LH
Remick
DG
Nitric oxide regulation of IL-8 expression in human endothelial cells.
Biochem Biophys Res Commun
211
1995
671
14
Villarete
LH
Remick
DG
Nitric oxide regulation of interleukin-8 gene expression.
Shock
7
1997
29
15
Bogdan
C
Vodovotz
Y
Paik
J
Xie
Q
Nathan
C
Mechanism of suppression of nitric oxide synthase expression by interleukin-4 in primary mouse macrophages.
J Leukoc Biol
55
1994
227
16
Vodovotz
Y
Bogdan
C
Paik
J
Xie
QW
Nathan
C
Mechanisms of suppression of macrophage nitric oxide release by transforming growth factor beta.
J Exp Med
178
1993
605
17
Galley
HF
Davies
MJ
Webster
NR
Xanthine oxidase activity and free radical generation in patients with sepsis syndrome.
Crit Care Med
24
1996
1649
18
Greenacre
S
Ridger
V
Wilsoncroft
P
Brain
SD
Peroxynitrite: A mediator of increased microvascular permeability?
Clin Exp Pharmacol Physiol
24
1997
880
19
Szabo
C
DNA strand breakage and activation of poly-ADP ribosyltransferase: A cytotoxic pathway triggered by peroxynitrite.
Free Radic Biol Med
21
1996
855
20
Hotchkiss
RS
Swanson
PE
Cobb
JP
Jacobson
A
Buchman
TG
Karl
IE
Apoptosis in lymphoid and parenchymal cells during sepsis: Findings in normal and T- and B-cell-deficient mice.
Crit Care Med
25
1997
1298
21
Jacob
AK
Hotchkiss
RS
DeMeester
SL
Hiramatsu
M
Karl
IE
Swanson
PE
Cobb
JP
Buchman
TG
Endothelial cell apoptosis is accelerated by inorganic iron and heat via an oxygen radical dependent mechanism.
Surgery
122
1997
243
22
Motoyama
S
Minamiya
Y
Saito
S
Saito
R
Matsuzaki
I
Abo
S
Inaba
H
Enomoto
K
Kitamura
M
Hydrogen peroxide derived from hepatocytes induces sinusoidal endothelial cell apoptosis in perfused hypoxic rat liver.
Gastroenterology
114
1998
153
23
Ayala
A
Herdon
CD
Lehman
DL
Ayala
CA
Chaudry
IH
Differential induction of apoptosis in lymphoid tissues during sepsis: Variation in onset, frequency, and the nature of the mediators.
Blood
15
1996
4261
24
Ayala
A
Karr
SM
Evans
TA
Chaudry
IH
Factors responsible for peritoneal granulocyte apoptosis during sepsis.
J Surg Res
69
1997
67
25
Rogy
MA
Oldenburg
HS
Coyle
S
Trousdale
R
Moldawer
LL
Lowry
SF
Correlation between Acute Physiology and Chronic Health Evaluation (APACHE) III score and immunological parameters in critically ill patients with sepsis.
Br J Surg
83
1996
396
26
Lynch
DH
Ramsdell
F
Alderson
MR
Fas and FasL in the homeostatic regulation of immune responses.
Immunol Today
16
1995
569
27
Suda
T
Okazaki
T
Naito
Y
Yokota
T
Arai
N
Ozaki
S
Nakao
K
Nagata
S
Expression of the Fas ligand in cells of T cell lineage.
J Immunol
154
1995
3806
28
Fischer
E
Van Zee
KJ
Marano
MA
Rock
CS
Kenney
JS
Poutsiaka
DD
Dinarello
CA
Lowry
SF
Moldawer
LL
Interleukin-1 receptor antagonist circulates in experimental inflammation and in human disease.
Blood
79
1992
2196
29
D’Andrea
A
Aste-Amezaga
M
Valiante
NM
Ma
X
Kubin
M
Trinchieri
G
Interleukin 10 (IL-10) inhibits human lymphocyte interferon gamma-production by suppressing natural killer cell stimulatory factor/IL-12 synthesis in accessory cells.
J Exp Med
178
1993
1041
30
Svenson
M
Hansen
MB
Heegaard
P
Abell
K
Bendtzen
K
Specific binding of interleukin 1 (IL-1) beta and IL-1 receptor antagonist (IL-1ra) to human serum. High-affinity binding of IL-1ra to soluble IL-1 receptor type I.
Cytokine
5
1993
427
31
Pruitt
JH
Copeland
EMr
Moldawer
LL
Interleukin-1 and interleukin-1 antagonism in sepsis, systemic inflammatory response syndrome, and septic shock.
Shock
3
1995
235
32
Pruitt
JH
Welborn
MB
Edwards
PD
Harward
TR
Seeger
JW
Martin
TD
Smith
C
Kenney
JA
Wesdorp
RI
Meijer
S
Cuesta
MA
Abouhanze
A
Copeland
EMr
Giri
J
Sims
JE
Moldawer
LL
Oldenburg
HS
Increased soluble interleukin-1 type II receptor concentrations in postoperative patients and in patients with sepsis syndrome.
Blood
87
1996
3282
33
Opal
SM
Fisher
CJ
Jr
Dhainaut
JF
Vincent
JL
Brase
R
Lowry
SF
Sadoff
JC
Slotman
GJ
Levy
H
Balk
RA
Shelly
MP
Pribble
JP
LaBrecque
JF
Lookabaugh
J
Donovan
H
Dubin
H
Baughman
R
Norman
J
DeMaria
E
Matzel
K
Abraham
E
Seneff
M
Confirmatory interleukin-1 receptor antagonist trial in severe sepsis: A phase III, randomized, double-blind, placebo-controlled, multicenter trial. The Interleukin-1 Receptor Antagonist Sepsis Investigator Group.
Crit Care Med
25
1997
1115
34
Brandtzaeg
P
Osnes
L
Ovstebo
R
Joo
GB
Westvik
AB
Kierulf
P
Net inflammatory capacity of human septic shock plasma evaluated by a monocyte-based target cell assay: Identification of interleukin-10 as a major functional deactivator of human monocytes.
J Exp Med
184
1996
51
35
Randow
F
Mechanism of endotoxin desensitization: Involvement of interleukin 10 and transforming growth factor beta.
J Exp Med
181
1995
1887
36
Marie
C
Muret
J
Fitting
C
Losser
MR
Payen
D
Cavaillon
JM
Reduced ex vivo interleukin-8 production by neutrophils in septic and nonseptic systemic inflammatory response syndrome.
Blood
91
1998
3439
37
Nunes
I
Munger
JS
Harpel
JG
Nagano
Y
Shapiro
RL
Gleizes
PE
Rifkin
DB
Structure and activation of the large latent transforming growth factor-beta complex.
Int J Obes Relat Metab Disord
20
1996
S4
(suppl 3)
38
Koeffler
HP
Gasson
J
Tobler
A
Transcriptional and posttranscriptional modulation of myeloid colony-stimulating factor expression by tumor necrosis factor and other agents.
Mol Cell Biol
8
1988
3432
39
Demetri
GD
Zenzie
BW
Rheinwald
JG
Griffin
JD
Expression of colony-stimulating factor genes by normal human mesothelial cells and human malignant mesothelioma cells lines in vitro.
Blood
74
1989
940
40
Demetri
GD
Ernst
TJ
Pratt
ES
2d
Zenzie
BW
Rheinwald
JG
Griffin
JD
Expression of ras oncogenes in cultured human cells alters the transcriptional and posttranscriptional regulation of cytokine genes.
J Clin Invest
86
1990
1261
41
DellaPuca
R
Gallicchio
VS
The regulation of phospholipase-A2 (PLA-2) by cytokines expressing hematopoietic growth-stimulating properties.
Proc Soc Exp Biol Med
212
1996
174
42
Nathan
CF
Respiratory burst in adherent human neutrophils: Triggering by colony-stimulating factors CSF-GM and CSF-G.
Blood
73
1989
301
43
Vincent
JL
Chamlou
R
Sepsis trials.
Curr Opin Anaesthesiol
9
1996
146
44
Zeni
F
Freeman
B
Natanson
C
Anti-inflammatory therapies to treat sepsis and septic shock: A reassessment.
Crit Care Med
25
1997
1095
45
Lieschke
GJ
Burgess
AW
Granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor.
N Engl J Med
327
1992
28
46
Wang
JM
Chen
ZG
Colella
S
Bonilla
MA
Welte
K
Bordignon
C
Mantovani
A
Chemotactic activity of recombinant human granulocyte colony-stimulating factor.
Blood
72
1988
1456
47
Roilides
E
Walsh
TJ
Pizzo
PA
Rubin
M
Granulocyte colony-stimulating factor enhances the phagocytic and bactericidal activity of normal and defective human neutrophils.
J Infect Dis
163
1991
579
48
Repp
R
Valerius
T
Sendler
A
Gramatzki
M
Iro
H
Kalden
JR
Platzer
E
Neutrophils express the high affinity receptor for IgG (FcγRI, CD64) after in vivo application of recombinant human granulocyte colony-stimulating factor.
Blood
78
1991
885
49
König
B
König
W
Effect of growth factors on Escherichia coli alpha-hemolysin-induced mediator release from human inflammatory cells: Involvement of the signal transduction pathway.
Infect Immun
62
1994
2085
50
Eichacker
PQ
Natanson
C
Rethinking the anti-inflammatory approach to septic shock
Yearbook of Intensive Care and Emergency Medicine 1996.
Vincent
JL
1996
165
Springer-Verlag
Berlin, Germany
51
Weiss
M
Gross-Weege
W
Schneider
M
Neidhardt
H
Liebert
S
Mirow
N
Wernet
P
Enhancement of neutrophil function by in vivo filgrastim treatment for prophylaxis of sepsis in surgical intensive care patients.
J Crit Care
10
1995
21
52
Liu
F
Poursine-Laurent
J
Wu
HY
Link
DC
Interleukin-6 and the granulocyte colony-stimulating factor receptor are major independent regulators of granulopoiesis in vivo but are not required for lineage commitment or terminal differentiation.
Blood
90
1997
2583
53
Marsh
JC
Long-term bone marrow cultures in aplastic anaemia.
Eur J Haematol Suppl
60
1996
75
54
Philpott
NJ
Prue
RL
Marsh
JC
Gordon Smith
EC
Gibson
FM
G-CSF-mobilized CD34 peripheral blood stem cells are significantly less apoptotic than unstimulated peripheral blood CD34 cells: role of G-CSF as survival factor.
Br J Haematol
97
1997
146
55
Pitrak
DL
Tsai
HC
Mullane
KM
Sutton
SH
Stevens
P
Accelerated neutrophil apoptosis in the acquired immunodeficiency syndrome.
J Clin Invest
98
1996
2714
56
Matute-Bello
G
Liles
WC
Radella
F
2nd
Steinberg
KP
Ruzinski
JT
Jonas
M
Chi
EY
Hudson
LD
Martin
TR
Neutrophil apoptosis in the acute respiratory distress syndrome.
Am J Respir Crit Care Med
156
1997
1969
57
Shimoda
K
Okamura
S
Harada
N
Niho
Y
Detection of the granulocyte colony-stimulating factor receptor using biotinylated granulocyte colony-stimulating factor: Presence of granulocyte colony-stimulating factor receptor on CD34-positive hematopoietic progenitor cells.
Res Exp Med Berl
192
1992
245
58
Pan
L
Delmonte
J
Jr
Jalonen
CK
Ferrara
JL
Pretreatment of donor mice with granulocyte colony-stimulating factor polarizes donor T lymphocytes toward type-2 cytokine production and reduces severity of experimental graft-versus-host disease.
Blood
86
1995
4422
59
Hartung
T
Docke
WD
Gantner
F
Krieger
G
Sauer
A
Stevens
P
Volk
HD
Wendel
A
Effect of granulocyte colony-stimulating factor treatment on ex vivo blood cytokine response in human volunteers.
Blood
85
1995
2482
60
Dinarello
CA
Wolff
SM
The role of interleukin-1 in disease.
N Engl J Med
328
1993
106
61
Weiss
M
Gross-Weege
W
Harms
B
Schneider
EM
Filgrastim (rhG-CSF) related modulation of the inflammatory response in patients at risk of sepsis or with sepsis.
Cytokine
8
1996
260
62
Pajkrt
D
Manten
A
van der Poll
T
Tiel van Buul
MM
Jansen
J
Wouter ten Cate
J
van Deventer
SJ
Modulation of cytokine release and neutrophil function by granulocyte colony-stimulating factor during endotoxemia in humans.
Blood
90
1997
1415
63
Weiss
M
Gross-Weege
W
Moldawer
LL
Schneider
EM
Relationship of IL-6 and IL-6R to sepsis and septic shock in postoperative/posttraumatic patients.
Shock Suppl
7
1997
98
(abstr)
64
Harms
B
Dumon
K
Gross-Weege
W
Lorenz
I
Röher
HD
Scharf
RE
Schneider
EM
Effect of G-CSF-treatment for the prophylaxis of sepsis on the reconstitution of CD14, on neutrophil pre-sensitization and on non-impaired phagocytic function in patients with extended surgery, trauma and hemorrhagic shock.
Shock Suppl
7
1997
79
(abstr)
65
Tian
SS
Lamb
P
Seidel
HM
Stein
RB
Rosen
J
Rapid activation of the STAT3 transcription factor by granulocyte colony-stimulating factor.
Blood
84
1994
1760
66
Zhong
Z
Wen
Z
Darnell
JE
Jr
Stat3: A STAT family member activated by tyrosine phosphorylation in response to epidermal growth factor and interleukin-6.
Science
264
1994
95
67
Akira
S
Nishio
Y
Inoue
M
Wang
XJ
Wei
S
Matsusaka
T
Yoshida
K
Sudo
T
Naruto
M
Kishimoto
T
Molecular cloning of APRF, a novel IFN-stimulated gene factor 3 p91-related transcription factor involved in the gp130-mediated signaling pathway.
Cell
77
1994
63
68
Tilg
H
Dinarello
CA
Mier
JW
IL-6 and APPs: Anti-inflammatory and immunosuppressive mediators.
Immunol Today
18
1997
428
69
Nagata
S
Fukunaga
R
Granulocyte colony-stimulating factor and its receptor.
Prog Growth Factor Res
3
1991
131
70
Omori
F
Okamura
S
Shimoda
K
Otsuka
T
Harada
M
Niho
Y
Levels of human serum granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor under pathological conditions.
Biotherapy
4
1992
147
71
Dunn
SM
Coles
LS
Lang
RK
Gerondakis
S
Vadas
MA
Shannon
MF
Requirement for nuclear factor (NF)-kappa B p65 and NF-interleukin-6 binding elements in the tumor necrosis factor response region of the granulocyte colony-stimulating factor promoter.
Blood
83
1994
2469
72
Smith
LT
Hohaus
S
Gonzalez
DA
Dziennis
SE
Tenen
DG
PU.1 (Spi-1) and C/EBP alpha regulate the granulocyte colony-stimulating factor receptor promotor in myeloid cells.
Blood
88
1996
1234
73
Watari
K
Asano
S
Shirafuji
N
Kodo
H
Ozawa
K
Takaku
F
Kamachi
S
Serum granulocyte colony-stimulating factor levels in healthy volunteers and patients with various disorders as estimated by enzyme immunoassay.
Blood
73
1989
117
74
Kawakami
M
Tsutsumi
H
Kumakawa
T
Abe
H
Hirai
M
Kurosawa
S
Mori
M
Fukushima
M
Levels of serum granulocyte colony-stimulating factor in patients with infections.
Blood
76
1990
1962
75
De Haas
M
Kerst
JM
van der Schoot
CE
Calafat
J
Hack
CE
Nuijens
JH
Roos
D
van Oers
RH
von dem Borne
AE
Granulocyte colony-stimulating factor administration to healthy volunteers: Analysis of the immediate activating effects on circulating neutrophils.
Blood
84
1994
3885
76
Aoygai
S
Arasawa
K
Matsuyuki
A
Kamatchi
S
Fukushima
M
Ohsawa
N
Chemiluminescence sandwich enzyme immunoassay for determination of human granulocyte colony stimulating factor (G-CSF).
J Biolumin Chemilumin
10
1995
345
77
Selig
C
Nothdurft
W
Cytokines and progenitor cells of granulocytopoiesis in peripheral blood of patients with bacterial infections.
Infect Immun
63
1995
104
78
Chen
YM
Whang-Peng
J
Chern
CH
Kuo
BIT
Wang
SY
Perng
RP
The prognostic value of serum cytokine levels in patients with acute infections.
Chin Med J (Taipei)
56
1995
75
79
Tanaka
H
Ishikawa
K
Nishino
M
Shimazu
T
Yoshioka
T
Changes in granulocyte colony-stimulating factor concentration in patients with trauma and sepsis.
J Trauma
40
1996
718
80
Kragsbjerg
P
Holmberg
H
Vikerfors
T
Dynamics of blood cytokine concentrations in patients with bacteremic infections.
Scand J Infect Dis
28
1996
391
81
Groopman
JE
Status of colony-stimulating factors in cancer and AIDS.
Semin Oncol
17
1990
31
82
Welte
K
Grabilove
J
Bronchud
MH
Platzer
E
Morstyn
G
Filgrastim (r-metHuG-CSF): The first 10 years.
Blood
88
1996
1907
83
Groopman
JE
Antiretroviral therapy and immunomodulators in patients with AIDS.
Am J Med
90
1991
18s
84
Sprikkelman
A
de Wolf
JTM
Vellenga
E
The application of hematopoietic growth factors in drug-induced agranulocytosis: A review of 70 cases.
Leukemia
8
1994
2031
85
Kasahara
Y
Iwai
K
Yachie
A
Ohta
K
Konno
A
Seki
H
Miyawaki
T
Taniguchi
N
Involvement of reactive oxygen intermediates in spontaneous and CD95 (Fas/APO-1)-mediated apoptosis of neutrophils.
Blood
89
1997
1748
86
Martin
MA
Pfaller
MA
Wenzel
RP
Coagulase-negative staphylococcal bacteremia. Mortality and hospital stay.
Ann Intern Med
110
1989
9
87
Vincent
JL
Moreno
R
Takala
J
Willatts
S
De Mendonca
A
Bruining
H
Reinhart
CK
Suter
PM
Thijs
LG
The SOFA (Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure. On behalf of the Working Group on Sepsis-Related Problems of the European Society of Intensive Care Medicine.
Intensive Care Med
22
1996
707
88
McMahon
MM
Bistrian
BR
Host defenses and susceptibility to infection in patients with diabetes mellitus.
Infect Dis Clin North Am
9
1995
1
89
Smitherman
KO
Peacock
JE
Jr
Infectious emergencies in patients with diabetes mellitus.
Med Clin North Am
79
1995
53
90
Eddleston
JM
Sharer
NM
Antioxidant therapy in critical illness
Yearbook of Intensive Care and Emergency Medicine 1996.
Vincent
JL
1996
96
Springer-Verlag
Berlin, Germany
91
Grimsley
EW
Granulocyte colony stimulating factor in the treatment of alcohol abuse, leukopenia, and pneumococcal sepsis.
South Med J
88
1995
220
92
Green
DR
Faist
E
Trauma and the immune response.
Immunol Today
9
1988
253
93
Dorman
T
Breslow
MJ
Altered immune function after trauma and hemorrhage: What does it all mean?
Crit Care Med
22
1994
1069
94
Schmand
JF
Ayala
A
Chaudry
IH
Effects of trauma, duration of hypotension, and resuscitation regimen on cellular immunity after hemorrhagic shock.
Crit Care Med
22
1994
1076
95
Brunson
ME
Alexander
JW
Mechanisms of tranfusion-induced immunosuppression.
Transfusion
30
1990
651
96
Weiss
M
Mirow
N
Birkhahn
A
Schneider
M
Wernet
P
Benzodiazepines and their solvents influence neutrophil granulocyte function.
Br J Anaesth
70
1993
317
97
Weiss
M
Buhl
R
Mirow
N
Birkhahn
A
Schneider
M
Wernet
P
Do barbiturates impair zymosan-induced granulocyte function?
J Crit Care
9
1994
83
98
Weiss
M
Schneider
EM
Tarnow
J
Mettler
S
Krone
M
Teschemacher
A
Lemoine
H
Is inhibition of oxygen radical production of neutrophils by sympathomimetics mediated via β2-adrenoceptors?
J Pharmacol Exp Ther
278
1996
1105
99
Lanser
ME
Mao
P
Brown
G
Coleman
B
Siegel
J
Serum-mediated depression of neutrophil chemiluminescence following blunt trauma.
Ann Surg
202
1985
111
100
Seekamp
A
Regel
G
Dwenger
A
Interaction of soft tissue trauma and unspecific immune system in polytraumatized patients.
Circ Shock
34
1991
34
101
Solomkin
JS
Bauman
MP
Nelson
RD
Simmons
RL
Neutrophils dysfunction during the course of intra-abdominal infection.
Ann Surg
194
1981
9
102
Vespasiano
MC
Lewandoski
JR
Zimmerman
JJ
Longitudinal analysis of neutrophil superoxide anion generation in patients with septic shock.
Crit Care Med
21
1993
666
103
Simms
HH
D’Amico
R
Polymorphonuclear leukocyte dysregulation during the systemic inflammatory response syndrome.
Blood
83
1994
1398
104
Kropec
A
Lemmen
SW
Grundmann
HJ
Engels
I
Daschner
FD
Synergy of simultaneous administration of ofloxacin and granulocyte colony-stimulating factor in killing of Escherichia coli by human neutrophils.
Infection
23
1995
298
105
Lundblad
R
Nesland
JM
Giercksky
KE
Granulocyte colony-stimulating factor improves survival rate and reduces concentrations of bacteria, endotoxin, tumor necrosis factor, and endothelin-1 in fulminant intra-abdominal sepsis in rats.
Crit Care Med
24
1996
820
106
Smith
WS
Sumnicht
GE
Sharpe
RW
Samuelson
D
Millard
FE
Granulocyte colony-stimulating factor versus placebo in addition to penicillin G in a randomized blinded study of gram-negative pneumonia sepsis: Analysis of survival and multisystem organ failure.
Blood
86
1995
1301
107
O’Reilly
M
Silver
GM
Greenhalgh
DG
Gamelli
RL
Davis
JH
Hebert
JC
Treatment of intra-abdominal infection with granulocyte colony-stimulating factor.
J Trauma
33
1992
679
108
Görgen
I
Hartung
T
Leist
M
Niehörster
M
Tiegs
G
Uhlig
S
Weitzel
F
Wendel
A
Granulocyte colony-stimulating factor treatment protects rodents against lipopolysaccharide-induced toxicity via suppression of systemic tumor necrosis factor-α.
J Immunol
149
1992
919
109
Endo
S
Inada
K
Inoue
Y
Yamada
Y
Takakuwa
T
Kasai
T
Nakae
H
Kuwata
Y
Hoshi
S
Evaluation of recombinant human granulocyte colony-stimulating factor (rhG-CSF) therapy in granulopoetic patients complicated with sepsis.
Curr Med Res Opin
13
1994
233
110
Tkatch
LS
Rubin
KA
Ziegler
SF
Tweardy
DJ
Modulation of human G-CSF receptor mRNA and protein in normal and leukemic myeloid cells by G-CSF and retinoic acid.
J Leukoc Biol
57
1995
964
111
Khwaja
A
Carver
J
Jones
HM
Paterson
D
Linch
DC
Expression and dynamic modulation of the human granulocyte colony-stimulating factor receptor in immature and differentiated myeloid cells.
Br J Haematol
85
1993
254
112
Tasaka
K
Doi
M
Nakaya
N
Mio
M
Reinforcement effect of histamine on the differentiation of murine myeloblasts and promyelocytes: Externalization of granulocyte colony-stimulating factor receptors induced by histamine.
Mol Pharmacol
45
1994
837
113
Nicola
NA
Vadas
MA
Lopez
AF
Down-modulation of receptors for granulocyte colony-stimulating factor on human neutrophils by granulocyte-activating agents.
J Cell Physiol
128
1986
501
114
Heard
SO
Fink
MP
Gamelli
RL
Solomkin
JS
Joshi
M
Trask
AL
Fabian
TC
Hudson
LD
Gerold
KB
Logan
ED
Effect of prophylactic administration of recombinant human granulocyte colony-stimulating factor (filgrastim) on the frequency of nosocomial infections in patients with acute traumatic brain injury or cerebral hemorrhage. The Filgrastim Study Group.
Crit Care Med
26
1998
748
115
Emminger
W
Emminger-Schmidmeier
W
Peters
C
Susani
M
Hawliczek
R
Höcker
P
Gadner
H
Capillary leak syndrome during low dose granulocyte-macrophage colony-stimulating factor (rhGM-CSF) treatment of a patient in a continuous febrile state.
Blut
61
1990
219
116
Denzlinger
C
Holler
E
Reisbach
G
Hiller
E
Wilmanns
W
Granulocyte colony-stimulating factor inhibits the endogenous leukotriene production in tumour patients.
Br J Haematol
86
1994
881
117
Squadrito
F
Altavilla
D
Squadrito
G
Campo
GM
Ioculano
M
Ammedolia
L
Arlotta
M
Saitta
A
Caputi
AP
The effects of recombinant human granulocyte colony-stimulating factor on vascular dysfunction and splanchnic ischemia-reperfusion injury.
Br J Pharmacol
120
1997
333
118
Kroncke
KD
Fehsel
K
Kolb-Bachofen
V
Inducible nitric oxide synthase and its product nitric oxide, a small molecule with complex biological activities.
Biol Chem Hoppe Seyler
376
1995
327
119
Liu
HW
Anand
A
Bloch
K
Christiani
D
Kradin
R
Expression of inducible nitric oxide synthase by macrophages in rat lung.
Am J Respir Crit Care Med
156
1997
223
120
Basu
S
Dunn
AR
Marino
MW
Savoia
H
Hodgson
G
Lieschke
GJ
Cebon
J
Increased tolerance to endotoxin by granulocyte-macrophage colony-stimulating factor-deficient mice.
J Immunol
159
1997
1412
121
Hill
AD
Naama
H
Shou
J
Calvano
SE
Daly
JM
Antimicrobial effects of granulocyte-macrophage colony-stimulating factor in protein-energy malnutrition.
Arch Surg
130
1995
1273
122
Feder
LS
Laskin
DL
Regulation of hepatic endothelial cell and macrophage proliferation and nitric oxide production by GM-CSF, M-CSF, and IL-1 beta following acute endotoxemia.
J Leukoc Biol
55
1994
507
123
Nagata
M
Sedgwick
JB
Busse
WW
Differential effects of granulocyte-macrophage colony-stimulating factor on eosinophil and neutrophil superoxide anion generation.
J Immunol
155
1995
4948
124
Stankova
J
Rola-Pleszczynski
M
Dubois
CM
Granulocyte-macrophage colony-stimulating factor increases 5-lipoxygenase gene transcription and protein expression in human neutrophils.
Blood
85
1995
3719
125
Brock
TG
McNish
RW
Coffey
MJ
Ojo
TC
Phare
SM
Peters-Golden
M
Effects of granulocyte-macrophage colony-stimulating factor on eicosanoid production by mononuclear phagocytes.
J Immunol
156
1996
2522
126
McDonald
PP
Pouliot
M
Borgeat
P
McColl
SR
Induction by chemokines of lipid mediator synthesis in granulocyte-macrophage colony-stimulating factor-treated human neutrophils.
J Immunol
151
1993
6399
127
Bober
LA
Grace
MJ
Pugliese-Sivo
C
Rojas-Triana
A
Waters
T
Sullivan
LM
Narula
SK
The effect of GM-CSF and G-CSF on human neutrophil function.
Immunopharmacology
29
1995
111
128
Eissner
G
Lindner
H
Reisbach
G
Klauke
I
Holler
E
Differential modulation of IL-1-induced endothelial adhesion molecules and transendothelial migration of granulocytes by G-CSF.
Br J Haematol
97
1997
726
129
Yong
KL
Granulocyte colony-stimulating factor (G-CSF) increases neutrophil migration across vascular endothelium independent of an effect on adhesion: Comparison with granulocyte-macrophage colony-stimulating factor (GM-CSF).
Br J Haematol
94
1996
40
130
Valerius
T
Repp
R
de Wit
TPM
Berthold
S
Platzer
E
Kalden
JR
Gramatzki
M
van de Winkel
JGJ
Involvement of the high-affinity receptor for IgG (FcγRI; CD64) in enhanced tumor cell cytotoxicity of neutrophils during granulocyte colony-stimulating factor therapy.
Blood
82
1993
931
131
Spiekermann
K
Emmendoerffer
A
Elsner
J
Raeder
E
Lohmann Matthes
ML
Prahst
A
Link
H
Freund
M
Welte
K
Roesler
J
Altered surface marker expression and function of G-CSF-induced neutrophils from test subjects and patients under chemotherapy.
Br J Haematol
87
1994
31
132
Gericke
GH
Ericson
SG
Pan
L
Mills
LE
Guyre
PM
Ely
P
Mature polymorphonuclear leukocytes express high-affinity recepors for IgG (FcγRI) after stimulation with granulocyte colony-stimulating factor (G-CSF).
J Leukoc Biol
57
1995
455
133
Carulli
G
Minnucci
S
Azzara
A
Angiolini
C
Sbrana
S
Caracciolo
F
Ambrogi
F
Granulocyte colony-stimulating factor (G-CSF) administration increases PMN CD32 (FcRII) expression and FcR-related functions.
Haematologica
80
1995
150
134
Erbe
DV
Collins
JE
Shen
L
Graziano
RF
Fange
MW
The effect of cytokines on the expression and function of Fc receptors for IgG on human myeloid cells.
Mol Immunol
27
1990
57
135
Ninomiya
H
Muraki
Y
Shibuya
K
Nagasawa
T
Abe
T
Induction of FcγR-III (CD16) expression on neutrophils affected by paroxysmal nocturnal haemoglobinuria by administration of granulocyte colony-stimulating factor.
Br J Haematol
84
1993
497
136
Gillan
ER
Christensen
RD
Suen
Y
Ellis
R
van de Ven
C
Cairo
MS
A randomized, placebo-controlled trial of recombinant human granulocyte colony-stimulating factor administration in newborn infants with presumed sepsis: Significant induction of peripheral and bone marrow neutrophilia.
Blood
84
1994
1427
137
Kerst
JM
de Haas
M
van der Schoot
CE
Slaper Cortenbach
IC
Kleijer
M
von dem Borne
AE
van Oers
RH
Recombinant granulocyte colony-stimulating factor administration to healthy volunteers: Induction of immunophenotypically and functionally altered neutrophils via an effect on myeloid progenitor cells.
Blood
82
1993
3265
138
Freeman
BD
Quezado
Z
Zeni
F
Natanson
C
Danner
RL
Banks
S
Quezado
M
Fitz
Y
Bacher
J
Eichacker
PQ
rG-CSF reduces endotoxemia and improves survival during E. coli pneumonia.
J Appl Physiol
83
1997
1467
139
Kitabayashi
A
Hirokawa
M
Hatano
Y
Lee
M
Kuroki
J
Niitsu
H
Miura
AB
Granulocyte colony-stimulating factor downregulates allogeneic immune responses by posttranscriptional inhibition of tumor necrosis factor-alpha production.
Blood
86
1995
2220
140
Aoki
Y
Hiromatsu
K
Kobayashi
N
Hotta
T
Saito
H
Igarashi
H
Niho
Y
Yoshikai
Y
Protective effect of granulocyte colony-stimulating factor against T-cell-mediated lethal shock triggered by superantigens.
Blood
86
1995
1420
141
Eichacker
PQ
Waisman
Y
Natanson
C
Farese
A
Hoffman
WD
Banks
SM
MacVittie
TJ
The cardiopulmonary effects of granulocyte colony-stimulating factor in a canine model of bacterial sepsis.
J Appl Physiol
77
1994
2366
142
Lorenz
W
Reimund
KP
Weitzel
F
Celik
I
Kurnatowski
M
Schneider
C
Mannheim
W
Heiske
A
Neumann
K
Sitter
H
Rothmund
M
Granulocyte colony-stimulating factor prophylaxis before operation protects against lethal consequences of postoperative peritonitis.
Surgery
116
1994
925
143
Haberstroh
J
Breuer
H
Lücke
I
Massarrat
K
Früh
R
Mand
U
Hagedorn
P
Brunnberg
L
von Specht
BU
Effect of recombinant human granulocyte colony-stimulating factor on hemodynamic and cytokine response in a porcine model of pseudomonas sepsis.
Shock
4
1995
216
144
Toki
S
Hiromatsu
K
Aoki
Y
Makino
M
Yoshikai
Y
Protective effects of granulocyte colony-stimulating factor on endotoxin shock in mice with retrovirus-induced immunodeficiency syndrome.
Cytokine
9
1997
781
145
Pollmächer
T
Korth
C
Mullington
J
Schreiber
W
Sauer
J
Vedder
H
Galanos
C
Holsboer
F
Effects of granulocyte colony-stimulating factor on plasma cytokine and cytokine receptor levels and on the in vivo host response to endotoxin in healthy men.
Blood
87
1996
900
146
Springer
TA
Adhesion receptors of the immune system.
Nature
346
1990
425
147
Hogg
N
Roll, roll, roll your leucocyte gently down the vein.
Immunol Today
13
1992
113
148
Strieter
RM
Kunkel
SL
Acute lung injury: The role of cytokines in the elicitation of neutrophils.
J Invest Med
42
1994
643
149
Korthuis
RJ
Anderson
DC
Granger
DN
Role of neutrophil-endothelial cell adhesion in inflammatory disorders.
J Crit Care
9
1994
47
150
Tedder
TF
Steeber
DA
Pizcueta
P
L-selectin-deficient mice have impaired leukocyte recruitment into inflammatory sites.
J Exp Med
181
1995
2259
151
Ohsaka
A
Saionji
K
Sato
N
Mori
T
Ishimoto
K
Inamatsu
T
Granulocyte colony-stimulating factor down-regulates the surface expression of the human leukocyte adhesion molecule-1 on human neutrophils in vitro and in vivo.
Br J Haematol
84
1993
574
152
Preece
G
Murphy
G
Ager
A
Metalloproteinase-mediated regulation of L-selectin levels on leucocytes.
J Biol Chem
271
1996
11634
153
Cowley
HC
Heney
D
Gearing
AJH
Hemingway
I
Webster
NR
Increased circulating adhesion molecule concentrations in patients with the systemic inflammatory response syndrome: A prospective cohort study.
Crit Care Med
22
1994
651
154
Weiss
M
Schneider
EM
Radermacher
P
Low dose G-CSF to augment host defense and counteract progression of sepsis
Yearbook of Intensive Care and Emergency Medicine 1996.
Vincent
JL
1996
150
Springer-Verlag
Berlin, Germany
155
Sessler
CN
Windsor
AC
Schwartz
M
Watson
L
Fisher
BJ
Sugerman
HJ
Fowler
AA
Circulating ICAM-1 is increased in septic shock.
Am J Respir Crit Care Med
151
1995
1420
156
Xu
BH
Gonzalo
JA
Pierre
YS
Williams
IR
Kupper
TS
Cotran
RS
Springer
TA
Gutierrez-Ramos
JC
Leukocytosis and resistance to septic shock in intercellular adhesion molecule 1-deficient mice.
J Exp Med
180
1994
95
157
Schilero
GJ
Oropello
J
Benjamin
E
Impairment in gas exchange after granulocyte colony-stimulating factor (G-CSF) in a patient with the adult respiratory distress syndrome.
Chest
107
1995
276
158
Demuynck
H
Zachee
P
Verhoef
GE
Schetz
M
van den Berghe
G
Lauwers
P
Boogaerts
MA
Risks of rhG-CSF treatment in drug-induced agranulocytosis.
Ann Hematol
70
1995
143
159
Nelson
S
Summer
W
Bagby
G
Nakamura
C
Stewart
L
Lipscomb
G
Andresen
J
Granulocyte colony-stimulating factor enhances pulmonary host defenses in normal and ethanol-treated rats.
J Infect Dis
164
1991
901
160
Hebert
JC
O’Reilly
M
Gamelli
RL
Protective effect of recombinant human granulocyte colony-stimulating factor against pneumococcal infections in splenectomized mice.
Arch Surg
125
1990
1075
161
Lister
PD
Gentry
MJ
Preheim
LC
Granulocyte colony-stimulating factor protects control rats but not ethanol-fed rats from fatal pneumococcal pneumonia.
J Infect Dis
168
1993
922
162
Abraham
E
Stevens
P
Effects of granulocyte colony-stimulating factor in modifying mortality from Pseudomonas aeruginosa pneumonia after hemorrhage.
Crit Care Med
20
1992
1127
163
Heyll
A
Aul
C
Gogolin
F
Thomas
M
Arning
M
Gehrt
A
Hadding
U
Granulocyte colony-stimulating factor (G-CSF) treatment in a neutropenic leukemia patient with diffuse interstitial pulmonary infiltrates.
Ann Hematol
63
1991
328
164
Dale
DC
The use of colony-stimulating factors for the prevention and treatment of infections.
Shock Suppl
7
1997
81
(abstr)
165
Gross-Weege
W
Weiss
M
Schneider
M
Wenning
M
Harms
B
Dumon
K
Ohmann
C
Röher
HD
Safety of a low-dosage filgrastim (rhG-CSF) treatment in non-neutropenic surgical intensive care patients with an inflammatory process.
Intensive Care Med
23
1997
16
166
Held
TK
Mielke
ME
Chedid
M
Unger
M
Trautmann
M
Huhn
D
Cross
AS
Granulocyte colony-stimulating factor worsens the outcome of experimental Klebsiella pneumoniae pneumonia through direct interaction with the bacteria.
Blood
91
1998
2525
167
Hierholzer
C
Kelly
E
Tsukada
K
Loeffert
E
Watkins
S
Billiar
TR
Tweardy
DJ
Hemorrhagic shock induces G-CSF expression in bronchial epithelium.
Am J Physiol
273
1997
L1058
168
Hierholzer
C
Kelly
E
Lyons
V
Roedling
E
Davies
P
Billiar
TR
Tweardy
DJ
G-CSF instillation into rat lungs mediates neutrophil recruitment, pulmonary edema, and hypoxia.
J Leukoc Biol
63
1998
169
169
Lundblad
R
Wang
MY
Kvalheim
G
Lingaas
E
Giercksky
KE
Granulocyte colony-stimulating factor improves myelopoiesis and host defense in fulminant intraabdominal sepsis in rats.
Shock
4
1995
68
170
Welborn
MBr
Douglas
WG
Abouhamze
Z
Auffenburg
T
Abouhamze
AS
Baumhofer
J
Seeger
JM
Pruitt
JH
Edwards
PD
Chizzonite
R
Martin
D
Moldawer
LL
Harward
TR
Visceral ischemia-reperfusion injury promotes tumor necrosis factor (TNF) and interleukin-1 (IL-1) dependent organ injury in the mouse.
Shock
6
1996
171
171
Ras
GJ
Anderson
R
Lecatsas
G
Sher
R
Acquired neutrophil dysfunction in male homosexuals with the acquired immunodeficiency syndrome.
S Afr Med J
65
1984
873
172
Takahashi
T
Wada
T
Mori
M
Kokai
Y
Ishii
S
Overexpression of the granulocyte colony-stimulating factor gene leads to osteoporosis in mice.
Lab Invest
74
1996
827
173
Hommes
DW
Meenan
J
Dijkhuizen
S
Ten-Kate
FJ
Tytgat
GN
van Deventer
SJ
Efficacy of recombinant granulocyte colony-stimulating factor (rhG-CSF) in experimental colitis.
Clin Exp Immunol
106
1996
529

Author notes

Address reprint requests to Manfred Weiss, MD, Department of Anesthesiology, Universitätsklinikum, Steinhövelstr. 9, D-89075 Ulm, Germany.

Sign in via your Institution