Macrophage-colony stimulating factor (CSF-1) signaling through its receptor (CSF-1R) promotes the differentiation of myeloid progenitors into heterogeneous populations of monocytes, macrophages, dendritic cells, and bone-resorbing osteoclasts. In the periphery, CSF-1 regulates the migration, proliferation, function, and survival of macrophages, which function at multiple levels within the innate and adaptive immune systems. Macrophage populations elicited by CSF-1 are associated with, and exacerbate, a broad spectrum of pathologies, including cancer, inflammation, and bone disease. Conversely, macrophages can also contribute to immunosuppression, disease resolution, and tissue repair. Recombinant CSF-1, antibodies against the ligand and the receptor, and specific inhibitors of CSF-1R kinase activity have been each been tested in a range of animal models and in some cases, in patients. This review examines the potential clinical uses of modulators of the CSF-1/CSF-1R system. We conclude that CSF-1 promotes a resident-type macrophage phenotype. As a treatment, CSF-1 has therapeutic potential in tissue repair. Conversely, inhibition of CSF-1R is unlikely to be effective in inflammatory disease but may have utility in cancer.

Cells of the mononuclear phagocyte system contribute to the pathology of major diseases and at the same time are essential for normal development, innate and acquired immunity, homeostasis, and tissue repair.1-6  They enter the circulation from the marrow as monocytes and leave the blood in response to a wide range of signals to either contribute to inflammatory processes or take up residence in specific locations in tissues.3,5  The function of macrophages in inflammation varies depending on the nature of the stimulus. Broadly speaking, 2 major pathways of macrophage activation have been described associated with the activation of distinct T lymphocyte immune responses. Classical activation is associated with the actions of IFN-γ and is directed toward killing of microbial pathogens.7  Alternative activation, involving responses to IL-4 or IL-13, has been associated with parasitic and allergic diseases.1  These states of macrophage activation have also been called M1 and M2, respectively, linked to the activation of Th1 and Th2 cells, and are considered mutually exclusive and antagonistic. M2 macrophages have also been ascribed functions in immunosuppression and vascularization in tumors.8,9  Biswas and Matonavani have extended the classification into further subclasses of M2-like macrophages.10  Markers of the different activation states have been proposed. For example, in the mouse, Arg1, Fizz, and Ym1 (chi3l3) have been considered M2 markers, whereas elevated MHC class II, CD86, and iNOS expression are associated with M1 polarization.1,7  However, the markers do not correlate very well with each other when analyzed across large datasets or in responses to different stimuli.1,11  In reality, each pathogen and each pathology probably generates a unique macrophage phenotype that also varies with time from the onset to the resolution (or chronic progression) of the response. Within macrophage populations, individual cells may also be infinitely heterogeneous because of the stochastic nature of transcription control.12  As Mosser and Edwards13  suggest, it is more appropriate to see macrophage heterogeneity in terms of the diversity of points on a color wheel, rather than on a linear scale between M1 and M2, or alternative and classical extremes. Within such a spectrum, it is also debatable whether the antigen-presenting dendritic cell (DC) can be considered as a separate entity distinguishable by function or markers from macrophages.4,14 

One essential regulator of macrophage homeostasis in vivo is macrophage colony-stimulating factor, or CSF-1, so named because it was the first of the hemopoietic growth factors to be isolated as a pure protein and because it can promote the growth of pure colonies of macrophages from bone marrow progenitors in semisolid media in vitro.15  Although CSF-1 has this activity, it is not the only factor that can promote macrophage growth from marrow cells, and indeed the numbers of colonies and their size are greatly increased when it acts in combination with other factors, including GM-CSF, IL-3, and IFN-γ.16-18  Nevertheless, natural mutations of the Csf-1 locus in mouse (op/op) and rat (tl/tl) confirmed that CSF-1 has a nonredundant function in controlling macrophage numbers in tissues and additionally revealed numerous pleiotropic consequences of CSF-1 deficiency, including severe growth retardation and low fertility.6,19,20  The osteopetrosis seen in CSF-1–deficient animals is a consequence of deficient production of bone-resorbing osteoclasts, which share a progenitor with macrophages and express the CSF-1 receptor.19,21,22 

Human CSF-1 cDNA was cloned independently by 2 groups in the late 1980s.23,24  We now recognize that there are actually 3 major forms of CSF-1 protein produced from alternatively spliced transcripts: the predominant secreted proteoglycan, a secreted glycoprotein, and cell-surface membrane-anchored form that can be released by proteolytic cleavage.19  Each shares an N-terminal region containing an active 149-amino acid fragment that forms a 4-helix bundle. The crystal structure of the active fragment was solved by Pandit et al.25  CSF-1 acts on its target cells by binding to CSF-1R (c-fms), a member of the type III protein tyrosine kinase receptor family. The crystal structure of the CSF-1/CSF-1R complex was described by Chen et al.26  Recently, a second ligand for the CSF-1R, IL-34, was described,27  potentially explaining some of the differences in severity between the op/op mouse and a CSF-1R mutation.28  The spatiotemporal expression of IL-34 differs from that of CSF-1, suggesting that they have distinct biologic functions.29  The two ligands acting on the same receptor are conserved across evolution to birds. CSF-1 and the binding sites on the receptor have evolved rapidly across species. By contrast, IL-34 is much more conserved across species, and computational modeling suggests that it binds to different parts of the receptor.30  In keeping with this view, Chihara et al identified mAbs binding to CSF-1R that can block CSF-1, but not IL-34 binding.31  The two proteins induced equivalent expression of chemokine genes when added to human whole blood,32  but Chihara et al found subtle difference in signal intensity between the two ligands on mouse cells.31  The very high level of conservation of IL-34 across species is atypical of immune-associated genes and could reflect an essential function. Thus far, no viable mouse knockout of IL-34 has been reported. If the knockout is lethal, where the CSF-1R knockout is known to be viable on some backgrounds,28  this would imply the existence of another receptor for IL-34. That, in turn, would have implications for the use of CSF-1R antagonists, which could lead to elevated IL-34 levels acting on an alternative target. The mRNA encoding the CSF-1R is expressed in a highly restricted manner in macrophage lineage cells in both mouse and human (www.biogps.gnf.org) and from a separate promoter (which differs between mouse and human33 ) in placental trophoblasts and in osteoclasts.34  The promoter region of the mouse locus has been characterized and used to produce a CSF-1R-EGFP transgenic mouse line, which enables the visualization of macrophages in tissues.35 

Shortly after recombinant CSF-1 became available, its ability to expand the mononuclear phagocyte system after administration in vivo was demonstrated in mice36  and subsequently in rats,37  nonhuman primates,38  and humans.39  Increased levels of circulating CSF-1 were detected in many different human disease states or animal models. Pharmacologic disruption of the CSF-1/CSF-1R axis to modulate macrophage populations has therapeutic potential in 4 broad clinical settings: inflammatory disease, cancer, autoimmunity, and bone disease (Figure 1). Accordingly, many different companies have explored different approaches to blocking CSF-1 action. In this review, we critically review animal and human studies of CSF-1 biology and explore the potential applications of both CSF-1 and CSF-1R antagonists in human medicine.

Figure 1

CSF-1 receptor signaling and blockade strategies. CSF-1 and IL-34 bind to the extracellular domain of the CSF-1R to induce dimerization and tyrosine kinase (TK)–mediated autophosphorylation of cytoplasmic tyrosine residues, leading to a cascade of intracellular signals, which regulate the production, survival, and function of macrophages. To date, no alternative receptor for IL-34 has been identified. Disruption of the CSF-1/CSF-1R axis can be achieved using neutralizing anti–CSF-1 mAbs or anti–CSF-1R mAbs (the latter can block binding of either CSF-1 or IL-34 or both cytokines) or inhibition of CSF-1R tyrosine kinase using small-molecule TK inhibitors.

Figure 1

CSF-1 receptor signaling and blockade strategies. CSF-1 and IL-34 bind to the extracellular domain of the CSF-1R to induce dimerization and tyrosine kinase (TK)–mediated autophosphorylation of cytoplasmic tyrosine residues, leading to a cascade of intracellular signals, which regulate the production, survival, and function of macrophages. To date, no alternative receptor for IL-34 has been identified. Disruption of the CSF-1/CSF-1R axis can be achieved using neutralizing anti–CSF-1 mAbs or anti–CSF-1R mAbs (the latter can block binding of either CSF-1 or IL-34 or both cytokines) or inhibition of CSF-1R tyrosine kinase using small-molecule TK inhibitors.

Close modal

CSF-1 is present in the circulation, predominantly as the proteoglycan form, at biologically active concentrations of approximately 10 ng/mL. It is produced constitutively by a wide variety of cells of mesenchymal and epithelial origin.6,19  The level in the circulation increases in many different pathologies, including infections, cancer, and chronic inflammatory disease, regardless of etiology.19,20,40  CSF-1 levels are also elevated in the circulation during pregnancy and contribute to placentation.41,42  In both mice and humans, there is a perinatal surge of tissue and circulating CSF-1.43,44  In inflammation, CSF-1 may also be produced by recruited macrophages themselves, although in the mouse at least, most macrophages do not produce CSF-1 and undergo cell death in the absence of the protein.45,46  Under normal steady-state conditions, the production of CSF-1 is balanced by its consumption by tissue macrophages, through receptor-mediated endocytosis by the CSF-1R followed by intracellular destruction.47 

Biologic effects of CSF-1 and the signaling pathways from the receptor have been reviewed in detail elsewhere.19  Addition of CSF-1 in cell culture can accelerate differentiation and maturation of monocytes into active phagocytes. In humans, CSF-1 is commonly used to generate monocyte-derived macrophages in vitro,48  and this process has been analyzed using cDNA microarrays.49  The phenotype of these monocyte-derived macrophages has been contrasted to the cells generated when monocytes are cultured in GM-CSF to produce monocyte-derived DCs, which have an increased capacity for antigen presentation.50  A meta-analysis of mouse microarray data indicates that CSF-1– and GM-CSF–stimulated cells are more similar than different, and both are clearly phagocytes.11  Nevertheless, CSF-1 as the sole stimulus polarizes macrophages away from an antigen-presenting phenotype and toward an immunosuppressive function in both mouse and human.4,14  Hence, macrophages generated in response to CSF-1 have been proposed by some to be alternatively activated or M2-like.49,51  Because CSF-1–stimulated macrophages can still respond to the M2-inducing lymphokine IL-4 with significant changes in gene expression,1,49,51,52  it is more appropriate to consider the CSF-1–stimulated cell as a distinct entity. One of the most studied and unique targets of CSF-1 signaling in mice and humans is the proteolytic enzyme urokinase plasminogen activator,53,54  a target of the ras-raf-MAPK pathway,55  which contributes to regulated fibrinolytic activity. But caution is needed when extrapolating from mouse experiments on CSF-1. In humans, but not in mice, continuous exposure to CSF-1 appears to drive a proatherogenic phenotype,48  in keeping with an inferred role in atherosclerosis.56  Broadly speaking, based on in vitro data, CSF-1 as a sole stimulus might be expected to generate actively phagocytic macrophages that promote extracellular proteolysis and tissue repair and suppress cell-mediated immunity.

The simple homeostatic mechanism that balances macrophage numbers with production and receptor-mediated clearance of CSF-147  is clearly disturbed when exogenous CSF-1 is administered, receptor-mediated clearance is saturated, and the concentration in the circulation rises. The administration of CSF-1 can generate a transient increase in c-fos mRNA in the spleen, providing a simple in vivo bioassay and suggesting that CSF-1 availability is not saturating for tissue macrophages.57  To generate a more sustained response leading to measurable changes in macrophage numbers, the molecular size of CSF-1 becomes critical because macrophages and their progenitors require continued exposure to CSF-1 to enter the cell cycle.58  The unglycosylated minimal active fragment of CSF-1, a disulphide-linked homodimer produced in bacteria by Chiron, is cleared rapidly by the kidney with a half-life of 1 to 2 hours.59  The recombinant protein produced by Genetics Institute was made in mammalian cells and was a secreted 70- to 90-kDa glycosylated protein, with a longer C terminus, closely resembling the form that can be isolated from human urine.24  In consequence, the larger protein has a much slower rate of renal clearance and a 5-fold lower dose of the mammalian-expressed protein (0.5-1 mg/kg per day) than the bacterial recombinant protein (4 mg/kg per day), was required to generate substantial responses in mice.

The first demonstrated efficacy of recombinant human CSF-1 in mice showed that treatment each day for 4 days caused a 10-fold increase in blood monocyte numbers.36  More recently, CSF-1 was shown to increase conventional and plasmacytoid DC numbers.60,61  The latter finding is consistent with the expression of CSF-1R (CD115) on the shared macrophage/DC progenitor cell and on mature DCs themselves.61,62  Prolonged CSF-1 administration to mice also altered osteoclastic function and generated an increase in serum markers of bone turnover.22  There was no net change in bone mineral density, presumably because of the coupling of osteoclast and osteoblast function. Indeed, there was an unexpected and unexplained increase in the trabecular bone, which would suggest the possible use of CSF-1 as an anabolic agent.

The maximal response to CSF-1 apparently required the repeated administration.36  Ulich et al conducted a single dose escalation study in rats, and a dose of 0.5 to 1 mg/kg caused a transient spike in monocyte count of approximately 5- to 10-fold after 24 hours, which returned to baseline by 36 hours.37  Prolonged treatment led to maintenance of the elevated monocyte count, and the only apparent toxicity was a thrombocytopenia. Subsequent studies progressed to rabbits and nonhuman primates38,63  and used continuous intravenous infusion or twice-daily subcutaneous injection. Repeated infusions after 2 weeks generated comparable increases in monocyte count. The same group subsequently noted a substantial decrease in plasma cholesterol in the treated animals63 ; precisely how this is related to the up-regulation of cholesterol biosynthesis in human monocyte-macrophages by CSF-148  is unclear.

This initial study of treatment of mice with CSF-1 did not distinguish among subpopulations of monocytes. Blood monocytes in mouse and human are heterogeneous in terms of surface markers. In humans, two populations have been distinguished based on the level of expression of the Fc receptor, CD16, which varies inversely with that of the lipopolysaccharide coreceptor, CD14.64,65  Similarly in mice, the expression of Ly6C and the fractalkine receptor CX3CR1 varies inversely to distinguish two subpopulations that are thought to be functionally equivalent to the human populations.64,66  One monocyte population (CD14 (hi) or Ly6C (hi) in human and mouse, respectively) is short-lived in the circulation and recruited in response to inflammatory stimuli, notably in response to chemokines that interact with the receptor CCR2.64,66,67  A recent paper from a consortium of investigators suggested these be called “classical” rather than “inflammatory” monocytes.65  By contrast, the precursors of resident tissue macrophages, subsets of which have also been reported to patrol vessel walls,68  were referred to as “nonclassical.” There was also an “intermediate” population identified. These designations are rather artificial; subdivision seems to require a rather arbitrary assignment of the location of gates on a flow cytometer. As discussed below in the context of anti–CSF-1R treatments, there is evidence that the monocyte “subpopulations” are actually a maturation series controlled by CSF-1.65,69  Accordingly, where CSF-1 caused a 5-fold increase in blood monocytes in nonhuman primates, the large majority expressed the “nonclassical” maturation marker, CD16.38  Furthermore, in keeping with the role of “nonclassical” cells as precursors of tissue macrophages, CSF-1 treatment of mice also caused a very large increase in resident tissue macrophage numbers and in expression of the maturation marker, F4/80.36 

CSF-1 has had limited application as a hemopoietic growth factor in the setting of bone marrow transplantation or during recovery from myelosuppressive chemotherapy, where other myeloid growth factors, such as G-CSF and GM-CSF, are in common use.70  One reported phase 3 trial by Masaoka et al, using purified CSF-1 from human urine, reported a reduction in circulating granulocyte recovery time and improved survival without retransplantation in marrow transplant patients.71  Similarly, Hidaka et al reported the use of CSF-1 (also known as Mirimostim) in patients with severe neutropenia after chemotherapy for ovarian cancer.72  Their data suggested an improvement in NK-cell function and numbers and T-cell maturation as well as an improvement in granulocyte function that was not improved by combination with G-CSF. Arguably, there is a rationale for exploring the efficacy of combined treatments CSF-1 and GM-CSF. In the mouse, the two factors act together on high proliferative potential precursors to generate very large colonies.16,17  In humans, very low concentrations of GM-CSF were required to generate large macrophage colonies in standard colony assays using CSF-1.18 

Phase 1 clinical trials of recombinant CSF-1 (produced by Genetics Institute) by continuous infusion in humans with advanced melanoma confirmed the ability to increase circulating monocyte numbers.73,74  The increases reversed rapidly on cessation of infusion and were reproduced on reinfusion. The transient thrombocytopenia and the decrease in circulating cholesterol seen in the previous animal studies were confirmed in these patients. The choice of melanoma as target for such trials was based in part on animal studies in which CSF-1 prevented metastasis of the B16 melanoma75  and evidence for the ability of CSF-1–stimulated cells to kill melanoma cells in vitro.38  Although there was anecdotal evidence of efficacy in a small number of the treated patients in these trials, to our knowledge there has not been a follow-up trial. A separate phase 1 study in cancer patients using rhCSF-1 produced by Chiron also observed mild thrombocytopenia in some persons and also unexplained hyperglycemia.76  This study did not identify any therapeutic effect on the cancers.

The other initial target for CSF-1 therapy was fungal diseases, where CSF-1–stimulated macrophages were purported to phagocytose organisms, such as Candida.77  There was no reported follow-up from the initial clinical studies in humans that suggested an increase in survival rate of bone marrow transplant patients with disseminated candidiasis78 ; it is not known whether this was the result of a lack of commercial interest or a lack of reproducibility. Around the time of these studies, it became clear that CSF-1 treatment could exacerbate the pathology of certain diseases in animal infectious and inflammatory disease models, not surprisingly, because pathology can be macrophage-mediated.79  Several studies have explored the role of CSF-1 in the pathology of autommune nephritis, combining studies of ectopic CSF-1 with increased or reduced CSF-1 levels in the op/op or transgenic mice overexpressing various forms of the protein.80-82  These studies indicated that CSF-1 drives increased production of inflammatory or “classical” macrophages from the marrow and also local proliferation within the kidney.83  Indeed, contrasting studies of candidiasis were informative. Whereas pretreatment of animals with CSF-1 was found to be protective,77  treatment of animals with an established disseminated candidiasis accelerated the disease.77,84  On initial exposure, CSF-1–stimulated macrophages are able to recognize and clear the pathogen. Once the disease is established, there is massive macrophage infiltration of the affected organs (the kidneys); these cells produce inflammatory cytokines that mediate the pathology. Accordingly, the CSF-1 treatment drastically accelerated weight loss in the Candida-infected mice.84 

The potential importance of CSF-1 in the regulatory balance of antigen presentation is supported by recent evidence of csf1r gene polymorphisms associated with altered CSF-1R expression being linked to asthma susceptibility.85  The gene encoding the ligand, CSF-1, also varies between persons and is strongly linked to Paget disease susceptibility.86  The ability of macrophages in certain activation states to suppress T-cell responses and to elicit tolerance has been known for a very long time.14,87  As noted in “Effect of CSF-1 in vitro,” in both mice and humans, populations of macrophages polarized with CSF-1 as the sole stimulus are generally relatively poor at stimulating T cells in vitro. In vivo, the F4/80 antigen, which is inducible by CSF-1,36  increased on maturation, and generally considered a macrophage marker,3,4  is required for the generation of oral tolerance. Furthermore, F4/80 KO mice are deficient in regulatory T cells (Tregs).88  An F4/80+ macrophage population, which expresses the transcription factor Foxp3, normally expressed by Treg, was recently identified and apparently suppressed T-cell responses and induce Treg conversion in vivo.89  This work was subsequently retracted. Nevertheless, in mice, treatment with CSF-1 has been shown to cause profound immunosuppression; T cells from the spleens of treated animals were unresponsive to T-cell mitogens or allogeneic cells.90  CSF-1 was also shown to directly suppress allogeneic T-cell responses in vitro91  and cutaneous hypersensitivity responses in vivo.92 

CSF-1 was reported to be substantially elevated in the circulation in acute GVHD in mice.93  Blazar et al were the first to assess the impact of CSF-1 treatment in bone marrow transplantation, examining T cell-depleted grafts, with and without a potential NK-cell contribution to their recognition by the recipient.94  They found an inhibitory effect of CSF-1 on engraftment, but only if the recipient NK cells were able to recognize and eliminate the graft. Indeed, others had reported that CSF-1 treatment led to increases in the numbers of NK cells and their mobilization from the marrow by an unknown mechanism.95  More recently, the effect of recipient CSF-1 pretreatment was investigated in an acute GVHD model after transfer of BALB/c bone marrow and spleen cells into irradiated C57Bl/6 recipients.96  Pretreatment reduced the severity of disease without apparently altering engraftment. These data were used to argue for distinct functions of “macrophages” versus “DCs” in GVHD pathology. We have pretreated B6D2F1 recipients for 4 days with CSF-1 at a 2× higher dose than used by Hashimoto et al,96  before irradiation and transplantation of parental B6 bone marrow supplemented with splenic T cells. Although we saw expansion of both macrophages and CD11c-positive cells in the recipient mice as expected from previous studies,36,61  the pretreatment had no effect on survival or GVHD severity (K.P.A.M. and S. Oliver, unpublished data, May 2008). Similarly, in a CD8 T cell–dependent model (C3H.SW into B6), we also found no effect of pretreatment (K.P.A.M. and S. Oliver, unpublished data, January 2009). We can only conclude that a limiting role for macrophage-mediated removal of alloreactive T cells is a specific feature of the model or strain combination used by Hashimoto et al.96  Pretreatment of a human bone marrow transplant recipient with CSF-1 has not been reported, and the impact is difficult to predict from the mouse studies, which are specifically designed to generate a substantial GVH response. There is, thus far, no reproducible evidence of clinical utility for CSF-1 as a treatment to suppress acute GVHD. Two separate studies in Japan71,97  and one in the United States98  found that CSF-1 (M-CSF) treatment had no effect on acute GVHD or relapse rate. The former study reported a small impact on extensive chronic GVHD.

The more promising trend in the recent studies of CSF-1 as a therapeutic agent relates to its trophic actions.6,21  Building on the evidence of the role of CSF-1 in reproduction and pregnancy mentioned above, and initial data on rats,99  Nishimura et al found a correlation between elevated CSF-1 in the serum and successful ovulation induction in an in vitro fertilization program.100  They subsequently reported that coadministration of CSF-1 with gonadotrophins could increase the numbers of oocytes, fertilized eggs, and transferred embryos101 

The pro-repair phenotype noted in “Effects of CSF-1 in vitro” includes the expression of numerous growth factors for other cells (eg, IGF-1, PDGFB, and VEGFA), endocytic receptors (eg, macrophage scavenger receptors), and proteolytic enzymes in addition to urokinase plasminogen activator.6,19-21  CSF-1 probably contributes to the local macrophage proliferation that is one hallmark of Th2-driven inflammation,102  especially because mouse inflammatory macrophages recruited by a sterile stimulus can be autocrine for CSF-1.45 

Among the systems that have been studied are fracture repair and macrophage-osteoblast interactions,103  repair after ischemia in the kidney104,105  and heart,106  promotion of angiogenesis,107  and elimination of amyloid deposits in the brain.108,109  Two biologic issues arise from these studies. First, there is the question of whether CSF-1–stimulated macrophages can interconvert into cells of mesenchymal lineages. We found some evidence using a CSF-1r–EGFP transgenic reporter that myeloid cells can give rise to myofibroblasts.110  In the case of angiogenesis, there is some evidence for bone marrow precursors of endothelial cells. Monocytes can apparently transdifferentiate into endothelium in vitro, but in vivo the major effect of CSF-1 is to promote production of proangiogenic growth factors.107  The second issue is whether the CSF-1R is expressed on cells outside of the mononuclear phagocyte lineage. Menke et al found that CSF-1 treatment can promote repair in a renal ischemia reperfusion model in mice and suggested that the effects were partly the result of direct effects on the tubular epithelial cells, which they claimed expressed significant levels of CSF-1R in response to the insult.105  Jose et al saw no evidence of CSF-1R protein expression by renal epithelial cells in rejecting renal allografts, where the ligand CSF-1 was also expressed very highly by tubular cells.83  According to Menke et al, a nonmacrophage-mediated mechanism of CSF-1 action was supported by depleting the animals of CD11b-positive cells using a CD11b-DTR transgene,105  but these authors did not apparently detect the resident macrophages, which are actually extremely numerous in the kidney and do not express CD11b.111  Macrophages may be a significant contaminant of primary renal epithelial cell cultures as they are in cultured calvarial osteoblasts; thus, contaminating macrophages could explain the apparent expression of CSF-1R.112  Alikhan et al recently published the same basic observation: that CSF-1 can promote renal repair after reversible ischemia.104  But using the same CSF-1R–EGFP transgene, they did not see any evidence of induced CSF-1R expression by tubular epithelial cells and instead demonstrated that the treatment-increased macrophage recruitment, polarized the cells toward an M2-like phenotype and promoted production of growth factors, such as IGF1. That finding built on an earlier observation: that CSF-1 can promote the growth of embryonic kidney in tissue culture,113  again a system in which there was no evidence of expression of CSF-1R in the renal parenchyma outside of the macrophages.

There are two approaches to blocking the action of CSF-1: the use of inhibitors directed against the protein tyrosine kinase activity of the receptor and the use of agents that block the binding of CSF-1 to its receptor (Figure 2). The latter category includes antibodies against the receptor, antibodies against the ligand, and soluble receptors. There are several complexities that arise from the use of CSF-1R inhibitors that are commonly neglected.

Figure 2

CSF-1 regulation of macrophage development in the mouse and the effects of prolonged M279 anti–CSF-1R mAb treatment. HSCs give rise to common myeloid precursors, which generate monocyte-DC precursors that in steady state give rise to DC precursors and CSF-1R+Ly6Chi and CSF-1R+Ly6Clo monocytes. Monocytes enter the circulation from the marrow and are signaled to exit the blood to contribute to inflammatory processes (CSF-1R+Ly6Chi) or to take up residence in specific locations (CSF-1R+Ly6Clo). CSF-1 is a critical regulator of the differentiation, proliferation, and survival of CSF-1R+Ly6Clo monocyte-derived tissue macrophages, alternatively activated macrophages, and tumor-associated macrophages. The cellular sources of CSF-1 are primarily mesenchymal in origin, but macrophages and tumors can also secrete this cytokine. Treatment with CSF1R blocking antibody M279 selectively depleted the CSF-1R+Ly6Clo monocyte precursor of resident tissue macrophages, whereas CSF-1R+Ly6Chi inflammatory monocytes were increased. Within tissue, the M279 mAb may prevent differentiation and proliferation of resident macrophage populations and tumor-associated macrophages.

Figure 2

CSF-1 regulation of macrophage development in the mouse and the effects of prolonged M279 anti–CSF-1R mAb treatment. HSCs give rise to common myeloid precursors, which generate monocyte-DC precursors that in steady state give rise to DC precursors and CSF-1R+Ly6Chi and CSF-1R+Ly6Clo monocytes. Monocytes enter the circulation from the marrow and are signaled to exit the blood to contribute to inflammatory processes (CSF-1R+Ly6Chi) or to take up residence in specific locations (CSF-1R+Ly6Clo). CSF-1 is a critical regulator of the differentiation, proliferation, and survival of CSF-1R+Ly6Clo monocyte-derived tissue macrophages, alternatively activated macrophages, and tumor-associated macrophages. The cellular sources of CSF-1 are primarily mesenchymal in origin, but macrophages and tumors can also secrete this cytokine. Treatment with CSF1R blocking antibody M279 selectively depleted the CSF-1R+Ly6Clo monocyte precursor of resident tissue macrophages, whereas CSF-1R+Ly6Chi inflammatory monocytes were increased. Within tissue, the M279 mAb may prevent differentiation and proliferation of resident macrophage populations and tumor-associated macrophages.

Close modal
  1. CSF-1R is part of a family of receptors, the intracellular domains of which are very closely related. Of the many available inhibitors,114  GW2580 has the greatest apparent specificity for CSF-1R versus related kinases, such as c-kit and Flt3.115  This inhibitor does not inhibit the avian CSF-1R,30  so specificity is not solely determined by the ATP-binding pocket (and the inhibitor is therefore more likely to be truly specific). But in vitro assays are not entirely predictive of the activity in whole cells.45 

  2. With the discovery of a second ligand for the CSF-1 receptor, IL-34, there is the question for antibodies of whether they inhibit both of the ligands.27,30  Both anti–mouse CSF-1R antibodies described in the paragraphs below inhibit both CSF-1 and IL-34 actions29  (and D.A.H. and L. Bonham, unpublished data, September 2006), although as noted earlier, the two ligands probably bind different parts of the receptor, and selective monoclonals have also been described.31 

  3. Because CSF-1 is cleared from the circulation by receptor-mediated endocytosis,47  CSF-1R blockade causes a massive elevation of circulating CSF-1 concentration. As the antibody concentration subsides, there is a consequential enhanced CSF-1 signal and rebound monocytopoiesis. This will also occur if CSF-1R–bearing cells are killed, for example, by toxic liposomes or with macrophage-directed toxic transgenes.116  However, it does not occur when CSF-1R kinase inhibitors are added, unless they are toxic, because receptor-mediated internalization of CSF-1 does not require the kinase activity of the receptor.45 

There has been relatively little use made of anti–CSF-1 antibodies. Wei et al generated a neutralizing rabbit anti-serum and examined the effect of PEGylated-antibody on postnatal development.117  They essentially recapitulated many of the growth retardation and developmental abnormalities of the op/op mouse, supporting the view that the postnatal surge of CSF-1 is important for organ maturation and somatic growth.6,21  Lokeshwar and Lin generated a rat anti–CSF-1 neutralizing mAb,118  which has been used to demonstrate a role for CSF-1 in inflammation and joint erosion in collagen-induced arthritis.119  The 5A1 mAb was also used in ovariectomized mice to demonstrate CSF-1 dependence in estrogen deficiency-induced bone loss,120  raising the possibility that such therapy could be applied in human osteoporosis. Anti–CSF-1 mAb has been tested in nonhuman primates,121  generating a selective depletion of the “mature” or resident CD16+ monocyte population and a time-dependent loss of Kupffer cells from the liver. These findings support the role of CSF-1 in monocyte maturation discussed in “The role of CSF-1 in monocyte maturation.” A phase 1/2 clinical trial of anti–CSF-1 antibody in prostate cancer with bone metastasis, to reduce osteolytic activity, was commenced by Novartis but discontinued for strategic reasons (http://clinicaltrials.gov/ct2/show/NCT00757757).

The first neutralizing mAb directed against CSF-1R, AFS98, was produced by Sudo et al, who used it to demonstrate the redundancy of CSF-1 in the generation of monocyte precursors,122  effectively supporting the conclusions from the op/op mouse in which there is no reduction of total circulating monocytes.28  Conversely, AFS98 has been used in several studies to address the functional role of CSF-1 signaling in pathology. For example, Murayama et al found that injection of the antibody (2 mg/day) on alternate days for 6 weeks reduced macrophage accumulation in atherosclerotic lesions in apoE-deficient mice.123  Similarly, Jose et al found that daily administration of AFS98 greatly reduced local proliferation of infiltrating macrophages in renal allografts.83  Lim et al injected similar doses for 6 weeks and observed suppression of macrophage accumulation in a model of diabetic nephropathy,124  and Segawa et al found that AFS98 administration reduced macrophage infiltration into damaged skeletal muscle.125  In the latter case, the effect was to increase fibrosis, in keeping with the antifibrotic actions of CSF-1–stimulated macrophages noted in the renal ischemia-reperfusion model discussed above. Kubota et al reported that daily AFS98 treatment (50 mg/kg) reduced macrophage numbers in an implanted osteosarcoma model and reduced vascularization, lymphangiogenesis, and tumor growth.126  Lenzo et al found that AFS98 treatment reduced the accumulation of exudate macrophages in two peritoneal and one lung inflammation model.127 

Results with AFS98 contrast to the conclusions obtained using a different monoclonal anti–CSF-1R antibody, M279, which binds the receptor with significantly greater affinity.69  Prolonged treatment with this antibody selectively removed tissue macrophage populations, including those found in growing tumors, but had no protective effect in several inflammatory models, including lipopolysaccharide-induced lung inflammation, tissue injury, GVHD, or the same peritoneal model studied by Lenzo et al.127  The tumor studies were rather differently designed than those of Kubota et al126  because the tumor was allowed to establish before treatment commenced and M279 anti–CSF-1R was still able to deplete the macrophages. CSF-1–dependent macrophages may have many roles in tumor progression and metastasis, and in humans the receptor may itself be expressed by tumor cells.6,128,129  In our studies, we saw no effect of anti–CSF-1R on tumor growth, despite the macrophage depletion, but that finding does not preclude possible applications in cancer therapy.

Interestingly, the M279 antibody treatment actually led to exacerbation of GVHD, an outcome later confirmed using AFS98 by others.96  In our study using M279,69  we argue that CSF-1–dependent tissue macrophages generate peripheral tolerance and their removal permits excessive alloreactive T-cell activation. In the subsequent study,96  the authors argue that CSF-1–dependent cells are involved in the clearance of the autoreactive T cells (host vs graft), a claim connected to the apparent protective effect of pretreatment with CSF-1 in the same model discussed above.

Consistent with the lack of effect on inflammation, prolonged M279 antibody treatment of mice had no effect on the total monocyte count. Treatment ablated the more mature F4/80hiLy6Clo monocyte subpopulation, with a corresponding increase in the Ly6Chi cells suggesting a maturation block (Figure 2). As expected, based on the function of these cells as precursors of resident macrophages,64  with time, the treatment removed the large majority of tissue macrophages. The exceptions were the macrophages of the lung, brain, uterus, and ovary, and subpopulations within lymphoid tissues. The lack of impact within the brain did not reflect CSF-1 independence of microglia because these cells were depleted in nervous tissue outside the blood-brain barrier, including the retina and spinal cord. The comparative lack of effect of M279 administration on the macrophages of the uterus contrasts with a study of the role of local CSF-1 in macrophage recruitment during pregnancy,130  where treatment with AFS98 caused almost complete macrophage depletion in the uterus within 3 days. There is certainly a substantial depletion of macrophages in the uterus of the op/op mouse,131  so the lack of effect of M279 is probably the result of the local production of CSF-1 in the uterus and long half-life of the resident macrophages in the virgin females.130  In other sites, the pattern of response to M279 anti–CSF-1R was entirely consistent with the CSF-1 dependence of tissue macrophage populations based on examination of the op/op and CSF-1r (−/−) mice, the known turnover rates of tissue macrophages, and the known function of ly6Chi monocytes in inflammation.64,65,69 

The acute loss of monocytes and tissue macrophages seen with AFS98 in the majority of studies suggests that this antibody, unlike M279, is either directly toxic, or it promotes clearance of monocytes/macrophages. Indeed, the effects of the antibody in the GVHD model of Hashimoto et al were mimicked by toxic liposomes.96  This finding immediately raises additional issues about interpretation and mechanism because macrophages on the surface of bone are a critical part of the stem niche, and their depletion leads to stem cell mobilization.132  Within days, AFS98 administration caused much greater depletion of splenic96  and uterine130  macrophages than we observed after prolonged treatment with M279. Among several differences between the antibodies, M279 is a rat IgG1, which is bound poorly by mouse FcRn, which regulates the half-life in the serum, whereas AFS98 is a rat IgG2A, which binds with high affinity.133  So, the doses of IgG2A used by Hashimoto et al (1-2 mg/injection) could generate sustained very high circulating concentrations of rat IgG2A.96  Second, IgG2A binds selectively to the high affinity cytophilic antibody receptor (CD64) on mouse macrophages134  and could trigger recognition by and/or aggregation with other macrophages. The effect of M279 seems more likely to represent the biology of CSF-1R signaling as opposed to less specific macrophage depletion seen with AFS98.

The major difference between antibodies and kinase inhibitors is that the latter are more likely to block autocrine actions of endogenous CSF-1, which is less accessible to antibodies. Irvine et al demonstrated that a novel CSF-1R kinase inhibitor could repress autocrine signaling in mouse inflammatory macrophages, leading to reduced expression of inflammatory cytokines.45  GW2580 is probably the most selective and best characterized of the available inhibitors. There are limited pharmacologic data on the inhibitor.115,135  Treatment of mice with the drug had a small effect on thioglycollate-elicited macrophage recruitment115 ; and in a later study, despite relatively high dosing twice daily, it had limited effects in a rat arthritis model.135  Rather remarkably, there was actually a dose-related increase in blood monocyte count in normal rats after prolonged treatment.135  Priceman et al treated mice with even higher doses of GW2580 (160 mg/kg) to test the role of CSF-1R in recruitment of macrophages into growing tumors.136  Despite this increased dose, there was a small decrease in tumor macrophages and no change in the numbers of monocytes/macrophages in marrow, peripheral blood, or spleen of naive mice, by contrast to the substantial effects on tissue macrophage numbers claimed by Hashimoto et al using a significantly lower dose (1.6 mg/mouse, ∼ 60 mg/kg) only once daily, after 6 days.96 

The only other CSF-1R inhibitor for which there are significant data in vivo is Ki20227. Kubota et al reported in their study of tumor angiogenesis that the inhibitor at 50 mg/kg daily subcutaneously had similar effects to AFS98 on the numbers of macrophages in tumors, and consequent angiogenesis.126  The same inhibitor was found to reduce macrophage numbers and associated pathology in models of inflammatory arthritis137  and encephalomyelitis.138  Even after prolonged treatment, the inhibitor caused only a marginal increase in circulating CSF-1 levels,138  by contrast to the massive escalation seen in CSF-1R–deficient mice.28  This implies that the inhibitor does not significantly reduce the numbers of CSF-1R–expressing tissue macrophages that normally clear the ligand from the circulation.47  Conversely, the inhibitor did greatly reduce the numbers of Ly6G-positive granulocytes which is not seen with either of the anti–CSF-1R antibodies, so there must be some concerns as to its specificity. There are several other less specific inhibitors that can block CSF-1R kinase activity but have broader specificity for other kinases. One orally available molecule, JNJ-28312141,139  has substantial activity against CSF-1R and the related Flt3. This inhibitor was found to decrease Kupffer cell numbers by approximately 40% and generated a somewhat larger increase in circulating CSF-1 (still < 2-fold) than Ki20227. Like the other molecule, Ki20227, the JNJ inhibitor reduced macrophage numbers in transplanted tumors and constrained tumor growth.139  Another molecule from the same company, dubbed fms-I, has been studied in the ureteric obstruction model of nephritis and in a glomerulonephritis model in rats.80  This inhibitor at low doses prevented local monocyte proliferation, and at higher doses drastically depleted blood monocytes and resident tissue macrophages, at least in the kidney. A molecule that inhibits CSF-1 kinase activity that is already in widespread clinical use is imatinib (STI571, Gleevec), which was originally developed as an inhibitor of ABL kinase for the treatment of chronic myeloid leukemia.140  Paniagua et al found that imatinib and GW2580 have similar efficacy in 3 different mouse models of autoimmune arthritis.141  We have tested the effect of maximally tolerated doses of imatinib using the MacGreen reporter mice. Even at doses that caused neutropenia and liver damage, we saw no evidence of the depletion of monocytes or resident tissue macrophages seen with M279 antibody (D.A.H., unpublished observations, September 2006). Dose-limiting toxicity probably occurs through inhibition of other kinases at concentrations below those required to inhibit CSF-1R kinase in vivo. Aside from these examples, none of the other patented CSF-1R inhibitors reviewed recently114  has made it into published animal trials, let alone human treatments.

In conclusion, CSF-1 as a candidate therapeutic agent is undergoing something of a renaissance in the context of tissue repair. IL-34 has not yet been tested, and we do not yet know whether it could target other receptor(s) as well as CSF-1R. A constraint on its development is that human IL-34 is not active on rodent cells,27  and it does not refold from bacterial expression. Extrapolation from animal studies of CSF-1 to humans needs to be approached with caution; a large animal model, such as the pig,142  could be a sensible intermediate. Furthermore, applications of CSF-1 must still be approached cautiously; there is the potential to make macrophage-mediated pathology considerably worse if it is applied early in the onset of disease.

The impact of the M279 anti–CSF-1R antibody in the mouse, combined with the effects of CSF-1 treatment and the phenotype of the CSF-1–deficient mouse and rat, suggests that the indispensible function of the CSF-1R signaling is to promote monocyte maturation to form the precursors of resident tissue macrophages. The available data clearly demonstrate that CSF-1R is not a good target for anti-inflammatory therapy. The high levels of CSF-1 seen in many inflammatory diseases are probably a part of a feedback damage repair response, and on that basis it could even be counterproductive to target the receptor. The exception may be where immunosuppression in certain infectious diseases and cancer is associated with very high levels of CSF-1, in which case anti–CSF-1R mAb treatments, in contrast to tyrosine kinase inhibitors, could restore effective immunity.

Anti–CSF-1R treatment could well provide a route to manipulation of macrophage numbers in tumors, where they are basically the resident tissue macrophages of a new organ. One application for anti–CSF-1R would be as an adjunct therapy to prevent regrowth after surgical or therapy-associated regression, a circumstance in which large numbers of macrophages are recruited and will otherwise contribute their trophic functions to regrowth of the tumor. The trophic role of macrophages in regrowth was first demonstrated in the op/op mouse, where transplantable tumor growth and vascularization are severely compromised.143  By analogy, CSF-1 deficiency in mice is associated with deficient hepatocyte proliferation after partial hepatectomy.144  Against that benchmark, we argue that the AFS98 antibody and the various kinase inhibitors probably elicit distinct effects through distinct mechanisms that may, or may not, involve CSF-1R signaling. In a sense, that is unimportant. As in the case of imatinib, which probably does not act solely as an Abl kinase inhibitor, the mechanism may end up being unrelated to the original hypothesis, but efficacy and therapeutic index are all that matters.

Contribution: D.A.H. and K.P.A.M. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: David A. Hume, Roslin Institute, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, Edinburgh, United Kingdom; e-mail: david.hume@roslin.ed.ac.uk.

1
Gordon
 
S
Martinez
 
FO
Alternative activation of macrophages: mechanism and functions.
Immunity
2010
, vol. 
32
 
5
(pg. 
593
-
604
)
2
Gordon
 
S
Taylor
 
PR
Monocyte and macrophage heterogeneity.
Nat Rev Immunol
2005
, vol. 
5
 
12
(pg. 
953
-
964
)
3
Hume
 
DA
The mononuclear phagocyte system.
Curr Opin Immunol
2006
, vol. 
18
 
1
(pg. 
49
-
53
)
4
Hume
 
DA
Differentiation and heterogeneity in the mononuclear phagocyte system.
Mucosal Immunol
2008
, vol. 
1
 
6
(pg. 
432
-
441
)
5
Hume
 
DA
Ross
 
IL
Himes
 
SR
Sasmono
 
RT
Wells
 
CA
Ravasi
 
T
The mononuclear phagocyte system revisited.
J Leukoc Biol
2002
, vol. 
72
 
4
(pg. 
621
-
627
)
6
Pollard
 
JW
Trophic macrophages in development and disease.
Nat Rev Immunol
2009
, vol. 
9
 
4
(pg. 
259
-
270
)
7
Schroder
 
K
Hertzog
 
PJ
Ravasi
 
T
Hume
 
DA
Interferon-gamma: an overview of signals, mechanisms and functions.
J Leukoc Biol
2004
, vol. 
75
 
2
(pg. 
163
-
189
)
8
Porta
 
C
Rimoldi
 
M
Raes
 
G
, et al. 
Tolerance and M2 (alternative) macrophage polarization are related processes orchestrated by p50 nuclear factor kappaB.
Proc Natl Acad Sci U S A
2009
, vol. 
106
 
35
(pg. 
14978
-
14983
)
9
Martinez
 
FO
Sica
 
A
Mantovani
 
A
Locati
 
M
Macrophage activation and polarization.
Front Biosci
2008
, vol. 
13
 (pg. 
453
-
461
)
10
Biswas
 
SK
Mantovani
 
A
Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm.
Nat Immunol
2010
, vol. 
11
 
10
(pg. 
889
-
896
)
11
Mabbott
 
NA
Kenneth Baillie
 
J
Hume
 
DA
Freeman
 
TC
Meta-analysis of lineage-specific gene expression signatures in mouse leukocyte populations.
Immunobiology
2010
, vol. 
215
 
9
(pg. 
724
-
736
)
12
Ravasi
 
T
Wells
 
C
Forest
 
A
, et al. 
Generation of diversity in the innate immune system: macrophage heterogeneity arises from gene-autonomous transcriptional probability of individual inducible genes.
J Immunol
2002
, vol. 
168
 
1
(pg. 
44
-
50
)
13
Mosser
 
DM
Edwards
 
JP
Exploring the full spectrum of macrophage activation.
Nat Rev Immunol
2008
, vol. 
8
 
12
(pg. 
958
-
969
)
14
Hume
 
DA
Macrophages as APC and the dendritic cell myth.
J Immunol
2008
, vol. 
181
 
9
(pg. 
5829
-
5835
)
15
Stanley
 
ER
Cifone
 
M
Heard
 
PM
Defendi
 
V
Factors regulating macrophage production and growth: identity of colony-stimulating factor and macrophage growth factor.
J Exp Med
1976
, vol. 
143
 
3
(pg. 
631
-
647
)
16
Breen
 
FN
Hume
 
DA
Weidemann
 
MJ
Interactions among granulocyte-macrophage colony-stimulating factor, macrophage colony-stimulating factor, and IFN-gamma lead to enhanced proliferation of murine macrophage progenitor cells.
J Immunol
1991
, vol. 
147
 
5
(pg. 
1542
-
1547
)
17
Breen
 
FN
Hume
 
DA
Weidemann
 
MJ
The effects of interleukin 3 (IL-3) on cells responsive to macrophage colony-stimulating factor (CSF-1) in liquid murine bone marrow culture.
Br J Haematol
1990
, vol. 
74
 
2
(pg. 
138
-
145
)
18
Caracciolo
 
D
Shirsat
 
N
Wong
 
GG
Lange
 
B
Clark
 
S
Rovera
 
G
Recombinant human macrophage colony-stimulating factor (M-CSF) requires subliminal concentrations of granulocyte/macrophage (GM)-CSF for optimal stimulation of human macrophage colony formation in vitro.
J Exp Med
1987
, vol. 
166
 
6
(pg. 
1851
-
1860
)
19
Chitu
 
V
Stanley
 
ER
Colony-stimulating factor-1 in immunity and inflammation.
Curr Opin Immunol
2006
, vol. 
18
 
1
(pg. 
39
-
48
)
20
Sweet
 
MJ
Hume
 
DA
CSF-1 as a regulator of macrophage activation and immune responses.
Arch Immunol Ther Exp (Warsz)
2003
, vol. 
51
 
3
(pg. 
169
-
177
)
21
Gow
 
DJ
Sester
 
DP
Hume
 
DA
CSF-1, IGF-1, and the control of postnatal growth and development.
J Leukoc Biol
2010
, vol. 
88
 
3
(pg. 
475
-
481
)
22
Lloyd
 
SA
Yuan
 
YY
Simske
 
SJ
Riffle
 
SE
Ferguson
 
VL
Bateman
 
TA
Administration of high-dose macrophage colony-stimulating factor increases bone turnover and trabecular volume fraction.
J Bone Miner Metab
2009
, vol. 
27
 
5
(pg. 
546
-
554
)
23
Kawasaki
 
ES
Ladner
 
MB
Wang
 
AM
, et al. 
Molecular cloning of a complementary DNA encoding human macrophage-specific colony-stimulating factor (CSF-1).
Science
1985
, vol. 
230
 
4723
(pg. 
291
-
296
)
24
Wong
 
GG
Temple
 
PA
Leary
 
AC
, et al. 
Human CSF-1: molecular cloning and expression of 4-kb cDNA encoding the human urinary protein.
Science
1987
, vol. 
235
 
4795
(pg. 
1504
-
1508
)
25
Pandit
 
J
Bohm
 
A
Jancarik
 
J
Halenbeck
 
R
Koths
 
K
Kim
 
SH
Three-dimensional structure of dimeric human recombinant macrophage colony-stimulating factor.
Science
1992
, vol. 
258
 
5086
(pg. 
1358
-
1362
)
26
Chen
 
X
Liu
 
H
Focia
 
PJ
Shim
 
AH
He
 
X
Structure of macrophage colony stimulating factor bound to FMS: diverse signaling assemblies of class III receptor tyrosine kinases.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
47
(pg. 
18267
-
18272
)
27
Lin
 
H
Lee
 
E
Hestir
 
K
, et al. 
Discovery of a cytokine and its receptor by functional screening of the extracellular proteome.
Science
2008
, vol. 
320
 
5877
(pg. 
807
-
811
)
28
Dai
 
XM
Ryan
 
GR
Hapel
 
AJ
, et al. 
Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects.
Blood
2002
, vol. 
99
 
1
(pg. 
111
-
120
)
29
Wei
 
S
Nandi
 
S
Chitu
 
V
, et al. 
Functional overlap but differential expression of CSF-1 and IL-34 in their CSF-1 receptor-mediated regulation of myeloid cells.
J Leukoc Biol
2010
, vol. 
88
 
3
(pg. 
495
-
505
)
30
Garceau
 
V
Smith
 
J
Paton
 
IR
, et al. 
Pivotal advance: avian colony-stimulating factor 1 (CSF-1), interleukin-34 (IL-34), and CSF-1 receptor genes and gene products.
J Leukoc Biol
2010
, vol. 
87
 
5
(pg. 
753
-
764
)
31
Chihara
 
T
Suzu
 
S
Hassan
 
R
, et al. 
IL-34 and M-CSF share the receptor Fms but are not identical in biological activity and signal activation.
Cell Death Differ
2010
, vol. 
17
 
12
(pg. 
1917
-
1927
)
32
Eda
 
H
Zhang
 
J
Keith
 
RH
Michener
 
M
Beidler
 
DR
Monahan
 
JB
Macrophage-colony stimulating factor and interleukin-34 induce chemokines in human whole blood.
Cytokine
2010
, vol. 
52
 
3
(pg. 
215
-
220
)
33
Bonifer
 
C
Hume
 
DA
The transcriptional regulation of the colony-stimulating factor 1 receptor (csf1r) gene during hematopoiesis.
Front Biosci
2008
, vol. 
13
 (pg. 
549
-
560
)
34
Ovchinnikov
 
DA
DeBats
 
CE
Sester
 
DP
Sweet
 
MJ
Hume
 
DA
A conserved distal segment of the mouse CSF-1 receptor promoter is required for maximal expression of a reporter gene in macrophages and osteoclasts of transgenic mice.
J Leukoc Biol
2010
, vol. 
87
 
5
(pg. 
815
-
822
)
35
Sasmono
 
RT
Oceandy
 
D
Pollard
 
JW
, et al. 
A macrophage colony-stimulating factor receptor-green fluorescent protein transgene is expressed throughout the mononuclear phagocyte system of the mouse.
Blood
2003
, vol. 
101
 
3
(pg. 
1155
-
1163
)
36
Hume
 
DA
Pavli
 
P
Donahue
 
RE
Fidler
 
IJ
The effect of human recombinant macrophage colony-stimulating factor (CSF-1) on the murine mononuclear phagocyte system in vivo.
J Immunol
1988
, vol. 
141
 
10
(pg. 
3405
-
3409
)
37
Ulich
 
TR
del Castillo
 
J
Watson
 
LR
Yin
 
SM
Garnick
 
MB
In vivo hematologic effects of recombinant human macrophage colony-stimulating factor.
Blood
1990
, vol. 
75
 
4
(pg. 
846
-
850
)
38
Munn
 
DH
Garnick
 
MB
Cheung
 
NK
Effects of parenteral recombinant human macrophage colony-stimulating factor on monocyte number, phenotype, and antitumor cytotoxicity in nonhuman primates.
Blood
1990
, vol. 
75
 
10
(pg. 
2042
-
2048
)
39
VandePol
 
CJ
Garnick
 
MB
Clinical applications of recombinant macrophage-colony stimulating factor (rhM-CSF).
Biotechnol Ther
1991
, vol. 
2
 
3
(pg. 
231
-
239
)
40
Hamilton
 
JA
Colony-stimulating factors in inflammation and autoimmunity.
Nat Rev Immunol
2008
, vol. 
8
 
7
(pg. 
533
-
544
)
41
Pollard
 
JW
Bartocci
 
A
Arceci
 
R
Orlofsky
 
A
Ladner
 
MB
Stanley
 
ER
Apparent role of the macrophage growth factor, CSF-1, in placental development.
Nature
1987
, vol. 
330
 
6147
(pg. 
484
-
486
)
42
Bartocci
 
A
Pollard
 
JW
Stanley
 
ER
Regulation of colony-stimulating factor 1 during pregnancy.
J Exp Med
1986
, vol. 
164
 
3
(pg. 
956
-
961
)
43
Roth
 
P
Stanley
 
ER
Colony stimulating factor-1 expression is developmentally regulated in the mouse.
J Leukoc Biol
1996
, vol. 
59
 
6
(pg. 
817
-
823
)
44
Roth
 
P
Stanley
 
ER
Colony-stimulating factor-1 expression in the human fetus and newborn.
J Leukoc Biol
1995
, vol. 
58
 
4
(pg. 
432
-
437
)
45
Irvine
 
KM
Burns
 
CJ
Wilks
 
AF
Su
 
S
Hume
 
DA
Sweet
 
MJ
A CSF-1 receptor kinase inhibitor targets effector functions and inhibits pro-inflammatory cytokine production from murine macrophage populations.
FASEB J
2006
, vol. 
20
 
11
(pg. 
1921
-
1923
)
46
Williams
 
GT
Smith
 
CA
Spooncer
 
E
Dexter
 
TM
Taylor
 
DR
Haemopoietic colony stimulating factors promote cell survival by suppressing apoptosis.
Nature
1990
, vol. 
343
 
6253
(pg. 
76
-
79
)
47
Bartocci
 
A
Mastrogiannis
 
DS
Migliorati
 
G
Stockert
 
RJ
Wolkoff
 
AW
Stanley
 
ER
Macrophages specifically regulate the concentration of their own growth factor in the circulation.
Proc Natl Acad Sci U S A
1987
, vol. 
84
 
17
(pg. 
6179
-
6183
)
48
Irvine
 
KM
Andrews
 
MR
Fernandez-Rojo
 
MA
, et al. 
Colony-stimulating factor-1 (CSF-1) delivers a proatherogenic signal to human macrophages.
J Leukoc Biol
2009
, vol. 
85
 
2
(pg. 
278
-
288
)
49
Martinez
 
FO
Gordon
 
S
Locati
 
M
Mantovani
 
A
Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression.
J Immunol
2006
, vol. 
177
 
10
(pg. 
7303
-
7311
)
50
Randolph
 
GJ
Jakubzick
 
C
Qu
 
C
Antigen presentation by monocytes and monocyte-derived cells.
Curr Opin Immunol
2008
, vol. 
20
 
1
(pg. 
52
-
60
)
51
Svensson
 
J
Jenmalm
 
MC
Matussek
 
A
Geffers
 
R
Berg
 
G
Ernerudh
 
J
Macrophages at the fetal-maternal interface express markers of alternative activation and are induced by M-CSF and IL-10.
J Immunol
2011
, vol. 
187
 
7
(pg. 
3671
-
3682
)
52
Rehli
 
M
Sulzbacher
 
S
Pape
 
S
, et al. 
Transcription factor Tfec contributes to the IL-4-inducible expression of a small group of genes in mouse macrophages including the granulocyte colony-stimulating factor receptor.
J Immunol
2005
, vol. 
174
 
11
(pg. 
7111
-
7122
)
53
Stacey
 
KJ
Fowles
 
LF
Colman
 
MS
Ostrowski
 
MC
Hume
 
DA
Regulation of urokinase-type plasminogen activator gene transcription by macrophage colony-stimulating factor.
Mol Cell Biol
1995
, vol. 
15
 
6
(pg. 
3430
-
3441
)
54
Hamilton
 
JA
Whitty
 
GA
Stanton
 
H
, et al. 
Macrophage colony-stimulating factor and granulocyte-macrophage colony-stimulating factor stimulate the synthesis of plasminogen-activator inhibitors by human monocytes.
Blood
1993
, vol. 
82
 
12
(pg. 
3616
-
3621
)
55
Fowles
 
LF
Martin
 
ML
Nelsen
 
L
, et al. 
Persistent activation of mitogen-activated protein kinases p42 and p44 and ets-2 phosphorylation in response to colony-stimulating factor 1/c-fms signaling.
Mol Cell Biol
1998
, vol. 
18
 
9
(pg. 
5148
-
5156
)
56
Zhao
 
B
Li
 
Y
Buono
 
C
, et al. 
Constitutive receptor-independent low density lipoprotein uptake and cholesterol accumulation by macrophages differentiated from human monocytes with macrophage-colony-stimulating factor (M-CSF).
J Biol Chem
2006
, vol. 
281
 
23
(pg. 
15757
-
15762
)
57
Wall
 
MJ
Chen
 
J
Meegalla
 
S
, et al. 
Synthesis and evaluation of novel 3,4,6-substituted 2-quinolones as FMS kinase inhibitors.
Bioorg Med Chem Lett
2008
, vol. 
18
 
6
(pg. 
2097
-
2102
)
58
Tushinski
 
RJ
Stanley
 
ER
The regulation of mononuclear phagocyte entry into S phase by the colony stimulating factor CSF-1.
J Cell Physiol
1985
, vol. 
122
 
2
(pg. 
221
-
228
)
59
Bauer
 
RJ
Gibbons
 
JA
Bell
 
DP
Luo
 
ZP
Young
 
JD
Nonlinear pharmacokinetics of recombinant human macrophage colony-stimulating factor (M-CSF) in rats.
J Pharmacol Exp Ther
1994
, vol. 
268
 
1
(pg. 
152
-
158
)
60
Fancke
 
B
Suter
 
M
Hochrein
 
H
O'Keeffe
 
M
M-CSF: a novel plasmacytoid and conventional dendritic cell poietin.
Blood
2008
, vol. 
111
 
1
(pg. 
150
-
159
)
61
MacDonald
 
KP
Rowe
 
V
Bofinger
 
HM
, et al. 
The colony-stimulating factor 1 receptor is expressed on dendritic cells during differentiation and regulates their expansion.
J Immunol
2005
, vol. 
175
 
3
(pg. 
1399
-
1405
)
62
Auffray
 
C
Fogg
 
DK
Narni-Mancinelli
 
E
, et al. 
CX3CR1+ CD115+ CD135+ common macrophage/DC precursors and the role of CX3CR1 in their response to inflammation.
J Exp Med
2009
, vol. 
206
 
3
(pg. 
595
-
606
)
63
Stoudemire
 
JB
Garnick
 
MB
Effects of recombinant human macrophage colony-stimulating factor on plasma cholesterol levels.
Blood
1991
, vol. 
77
 
4
(pg. 
750
-
755
)
64
Geissmann
 
F
Auffray
 
C
Palframan
 
R
, et al. 
Blood monocytes: distinct subsets, how they relate to dendritic cells, and their possible roles in the regulation of T-cell responses.
Immunol Cell Biol
2008
, vol. 
86
 
5
(pg. 
398
-
408
)
65
Ziegler-Heitbrock
 
L
Ancuta
 
P
Crowe
 
S
, et al. 
Nomenclature of monocytes and dendritic cells in blood.
Blood
2010
, vol. 
116
 
16
(pg. 
e74
-
e80
)
66
Ingersoll
 
MA
Spanbroek
 
R
Lottaz
 
C
, et al. 
Comparison of gene expression profiles between human and mouse monocyte subsets.
Blood
2010
, vol. 
115
 
3
(pg. 
e10
-
e19
)
67
Geissmann
 
F
Jung
 
S
Littman
 
DR
Blood monocytes consist of two principal subsets with distinct migratory properties.
Immunity
2003
, vol. 
19
 
1
(pg. 
71
-
82
)
68
Auffray
 
C
Fogg
 
D
Garfa
 
M
, et al. 
Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior.
Science
2007
, vol. 
317
 
5838
(pg. 
666
-
670
)
69
MacDonald
 
KP
Palmer
 
JS
Cronau
 
S
, et al. 
An antibody against the colony-stimulating factor 1 receptor depletes the resident subset of monocytes and tissue- and tumor-associated macrophages but does not inhibit inflammation.
Blood
2010
, vol. 
116
 
19
(pg. 
3955
-
3963
)
70
Heuser
 
M
Ganser
 
A
Bokemeyer
 
C
Use of colony-stimulating factors for chemotherapy-associated neutropenia: review of current guidelines.
Semin Hematol
2007
, vol. 
44
 
3
(pg. 
148
-
156
)
71
Masaoka
 
T
Shibata
 
H
Ohno
 
R
, et al. 
Double-blind test of human urinary macrophage colony-stimulating factor for allogeneic and syngeneic bone marrow transplantation: effectiveness of treatment and 2-year follow-up for relapse of leukaemia.
Br J Haematol
1990
, vol. 
76
 
4
(pg. 
501
-
505
)
72
Hidaka
 
T
Akada
 
S
Teranishi
 
A
, et al. 
Mirimostim (macrophage colony-stimulating factor; M-CSF) improves chemotherapy-induced impaired natural killer cell activity, Th1/Th2 balance, and granulocyte function.
Cancer Sci
2003
, vol. 
94
 
9
(pg. 
814
-
820
)
73
Cole
 
DJ
Sanda
 
MG
Yang
 
JC
, et al. 
Phase I trial of recombinant human macrophage colony-stimulating factor administered by continuous intravenous infusion in patients with metastatic cancer.
J Natl Cancer Inst
1994
, vol. 
86
 
1
(pg. 
39
-
45
)
74
Jakubowski
 
AA
Bajorin
 
DF
Templeton
 
MA
, et al. 
Phase I study of continuous-infusion recombinant macrophage colony-stimulating factor in patients with metastatic melanoma.
Clin Cancer Res
1996
, vol. 
2
 
2
(pg. 
295
-
302
)
75
Hume
 
DA
Donahue
 
RE
Fidler
 
IJ
The therapeutic effect of human recombinant macrophage colony stimulating factor (CSF-1) in experimental murine metastatic melanoma.
Lymphokine Res
1989
, vol. 
8
 
1
(pg. 
69
-
77
)
76
Bukowski
 
RM
Budd
 
GT
Gibbons
 
JA
, et al. 
Phase I trial of subcutaneous recombinant macrophage colony-stimulating factor: clinical and immunomodulatory effects.
J Clin Oncol
1994
, vol. 
12
 
1
(pg. 
97
-
106
)
77
Cenci
 
E
Bartocci
 
A
Puccetti
 
P
Mocci
 
S
Stanley
 
ER
Bistoni
 
F
Macrophage colony-stimulating factor in murine candidiasis: serum and tissue levels during infection and protective effect of exogenous administration.
Infect Immun
1991
, vol. 
59
 
3
(pg. 
868
-
872
)
78
Nemunaitis
 
J
Shannon-Dorcy
 
K
Appelbaum
 
FR
, et al. 
Long-term follow-up of patients with invasive fungal disease who received adjunctive therapy with recombinant human macrophage colony-stimulating factor.
Blood
1993
, vol. 
82
 
5
(pg. 
1422
-
1427
)
79
Abd
 
AH
Savage
 
NW
Halliday
 
WJ
Hume
 
DA
The role of macrophages in experimental arthritis induced by Streptococcus agalactiae sonicate: actions of macrophage colony-stimulating factor (CSF-1) and other macrophage-modulating agents.
Lymphokine Cytokine Res
1991
, vol. 
10
 
1
(pg. 
43
-
50
)
80
Han
 
Y
Ma
 
FY
Tesch
 
GH
Manthey
 
CL
Nikolic-Paterson
 
DJ
c-fms blockade reverses glomerular macrophage infiltration and halts development of crescentic anti–GBM glomerulonephritis in the rat.
Lab Invest
2011
, vol. 
91
 (pg. 
978
-
991
)
81
Jose
 
MD
Ikezumi
 
Y
van Rooijen
 
N
Atkins
 
RC
Chadban
 
SJ
Macrophages act as effectors of tissue damage in acute renal allograft rejection.
Transplantation
2003
, vol. 
76
 
7
(pg. 
1015
-
1022
)
82
Menke
 
J
Rabacal
 
WA
Byrne
 
KT
, et al. 
Circulating CSF-1 promotes monocyte and macrophage phenotypes that enhance lupus nephritis.
J Am Soc Nephrol
2009
, vol. 
20
 
12
(pg. 
2581
-
2592
)
83
Jose
 
MD
Le Meur
 
Y
Atkins
 
RC
Chadban
 
SJ
Blockade of macrophage colony-stimulating factor reduces macrophage proliferation and accumulation in renal allograft rejection.
Am J Transplant
2003
, vol. 
3
 
3
(pg. 
294
-
300
)
84
Hume
 
DA
Denkins
 
Y
The deleterious effect of macrophage colony-stimulating factor (CSF-1) on the pathology of experimental candidiasis in mice.
Lymphokine Cytokine Res
1992
, vol. 
11
 
2
(pg. 
95
-
98
)
85
Shin
 
EK
Lee
 
SH
Cho
 
SH
, et al. 
Association between colony-stimulating factor 1 receptor gene polymorphisms and asthma risk.
Hum Genet
2010
, vol. 
128
 
3
(pg. 
293
-
302
)
86
Albagha
 
OM
Visconti
 
MR
Alonso
 
N
, et al. 
Genome-wide association study identifies variants at CSF1, OPTN and TNFRSF11A as genetic risk factors for Paget's disease of bone.
Nat Genet
2010
, vol. 
42
 
6
(pg. 
520
-
524
)
87
Steinman
 
RM
Hawiger
 
D
Nussenzweig
 
MC
Tolerogenic dendritic cells.
Annu Rev Immunol
2003
, vol. 
21
 (pg. 
685
-
711
)
88
Lin
 
HH
Faunce
 
DE
Stacey
 
M
, et al. 
The macrophage F4/80 receptor is required for the induction of antigen-specific efferent regulatory T cells in peripheral tolerance.
J Exp Med
2005
, vol. 
201
 
10
(pg. 
1615
-
1625
)
89
Manrique
 
SZ
Duque Correa
 
MA
Hoelzinger
 
DB
, et al. 
Foxp3-positive macrophages display immunosuppressive properties and promote tumor growth.
J Exp Med
2011
, vol. 
208
 
7
(pg. 
1485
-
1499
)
90
Doyle
 
AG
Halliday
 
WJ
Barnett
 
CJ
Dunn
 
TL
Hume
 
DA
Effect of recombinant human macrophage colony-stimulating factor 1 on immunopathology of experimental brucellosis in mice.
Infect Immun
1992
, vol. 
60
 
4
(pg. 
1465
-
1472
)
91
Sakurai
 
T
Yamada
 
M
Simamura
 
S
Motoyoshi
 
K
Recombinant human macrophage-colony stimulating factor suppresses the mouse mixed lymphocyte reaction.
Cell Immunol
1996
, vol. 
171
 
1
(pg. 
87
-
94
)
92
Sakurai
 
T
Wakimoto
 
N
Yamada
 
M
Shimamura
 
S
Motoyoshi
 
K
Effect of macrophage colony-stimulating factor (M-CSF) on mouse immune responses in vivo.
Immunopharmacol Immunotoxicol
1998
, vol. 
20
 
1
(pg. 
79
-
102
)
93
Praloran
 
V
Raventos-Suarez
 
C
Bartocci
 
A
Lucas
 
J
Stanley
 
ER
Gibbons
 
JJ
Alterations in the expression of colony-stimulating factor-1 and its receptor during an acute graft-vs-host reaction in mice.
J Immunol
1990
, vol. 
145
 
10
(pg. 
3256
-
3261
)
94
Blazar
 
BR
Aukerman
 
SL
Vallera
 
DA
Effect of recombinant human macrophage colony-stimulating factor in irradiated murine recipients of T-cell-depleted allogeneic or non-depleted syngeneic bone marrow transplants.
Blood
1992
, vol. 
79
 
6
(pg. 
1636
-
1642
)
95
Sakurai
 
T
Misawa
 
E
Yamada
 
M
Hayasawa
 
H
Motoyoshi
 
K
Comparative study and effects of macrophage colony-stimulating factor (M-CSF) administration on NK1.1+ cells in mouse spleen and bone marrow.
Immunopharmacol Immunotoxicol
2002
, vol. 
24
 
4
(pg. 
547
-
566
)
96
Hashimoto
 
D
Chow
 
A
Greter
 
M
, et al. 
Pretransplant CSF-1 therapy expands recipient macrophages and ameliorates GVHD after allogeneic hematopoietic cell transplantation.
J Exp Med
2011
, vol. 
208
 
5
(pg. 
1069
-
1082
)
97
Kimura
 
F
Sato
 
K
Akiyama
 
H
, et al. 
M-CSF attenuates severity of chronic GVHD after unrelated BMT [published online ahead of print April 18, 2011].
Bone Marrow Transplant
 
98
Nemunaitis
 
J
Meyers
 
JD
Buckner
 
CD
, et al. 
Phase I trial of recombinant human macrophage colony-stimulating factor in patients with invasive fungal infections.
Blood
1991
, vol. 
78
 
4
(pg. 
907
-
913
)
99
Nishimura
 
K
Tanaka
 
N
Ohshige
 
A
Fukumatsu
 
Y
Matsuura
 
K
Okamura
 
H
Effects of macrophage colony-stimulating factor on folliculogenesis in gonadotrophin-primed immature rats.
J Reprod Fertil
1995
, vol. 
104
 
2
(pg. 
325
-
330
)
100
Nishimura
 
K
Tanaka
 
N
Kawano
 
T
Matsuura
 
K
Okamura
 
H
Changes in macrophage colony-stimulating factor concentration in serum and follicular fluid in in-vitro fertilization and embryo transfer cycles.
Fertil Steril
1998
, vol. 
69
 
1
(pg. 
53
-
57
)
101
Takasaki
 
A
Ohba
 
T
Okamura
 
Y
, et al. 
Clinical use of colony-stimulating factor-1 in ovulation induction for poor responders.
Fertil Steril
2008
, vol. 
90
 
6
(pg. 
2287
-
2290
)
102
Jenkins
 
SJ
Ruckerl
 
D
Cook
 
PC
, et al. 
Local macrophage proliferation, rather than recruitment from the blood, is a signature of TH2 inflammation.
Science
2011
, vol. 
332
 
6035
(pg. 
1284
-
1288
)
103
Alexander
 
KA
Chang
 
MK
Maylin
 
ER
, et al. 
Osteal macrophages promote in vivo intramembranous bone healing in a mouse tibial injury model.
J Bone Miner Res
2011
, vol. 
26
 
7
(pg. 
1517
-
1532
)
104
Alikhan
 
MA
Jones
 
CV
Williams
 
TM
, et al. 
Colony-stimulating factor-1 promotes kidney growth and repair via alteration of macrophage responses.
Am J Pathol
2011
, vol. 
179
 
3
(pg. 
1243
-
1256
)
105
Menke
 
J
Iwata
 
Y
Rabacal
 
WA
, et al. 
CSF-1 signals directly to renal tubular epithelial cells to mediate repair in mice.
J Clin Invest
2009
, vol. 
119
 
8
(pg. 
2330
-
2342
)
106
Okazaki
 
T
Ebihara
 
S
Asada
 
M
, et al. 
Macrophage colony-stimulating factor improves cardiac function after ischemic injury by inducing vascular endothelial growth factor production and survival of cardiomyocytes.
Am J Pathol
2007
, vol. 
171
 
4
(pg. 
1093
-
1103
)
107
Okuno
 
Y
Nakamura-Ishizu
 
A
Kishi
 
K
Suda
 
T
Kubota
 
Y
Bone marrow-derived cells serve as proangiogenic macrophages but not endothelial cells in wound healing.
Blood
2011
, vol. 
117
 
19
(pg. 
5264
-
5272
)
108
Boissonneault
 
V
Filali
 
M
Lessard
 
M
Relton
 
J
Wong
 
G
Rivest
 
S
Powerful beneficial effects of macrophage colony-stimulating factor on beta-amyloid deposition and cognitive impairment in Alzheimer's disease.
Brain
2009
, vol. 
132
 
4
(pg. 
1078
-
1092
)
109
Mitrasinovic
 
OM
Vincent
 
VA
Simsek
 
D
Murphy
 
GM
Macrophage colony stimulating factor promotes phagocytosis by murine microglia.
Neurosci Lett
2003
, vol. 
344
 
3
(pg. 
185
-
188
)
110
Mooney
 
JE
Rolfe
 
BE
Osborne
 
GW
, et al. 
Cellular plasticity of inflammatory myeloid cells in the peritoneal foreign body response.
Am J Pathol
2010
, vol. 
176
 
1
(pg. 
369
-
380
)
111
Hume
 
DA
Gordon
 
S
Mononuclear phagocyte system of the mouse defined by immunohistochemical localization of antigen F4/80: identification of resident macrophages in renal medullary and cortical interstitium and the juxtaglomerular complex.
J Exp Med
1983
, vol. 
157
 
5
(pg. 
1704
-
1709
)
112
Chang
 
MK
Raggatt
 
LJ
Alexander
 
KA
, et al. 
Osteal tissue macrophages are intercalated throughout human and mouse bone lining tissues and regulate osteoblast function in vitro and in vivo.
J Immunol
2008
, vol. 
181
 
2
(pg. 
1232
-
1244
)
113
Rae
 
F
Woods
 
K
Sasmono
 
T
, et al. 
Characterisation and trophic functions of murine embryonic macrophages based upon the use of a Csf1r-EGFP transgene reporter.
Dev Biol
2007
, vol. 
308
 
1
(pg. 
232
-
246
)
114
Burns
 
CJ
Wilks
 
AF
c-FMS inhibitors: a patent review.
Expert Opin Ther Pat
2011
, vol. 
21
 
2
(pg. 
147
-
165
)
115
Conway
 
JG
McDonald
 
B
Parham
 
J
, et al. 
Inhibition of colony-stimulating-factor-1 signaling in vivo with the orally bioavailable cFMS kinase inhibitor GW2580.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 
44
(pg. 
16078
-
16083
)
116
Hume
 
DA
Applications of myeloid-specific promoters in transgenic mice support in vivo imaging and functional genomics but do not support the concept of distinct macrophage and dendritic cell lineages or roles in immunity.
J Leukoc Biol
2011
, vol. 
89
 
4
(pg. 
525
-
538
)
117
Wei
 
S
Lightwood
 
D
Ladyman
 
H
, et al. 
Modulation of CSF-1-regulated post-natal development with anti–CSF-1 antibody.
Immunobiology
2005
, vol. 
210
 
2
(pg. 
109
-
119
)
118
Lokeshwar
 
BL
Lin
 
HS
Development and characterization of monoclonal antibodies to murine macrophage colony-stimulating factor.
J Immunol
1988
, vol. 
141
 
2
(pg. 
483
-
488
)
119
Campbell
 
IK
Rich
 
MJ
Bischof
 
RJ
Hamilton
 
JA
The colony-stimulating factors and collagen-induced arthritis: exacerbation of disease by M-CSF and G-CSF and requirement for endogenous M-CSF.
J Leukoc Biol
2000
, vol. 
68
 
1
(pg. 
144
-
150
)
120
Cenci
 
S
Weitzmann
 
MN
Gentile
 
MA
Aisa
 
MC
Pacifici
 
R
M-CSF neutralization and egr-1 deficiency prevent ovariectomy-induced bone loss.
J Clin Invest
2000
, vol. 
105
 
9
(pg. 
1279
-
1287
)
121
Radi
 
ZA
Koza-Taylor
 
PH
Bell
 
RR
, et al. 
Increased serum enzyme levels associated with kupffer cell reduction with no signs of hepatic or skeletal muscle injury.
Am J Pathol
2011
, vol. 
179
 
1
(pg. 
240
-
247
)
122
Sudo
 
T
Nishikawa
 
S
Ogawa
 
M
, et al. 
Functional hierarchy of c-kit and c-fms in intramarrow production of CFU-M.
Oncogene
1995
, vol. 
11
 
12
(pg. 
2469
-
2479
)
123
Murayama
 
T
Yokode
 
M
Kataoka
 
H
, et al. 
Intraperitoneal administration of anti–c-fms monoclonal antibody prevents initial events of atherogenesis but does not reduce the size of advanced lesions in apolipoprotein E-deficient mice.
Circulation
1999
, vol. 
99
 
13
(pg. 
1740
-
1746
)
124
Lim
 
AK
Ma
 
FY
Nikolic-Paterson
 
DJ
Thomas
 
MC
Hurst
 
LA
Tesch
 
GH
Antibody blockade of c-fms suppresses the progression of inflammation and injury in early diabetic nephropathy in obese db/db mice.
Diabetologia
2009
, vol. 
52
 
8
(pg. 
1669
-
1679
)
125
Segawa
 
M
Fukada
 
S
Yamamoto
 
Y
, et al. 
Suppression of macrophage functions impairs skeletal muscle regeneration with severe fibrosis.
Exp Cell Res
2008
, vol. 
314
 
17
(pg. 
3232
-
3244
)
126
Kubota
 
Y
Takubo
 
K
Shimizu
 
T
, et al. 
M-CSF inhibition selectively targets pathological angiogenesis and lymphangiogenesis.
J Exp Med
2009
, vol. 
206
 
5
(pg. 
1089
-
1102
)
127
Lenzo
 
JC
Turner
 
AL
Cook
 
AD
, et al. 
Control of macrophage lineage populations by CSF-1 receptor and GM-CSF in homeostasis and inflammation [published online ahead of print July15, 2011].
Immunol Cell Biol
 
128
Condeelis
 
J
Pollard
 
JW
Macrophages: obligate partners for tumor cell migration, invasion, and metastasis.
Cell
2006
, vol. 
124
 
2
(pg. 
263
-
266
)
129
Wyckoff
 
JB
Wang
 
Y
Lin
 
EY
, et al. 
Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors.
Cancer Res
2007
, vol. 
67
 
6
(pg. 
2649
-
2656
)
130
Tagliani
 
E
Shi
 
C
Nancy
 
P
Tay
 
CS
Pamer
 
EG
Erlebacher
 
A
Coordinate regulation of tissue macrophage and dendritic cell population dynamics by CSF-1.
J Exp Med
2011
, vol. 
208
 
9
(pg. 
1901
-
1916
)
131
Pollard
 
JW
Hunt
 
JS
Wiktor-Jedrzejczak
 
W
Stanley
 
ER
A pregnancy defect in the osteopetrotic (op/op) mouse demonstrates the requirement for CSF-1 in female fertility.
Dev Biol
1991
, vol. 
148
 
1
(pg. 
273
-
283
)
132
Winkler
 
IG
Sims
 
NA
Pettit
 
AR
, et al. 
Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs.
Blood
2010
, vol. 
116
 
23
(pg. 
4815
-
4828
)
133
Ober
 
RJ
Radu
 
CG
Ghetie
 
V
Ward
 
ES
Differences in promiscuity for antibody-FcRn interactions across species: implications for therapeutic antibodies.
Int Immunol
2001
, vol. 
13
 
12
(pg. 
1551
-
1559
)
134
Unkeless
 
JC
Scigliano
 
E
Freedman
 
VH
Structure and function of human and murine receptors for IgG.
Annu Rev Immunol
1988
, vol. 
6
 (pg. 
251
-
281
)
135
Conway
 
JG
Pink
 
H
Bergquist
 
ML
, et al. 
Effects of the cFMS kinase inhibitor 5-(3-methoxy-4-((4-methoxybenzyl)oxy)benzyl)pyrimidine-2,4-diamine (GW2580) in normal and arthritic rats.
J Pharmacol Exp Ther
2008
, vol. 
326
 
1
(pg. 
41
-
50
)
136
Priceman
 
SJ
Sung
 
JL
Shaposhnik
 
Z
, et al. 
Targeting distinct tumor-infiltrating myeloid cells by inhibiting CSF-1 receptor: combating tumor evasion of antiangiogenic therapy.
Blood
2010
, vol. 
115
 
7
(pg. 
1461
-
1471
)
137
Ohno
 
H
Uemura
 
Y
Murooka
 
H
, et al. 
The orally-active and selective c-Fms tyrosine kinase inhibitor Ki20227 inhibits disease progression in a collagen-induced arthritis mouse model.
Eur J Immunol
2008
, vol. 
38
 
1
(pg. 
283
-
291
)
138
Uemura
 
Y
Ohno
 
H
Ohzeki
 
Y
, et al. 
The selective M-CSF receptor tyrosine kinase inhibitor Ki20227 suppresses experimental autoimmune encephalomyelitis.
J Neuroimmunol
2008
, vol. 
195
 
1
(pg. 
73
-
80
)
139
Manthey
 
CL
Johnson
 
DL
Illig
 
CR
, et al. 
JNJ-28312141, a novel orally active colony-stimulating factor-1 receptor/FMS-related receptor tyrosine kinase-3 receptor tyrosine kinase inhibitor with potential utility in solid tumors, bone metastases, and acute myeloid leukemia.
Mol Cancer Ther
2009
, vol. 
8
 
11
(pg. 
3151
-
3161
)
140
Dewar
 
AL
Cambareri
 
AC
Zannettino
 
AC
, et al. 
Macrophage colony-stimulating factor receptor c-fms is a novel target of imatinib.
Blood
2005
, vol. 
105
 
8
(pg. 
3127
-
3132
)
141
Paniagua
 
RT
Chang
 
A
Mariano
 
MM
, et al. 
c-Fms-mediated differentiation and priming of monocyte lineage cells play a central role in autoimmune arthritis.
Arthritis Res Ther
2010
, vol. 
12
 
1
pg. 
R32
 
142
Fairbairn
 
L
Kapetanovic
 
R
Sester
 
DP
Hume
 
DA
The mononuclear phagocyte system of the pig as a model for understanding human innate immunity and disease.
J Leukoc Biol
2011
, vol. 
89
 
6
(pg. 
855
-
871
)
143
Nowicki
 
A
Szenajch
 
J
Ostrowska
 
G
, et al. 
Impaired tumor growth in colony-stimulating factor 1 (CSF-1)-deficient, macrophage-deficient op/op mouse: evidence for a role of CSF-1-dependent macrophages in formation of tumor stroma.
Int J Cancer
1996
, vol. 
65
 
1
(pg. 
112
-
119
)
144
Amemiya
 
H
Kono
 
H
Fujii
 
H
Liver regeneration is impaired in macrophage colony stimulating factor deficient mice after partial hepatectomy: the role of M-CSF-induced macrophages.
J Surg Res
2011
, vol. 
165
 
1
(pg. 
59
-
67
)
Sign in via your Institution