G-CSF and GM-CSF are used widely to promote the production of granulocytes or APCs. The U.S. Food and Drug Administration approved G-CSF (filgrastim) for the treatment of congenital and acquired neutropenias and for mobilization of peripheral hematopoietic progenitor cells for stem cell transplantation. A polyethylene glycol–modified form of G-CSF is approved for the treatment of neutropenias. Clinically significant neutropenia, rendering an individual immunocompromised, occurs when their number is <1500/μl. Current guidelines recommend their use when the risk for febrile neutropenia is >20%. GM-CSF (sargramostim) is approved for neutropenia associated with stem cell transplantation. Because of its promotion of APC function, GM-CSF is being evaluated as an immunostimulatory adjuvant in a number of clinical trials. More than 20 million persons have benefited worldwide, and >$5 billion in sales occur annually in the United States.

Few physician-scientists have made as great an impact on our understanding of hematology or improved the lives of patients, estimated at >20 million (1), with blood and cancer disorders as Don Metcalf, who died in December of 2014. Laboring at the Walter and Eliza Hall Institute in Melbourne, Australia throughout his 50-year career, Metcalf used semisolid medium and cell culture supernatants to discover hematopoietic progenitor cells (e.g., granulocyte/macrophage colonies) and their growth factors (e.g., G-CSF and GM-CSF). Increased purification of these and related growth factors, sometimes from hundreds of mice injected with endotoxin, led to the molecular characterization and cloning of G-CSF, GM-CSF, M-CSF, stem cell factor, and IL-3 in the 1980s (2).

Hematopoiesis is a highly proliferative (∼1010 cells/d) dynamic process driven by multiple hematopoietic growth factors/cytokines (Fig. 1A). The hematopoietic growth factors are multifunctional and are critical for proliferation, survival, and differentiation of hematopoietic stem, progenitor, and precursor cells to a terminally differentiated, functional cell type. Colony-forming assays identified the ability of first crude supernatants, and then highly purified cytokines, to drive multi-lineage and single-lineage differentiation. After coculturing for 7–14 d, colonies from mononuclear cells obtained from the mouse spleen or bone marrow were measured in semisolid medium. Based on the characteristics of cells within a single colony, the lineage(s) governed by the cytokine was determined. Granulocytes make up the majority of WBCs in human circulation and play an integral role in innate and adaptive immunity. In granulopoiesis, their production is mediated by a number of growth factors, especially G-CSF and GM-CSF (3, 4). Due to asymmetric division, some daughter cells of the hematopoietic stem cell (HSC) remain as HSCs, preventing the depletion of the stem cell pool (5). Multiparameter immunophenotyping has transformed our ability to identify different cell types in hematopoiesis. Murine HSCs are characterized as linsca1+c-kit+, and human HSCs display CD34 in the absence of lineage markers. The differentiation pathway from HSCs to granulocytes is dependent on G-CSF and, less so, on GM-CSF. The HSC gives rise to a common myeloid progenitor and a common lymphoid progenitor cell (6). The common myeloid progenitor cells differentiate into myeloblasts, erythrocytes, and megakaryocytes via at least two intermediates: the granulocyte/monocyte progenitor cell and the erythrocyte/megakaryocyte progenitor cell. In the granulocytic series, myeloblasts (15–20 μm) are the first recognizable cells by their scant cytoplasm, absence of granules, and fine nucleus with nucleoli in the bone marrow clearly committed to differentiation to granulocytes. Myeloblasts differentiate into promyelocytes, which are larger (20 μm) and begin to possess granules (Fig. 1B). Promyelocytes give rise to neutrophilic, basophilic, and eosinophilic precursor cells. Cell division continues through the promyelocyte stage. Fine specific granules containing inflammatory-related proteins appear during myelocyte maturation. For neutrophils, their size and nuclei become increasingly more condensed as the cells mature through myelocyte, metamyelocyte, band, and the terminally differentiated neutrophil (polymorphonuclear and ∼15 μm). During episodes of stress, such as infection, band cells can be found in the peripheral blood and are used as a measure of inflammation. The above process is complex, dynamic, and orchestrated by multiple cytokines and their receptors, most notably G-CSF and GM-CSF.

FIGURE 1.

(A) Schematic diagram of hematopoiesis from the multipotential HSC to fully differentiated cell types. Principal cytokines that determine differentiation patterns are shown in red. (B) The stages of granulopoiesis from myeloblast to the mature granulocyte. During neutrophil maturation, which is driven primarily by G-CSF, granulocytic cells change shape, acquire primary and specific granules, and undergo nuclear condensation. Epo, erythropoietin; SCF, stem cell factor; SDF-1, stromal cell–derived factor-1; TPO, thrombopoietin.

FIGURE 1.

(A) Schematic diagram of hematopoiesis from the multipotential HSC to fully differentiated cell types. Principal cytokines that determine differentiation patterns are shown in red. (B) The stages of granulopoiesis from myeloblast to the mature granulocyte. During neutrophil maturation, which is driven primarily by G-CSF, granulocytic cells change shape, acquire primary and specific granules, and undergo nuclear condensation. Epo, erythropoietin; SCF, stem cell factor; SDF-1, stromal cell–derived factor-1; TPO, thrombopoietin.

Close modal

Following Ag stimulation or activation by cytokines, such as IL-1, IL-6, and TNF-α, macrophages, T cells, endothelial cells, and fibroblasts produce and secrete G-CSF and GM-CSF. Of unknown significance, a variety of tumor cells also produce these paracrine growth factors. Glycoproteins with a molecular mass ∼ 23 kDa, G-CSF and GM-CSF, are now produced through recombinant technology in either Escherichia coli or yeast. G-CSF induces the appearance of colonies containing only granulocytes, whereas GM-CSF gives colonies containing both granulocytes and macrophages. Generation of G-CSF (Csf3)- and G-CSFR (Csf3r)-knockout mice confirmed that G-CSF critically drives granulopoiesis (7). The cognate receptor for G-CSF is a single-transmembrane receptor that homodimerizes upon G-CSF binding. Unlike G-CSF, GM-CSF functions via a two-receptor system involving a specific α-chain and a common β-chain shared by IL-3 and IL-5 (8). However, GM-CSF–knockout mice did not display a perturbation in hematopoiesis (9, 10). Both G-CSF and GM-CSF signal through pathways involving JAK/STAT, SRC family kinases, PI3K/AKT, and Ras/ERK1/2. The receptor complexes are characterized by high-affinity (apparent KD ∼ 100–500 pM) and low-density (50–1000 copies/cell). Interestingly, human G-CSF is functionally active on murine myeloid cells, but human GM-CSF is not. The signaling specificity likely involves nuances in the proximal postreceptor phosphoprotein networks and the distal gene regulatory networks. The molecular pathways and their cross-interactions in determining lineage specificity are critical to the development of more specific therapies.

Cloning of human GM-CSF and its expression in bacterial and eukaryotic cells were achieved in 1985 at the Genetics Institute (11), and cloning of G-CSF and its expression in E. coli was achieved a year later at Amgen (12). Commercialized by these biotechnology start-ups, G-CSF and GM-CSF revolutionized the treatment of patients with congenital or acquired neutropenias and those undergoing stem cell transplantation. Sidelined from the treatment of neutropenias by its toxicity profile, GM-CSF is now undergoing a renaissance as an immunomodulatory agent.

G-CSF is approved by the U.S. Food and Drug Administration (FDA) for use to decrease the incidence of infection in patients with nonmyeloid malignancies receiving myelosuppressive anticancer drugs associated with a significant incidence of severe neutropenia with fever; reduce the time to neutrophil recovery and the duration of fever following induction or consolidation chemotherapy treatment of patients with acute myeloid leukemia (AML); reduce the duration of neutropenia and febrile neutropenia in patients with nonmyeloid malignancies undergoing myeloablative chemotherapy followed by stem cell transplantation; mobilize hematopoietic progenitor cells into the peripheral blood for collection by leukapheresis; and reduce the incidence and duration of complications of severe neutropenia in symptomatic patients with congenital neutropenia, cyclic neutropenia, or idiopathic neutropenia. Forms of G-CSF available worldwide include filgrastim, pegfilgrastim, and lenograstim.

GM-CSF is approved by the FDA to accelerate myeloid recovery in patients with non-Hodgkin’s lymphoma (NHL), acute lymphoblastic leukemia, and Hodgkin’s disease undergoing autologous stem cell transplantation; following induction chemotherapy in older adult patients with AML to shorten time to neutrophil recovery and reduce the incidence of life-threatening infections; to accelerate myeloid recovery in patients undergoing allogeneic stem cell transplantation from HLA-matched related donors; for patients who have undergone allogeneic or autologous stem cell transplantation in whom engraftment is delayed or failed; and to mobilize hematopoietic progenitor cells into peripheral blood for collection by leukapheresis. Forms of GM-CSF available worldwide are sargramostim and molgramostim.

The recommended dosage for G-CSF is 5 μg/kg/d and for GM-CSF is 250 μg/m2/d. Both drugs may be given s.c. or i.v., although randomized clinical trials demonstrate greater efficacy (i.e., decreased duration of neutropenia) without a difference in toxicity for the s.c. route (13). For chemotherapy-induced neutropenia, G-CSF is administered until there are >1000 neutrophils/μl. For congenital neutropenias, the goal is to maintain neutrophil counts ∼ 750/μl. G-CSF is well tolerated. Transient fever and bone pain are more commonly observed in those receiving GM-CSF. Pleural and/or pericardial effusions can also occur in those receiving GM-CSF. Long-term side effects of G-CSF administration, such as osteopenia, are being monitored in patients with severe congenital neutropenia (SCN). One concern is that G-CSF may accelerate the transformation of SCN to myelodysplastic syndromes (MDSs) or AML, associated with acquired mutations in G-CSFR.

The receptors for both GM-CSF and G-CSF belong to the hematopoietin/cytokine receptor superfamily. G-CSFR acts as a homodimer, whereas GM-CSFR is a heterodimer with a shared β-chain with the IL-3R and IL-5R complexes. G-CSFR is expressed primarily on neutrophils and bone marrow precursor cells, which undergo proliferation and eventually differentiate into mature granulocytes. G-CSF binds to G-CSFR, resulting in its dimerization, with a stoichiometry of 2:2 and with a high affinity (KD = 500 pM) (14, 15). Among the activated downstream signal-transduction pathways are JAK/STAT, Src kinases, such as Lyn, Ras/ERK, and PI3K (16). The cytoplasmic domain of G-CSFR possesses four tyrosine residues (Y704, Y729, Y744, Y764) serving as phospho-acceptor sites (17, 18). Src homology 2–containing proteins STAT5 and STAT3 bind to Y704 and Gab2 to Y764. Grb2 couples to both Gab2 and to SOS, permitting signaling diversification, such as Ras/ERK, PI3K/Akt, and Shp2 (19, 20). An alternatively spliced isoform of G-CSFR elicits activation of a JAK/SHP2 pathway (15). The precise physiological roles of protein kinases and their downstream events in G-CSF–induced signaling remain unclear, although some clues are beginning to emerge (21, 22).

GM-CSF binds to the α-chain of the GM-CSFR with a low affinity (KD = 0.2–100 nM), but a higher affinity (KD = 100 pM) occurs in the presence of both subunits. GM-CSF signaling involves the formation of dodecameric supercomplex that is required for JAK activation (23). In addition to the JAK/STAT pathway, GM-CSF activates the ERK1/2, PI3K/Akt, and IκB/NF-κB pathways. Although the α-chain is primarily considered a ligand-recognition unit, it interacts with Lyn, resulting in JAK-independent Akt activation of the survival pathway (24). Thus, differences in receptor expression patterns and known and unknown nuances in signaling pathway circuits account for the functional differences between G-CSF and GM-CSF.

G-CSF and GM-CSF are pleiotropic growth factors, with overlapping functions. GM-CSF also shares properties with M-CSF on monocyte function (25). Both GM-CSF and G-CSF increase chemotaxis and migration of neutrophils, but response kinetics may differ. GM-CSF may be considered to be more proinflammatory than G-CSF. GM-CSF increases cytotoxic killing of Candida albicans, surface expression of Fc- and complement-mediated cell-binding (FcγR1, CR-1, CR-3), and adhesion receptor (14). Yet, both cytokines promote neutrophil phagocytosis (26). More extensive reviews on G-CSF and GM-CSF function in neutrophils may be found (27, 28).

An absolute neutrophil count (ANC) < 1500/μl is defined as neutropenia, which is graded on the severity of decreased ANC (Table I). Causes for neutropenia may be congenital or, more commonly, acquired. Neutropenia may be asymptomatic until an infection occurs. Benign neutropenia exists, and the individuals are not at risk for serious infection. However, onset of fever with neutropenia, termed febrile neutropenia, commonly occurs as a potentially life-threatening complication of chemotherapy and involves considerable cost as a result of treatment with i.v. antibiotics and prolonged hospitalization. In addition, febrile neutropenia prevents continuation of chemotherapy until recovery from it occurs. According to the Norton–Simon hypothesis (29), the efficacy of chemotherapy is reduced if stopped midway. A pause in treatment allows recovery of the cancer cells and facilitates the emergence of chemoresistant clones (2931). Neutropenia also occurs secondary to bone marrow infiltration with leukemic or myelodysplastic cells.

Table I.
Correlation of neutropenia with absolute neutrophil count
Neutropenia GradeAbsolute Neutrophil Count
Grade 1 ≥1.5 to <2 × 109/ml 
Grade 2 ≥1 to <1.5 × 109/ml 
Grade 3 ≥0.5 to <1 × 109/ml 
Grade 4 <0.5 × 109/ml 
Neutropenia GradeAbsolute Neutrophil Count
Grade 1 ≥1.5 to <2 × 109/ml 
Grade 2 ≥1 to <1.5 × 109/ml 
Grade 3 ≥0.5 to <1 × 109/ml 
Grade 4 <0.5 × 109/ml 

Neutropenia results from a growing list of germline mutations in genes, such as ELANE, HAX1, GFI1, G6PC3, WAS, and CSF3R (32). Soon after birth, children with SCN develop a grade 4 neutropenia. SCN is a lifetime condition resulting from increased apoptosis of granulocytic progenitors in the marrow. As a result of the severity and chronic nature of SCN, individuals are prone to recurrent infections, especially from the endogenous flora in the gut, mouth, and skin. Most cases of SCN are due to de novo mutations. Transmission may be autosomal dominant, recessive, or X-linked. The most common mutation involves ELANE and is autosomal dominant (33, 34). Mutations in ELANE encode the neutrophil elastase, a serine protease. ELANE is expressed during granulopoiesis, maximally at the promyelocyte stage. It is hypothesized that mutations in ELANE cause neutropenia via improper folding of the protein that triggers the unfolded protein response. Unfolded protein response–generated stress drives apoptosis due to an overload of unfolded protein, and an arrest in differentiation at the promyelocyte stage is observed. Fascinatingly, ELANE mutations are also associated with cyclic neutropenia. Cyclic neutropenia is characterized by granulocyte nadirs < 200/μl occurring every 21 d.

Patients with SCN are always at risk for life-threatening infections. Early phase 1 clinical trials held in 1989 (35, 36) evaluated G-CSF therapy for SCN and cyclic neutropenia. Both trials demonstrated a ≥10-fold increase in neutrophil counts, reducing the severity of neutropenia from grade 4 to grade 1 to normal counts. A reduction in the days of cyclic neutropenia from 21 to 14 d was observed, and a consistent increase in ANC was observed in SCN. In 1990, two studies explored the benefit of G-CSF versus GM-CSF in treating congenital neutropenia. Gray collie dogs with cyclic neutropenia due to mutations in the endocytosis gene AP3B1 (37) were studied with three cytokines: G-CSF, GM-CSF, and IL-3. GM-CSF and G-CSF showed an expansion of neutrophil counts, but only G-CSF prevented the cycling of hematopoiesis (10). Similar to the dog study, G-CSF therapy increased ANC, whereas GM-CSF therapy increased eosinophil counts but not neutrophil counts (38). Following the beneficial effects of G-CSF in the above phase 1/2 studies, a phase 3 clinical trial was performed in 1993 (39). Patients with SCN, cyclic neutropenia, and idiopathic neutropenia (n = 123) were included in the study. Patients were randomly treated immediately or after a 4-mo observation period. Almost all of the patients (108/120) receiving G-CSF therapy displayed a restoration of ANC from grade 4 to normal levels. The increase in ANC resulted from increased production of neutrophils in bone marrow. Infection-related incidents were reduced by ∼50% (p < 0.05), and antibiotic use was reduced by 70%.

One particular form of inherited neutropenia is WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome (40). Myelokathexis refers to a build-up of mature neutrophils in the bone marrow. Mutations in CXCR4 result in the syndrome (41). CXCR4 and its ligand SDF-1 mediate the retention of neutrophils. G-CSF administration leads to upregulation of SDF-1 and the subsequent release of neutrophils into the peripheral circulation (42). A recently published phase 1 study demonstrated the safety and efficacy of low-dose plerixafor, a CXCR4 antagonist (43). One widely used indication for G-CSF is to mobilize and harvest hematopoietic progenitor cells into the periphery for stem cell transplantation (44), and the concomitant use of plerixafor enhances the mobilization (45).

Severe aplastic anemia (SAA) is a disease in which stem cells residing in the bone marrow are damaged, leading to a deficiency in all hematopoietic cell lines. SAA has a high mortality, but the 5-y mortality is reduced to <10% with matched sibling stem cell transplantation or to 30% with immunosuppressive therapy (IST) (46). IST includes antithymocyte globulin, cyclosporine, and glucocorticoids. The addition of G-CSF to IST was studied in a number of randomized studies. It was shown that G-CSF reduces the number of infectious complications and hospital days compared with standard therapy alone; however, its addition did not affect overall survival rates (47, 48). Although treatment with G-CSF or GM-CSF results in a neutrophil response, a sustained trilineage response was uncommon when used alone or in combination with other hematopoietic growth factors (49, 50). The response to G-CSF may have prognostic value. Patients treated with IST plus G-CSF who did not achieve a WBC count ≥ 5000/μl had a low probability of response and high mortality (5153). Similarly, GM-CSF was studied as a potential adjunct to IST with similar results (48). These finding suggest that G-CSF and GM-CSF may be useful adjuncts to standard IST for SAA.

In 1991, the FDA approved the use of recombinant human G-CSF (filgrastim) to treat cancer patients undergoing myelotoxic chemotherapy. Multiple factors affect the severity of neutropenia, with the most important being the type and severity of chemotherapy dosage and the underlying disease (54, 55). In 1994, the American Society of Clinical Oncology (ASCO) recommended primary prophylaxis with G-CSF or GM-CSF for the expected incidence of neutropenia ≥ 40% (56). The purpose of the guidelines was to reduce the incidence and length of neutropenia and, thus, the time of hospitalization, which would reduce costs significantly. Three prospective, randomized, placebo-controlled trials formed the basis of the recommendations. The first phase 3 trial tested the applicability of G-CSF as an adjunct to chemotherapy in patients treated for small cell lung cancer with cyclophosphamide, doxorubicin, and etoposide (57). A major outcome of the study was the significant reduction by at least one episode of febrile neutropenia in 77% of those treated with G-CSF versus 40% in the placebo group. A reduction in the median duration of grade 4 neutropenia was observed in all cycles of chemotherapy (1 d in the G-CSF group versus 6 d in the placebo group). From a cost-benefit perspective, the data translated into a reduction in the 50% incidence of infection, antibiotic treatment, and days of hospitalization with G-CSF treatment versus placebo. A similar study performed in Europe in patients with small cell lung cancer also found that prophylactic G-CSF treatment reduced the incidence of febrile neutropenia (53% in placebo group versus 26% in G-CSF group) (58). A reduction in chemotherapy dose by 15% was indicated in 61% of the placebo group versus 29% of the G-CSF group. A gap ≥ 2 d in the chemotherapy treatment group was observed for 47% of patients in the placebo group and 29% of the patients in the G-CSF group. The third trial investigated G-CSF therapy in NHL treated with vincristine, doxorubicin, prednisolone, etoposide, cyclophosphamide, and bleomycin (59). The incidence of neutropenia was reduced for the G-CSF group (23%) versus the placebo group (44%), with fewer delays and shorter duration of treatment in the G-CSF–treated group. In comparison, GM-CSF trials provided less convincing data. In a trial for cyclophosphamide, vincristine, procarbazine, bleomycin, prednisolone, doxorubicin, and mesna administered as therapy for NHL, the use of molgramostim (GM-CSF) resulted in faster recovery from neutropenia and reduced hospitalization, but the benefit was limited to only 72% of the patients that could tolerate GM-CSF (60). Another trial with small cell lung cancer did not show any significant effect with molgramostim treatment (61).

The development of better chemotherapeutic regimens that were less myelotoxic provided more cost-effective options compared with CSF therapy. In many cases, the incidence of neutropenia was reduced to ≤10%. However, the advantage of CSF therapy in both increasing the intensity and maintenance of dose versus the cost of the growth factors was actively debated. In 2003, a large randomized study showed the benefit of G-CSF therapy with dose-dense chemotherapy (cyclophosphamide, paclitaxel, and doxorubicin) in patients with node-positive breast cancer (62). Significantly improved disease-free survival (risk ratio = 0.74; p = 0.01) and overall survival (risk ratio = 0.69, p = 0.013) were observed in patients receiving G-CSF. Fewer patients reported grade 4 neutropenia in the G-CSF group (6%) compared with the non–G-CSF group (33%). In 2004, two additional studies with old (60–75 y) and young (<60 y) NHL patients observed a reduction in chemotherapy regimens from 3 to 2 wk combined with an improvement in the rate of progressive disease and overall survival (63, 64). In 2005, two large trials supported the use of G-CSF in reducing the incidence of fever and neutropenia and suggested its use with the first cycle of chemotherapy (65, 66). The first study compared the effect of antibiotics versus antibiotics + G-CSF in small cell lung cancer patients undergoing cyclophosphamide, doxorubicin, and etoposide treatment. A significant reduction in the incidence of febrile neutropenia was observed for the antibiotics + G-CSF group (10%) compared with the antibiotics-only group (24%) (66). The second study investigated the effect of pegfilgrastim in breast cancer patients treated with docetaxel (65). Approved in 2001, pegfilgrastim was developed to improve the renal clearance rate (67), and a single dose provided similar or greater improvement in the ANC after chemotherapy compared with daily doses of filgrastim (68). The randomized, placebo-controlled trial conducted with 928 patients demonstrated a lower incidence of febrile neutropenia in patients receiving pegfilgrastim (1%) compared with placebo (17%). Hospitalization also was reduced in the pegfilgrastim group (1%) compared with the placebo group (14%). In 2005 and 2006, the National Comprehensive Cancer Network (http://www.nccn.org) and ASCO changed the risk threshold for contracting neutropenia from 40 to 20% to justify the use of myeloid growth factors as an adjuvant to chemotherapy in treating neutropenia (69). The use of myeloid growth factors, their cost effectiveness, and the duration of their use during chemotherapy remain of great interest to clinical oncologists. A randomized phase 3 study with a noninferiority design demonstrated the efficacy of G-CSF prophylaxis against febrile neutropenia in women with breast cancer for the entirety of their myelosuppressive treatment (70). Current guidelines from ASCO, the National Comprehensive Cancer Network, and the European Organization for Research and Treatment of Cancer recommend the use of myeloid growth factors when the risk for febrile neutropenia is ≥20% (71, 72).

Neutropenia in patients with leukemia results from both the underlying disease and aggressive chemotherapy. The ASCO guidelines developed in 1994, like for solid tumors, considered data obtained from three large randomized trials. Unlike the solid tumor trials, two of the three trials used GM-CSF versus G-CSF. The two GM-CSF trials reported conflicting findings, with some statistical significance in the recovery of ANC but no significant reduction in hospitalization or the incidence of serious infections (73, 74). The G-CSF trial showed a recovery in ANC, reduction in days of neutropenia, and a trend toward better recovery rates. However, like the GM-CSF trials, no improvement in days of hospitalization or usage of antibiotics was observed (75). Thus, a beneficial response by the growth factors was not observed in leukemia. However at the time of ASCO’s 2000 guidelines, newer placebo-controlled trials demonstrated a reduction in neutrophil recovery time from 6 to 2 d and reduced hospitalization times in the setting of induction chemotherapy (76). The 2000 ASCO guidelines also identified a potential benefit for growth factor therapy in consolidation chemotherapy. The 2006 update did not introduce any significant changes and recommended the application of CSF therapy postinduction and consolidation therapy (69).

Unlike chemotherapy-induced neutropenia, congenital neutropenia patients experience neutropenia for life and require long-term treatment with G-CSF. The long-term effects of G-CSF therapy have become important in the management of congenital neutropenia. Patients receiving G-CSF therapy for as long as 8 y were evaluated for safety and efficacy (77). Neutrophil counts were maintained without exhaustion of myelopoiesis. A significant improvement in the quality of life was achieved by the reduction in antibiotic treatment and hospitalization time, allowing for normal growth, development, and participation in normal daily activities. The SCN international registry was formed in 1994 to further assess the progress of SCN patients being treated with G-CSF. A 10-y report that followed patients with SCN (n = 526) who were being treated with G-CSF was released in 2006 (78). Consistent with previous reports, an increase in ANC was observed in majority of the patients with an overall improvement in quality of life.

Leukemia transformation is significantly higher in SCN patients, and the SCN international registry reported that 21% of patients with SCN developed leukemia while being treated with G-CSF. Although leukemic transformation has been reported in SCN patients before the development of G-CSF therapy (79), the precise role of G-CSF therapy in leukemic transformation remains unknown. Almost all SCN patients undergo G-CSF therapy; thus, it is difficult to assess leukemic transformation in the absence of G-CSF treatment. However, patients who require higher doses of G-CSF are at a higher risk for developing MDS/AML (80).

Germline mutations in CSF3R, which encodes G-CSFR, are infrequent causes of SCN and result in refractoriness to filgrastim (81). Acquired nonsense mutations in CSF3R were observed in ∼80% of SCN patients who progressed to secondary MDS/AML (8286). The nonsense mutations result in deletion of the C terminus of G-CSFR, resulting in the loss of one to all four tyrosine residues and the inability to undergo normal ligand-induced internalization and endosomal routing (87, 88). The truncated receptor mutants produce a phenotype of enhanced proliferation and impaired differentiation in response to G-CSF. Furthermore, knock-in mice harboring a similar mutation showed hyperproliferative responses to G-CSF administration and strongly prolonged activation of STAT5, which are implicated in increased hematopoietic progenitor stem cell expansion in vivo (89). This prediction was validated in a patient with SCN who developed secondary AML concomitant with a nonsense mutation of G-CSFR. Upon discontinuation of G-CSF and without chemotherapy, the blast count in the blood and bone marrow disappeared, although the mutation remained detectable (90). The tight correlation between the acquisition of G-CSFR mutations and progression of SCN to secondary MDS/AML and the abnormal signaling features in vitro and in vivo strongly suggested that mutated CSF3R could be a driver of myelodysplasia. Data from recent studies, which showed that the CSF3R T595I mutation is the most prevalent mutation found in chronic neutrophilic leukemia and that treatment with the Jak2 inhibitor ruxolitinib resulted in marked clinical improvement, support the hypothesis that mutations in G-CSFR are indeed drivers of myeloproliferative disease (91, 92). A low frequency of CSF3R mutations also occurs in AML and chronic myelomonocytic leukemia (93).

More than 20 y after its registration by the FDA, biosimilars (94) of G-CSF are being developed (95). (Amgen lost its patent protection for filgrastim in 2008, after developing worldwide sales ∼$4.5 billion.) The European Medicine Agency approved six biosimilars to G-CSF, and in February 2015, the FDA approved the first biosimilar, filgrastim-sndz, in the United States since passage of the Biologics Price Competition and Innovation Act. Teva’s tbo-filgrastim was approved in the United States in 2012 before that legislation. These drugs must demonstrate “high similarity” in the molecular characterization, purity, stability, pharmacokinetics, pharmacodynamics, clinical efficacy, tolerability, and safety to the original agent (69). Pricing analysis suggests that that use of biosimilars to filgrastim, bevacizumab, trastuzumab, and rituximab may save up to $44.2 billion over the next 10 y (96).

G-CSF and/or GM-CSF may improve chemotherapy and immunotherapy of hematologic malignancies and nonblood cancers. For instance, these myeloid growth factors can recruit quiescent leukemic cells into the cell cycle for enhanced killing from cell cycle–specific chemotherapy (74, 97). As a proinflammatory cytokine, GM-CSF is being used to promote dendritic cell activity in a variety of anticancer trials. Indeed, GM-CSF is approved as part of the sipuleucel-T regimen for the treatment of hormone-resistant prostate cancer. There, dendritic cells are incubated with a fusion protein consisting of prostatic acid phosphatase and GM-CSF. Although sipuleucel-T has been underused, in part because of its expense, GM-CSF is being studied in the context of other immunotherapeutic interventions (https://clinicaltrials.gov).

G-CSF has immunomodulatory effects on immune cells. G-CSF enhances Ab-dependent cellular cytotoxicity and cytokine production in neutrophils (98). However, it also inhibits TLR-induced proinflammatory cytokines produced by monocytes and macrophages (99). CD34+ monocytes that inhibit graft-versus-host disease are mobilized in response to G-CSF (100). In addition, G-CSF inhibits LPS-induced IL-12 production from bone marrow–derived dendritic cells cultured in vitro (101). Interestingly, administration of GM-CSF has the opposite effect, inducing cytokine production in the circulation in response to LPS (102).

GM-CSF pathways may be high-value targets in autoimmune diseases. For example, inflammatory bowel disease (IBD) is a chronic inflammatory condition of the gastrointestinal tract caused by a combination of environmental and genetic factors. Crohn’s disease and ulcerative colitis can be difficult to treat, and relapse of disease can occur at any time. Biochemical markers identifying patients at risk for relapse are currently lacking. GM-CSF signaling was recently implicated in the pathogenesis of Crohn’s disease. GM-CSF is required for myeloid cell antimicrobial functions and homeostatic responses to tissue injury in the intestine (103). Preliminary studies found that GM-CSF reduces chemically induced gut injury in mice (104). In human studies, higher concentrations of circulating Abs against GM-CSF are found in patients with active IBD compared with those with inactive disease (103). Currently, several studies and clinical trials are looking at the use of GM-CSF in the treatment of IBD and anti–GM-CSF Ab for the monitoring of disease activity and assessing the risk for recurrence (105, 106).

Pulmonary alveolar proteinosis (PAP) is a rare disorder characterized by accumulation of periodic acid–Schiff+ lipoproteinaceous material in the alveoli of the lung, leading to impaired gas exchange, respiratory insufficiency, and, in severe cases, respiratory failure (107). Autoimmune PAP accounts for 90% of cases and is due to the presence of autoantibodies against GM-CSF. Hereditary PAP is caused by mutations in the genes CSF2RA and CSF2RB that code for the α and β subunit of GM-CSFR, respectively (108, 109). In autoimmune PAP, the presence of anti–GM-CSF Abs leads to aberrant in vivo GM-CSF signaling that is required for the macrophage-mediated clearance, but not uptake, of pulmonary surfactant. This results in the progressive accumulation of foamy surfactant-laden macrophages and intra-alveolar surfactant in the alveoli of the lung (110). The gold standard of therapy has been whole-lung lavage. Although an effective therapy, it often needs to be repeated as a result of reaccumulation of lipoproteinaceous sediment and is not without complications. Newer therapies have been studied, including pulmonary macrophage transplantation, plasmapheresis to remove the GM-CSF autoantibody, and inhaled GM-CSF (111113). Inhaled GM-CSF is of particular interest because it was shown to be safe and effective in animal studies and phase 1 and 2 clinical trials (114, 115).

This work was supported by the National Institutes of Health, the American Society of Hematology, the Leukemia and Lymphoma Society, and the Cures Within Reach Foundation (all to S.J.C.).

Abbreviations used in this article:

AML

acute myeloid leukemia

ANC

absolute neutrophil count

ASCO

American Society of Clinical Oncology

FDA

U.S. Food and Drug Administration

HSC

hematopoietic stem cell

IBD

inflammatory bowel disease

IST

immunosuppressive therapy

MDS

myelodysplastic syndrome

NHL

non-Hodgkin’s lymphoma

PAP

pulmonary alveolar proteinosis

SAA

severe aplastic anemia

SCN

severe congenital neutropenia.

1
Hilton
D. J.
,
Nicola
N. A.
,
Alexander
W. S.
,
Roberts
A. W.
,
Dunn
A. R.
.
2015
.
Donald Metcalf (1929-2014)
.
Cell
160
:
361
362
.
2
Metcalf
D.
1988
.
The Molecular Control of Blood Cells
.
Harvard University Press
,
Cambridge, MA
.
3
Kawamoto
H.
,
Ikawa
T.
,
Masuda
K.
,
Wada
H.
,
Katsura
Y.
.
2010
.
A map for lineage restriction of progenitors during hematopoiesis: the essence of the myeloid-based model
.
Immunol. Rev.
238
:
23
36
.
4
Theilgaard-Mönch
K.
,
Jacobsen
L. C.
,
Borup
R.
,
Rasmussen
T.
,
Bjerregaard
M. D.
,
Nielsen
F. C.
,
Cowland
J. B.
,
Borregaard
N.
.
2005
.
The transcriptional program of terminal granulocytic differentiation
.
Blood
105
:
1785
1796
.
5
Morrison
S. J.
,
Kimble
J.
.
2006
.
Asymmetric and symmetric stem-cell divisions in development and cancer
.
Nature
441
:
1068
1074
.
6
Doulatov
S.
,
Notta
F.
,
Laurenti
E.
,
Dick
J. E.
.
2012
.
Hematopoiesis: a human perspective
.
Cell Stem Cell
10
:
120
136
.
7
Lieschke
G. J.
,
Grail
D.
,
Hodgson
G.
,
Metcalf
D.
,
Stanley
E.
,
Cheers
C.
,
Fowler
K. J.
,
Basu
S.
,
Zhan
Y. F.
,
Dunn
A. R.
.
1994
.
Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization
.
Blood
84
:
1737
1746
.
8
Geijsen
N.
,
Koenderman
L.
,
Coffer
P. J.
.
2001
.
Specificity in cytokine signal transduction: lessons learned from the IL-3/IL-5/GM-CSF receptor family
.
Cytokine Growth Factor Rev.
12
:
19
25
.
9
Stanley
E.
,
Lieschke
G. J.
,
Grail
D.
,
Metcalf
D.
,
Hodgson
G.
,
Gall
J. A.
,
Maher
D. W.
,
Cebon
J.
,
Sinickas
V.
,
Dunn
A. R.
.
1994
.
Granulocyte/macrophage colony-stimulating factor-deficient mice show no major perturbation of hematopoiesis but develop a characteristic pulmonary pathology
.
Proc. Natl. Acad. Sci. USA
91
:
5592
5596
.
10
Hammond
W. P.
,
Boone
T. C.
,
Donahue
R. E.
,
Souza
L. M.
,
Dale
D. C.
.
1990
.
A comparison of treatment of canine cyclic hematopoiesis with recombinant human granulocyte-macrophage colony-stimulating factor (GM-CSF), G-CSF interleukin-3, and canine G-CSF
.
Blood
76
:
523
532
.
11
Wong
G. G.
,
Witek
J. S.
,
Temple
P. A.
,
Wilkens
K. M.
,
Leary
A. C.
,
Luxenberg
D. P.
,
Jones
S. S.
,
Brown
E. L.
,
Kay
R. M.
,
Orr
E. C.
, et al
.
1985
.
Human GM-CSF: molecular cloning of the complementary DNA and purification of the natural and recombinant proteins
.
Science
228
:
810
815
.
12
Souza
L. M.
,
Boone
T. C.
,
Gabrilove
J.
,
Lai
P. H.
,
Zsebo
K. M.
,
Murdock
D. C.
,
Chazin
V. R.
,
Bruszewski
J.
,
Lu
H.
,
Chen
K. K.
, et al
.
1986
.
Recombinant human granulocyte colony-stimulating factor: effects on normal and leukemic myeloid cells
.
Science
232
:
61
65
.
13
Paul
M.
,
Ram
R.
,
Kugler
E.
,
Farbman
L.
,
Peck
A.
,
Leibovici
L.
,
Lahav
M.
,
Yeshurun
M.
,
Shpilberg
O.
,
Herscovici
C.
, et al
.
2014
.
Subcutaneous versus intravenous granulocyte colony stimulating factor for the treatment of neutropenia in hospitalized hemato-oncological patients: randomized controlled trial
.
Am. J. Hematol.
89
:
243
248
.
14
Horan
T.
,
Wen
J.
,
Narhi
L.
,
Parker
V.
,
Garcia
A.
,
Arakawa
T.
,
Philo
J.
.
1996
.
Dimerization of the extracellular domain of granuloycte-colony stimulating factor receptor by ligand binding: a monovalent ligand induces 2:2 complexes
.
Biochemistry
35
:
4886
4896
.
15
Mehta
H. M.
,
Futami
M.
,
Glaubach
T.
,
Lee
D. W.
,
Andolina
J. R.
,
Yang
Q.
,
Whichard
Z.
,
Quinn
M.
,
Lu
H. F.
,
Kao
W. M.
, et al
.
2014
.
Alternatively spliced, truncated GCSF receptor promotes leukemogenic properties and sensitivity to JAK inhibition
.
Leukemia
28
:
1041
1051
.
16
Touw
I. P.
,
van de Geijn
G. J.
.
2007
.
Granulocyte colony-stimulating factor and its receptor in normal myeloid cell development, leukemia and related blood cell disorders
.
Front. Biosci.
12
:
800
815
.
17
Hermans
M. H.
,
van de Geijn
G. J.
,
Antonissen
C.
,
Gits
J.
,
van Leeuwen
D.
,
Ward
A. C.
,
Touw
I. P.
.
2003
.
Signaling mechanisms coupled to tyrosines in the granulocyte colony-stimulating factor receptor orchestrate G-CSF-induced expansion of myeloid progenitor cells
.
Blood
101
:
2584
2590
.
18
Akbarzadeh
S.
,
Ward
A. C.
,
McPhee
D. O.
,
Alexander
W. S.
,
Lieschke
G. J.
,
Layton
J. E.
.
2002
.
Tyrosine residues of the granulocyte colony-stimulating factor receptor transmit proliferation and differentiation signals in murine bone marrow cells
.
Blood
99
:
879
887
.
19
Zhu
Q. S.
,
Robinson
L. J.
,
Roginskaya
V.
,
Corey
S. J.
.
2004
.
G-CSF-induced tyrosine phosphorylation of Gab2 is Lyn kinase dependent and associated with enhanced Akt and differentiative, not proliferative, responses
.
Blood
103
:
3305
3312
.
20
Futami
M.
,
Zhu
Q. S.
,
Whichard
Z. L.
,
Xia
L.
,
Ke
Y.
,
Neel
B. G.
,
Feng
G. S.
,
Corey
S. J.
.
2011
.
G-CSF receptor activation of the Src kinase Lyn is mediated by Gab2 recruitment of the Shp2 phosphatase
.
Blood
118
:
1077
1086
.
21
Jack
G. D.
,
Zhang
L.
,
Friedman
A. D.
.
2009
.
M-CSF elevates c-Fos and phospho-C/EBPalpha(S21) via ERK whereas G-CSF stimulates SHP2 phosphorylation in marrow progenitors to contribute to myeloid lineage specification
.
Blood
114
:
2172
2180
.
22
Laslo
P.
,
Spooner
C. J.
,
Warmflash
A.
,
Lancki
D. W.
,
Lee
H. J.
,
Sciammas
R.
,
Gantner
B. N.
,
Dinner
A. R.
,
Singh
H.
.
2006
.
Multilineage transcriptional priming and determination of alternate hematopoietic cell fates
.
Cell
126
:
755
766
.
23
Hansen
G.
,
Hercus
T. R.
,
McClure
B. J.
,
Stomski
F. C.
,
Dottore
M.
,
Powell
J.
,
Ramshaw
H.
,
Woodcock
J. M.
,
Xu
Y.
,
Guthridge
M.
, et al
.
2008
.
The structure of the GM-CSF receptor complex reveals a distinct mode of cytokine receptor activation
.
Cell
134
:
496
507
.
24
Perugini
M.
,
Brown
A. L.
,
Salerno
D. G.
,
Booker
G. W.
,
Stojkoski
C.
,
Hercus
T. R.
,
Lopez
A. F.
,
Hibbs
M. L.
,
Gonda
T. J.
,
D’Andrea
R. J.
.
2010
.
Alternative modes of GM-CSF receptor activation revealed using activated mutants of the common beta-subunit
.
Blood
115
:
3346
3353
.
25
Hamilton
J. A.
,
Achuthan
A.
.
2013
.
Colony stimulating factors and myeloid cell biology in health and disease
.
Trends Immunol.
34
:
81
89
.
26
Pitrak
D. L.
1997
.
Effects of granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor on the bactericidal functions of neutrophils
.
Curr. Opin. Hematol.
4
:
183
190
.
27
Kolaczkowska
E.
,
Kubes
P.
.
2013
.
Neutrophil recruitment and function in health and inflammation
.
Nat. Rev. Immunol.
13
:
159
175
.
28
Manz
M. G.
,
Boettcher
S.
.
2014
.
Emergency granulopoiesis
.
Nat. Rev. Immunol.
14
:
302
314
.
29
Simon
R.
,
Norton
L.
.
2006
.
The Norton-Simon hypothesis: designing more effective and less toxic chemotherapeutic regimens
.
Nat. Clin. Pract. Oncol.
3
:
406
407
.
30
Norton
L.
,
Simon
R.
,
Brereton
H. D.
,
Bogden
A. E.
.
1976
.
Predicting the course of Gompertzian growth
.
Nature
264
:
542
545
.
31
Norton
L.
,
Simon
R.
.
1986
.
The Norton-Simon hypothesis revisited
.
Cancer Treat. Rep.
70
:
163
169
.
32
Glaubach
T.
,
Minella
A. C.
,
Corey
S. J.
.
2014
.
Cellular stress pathways in pediatric bone marrow failure syndromes: many roads lead to neutropenia
.
Pediatr. Res.
75
:
189
195
.
33
Gilman
P. A.
,
Jackson
D. P.
,
Guild
H. G.
.
1970
.
Congenital agranulocytosis: prolonged survival and terminal acute leukemia
.
Blood
36
:
576
585
.
34
Welte
K.
,
Dale
D.
.
1996
.
Pathophysiology and treatment of severe chronic neutropenia
.
Ann. Hematol.
72
:
158
165
.
35
Hammond IV
W. P.
,
Price
T. H.
,
Souza
L. M.
,
Dale
D. C.
.
1989
.
Treatment of cyclic neutropenia with granulocyte colony-stimulating factor
.
N. Engl. J. Med.
320
:
1306
1311
.
36
Bonilla
M. A.
,
Gillio
A. P.
,
Ruggeiro
M.
,
Kernan
N. A.
,
Brochstein
J. A.
,
Abboud
M.
,
Fumagalli
L.
,
Vincent
M.
,
Gabrilove
J. L.
,
Welte
K.
, et al
.
1989
.
Effects of recombinant human granulocyte colony-stimulating factor on neutropenia in patients with congenital agranulocytosis
.
N. Engl. J. Med.
320
:
1574
1580
.
37
Benson
K. F.
,
Li
F. Q.
,
Person
R. E.
,
Albani
D.
,
Duan
Z.
,
Wechsler
J.
,
Meade-White
K.
,
Williams
K.
,
Acland
G. M.
,
Niemeyer
G.
, et al
.
2003
.
Mutations associated with neutropenia in dogs and humans disrupt intracellular transport of neutrophil elastase
.
Nat. Genet.
35
:
90
96
.
38
Welte
K.
,
Zeidler
C.
,
Reiter
A.
,
Müller
W.
,
Odenwald
E.
,
Souza
L.
,
Riehm
H.
.
1990
.
Differential effects of granulocyte-macrophage colony-stimulating factor and granulocyte colony-stimulating factor in children with severe congenital neutropenia
.
Blood
75
:
1056
1063
.
39
Dale
D. C.
,
Bonilla
M. A.
,
Davis
M. W.
,
Nakanishi
A. M.
,
Hammond
W. P.
,
Kurtzberg
J.
,
Wang
W.
,
Jakubowski
A.
,
Winton
E.
,
Lalezari
P.
, et al
.
1993
.
A randomized controlled phase III trial of recombinant human granulocyte colony-stimulating factor (filgrastim) for treatment of severe chronic neutropenia
.
Blood
81
:
2496
2502
.
40
Gorlin
R. J.
,
Gelb
B.
,
Diaz
G. A.
,
Lofsness
K. G.
,
Pittelkow
M. R.
,
Fenyk
J. R.
 Jr
.
2000
.
WHIM syndrome, an autosomal dominant disorder: clinical, hematological, and molecular studies
.
Am. J. Med. Genet.
91
:
368
376
.
41
Hernandez
P. A.
,
Gorlin
R. J.
,
Lukens
J. N.
,
Taniuchi
S.
,
Bohinjec
J.
,
Francois
F.
,
Klotman
M. E.
,
Diaz
G. A.
.
2003
.
Mutations in the chemokine receptor gene CXCR4 are associated with WHIM syndrome, a combined immunodeficiency disease
.
Nat. Genet.
34
:
70
74
.
42
Semerad
C. L.
,
Liu
F.
,
Gregory
A. D.
,
Stumpf
K.
,
Link
D. C.
.
2002
.
G-CSF is an essential regulator of neutrophil trafficking from the bone marrow to the blood
.
Immunity
17
:
413
423
.
43
McDermott
D. H.
,
Liu
Q.
,
Velez
D.
,
Lopez
L.
,
Anaya-O’Brien
S.
,
Ulrick
J.
,
Kwatemaa
N.
,
Starling
J.
,
Fleisher
T. A.
,
Priel
D. A.
, et al
.
2014
.
A phase 1 clinical trial of long-term, low-dose treatment of WHIM syndrome with the CXCR4 antagonist plerixafor
.
Blood
123
:
2308
2316
.
44
Greenbaum
A. M.
,
Link
D. C.
.
2011
.
Mechanisms of G-CSF-mediated hematopoietic stem and progenitor mobilization
.
Leukemia
25
:
211
217
.
45
To
L. B.
,
Levesque
J. P.
,
Herbert
K. E.
.
2011
.
How I treat patients who mobilize hematopoietic stem cells poorly
.
Blood
118
:
4530
4540
.
46
Scheinberg
P.
,
Young
N. S.
.
2012
.
How I treat acquired aplastic anemia
.
Blood
120
:
1185
1196
.
47
Tichelli
A.
,
Schrezenmeier
H.
,
Socié
G.
,
Marsh
J.
,
Bacigalupo
A.
,
Dührsen
U.
,
Franzke
A.
,
Hallek
M.
,
Thiel
E.
,
Wilhelm
M.
, et al
.
2011
.
A randomized controlled study in patients with newly diagnosed severe aplastic anemia receiving antithymocyte globulin (ATG), cyclosporine, with or without G-CSF: a study of the SAA Working Party of the European Group for Blood and Marrow Transplantation
.
Blood
117
:
4434
4441
.
48
Jeng
M. R.
,
Naidu
P. E.
,
Rieman
M. D.
,
Rodriguez-Galindo
C.
,
Nottage
K. A.
,
Thornton
D. T.
,
Li
C. S.
,
Wiang
W. C.
.
2005
.
Granulocyte-macrophage colony stimulating factor and immunosuppression in the treatment of pediatric acquired severe aplastic anemia
.
Pediatr. Blood Cancer
45
:
170
175
.
49
Kojima
S.
1996
.
Use of hematopoietic growth factors for treatment of aplastic anemia
.
Bone Marrow Transplant.
18
(
Suppl. 3
):
S36
S38
.
50
Marsh
J. C.
2000
.
Hematopoietic growth factors in the pathogenesis and for the treatment of aplastic anemia
.
Semin. Hematol.
37
:
81
90
.
51
Bacigalupo
A.
,
Broccia
G.
,
Corda
G.
,
Arcese
W.
,
Carotenuto
M.
,
Gallamini
A.
,
Locatelli
F.
,
Mori
P. G.
,
Saracco
P.
,
Todeschini
G.
, et al
.
1995
.
Antilymphocyte globulin, cyclosporin, and granulocyte colony-stimulating factor in patients with acquired severe aplastic anemia (SAA): a pilot study of the EBMT SAA Working Party
.
Blood
85
:
1348
1353
.
52
Bacigalupo
A.
,
Bruno
B.
,
Saracco
P.
,
Di Bona
E.
,
Locasciulli
A.
,
Locatelli
F.
,
Gabbas
A.
,
Dufour
C.
,
Arcese
W.
,
Testi
G.
, et al
European Group for Blood and Marrow Transplantation (EBMT) Working Party on Severe Aplastic Anemia and the Gruppo Italiano Trapianti di Midolio Osseo (GITMO)
.
2000
.
Antilymphocyte globulin, cyclosporine, prednisolone, and granulocyte colony-stimulating factor for severe aplastic anemia: an update of the GITMO/EBMT study on 100 patients
.
Blood
95
:
1931
1934
.
53
Kojima
S.
,
Matsuyama
T.
,
Kodera
Y.
,
Nishihira
H.
,
Ueda
K.
,
Shimbo
T.
,
Nakahata
T.
.
1996
.
Measurement of endogenous plasma granulocyte colony-stimulating factor in patients with acquired aplastic anemia by a sensitive chemiluminescent immunoassay
.
Blood
87
:
1303
1308
.
54
Bodey
G. P.
,
Buckley
M.
,
Sathe
Y. S.
,
Freireich
E. J.
.
1966
.
Quantitative relationships between circulating leukocytes and infection in patients with acute leukemia
.
Ann. Intern. Med.
64
:
328
340
.
55
Talcott
J. A.
,
Siegel
R. D.
,
Finberg
R.
,
Goldman
L.
.
1992
.
Risk assessment in cancer patients with fever and neutropenia: a prospective, two-center validation of a prediction rule
.
J. Clin. Oncol.
10
:
316
322
.
56
American Society of Clinical Oncology
.
1994
.
Recommendations for the use of hematopoietic colony-stimulating factors: evidence-based, clinical practice guidelines
.
J. Clin. Oncol.
12
:
2471
2508
.
57
Crawford
J.
,
Ozer
H.
,
Stoller
R.
,
Johnson
D.
,
Lyman
G.
,
Tabbara
I.
,
Kris
M.
,
Grous
J.
,
Picozzi
V.
,
Rausch
G.
, et al
.
1991
.
Reduction by granulocyte colony-stimulating factor of fever and neutropenia induced by chemotherapy in patients with small-cell lung cancer
.
N. Engl. J. Med.
325
:
164
170
.
58
Trillet-Lenoir
V.
,
Green
J.
,
Manegold
C.
,
Von Pawel
J.
,
Gatzemeier
U.
,
Lebeau
B.
,
Depierre
A.
,
Johnson
P.
,
Decoster
G.
,
Tomita
D.
, et al
.
1993
.
Recombinant granulocyte colony stimulating factor reduces the infectious complications of cytotoxic chemotherapy
.
Eur. J. Cancer
29A
:
319
324
.
59
Pettengell
R.
,
Gurney
H.
,
Radford
J. A.
,
Deakin
D. P.
,
James
R.
,
Wilkinson
P. M.
,
Kane
K.
,
Bentley
J.
,
Crowther
D.
.
1992
.
Granulocyte colony-stimulating factor to prevent dose-limiting neutropenia in non-Hodgkin’s lymphoma: a randomized controlled trial
.
Blood
80
:
1430
1436
.
60
Gerhartz
H. H.
,
Engelhard
M.
,
Meusers
P.
,
Brittinger
G.
,
Wilmanns
W.
,
Schlimok
G.
,
Mueller
P.
,
Huhn
D.
,
Musch
R.
,
Siegert
W.
, et al
.
1993
.
Randomized, double-blind, placebo-controlled, phase III study of recombinant human granulocyte-macrophage colony-stimulating factor as adjunct to induction treatment of high-grade malignant non-Hodgkin’s lymphomas
.
Blood
82
:
2329
2339
.
61
Bajorin
D. F.
,
Nichols
C. R.
,
Schmoll
H. J.
,
Kantoff
P. W.
,
Bokemeyer
C.
,
Demetri
G. D.
,
Einhorn
L. H.
,
Bosl
G. J.
.
1995
.
Recombinant human granulocyte-macrophage colony-stimulating factor as an adjunct to conventional-dose ifosfamide-based chemotherapy for patients with advanced or relapsed germ cell tumors: a randomized trial
.
J. Clin. Oncol.
13
:
79
86
.
62
Citron
M. L.
,
Berry
D. A.
,
Cirrincione
C.
,
Hudis
C.
,
Winer
E. P.
,
Gradishar
W. J.
,
Davidson
N. E.
,
Martino
S.
,
Livingston
R.
,
Ingle
J. N.
, et al
.
2003
.
Randomized trial of dose-dense versus conventionally scheduled and sequential versus concurrent combination chemotherapy as postoperative adjuvant treatment of node-positive primary breast cancer: first report of Intergroup Trial C9741/Cancer and Leukemia Group B Trial 9741
.
J. Clin. Oncol.
21
:
1431
1439
.
63
Pfreundschuh
M.
,
Trümper
L.
,
Kloess
M.
,
Schmits
R.
,
Feller
A. C.
,
Rübe
C.
,
Rudolph
C.
,
Reiser
M.
,
Hossfeld
D. K.
,
Eimermacher
H.
, et al
German High-Grade Non-Hodgkin’s Lymphoma Study Group
.
2004
.
Two-weekly or 3-weekly CHOP chemotherapy with or without etoposide for the treatment of elderly patients with aggressive lymphomas: results of the NHL-B2 trial of the DSHNHL
.
Blood
104
:
634
641
.
64
Pfreundschuh
M.
,
Trümper
L.
,
Kloess
M.
,
Schmits
R.
,
Feller
A. C.
,
Rudolph
C.
,
Reiser
M.
,
Hossfeld
D. K.
,
Metzner
B.
,
Hasenclever
D.
, et al
German High-Grade Non-Hodgkin’s Lymphoma Study Group
.
2004
.
Two-weekly or 3-weekly CHOP chemotherapy with or without etoposide for the treatment of young patients with good-prognosis (normal LDH) aggressive lymphomas: results of the NHL-B1 trial of the DSHNHL
.
Blood
104
:
626
633
.
65
Vogel
C. L.
,
Wojtukiewicz
M. Z.
,
Carroll
R. R.
,
Tjulandin
S. A.
,
Barajas-Figueroa
L. J.
,
Wiens
B. L.
,
Neumann
T. A.
,
Schwartzberg
L. S.
.
2005
.
First and subsequent cycle use of pegfilgrastim prevents febrile neutropenia in patients with breast cancer: a multicenter, double-blind, placebo-controlled phase III study
.
J. Clin. Oncol.
23
:
1178
1184
.
66
Timmer-Bonte
J. N.
,
de Boo
T. M.
,
Smit
H. J.
,
Biesma
B.
,
Wilschut
F. A.
,
Cheragwandi
S. A.
,
Termeer
A.
,
Hensing
C. A.
,
Akkermans
J.
,
Adang
E. M.
, et al
.
2005
.
Prevention of chemotherapy-induced febrile neutropenia by prophylactic antibiotics plus or minus granulocyte colony-stimulating factor in small-cell lung cancer: a Dutch Randomized Phase III Study
.
J. Clin. Oncol.
23
:
7974
7984
.
67
Yang
B. B.
,
Lum
P. K.
,
Hayashi
M. M.
,
Roskos
L. K.
.
2004
.
Polyethylene glycol modification of filgrastim results in decreased renal clearance of the protein in rats
.
J. Pharm. Sci.
93
:
1367
1373
.
68
Johnston
E.
,
Crawford
J.
,
Blackwell
S.
,
Bjurstrom
T.
,
Lockbaum
P.
,
Roskos
L.
,
Yang
B. B.
,
Gardner
S.
,
Miller-Messana
M. A.
,
Shoemaker
D.
, et al
.
2000
.
Randomized, dose-escalation study of SD/01 compared with daily filgrastim in patients receiving chemotherapy
.
J. Clin. Oncol.
18
:
2522
2528
.
69
Smith
T. J.
,
Khatcheressian
J.
,
Lyman
G. H.
,
Ozer
H.
,
Armitage
J. O.
,
Balducci
L.
,
Bennett
C. L.
,
Cantor
S. B.
,
Crawford
J.
,
Cross
S. J.
, et al
.
2006
.
2006 update of recommendations for the use of white blood cell growth factors: an evidence-based clinical practice guideline
.
J. Clin. Oncol.
24
:
3187
3205
.
70
Aarts
M. J.
,
Grutters
J. P.
,
Peters
F. P.
,
Mandigers
C. M.
,
Dercksen
M. W.
,
Stouthard
J. M.
,
Nortier
H. J.
,
van Laarhoven
H. W.
,
van Warmerdam
L. J.
,
van de Wouw
A. J.
, et al
.
2013
.
Cost effectiveness of primary pegfilgrastim prophylaxis in patients with breast cancer at risk of febrile neutropenia
.
J. Clin. Oncol.
31
:
4283
4289
.
71
Crawford
J.
,
Armitage
J.
,
Balducci
L.
,
Becker
P. S.
,
Blayney
D. W.
,
Cataland
S. R.
,
Heaney
M. L.
,
Hudock
S.
,
Kloth
D. D.
,
Kuter
D. J.
, et al
National comprehensive cancer network
.
2013
.
Myeloid growth factors
.
J. Natl. Compr. Canc. Netw.
11
:
1266
1290
.
72
Dinan
M. A.
,
Hirsch
B. R.
,
Lyman
G. H.
.
2015
.
Management of chemotherapy-induced neutropenia: measuring quality, cost, and value
.
J. Natl. Compr. Canc. Netw.
13
:
e1
e7
.
73
Rowe
J. M.
,
Andersen
J. W.
,
Mazza
J. J.
,
Bennett
J. M.
,
Paietta
E.
,
Hayes
F. A.
,
Oette
D.
,
Cassileth
P. A.
,
Stadtmauer
E. A.
,
Wiernik
P. H.
.
1995
.
A randomized placebo-controlled phase III study of granulocyte-macrophage colony-stimulating factor in adult patients (> 55 to 70 years of age) with acute myelogenous leukemia: a study of the Eastern Cooperative Oncology Group (E1490)
.
Blood
86
:
457
462
.
74
Stone
R. M.
,
Berg
D. T.
,
George
S. L.
,
Dodge
R. K.
,
Paciucci
P. A.
,
Schulman
P.
,
Lee
E. J.
,
Moore
J. O.
,
Powell
B. L.
,
Schiffer
C. A.
Cancer and Leukemia Group B
.
1995
.
Granulocyte-macrophage colony-stimulating factor after initial chemotherapy for elderly patients with primary acute myelogenous leukemia
.
N. Engl. J. Med.
332
:
1671
1677
.
75
Ohno
R.
,
Tomonaga
M.
,
Kobayashi
T.
,
Kanamaru
A.
,
Shirakawa
S.
,
Masaoka
T.
,
Omine
M.
,
Oh
H.
,
Nomura
T.
,
Sakai
Y.
, et al
.
1990
.
Effect of granulocyte colony-stimulating factor after intensive induction therapy in relapsed or refractory acute leukemia
.
N. Engl. J. Med.
323
:
871
877
.
76
Ozer
H.
,
Armitage
J. O.
,
Bennett
C. L.
,
Crawford
J.
,
Demetri
G. D.
,
Pizzo
P. A.
,
Schiffer
C. A.
,
Smith
T. J.
,
Somlo
G.
,
Wade
J. C.
, et al
American Society of Clinical Oncology
.
2000
.
2000 update of recommendations for the use of hematopoietic colony-stimulating factors: evidence-based, clinical practice guidelines
.
J. Clin. Oncol.
18
:
3558
3585
.
77
Bonilla
M. A.
,
Dale
D.
,
Zeidler
C.
,
Last
L.
,
Reiter
A.
,
Ruggeiro
M.
,
Davis
M.
,
Koci
B.
,
Hammond
W.
,
Gillio
A.
, et al
.
1994
.
Long-term safety of treatment with recombinant human granulocyte colony-stimulating factor (r-metHuG-CSF) in patients with severe congenital neutropenias
.
Br. J. Haematol.
88
:
723
730
.
78
Dale
D. C.
,
Bolyard
A. A.
,
Schwinzer
B. G.
,
Pracht
G.
,
Bonilla
M. A.
,
Boxer
L.
,
Freedman
M. H.
,
Donadieu
J.
,
Kannourakis
G.
,
Alter
B. P.
, et al
.
2006
.
The Severe Chronic Neutropenia International Registry: 10-Year Follow-up Report
.
Support. Cancer Ther.
3
:
220
231
.
79
Rosen
R. B.
,
Kang
S. J.
.
1979
.
Congenital agranulocytosis terminating in acute myelomonocytic leukemia
.
J. Pediatr.
94
:
406
408
.
80
Rosenberg
P. S.
,
Alter
B. P.
,
Bolyard
A. A.
,
Bonilla
M. A.
,
Boxer
L. A.
,
Cham
B.
,
Fier
C.
,
Freedman
M.
,
Kannourakis
G.
,
Kinsey
S.
, et al
Severe Chronic Neutropenia International Registry
.
2006
.
The incidence of leukemia and mortality from sepsis in patients with severe congenital neutropenia receiving long-term G-CSF therapy
.
Blood
107
:
4628
4635
.
81
Sinha
S.
,
Zhu
Q. S.
,
Romero
G.
,
Corey
S. J.
.
2003
.
Deletional mutation of the external domain of the human granulocyte colony-stimulating factor receptor in a patient with severe chronic neutropenia refractory to granulocyte colony-stimulating factor
.
J. Pediatr. Hematol. Oncol.
25
:
791
796
.
82
Germeshausen
M.
,
Ballmaier
M.
,
Welte
K.
.
2007
.
Incidence of CSF3R mutations in severe congenital neutropenia and relevance for leukemogenesis: Results of a long-term survey
.
Blood
109
:
93
99
.
83
Dong
F.
,
Brynes
R. K.
,
Tidow
N.
,
Welte
K.
,
Löwenberg
B.
,
Touw
I. P.
.
1995
.
Mutations in the gene for the granulocyte colony-stimulating-factor receptor in patients with acute myeloid leukemia preceded by severe congenital neutropenia
.
N. Engl. J. Med.
333
:
487
493
.
84
Dong
F.
,
Dale
D. C.
,
Bonilla
M. A.
,
Freedman
M.
,
Fasth
A.
,
Neijens
H. J.
,
Palmblad
J.
,
Briars
G. L.
,
Carlsson
G.
,
Veerman
A. J.
, et al
.
1997
.
Mutations in the granulocyte colony-stimulating factor receptor gene in patients with severe congenital neutropenia
.
Leukemia
11
:
120
125
.
85
Dong
F.
,
Hoefsloot
L. H.
,
Schelen
A. M.
,
Broeders
C. A.
,
Meijer
Y.
,
Veerman
A. J.
,
Touw
I. P.
,
Löwenberg
B.
.
1994
.
Identification of a nonsense mutation in the granulocyte-colony-stimulating factor receptor in severe congenital neutropenia
.
Proc. Natl. Acad. Sci. USA
91
:
4480
4484
.
86
Dong
F.
,
van Paassen
M.
,
van Buitenen
C.
,
Hoefsloot
L. H.
,
Löwenberg
B.
,
Touw
I. P.
.
1995
.
A point mutation in the granulocyte colony-stimulating factor receptor (G-CSF-R) gene in a case of acute myeloid leukemia results in the overexpression of a novel G-CSF-R isoform
.
Blood
85
:
902
911
.
87
Hunter
M. G.
,
Avalos
B. R.
.
1999
.
Deletion of a critical internalization domain in the G-CSFR in acute myelogenous leukemia preceded by severe congenital neutropenia
.
Blood
93
:
440
446
.
88
Ward
A. C.
,
van Aesch
Y. M.
,
Schelen
A. M.
,
Touw
I. P.
.
1999
.
Defective internalization and sustained activation of truncated granulocyte colony-stimulating factor receptor found in severe congenital neutropenia/acute myeloid leukemia
.
Blood
93
:
447
458
.
89
Hermans
M. H.
,
Antonissen
C.
,
Ward
A. C.
,
Mayen
A. E.
,
Ploemacher
R. E.
,
Touw
I. P.
.
1999
.
Sustained receptor activation and hyperproliferation in response to granulocyte colony-stimulating factor (G-CSF) in mice with a severe congenital neutropenia/acute myeloid leukemia-derived mutation in the G-CSF receptor gene
.
J. Exp. Med.
189
:
683
692
.
90
Jeha
S.
,
Chan
K. W.
,
Aprikyan
A. G.
,
Hoots
W. K.
,
Culbert
S.
,
Zietz
H.
,
Dale
D. C.
,
Albitar
M.
.
2000
.
Spontaneous remission of granulocyte colony-stimulating factor-associated leukemia in a child with severe congenital neutropenia
.
Blood
96
:
3647
3649
.
91
Maxson
J. E.
,
Gotlib
J.
,
Pollyea
D. A.
,
Fleischman
A. G.
,
Agarwal
A.
,
Eide
C. A.
,
Bottomly
D.
,
Wilmot
B.
,
McWeeney
S. K.
,
Tognon
C. E.
, et al
.
2013
.
Oncogenic CSF3R mutations in chronic neutrophilic leukemia and atypical CML
.
N. Engl. J. Med.
368
:
1781
1790
.
92
Pardanani
A.
,
Lasho
T. L.
,
Laborde
R. R.
,
Elliott
M.
,
Hanson
C. A.
,
Knudson
R. A.
,
Ketterling
R. P.
,
Maxson
J. E.
,
Tyner
J. W.
,
Tefferi
A.
.
2013
.
CSF3R T618I is a highly prevalent and specific mutation in chronic neutrophilc leukemia
.
Leukemia.
27
:
1870
1830
.
93
Mehta
H. M.
,
Glaubach
T.
,
Long
A.
,
Lu
H.
,
Przychodzen
B.
,
Makishima
H.
,
McDevitt
M. A.
,
Cross
N. C.
,
Maciejewski
J.
,
Corey
S. J.
.
2013
.
Granulocyte colony-stimulating factor receptor T595I (T618I) mutation confers ligand independence and enhanced signaling
.
Leukemia
27
:
2407
2410
.
94
Weise
M.
,
Bielsky
M. C.
,
De Smet
K.
,
Ehmann
F.
,
Ekman
N.
,
Giezen
T. J.
,
Gravanis
I.
,
Heim
H. K.
,
Heinonen
E.
,
Ho
K.
, et al
.
2012
.
Biosimilars: what clinicians should know
.
Blood
120
:
5111
5117
.
95
Mellstedt
H.
,
Niederwieser
D.
,
Ludwig
H.
.
2008
.
The challenge of biosimilars
.
Ann. Oncol.
19
:
411
419
.
96
Sun
D.
,
Andayani
T. M.
,
Altyar
A.
,
MacDonald
K.
,
Abraham
I.
.
2015
.
Potential cost savings from chemotherapy-induced febrile neutropenia with biosimilar filgrastim and expanded access to targeted antineoplastic treatment across the European Union G5 countries: a simulation study
.
Clin. Ther.
37
:
842
857
.
97
Löwenberg
B.
,
van Putten
W.
,
Theobald
M.
,
Gmür
J.
,
Verdonck
L.
,
Sonneveld
P.
,
Fey
M.
,
Schouten
H.
,
de Greef
G.
,
Ferrant
A.
, et al
Dutch-Belgian Hemato-Oncology Cooperative Group
; 
Swiss Group for Clinical Cancer Research
.
2003
.
Effect of priming with granulocyte colony-stimulating factor on the outcome of chemotherapy for acute myeloid leukemia
.
N. Engl. J. Med.
349
:
743
752
.
98
Carulli
G.
1997
.
Effects of recombinant human granulocyte colony-stimulating factor administration on neutrophil phenotype and functions
.
Haematologica
82
:
606
616
.
99
Kim
S. O.
,
Sheikh
H. I.
,
Ha
S. D.
,
Martins
A.
,
Reid
G.
.
2006
.
G-CSF-mediated inhibition of JNK is a key mechanism for Lactobacillus rhamnosus-induced suppression of TNF production in macrophages
.
Cell. Microbiol.
8
:
1958
1971
.
100
D’Aveni
M.
,
Rossignol
J.
,
Coman
T.
,
Sivakumaran
S.
,
Henderson
S.
,
Manzo
T.
,
Santos e Sousa
P.
,
Bruneau
J.
,
Fouquet
G.
,
Zavala
F.
, et al
.
2015
.
G-CSF mobilizes CD34+ regulatory monocytes that inhibit graft-versus-host disease
.
Sci. Transl. Med.
7
:
281ra42
.
101
Martins
A.
,
Han
J.
,
Kim
S. O.
.
2010
.
The multifaceted effects of granulocyte colony-stimulating factor in immunomodulation and potential roles in intestinal immune homeostasis
.
IUBMB Life
62
:
611
617
.
102
Brissette
W. H.
,
Baker
D. A.
,
Stam
E. J.
,
Umland
J. P.
,
Griffiths
R. J.
.
1995
.
GM-CSF rapidly primes mice for enhanced cytokine production in response to LPS and TNF
.
Cytokine
7
:
291
295
.
103
Däbritz
J.
,
Bonkowski
E.
,
Chalk
C.
,
Trapnell
B. C.
,
Langhorst
J.
,
Denson
L. A.
,
Foell
D.
.
2013
.
Granulocyte macrophage colony-stimulating factor auto-antibodies and disease relapse in inflammatory bowel disease
.
Am. J. Gastroenterol.
108
:
1901
1910
.
104
Egea
L.
,
McAllister
C. S.
,
Lakhdari
O.
,
Minev
I.
,
Shenouda
S.
,
Kagnoff
M. F.
.
2013
.
GM-CSF produced by nonhematopoietic cells is required for early epithelial cell proliferation and repair of injured colonic mucosa
.
J. Immunol.
190
:
1702
1713
.
105
Roth
L.
,
MacDonald
J. K.
,
McDonald
J. W.
,
Chande
N.
.
2012
.
Sargramostim (GM-CSF) for induction of remission in Crohn’s disease: a Cochrane inflammatory bowel disease and functional bowel disorders systematic review of randomized trials
.
Inflamm. Bowel Dis.
18
:
1333
1339
.
106
Bonneau
J.
,
Dumestre-Perard
C.
,
Rinaudo-Gaujous
M.
,
Genin
C.
,
Sparrow
M.
,
Roblin
X.
,
Paul
S.
.
2015
.
Systematic review: new serological markers (anti-glycan, anti-GP2, anti-GM-CSF Ab) in the prediction of IBD patient outcomes
.
Autoimmun. Rev.
14
:
231
245
.
107
Rosen
S. H.
,
Castleman
B.
,
Liebow
A. A.
.
1958
.
Pulmonary alveolar proteinosis
.
N. Engl. J. Med.
258
:
1123
1142
.
108
Suzuki
T.
,
Sakagami
T.
,
Young
L. R.
,
Carey
B. C.
,
Wood
R. E.
,
Luisetti
M.
,
Wert
S. E.
,
Rubin
B. K.
,
Kevill
K.
,
Chalk
C.
, et al
.
2010
.
Hereditary pulmonary alveolar proteinosis: pathogenesis, presentation, diagnosis, and therapy
.
Am. J. Respir. Crit. Care Med.
182
:
1292
1304
.
109
Trapnell
B. C.
,
Whitsett
J. A.
,
Nakata
K.
.
2003
.
Pulmonary alveolar proteinosis
.
N. Engl. J. Med.
349
:
2527
2539
.
110
Jouneau
S.
,
Kerjouan
M.
,
Briens
E.
,
Lenormand
J. P.
,
Meunier
C.
,
Letheulle
J.
,
Chiforeanu
D.
,
Lainé-Caroff
C.
,
Desrues
B.
,
Delaval
P.
.
2014
.
[Pulmonary alveolar proteinosis]
.
Rev. Mal. Respir.
31
:
975
991
.
111
Suzuki
T.
,
Arumugam
P.
,
Sakagami
T.
,
Lachmann
N.
,
Chalk
C.
,
Sallese
A.
,
Abe
S.
,
Trapnell
C.
,
Carey
B.
,
Moritz
T.
, et al
.
2014
.
Pulmonary macrophage transplantation therapy
.
Nature
514
:
450
454
.
112
Garber
B.
,
Albores
J.
,
Wang
T.
,
Neville
T. H.
.
2015
.
A plasmapheresis protocol for refractory pulmonary alveolar proteinosis
.
Lung
193
:
209
211
.
113
Tazawa
R.
,
Trapnell
B. C.
,
Inoue
Y.
,
Arai
T.
,
Takada
T.
,
Nasuhara
Y.
,
Hizawa
N.
,
Kasahara
Y.
,
Tatsumi
K.
,
Hojo
M.
, et al
.
2010
.
Inhaled granulocyte/macrophage-colony stimulating factor as therapy for pulmonary alveolar proteinosis
.
Am. J. Respir. Crit. Care Med.
181
:
1345
1354
.
114
Reed
J. A.
,
Ikegami
M.
,
Cianciolo
E. R.
,
Lu
W.
,
Cho
P. S.
,
Hull
W.
,
Jobe
A. H.
,
Whitsett
J. A.
.
1999
.
Aerosolized GM-CSF ameliorates pulmonary alveolar proteinosis in GM-CSF-deficient mice
.
Am. J. Physiol.
276
:
L556
L563
.
115
Tazawa
R.
,
Hamano
E.
,
Arai
T.
,
Ohta
H.
,
Ishimoto
O.
,
Uchida
K.
,
Watanabe
M.
,
Saito
J.
,
Takeshita
M.
,
Hirabayashi
Y.
, et al
.
2005
.
Granulocyte-macrophage colony-stimulating factor and lung immunity in pulmonary alveolar proteinosis
.
Am. J. Respir. Crit. Care Med.
171
:
1142
1149
.

The authors have no financial conflicts of interest.