Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/258829907

Granulocyte-macrophage colony-stimulating factor: Not just another


haematopoietic growth factor

Article in Medical Oncology · November 2014


DOI: 10.1007/s12032-013-0774-6 · Source: PubMed

CITATIONS READS

112 1,209

7 authors, including:

Alejandro Francisco-Cruz Miguel aguilar santelises


University of Texas MD Anderson Cancer Center Universidad Nacional Autónoma de México
41 PUBLICATIONS 1,256 CITATIONS 66 PUBLICATIONS 1,865 CITATIONS

SEE PROFILE SEE PROFILE

Octavio Ramos-Espinosa Dulce Mata-Espinosa


Saint Louis University Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
14 PUBLICATIONS 222 CITATIONS 45 PUBLICATIONS 941 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Immunopathology View project

The role of vasopressin in active pulmonary tuberculosis immunopathology View project

All content following this page was uploaded by Miguel aguilar santelises on 24 November 2014.

The user has requested enhancement of the downloaded file.


Med Oncol (2013) 30:774
DOI 10.1007/s12032-013-0774-6

REVIEW ARTICLE

Granulocyte–macrophage colony-stimulating factor: not just


another haematopoietic growth factor
Alejandro Francisco-Cruz • Miguel Aguilar-Santelises • Octavio Ramos-Espinosa •

Dulce Mata-Espinosa • Brenda Marquina-Castillo • Jorge Barrios-Payan •


Rogelio Hernandez-Pando

Received: 25 September 2013 / Accepted: 13 November 2013


Ó Springer Science+Business Media New York 2013

Abstract Granulocyte–macrophage colony-stimulating Abbreviations


factor (GM-CSF) is often used to treat leucopenia. Other AML Acute myeloid leukaemia
haematopoietins may increase the number of circulating AMØ Alveolar macrophage
leucocytes with higher efficiency, but GM-CSF has addi- BM Bone marrow
tional effects that may be far more relevant than its hae- CISH Cytokine inducible SH2-domain
matopoietic activity. GM-CSF induces differentiation, DC Dendritic cell
proliferation and activation of macrophages and dendritic GRAP GM-CSFRa subunit-associated protein
cells which are necessary for the subsequent T helper cell iNKT Invariant natural killer T
type 1 and cytotoxic T lymphocyte activation. GM-CSF IjK IjB kinase
haematopoietic and non-haematopoietic functions have MØ Macrophage
pro-inflammatory and immune regulatory potential to treat MAPK Mitogen-activated kinase-like protein
a variety of autoimmune diseases and tumours. On the NFjB Nuclear factor kappa-light-chain-enhancer of
other hand, GM-CSF deficiency leads to various immune activated B cells
dysfunctions and the current utilization of GM-CSF as IkB Nuclear factor of kappa light polypeptide
haematopoietic factor might be an accurate but very gene enhancer in B cells inhibitor b
incomplete indication for a cytokine with vast clinical PI3K Phosphatidylinositol 3 kinase
potential. PKC Protein kinase C
PAP Pulmonary alveolar proteinosis
Keywords Haematopoietic growth factor  rhGM-CSF Recombinant human GM-CSF
Granulocyte–macrophage colony-stimulating factor  rhG-CSF Recombinant human granulocyte colony-
Innate immunity  Adaptive immunity stimulating factor
STAT Signal transducers and activators of
transcription
Electronic supplementary material The online version of this SLAP SRC-like adapter protein
article (doi:10.1007/s12032-013-0774-6) contains supplementary
material, which is available to authorized users.
TLR Toll-like receptor

A. Francisco-Cruz  O. Ramos-Espinosa  D. Mata-Espinosa 


B. Marquina-Castillo  J. Barrios-Payan 
R. Hernandez-Pando (&)
Introduction
Department of Pathology, National Institute of Medical Sciences
and Nutrition ‘‘Salvador Zubiran’’, Vasco de Quiroga 15,
Tlalpan, 14000 México City, Mexico Colony-stimulating factors (CSF) are crucial for survival,
e-mail: rhdezpando@hotmail.com proliferation, differentiation, maturation and functional
activation of haematopoietic cells [1]. There are three types
A. Francisco-Cruz  M. Aguilar-Santelises
Department of Immunology, National School of Biological of CSF, namely macrophage colony-stimulating factor (M-
Sciences, National Polytechnic Institute, México City, Mexico CSF) or CSF1, granulocyte–macrophage colony-stimulating

123
Page 2 of 14 Med Oncol (2013) 30:774

factor (GM-CSF) or CSF2 and granulocyte colony-stimu- (Flt3L), erythropoietin (EPO), thrombopoietin (TPO) and
lating factor (G-CSF) or CSF3 (Fig. 1). CSF were uninten- IL-3, IL-5, IL-11 also have haematopoietic activity [1].
tionally discovered when solid-state culture systems allowed GM-CSF was first described for its ability to induce
specific precursors of granulocytes and macrophages to mouse myelopoiesis, in addition to its capacity to stimulate
proliferate and form colonies of maturing but non-autono- granulocyte, macrophage, eosinophil, megakaryocyte and
mous proliferating cells [1, 2]. The newly found cell product erythroid colony formation. However, there was not a clear
that needed to be added to stimulate colony formation functional distinction between CSF and IL-3 until the
received the operational name ‘‘colony-stimulating factor’’ human GM-CSF was sequenced in 1985. The murine GM-
[1, 2]. CSF was sequenced in 1994. Cloning techniques also
As part of the haematopoietin family, GM-CSF is a revealed in 1986 that the hypothetical human ‘‘pluripoie-
four-helix packing cytokine (Fig. 2a). The GM-CSF gene tin’’ was actually G-CSF which has since then been used as
is located in the 5q31 region. Deletion of 5q is associated the most important haematopoietic factor in humans [2–5].
with acute myeloid leukaemia (AML), but AML is not Deletion of genes encoding for EPO, G-CSF, TPO or
always generated by deletion of 5q. The same chromo- M-CSF produces dramatic reductions in the amount of
somal location is associated with other genes encoding cells that are normally stimulated by each one of these
cytokines such as IL-4, IL-5 and IL-13 [2–4]. The stem cell cytokines. However, deletion of the GM-CSF gene only
factor (SCF), leukaemia inhibitory factor, Flt3 ligand reduces neutrophils function without significantly affecting

Fig. 1 GM–CSF relationship Multipotent stem cell


with haematopoiesis. Formation
of blood cells progress from a
Self-renewal
haematopoietic stem cell, which HSC
can undergo either self-renewal
or differentiation into a
multilineage committed
SCF
progenitor cell named common
TPO
myeloid progenitor. This cell
gives rise to a more Primitive progenitor cells
differentiated progenitor by
stimulation with GM–CSF
committed to the granulocytes
CMP
and macrophages (GM) linage.
Finally, these cells give rise to
uni-lineage committed
progenitors for granulocytes or GM-CSF
monocytes. SCF stem cell
factor, TPO thrombopoietin Committed precursor cells
[115]

GM

Linage commited cells

MP GP

M-CSF G-CSF, IL-5, SCF

Monocytes Neutrophils, eosinophils, basophils

123
Med Oncol (2013) 30:774 Page 3 of 14

Fig. 2 Structure, cellular sources and cellular targets of GM–CSF. neutrophils, endothelial cells, CD56hiCD16- NK cells, bone-forming
a Type I cytokine or alpha short chain with four packing helix, native osteoblasts, mast cells, multipotent mesenchymal stromal cells and
GM–CSF is a monomeric glycosylated protein of 127 aa. rhGM-CSF Paneth’s cell. Upon appropriate stimulation, much of the production
is associated with dimeric complexes. Structure obtained from Protein and action of GM-CSF occurs locally at the site of inflammation.
Data Bank PDB: 2GMF. b GM-CSF production usually requires c The major target cells and effects of GM-CSF are maturation in DC,
stimulation with cytokines, antigens, microbial products or inflam- activation, survival and differentiation in granulocytes, steady-state
matory agents (IL-1, TNF-a or LPS). Both in steady state and induced differentiation in alveolar MØ (AMØ) and invariant natural killer T
conditions GM-CSF may be produced by CD4? T cells (TH1, TH2 (iNKT) cells and proliferation in microglia and pneumocytes type I
and TH17), B lymphocytes, macrophages, keratinocytes, eosinophils,

their number in circulation [1]. Recombinant human GM- neutrophils, macrophages (MØ) and dendritic cells (DC)
CSF (rhGM-CSF) has been available in USA for thera- [1, 5, 6].
peutic use since the early nineties. Initially prescribed only
to enhance myeloid reconstitution in transplanted patients,
it is now also used as myeloproliferative agent [1, 5, 6]. GM-CSF expression and target cells
Clinical trials revealed its pro-inflammatory effect and, in
consequence, its potential usefulness as a prophylactic or GM-CSF can be detected under physiological conditions at
adjuvant therapeutic agent for patients at high risk of serum concentrations ranging from 20 to 100 pg/ml [8].
infection [7]. In 1996, the Food and Drug Administration Higher serum levels of GM-CSF require stimulation with
approved GM-CSF for treatment for leucopenia associated cytokines, antigens, microbial products or inflammatory
with fungal infections [7]. GM-CSF applications will be agents such as IL-1, TNF-a or lipopolysaccharide (LPS) [8].
most probably further extended due to its ability to induce In fact, GM-CSF was first purified from the conditioned
inflammatory responses of differentiated eosinophils, medium of lung tissue from mice after LPS administration

123
Page 4 of 14 Med Oncol (2013) 30:774

[8]. GM-CSF is produced by a variety of cells, including vascular endothelial cells, uterine cells, gastrointestinal
stromal cells, Paneth’s cells, MØ, DC, mast cells, endothelial tract epithelial cells, astrocytes, oligodendrocytes and
cells, smooth muscle cells, fibroblasts, chondrocytes, as well microglia cells [9, 15, 16].
as IL-23-stimulated TH17 cells and IL-1b-stimulated TH1
and TH17 cells (Fig. 2b) [8, 9]. Among these, T cell-derived
GM-CSF/GM-CSFR interactions
GM-CSF has a particularly important role in the crosstalk
between antigen-presenting cells and T cells [8, 9]. Although
GM-CSF is able to bind GM-CSFRbc in the absence of
the major role for GM-CSF seems to induce DC maturation,
GM-CSFRa, but the heterodimerization with both subunits
GM-CSF also induces granulocytes activation, microglia
is required for intracellular signal transduction. GM-CSFR
proliferation, steady-state differentiation in invariant natural
activation follows general rules regarding receptor dimer-
killer T (iNKT) cells and survival and differentiation of
ization and transphosphorylation of tyrosine residues in the
alveolar MØ (AMØ; Fig. 2c). Moreover, lymphocytes and
receptor cytoplasmic domains. GM-CSFR does not have
endothelial cells have GM-CSF receptors (GM-CSFR)
intrinsic tyrosine kinase activity. Instead, it requires the
which makes them susceptible to GM-CSF stimulation, but
association of GM-CSFRbc with JAK-2 for GM-CSFRbc
the consequences of such stimulation are not clear [8–11].
transphosphorylation, involving two bc chains closely sit-
GM-CSF knockout (KO) mice do not suffer abnormal-
uated in the cytoplasmic region [17]. Crystallographic
ities in myelopoiesis, but they do have the characteristic
studies have shown that GM-CSFRbc is in fact homodi-
abnormalities of pulmonary alveolar proteinosis (PAP),
meric, but its cytoplasmic regions are quite separated
indicating that GM-CSF might have a significant role in
(120 Å), which makes transphosphorylation difficult to
maintenance of the normal lung physiology [5, 10]. GM-
occur [17–19].
CSF deficient mice also have a deficient iNKT cells dif-
A unique tertiary structure of the GM-CSF/GM-CSFR
ferentiation during thymic ontogeny since GM-CSF acts at
complex has been described. It corresponds to a coordi-
the early thymic iNKT cell precursor stage, after lineage
nated dodecamer structure, which is necessary for receptor
commitment and positive selection by regulating the
activation (Fig. 3a). The association between GM-CSF and
acquisition of secretory function for immune synapse-
GM-CSFR involves three sites of interaction. The first
directed release of immunoregulatory cytokines [5, 10].
interaction is between GM-CSF and GM-CSFRa, the sec-
ond is between GM-CSF and two domains of two different
GM-CSF receptor
GM-CSFRbc molecules and the third one is a stabilizing
site formed between GM-CSFRa and GM-CSFRbc. These
The GM-CSFR is a heterodimer formed by a ligand-spe-
three combinations determine the formation of a higher-
cific a subunit (GM-CSFRa or CD116; 60–80 kDa) and a
order dodecamer complex composed by two hexameric
b subunit (GM-CSFRbc or CD131; 120–140 kDa) shared
complexes linked by a fourth site of interaction. Antibodies
with IL-3 and IL-5 receptors [12]. Both subunits are type I
and mutations directed to the fourth site of interaction
transmembrane glycoprotein structurally characterized by
significantly decrease GM-CSF transduction and initiate
the presence of cytokine-receptor homology modules,
the loss of the dodecamer complex [13]. These interactions,
consisting of two domains of fibronectin type III [13]. GM-
which are only observed in GM-CSFR, explain the trans-
CSFRa binds its ligand with low affinity (KD =
phosphorylation by GM-CSF/GM-CSFR in which JAK-2 is
0.2–100 nM), but the expression of GM-CSFRbc increases
associated with the beta chain. The dodecamer complex
GM-CSFRa affinity to KD = 100 pM [13]. Eight confor-
product relocates closer two GM-CSFRbc, at a distance of
mational variants have been described for GM-CSFRa, but
10 Å permitting transphosphorylation and the activation of
only two are biologically relevant for its transductional
subsequent signalling pathways [13, 19].
effect. The a-1 and a-2 isoforms contain transmembrane
and cytoplasmic regions with abundant serine and proline
residues. The importance of GM-CSFRa is illustrated by GM-CSF signalling
the fact that the complete deletion of its cytoplasmic region
results in lack of cell growth and differentiation. GM- GM-CSFR transduction is similar to the activation carried
CSFRbc is constitutively expressed on the cell surface on by the interferon production regulator family. Binding
[14]. Similarly to IL-3R and IL-5R, GM-CSFR is expres- of GM-CSF with its receptor leads to the activation of
sed at very low levels on the surface of haematopoietic kinases of the JAK family which then phosphorylate signal
cells, with only 100–1,000 molecules per cell [13]. GM- transducers and activators of transcription (STAT) that
CSFR is also expressed on granulocytes and macrophages migrate to the nucleus and bind specific DNA elements
progenitor cells, mature cells such as monocytes, MØ, DC, directing the transcription of specific genes related to cell
megakaryocytes, neutrophils, plasma cells, T lymphocytes, differentiation. GM-CSF induces cell proliferation mainly

123
Med Oncol (2013) 30:774 Page 5 of 14

by activation and signalling of protein kinase C, preventing Chinese hamster ovary cells is named regramostim (Lan-
cell death by phosphatidylinositol 3 kinase and JAK/ ospharma Laboratories, China) [7]. Sargramostim may help
STAT5-Bcl-2 signalling. GM-CSF induces proliferating to prevent infections by enhancing the immune cell func-
and inflammatory responses by mitogen-activated protein tion. It is the only growth factor approved in USA for
kinases (MAPK) and NFjB activation (Fig. 3B) [14, 20]. treatment of older adults with AML, after induction che-
GM-CSF increases AMØ and neutrophils function. motherapy, to shorten the time to neutrophil recovery and
Proliferating AMØ increase their own expression of toll- to reduce the incidence of life-threatening infections [7,
like receptor 4 (TLR4) and nuclear factor kappa-light- 29]. Sargramostim is also approved in USA for myeloid
chain-enhancer of activated B cells (NFjB) by direct GM- reconstitution following allogeneic bone marrow trans-
CSFR and IjB kinase (IjK) interaction. GM-CSF regulates plantation (BMT), autologous BMT or peripheral blood
the expression of TLR2 and TLR4 in neutrophils and TLR2 stem cell transplantation. Furthermore, sargramostim is
in monocytes. Additionally, GM-CSF induces the release also approved for peripheral blood stem cell mobilization,
of IL-12 and TNF-a and enhances the expression of BMT failure and engraftment delay [29].
monocyte chemoattractant protein (MCP)-1 by JAK2-
STAT5 signalling in monocytes [21, 22]. Recently, a spe- Pharmacokinetics and pharmacodynamics of rhGM-
cific signalling pathway has been described for inducing CSF
classic MØ phenotype or M1 and inflammatory DC, which
is described below. SRC-like adapter protein (SLAP) When sargramostim is administered to the patients by
functions as a negative regulator for DC maturation intravenous infusion during 2 h, it reaches a mean beta
induced by GM-CSF. The need for SLAP action suggests a half-life of approximately 60 min. The peak concentra-
specific threshold for correct stimulation by GM-CSF for tions of GM-CSF are observed in blood samples
monocytic DC maturation [23]. obtained during or immediately after completion of the
GM-CSF binding to GM-CSFRbc triggers most of the sargramostim infusion, while minor concentrations are
intracellular signals that induce inflammation, cell survival, still detected in blood 6 h after the beginning of the
differentiation and proliferation in monocytes and granu- infusion [7, 29]. GM-CSF is also detected in serum first
locytes. c-Kit and GM-CSFRa are well-known markers of after 15 min of subcutaneous injection of sargramostim
haematopoietic cells differentiation. Haematopoietic stem to healthy volunteers. Then, the mean beta half-life is
cells down-regulate c-Kit and up-regulate GM-CSFRa approximately 162 min, and peak levels are reached after
when they enter to the granulocyte/macrophage lineage one to 3 h post-injection and remain detectable for up to
[24, 25]. GM-CSFRa has two isoforms. Stimulation of the 6 h. Parenteral administration of rhGM-CSF alters the
GM-CSFRa-1 isoform induces overexpression of CD86 kinetics of myeloid progenitor cells in bone marrow
and F4/80 cell surface proteins, and it may also induce accelerating the cell-cycle entry and reducing the cell-
intense phosphorylation of JAK-2 [26]. GM-CSFRa-2 cycle time. These effects are reversible once adminis-
isoform has a different cytoplasmic domain, but it associ- tration is discontinued. However, the numbers of mobi-
ates with GM-CSFRbc in the same manner than GM- lized progenitor cells that are released in peripheral
CSFRa-1 does it [27]. There also exist soluble isoforms of blood by GM-CSF are 10 times lower than those
both a-subunits derived from alternative mRNA splicing, released by G-CSF [6, 30].
but their functions are not known [27, 28].
rhGM-CSF and rhG-CSF in leucopenia

GM-CSF clinical applications as haematopoietin rhGM-CSF produces small increases in the number of
peripheral neutrophils and circulating monocytes. It is
GM-CSF is the growth factor most extensively used as currently indicated as second-line treatment for patients
hemopoietin in the clinical practice. rhGM-CSF is pro- with severe neutropenia [7, 31]. rhGM-CSF also induces a
duced in S. cerevisiae, and it is named sargramostim or mild increase in the number of circulating eosinophils and
LeukineÒ (Bayer HealthCare Pharmaceuticals, USA). Its basophils and enhances the phagocytic function of neu-
amino acid sequence is homologous to the endogenous trophils. Filgrastim (rhG-CSF) and pegfilgrastim (pegylat-
human GM-CSF, except for a leucine instead of a proline ed rhG-CSF) are currently approved by FDA for prevention
at the 23rd position and different glycosylated motives. The of chemotherapy-induced neutropenia [1, 7].
degree of glycosylation affects the biological activity, The therapeutic application of rhGM-CSF for leuco-
antigenicity, toxicity and pharmacokinetics. When rhGM- penia could be overestimated. G-CSF recommendations
CSF is produced in E. coli is named molgramostim for neutropenia are supported by various results from
(Zenotech Laboratories, India) and when it is produced in clinical trials and head-to-head comparative studies about

123
Page 6 of 14 Med Oncol (2013) 30:774

clinical benefits of G-CSF compared with GM-CSF are Network (NCCN) and the American Society of Clinical
lacking. Moreover, G-CSF has higher myeloproliferative Oncology (ASCO) recommending rhG-CSF as first-line
activity than GM-CSF [30]. CD34? mononuclear cells therapy for febrile neutropenia [31].
mobilization into the peripheral blood by GM-CSF
administration is approximately 10 times smaller than
G-CSF (12.6 9 106 cells vs. 119 9 106 cells, respec- Toxicological profile of rhGM-CSF
tively). These effects are not synergistic when both CSF
are combined [6, 7, 30]. Besides, there are ample differ- Sargramostim is usually well tolerated by healthy subjects,
ences in therapeutic doses between G-CSF (5 lg/kg) and who have not shown clinical alterations in their clinical
GM-CSF (250 lg/m2), which are considered in the clin- analysis, as compared to placebo-treated individuals [29].
ical guidelines from the National Comprehensive Cancer NCCN guidelines recommend sargramostim administration

123
Med Oncol (2013) 30:774 Page 7 of 14

b Fig. 3 Dodecameric complex of GM-CSFR activation and signalling Non-haematopoietic functions of GM-CSF
pathways induced by GM–CSF. a Schematic representation of GM-
CSFR and steps for dodecameric complex formation and activation.
Low-affinity binary complex consists of linked GM–CSF and GM- GM-CSF has significant non-haematopoietic effects in
CSFRa. Interaction with free dimeric forms of GM-CSFRbc chains various physiological processes other than haematopoiesis.
makes possible the formation of a hexameric complex formed by two GM-CSF KO mice do not suffer alterations in the steady
GM-CSFRbc chains. Each one of these two chains is joined to one state of haematopoiesis. Instead, GM-CSF KO animals
GM-CSFRa chain and carries a GM-CSF molecule (2:2:2). The
dodecameric complex, the highest order complex, is formed by the acquire a particular lung injury phenotype characterized by
lateral aggregation of two hexameric complexes thanks to a fourth site accumulation of lipids and surfactant proteins in alveolar
of interaction that forms a signalling competent structure, diminishing spaces, similar to the human condition described as PAP
the distance between bc chains and allowing the transphosphorylation [3, 4, 34].
of JAK-2. b GM–CSF exerts its biological functions by phosphory-
lating at least two domains in the b-chain of its receptor. Once GM-
CSFRbc is transphosphorylated, adapter proteins and kinases are Inflammation
recruited for inducing activation of nuclear factors that up-regulate the
expression of specific genes related to inflammation, survival, The contribution of GM-CSF to the inflammatory response
differentiation and cell proliferation. One of the GM-CSFRbc trans-
phosphorylated domains induces the activation of Ras and mitogen- has been evaluated in vitro. rhGM-CSF induces longer
activated protein kinases (MAPK) with the consequent induction of survival in monocytes, MØ and neutrophils, augments the
c-fos and c-jun and PI3 K/Akt/p21waf-1. The other phosphorylated amount of inflammatory mediators released by these cells
domain mediates the activation of Janus kinase 2 (JAK2)/signal and contributes to the elimination of pathogens and
transducer and activator of transcription 5 (STAT5) signalling
pathway. Activated STAT5 migrates into the nucleus after mutual tumours [35, 36]. MØ are more efficiently stimulated by
phosphorylation and dimerization and binds to specific DNA-binding secondary stimuli such as LPS and IFN-c, when they are
sites, followed by the expression of STAT5 target genes. Jak kinase– initially stimulated with GM-CSF. Intra-peritoneal admin-
STAT5 transcription factor pathway induces the expression of istration of GM-CSF induces a strong recruitment of
interferon-regulating factor 5 (IRF5). High expression of IRF5 results
in classical MØ activation (M1) or inflammatory MØ induction. GM– human MØ, and both human and murine monocytes show
CSF supports differentiation, survival and proliferation in part through higher pro-inflammatory response when they are primary
phosphorylated STAT5 (pSTAT5) signalling. In parallel, pSTAT stimulated with GM-CSF and then restimulated with LPS
activity leads to an accumulation of cytoplasmic cytokine inducible [37, 38].
SH2-domain protein (CISH). CISH induces feedback inhibition of
STAT5 activation, triggering the differentiation of type 1 polarized One of the most interesting features of GM-CSF was
DC. GM-CSFRa subunit-associated protein (GRAP), a protein that observed in patients with Felty’s syndrome (rheumatoid
binds to the cytoplasmic region of the alpha chain, is highly expressed arthritis, splenomegaly and neutropenia) that received
in neoplastic cells rhGM-CSF and experienced worsening of arthritis [39].
Additionally, patients with rheumatoid arthritis (RA) also
to 55 years old and older AML patients 24 h after induc- experienced symptoms exacerbation when treated with
tion chemotherapy [31]. Although sargramostim adminis- rhGM-CSF after chemotherapy [40]. Moreover, the exac-
tration increases cellularity in haematological, heart and erbation of RA symptoms may be reproduced with GM-
lung tissue on monkeys, these effects have not been serious CSF administration in experimental models [41, 42]. On
enough to fulfil toxicity standard criteria [29]. However, the other hand, administration of mavrilimumab, a human
sargramostim-treated patients with febrile neutropenia monoclonal antibody targeting GM-CSFRa, induces rapid
secondary to chemotherapy, risk to develop fluid retention, and significant responses in RA subjects [43].
respiratory symptoms due to sequestration of granulocytes
in pulmonary circulation, cardiovascular symptoms, renal Activation and regulation of the immune system
and hepatic dysfunction, as well as adverse reactions
related to bone marrow transplant or peripheral blood GM-CSF stimulates the expression of adhesion molecules,
progenitor cell transplant [31]. Paediatric patients suffering IgGFcR and activated complemented receptors on neutro-
Crohn’s disease have been treated with subcutaneous doses phils, enhancing their response to chemotactic factors,
of 4 or 6 lg/kg sargramostim every day during 8 weeks, phagocytosis, synthesis of leukotriene B4, arachidonic acid
and 90 % of them only had mild injection site reactions, release and superoxide anion generation. Moreover, GM-
whereas the resting 10 % required dose reductions due to CSF increases survival of neutrophils and induces its
elevated absolute neutrophil counts [32]. Syncope is the MHC-II expression enabling them to activate T cells
worst side effect triggered thus far by sargramostim response to superantigens [44, 45].
administration to human subjects. Since it was attributed to AMØ orchestrate structural and functional parenchyma
EDTA that was included in the liquid form, the FDA repair processes that are essential for homoeostasis [46].
prohibited the preparation of LeukineÒ in its liquid for- Different extracellular signals are integrated to shape up
mulation [33]. pulmonary MØ phenotypes, during lung inflammation

123
Page 8 of 14 Med Oncol (2013) 30:774

TNF-a released from activated AMØ induces epithelial has been related to the pathogenesis of autoimmune
GM-CSF expression, which initiates alveolar epithelial cell encephalomyelitis [53].
proliferation and helps to restore the alveolar barrier
function [47]. Pneumocytes type II express GM-CSFRa GM–CSF relationships with disease
and GM-CSFRbc. Proliferating type II pneumocytes may
then down-regulate GM-CSFR expression during its trans- The ability of GM-CSF to regulate the immune system has
differentiation into type I pneumocytes occurring in been related to Felty’s syndrome, RA and various other
response to acute lesion [46, 47]. autoimmune, cardiovascular and metabolic diseases [43,
GM-CSF regulates the inflammatory response by acti- 54]. Local over expression of GM-CSF in the stomach of
vation of the JAK kinase–STAT5 transcription factor mice leads to autoimmune gastritis [55]. On the other hand,
pathway, inducing the expression of the interferon-regu- GM-CSF deficient mice are less prone to develop experi-
lating factor 5 (IRF5). A high IRF5 expression by GM-CSF mental allergic encephalitis, myocarditis and collagen-
or IFN-c results in M1 MØ activation. IRF5 acts together induced arthritis [5, 8, 10]. GM-CSF promotes autoim-
with RelA (NFjB protein) inducing TNF-a gene expres- munity by enhancing IL-6-dependent survival of antigen-
sion and IRF5 inhibits the transcription of IL-10 and pro- specific T CD4? cells. It has been described as the en-
motes IFN-c expression [48]. In contrast, when M-CSF cephalitogenic factor produced by IL-23-driven pathogenic
predominates, IRF4 is overexpressed inducing a M2 MØ TH cells. GM-CSF is required for the response to IL-6 and
phenotype. Several cell types, including fibroblasts, endo- IL-23 by DC and subsequent TH17 cell generation [54].
thelial cells, stromal cells and osteoblasts, constitutively Moreover, GM-CSF promotes autoimmunity by enhancing
produce M-CSF. There is a steady-state production of IL-6-dependent survival of antigen-specific T CD4? cells
M-CSF that turns MØ towards the M2 phenotype trough [54, 55].
IRF4, probably balancing the pro-inflammatory microen- GM-CSF is expressed by adipose tissue, but its role in
vironment achieved by the expression of IL-12, IL-6 and there remains unclear. GM-CSF KO mice are obese,
IL-23 that are produced by GM-CSF-stimulated MØ [46, hyperphagic and show larger mesenteric fat adipocytes,
48]. decreased number of MØ, lower transcription of pro-
GM-CSF has an important role in the production of BM- inflammatory cytokines and enhanced peripheral uptake of
derived type 1 DC. BM-derived DC acquires potent anti- glucose when receive a high-fat diet [56]. iNKT cells
gen-presenting capacity, representing a crucial reservoir of constitute up to 50 % of liver lymphocytes, and the relation
professional antigen-presenting cells (APC). In type 1 DC between GM-CSF, liver iNKT cells and metabolism has
differentiation, the expression and activation of STAT5 is not yet been investigated [57]. On the other hand, M-CSF
gradually enhanced by GM-CSF during the early stage of production in CD68? MØ found in human fatty streaks and
DC development. In parallel, pSTAT activity leads to the atherosclerotic plaques suggests that the ratio between
accumulation of cytoplasmic cytokine inducible SH2- M-CSF and GM-CSF is important to determine MØ het-
domain (CISH) protein, at the later stage [49]. When a erogeneity in atherosclerotic lesions [58]. The antagonism
threshold concentration of CISH is reached, CISH inhibits between M-CSF and GM-CSF is also important to define
the GM-CSF-mediated STAT5 phosphorylation, triggering the functional polarization of M2 and M1 MØ during the
its differentiation to strong type 1 polarized DC, promoting course of diverse inflammatory and metabolic disorders
MHC class I expression. Thus, CISH can act as a molecular [59, 60]. MØ migration to areas of myocardial infarction is
switch from the DC progenitor stage to immature DC and a associated with tissue damage, whereas the treatment with
control point for cytotoxic T lymphocytes activation [49]. anti-GM-CSF antibodies decreases the amount of damaged
GM-CSF is a DC-activating cytokine inducing a strong tissue after infarction [61]. A model of cerebral ischaemia
differentiation of TH1 type cells. GM-CSF enhances the also shows that GM-CSF promotes collateral arteries
oxidative metabolism, cytotoxicity and antibody-dependent growth and reduces cerebral ischaemia [62].
phagocytosis [7, 9]. Human monocytes and monocyte-
derived DC treated with GM-CSF exhibited higher GM–CSF relationship with tumour cells
expression of MHC-II molecules, CD80, CD86 and CD40,
increasing the immune response against antimicrobial The presence of GM-CSFR on tumour cells makes them
agents [50, 51]. However, the endometrium originated susceptible to GM-CSF stimulation. GM-CSFRa binding
GM-CSF induces tolerance during early pregnancy [52], may be particularly important for signal transduction in
GM-CSF directs DC from the central nervous system tumour cells overexpressing the GM-CSFRa subunit-
(CNS) towards an inhibitory phenotype. In contrast, DC associated protein [63]. The interaction between tumour
recruited by GM-CSF from the peripheral circulation into cells and sensory neurons seems to be part of the patho-
the CNS tissue has a pro-inflammatory phenotype, which physiology of cancer-induced pain [64, 65]. The intense

123
Med Oncol (2013) 30:774 Page 9 of 14

bone pain occurring as side effect in rhGM-CSF-treated tumour cells proliferation may not be the predominant
patients that suffer myelodysplastic syndromes may be result, due to the presence of GM-CSF and interaction with
caused by a two-way interaction between GM-CSFRa other GM-CSF responder cells. If tumour growth promo-
bearing tumour cells and neurons [66]. GM-CSFRa is also tion occurs, it may be rather due to M-CSF stimulation,
expressed on pancreatic nerves from healthy subjects and leading to activation of M2 MØ and other tumour-associ-
in large hypertrophic nerves close to tumours in pancreatic ated macrophages that produce anti-inflammatory cyto-
carcinoma patients [67]. GM-CSFR on nerve fibres may kines which are linked to in vivo growth, migration and
contribute to tumour-induced pain through the JAK- spreading of a variety of cancers [48, 71].
STAT3 pathway and ERK1/2 signalling involving both
GM-CSF and G-CSF (Fig. 4) [66, 67].
Myeloid-derived suppressive cells (MDSC) are a het- Therapeutic applications of GM-CSF
erogeneous population of immature granulocytic and
monocytic cells, not expressing cell surface markers GM-CSF KO mice are more susceptible than wild-type
associated with differentiated monocytes, MØ or DC. mice to several infections and intestinal inflammation [72].
MDSC exert an immunosuppressive effect of the immune Whereas GM-CSF production is necessary to maintain a
response against cancer [68]. Although the suppression fully functional intestinal innate immunity and to facilitate
mechanism is not clear, it has been strongly associated with recruitment and survival of intestinal DC, lack of GM-CSF
the local presence of MDSC and M2 MØ in mice and may be related to an immune deficiency associated with
humans [68, 69]. In humans, MØ can be differentiated Crohn’s disease [73]. Mutations in the intracellular pattern
either towards pro-inflammatory M1 MØ or anti-inflam- recognition receptor NOD2, also known as inflammatory
matory M2 MØ. M1 MØ express IL-12high, IL- 23high, IL- bowel disease protein 1, have been identified in patients
10low and have high bactericidal and tumoricidal capacity, with Crohn’s disease. These patients produce normal levels
whereas M2 MØ expressing IL-12low, IL-23low, IL-10high of TNF-a, but they do not produce enough GM-CSF [74].
are involved with promotion of tissue remodelling, tumour Multicentre randomized clinical trials have demonstrated
growth and immunoregulation [70]. The possibility exists that subcutaneous administration of GM-CSF induces sig-
that tumour cells may be favoured by GM-CSF since they nificant remission of Crohn’s disease [75, 76]. PAP is
can be stimulated to in vitro proliferation. However, another disease in which GM-CSF production is relevant.

Inflammatory cells
Release of proinflammatory
cytokines

Tumor cells Sensory nerves


Proliferation Hyperalgesia
Nerver emodelling

Fig. 4 Tumour–nerve interactions and cancer-induced pain. GM– endings in the skin mediating the sensitization of nociceptors
CSF may contribute to autocrine and paracrine mechanisms involving following exposure to CSF trough JAK-STAT3 and ERK1/2 signal-
proliferation of tumour cells. GM–CSF sensitizes nerves to mechan- ling. GM–CSF production can lead to recruitment and activation of
ical stimuli in vitro and in vivo, potentiates calcitonin gene-related inflammatory MØs and monocytes and subsequent release of pro-
peptide (CGRP) release and causes sprouting of sensory nerve inflammatory cytokines such as TNF-a

123
Page 10 of 14 Med Oncol (2013) 30:774

PAP is a fatal lung disease characterized by respiratory S. aureus in children with HIV infection [93]. The use of
failure secondary to a deficient AMØ clearance, abnor- GM-CSF in haematopoietic stem cell transplanted patients
malities in host defence and accumulation of surfactant reduces the incidence of bacterial infections and decreases
proteins [34]. Autoantibodies against GM-CSF and defi- the infection-related hospitalization rates [94]. Local
cient expression of the GM-CSFRa or bc chain have been administration of GM-CSF protects mice against lethal
described in patients with PAP [34]. rhGM-CSF inhalation pneumococcal pneumonia and Chlamydia trachomatis
is beneficial for PAP patients, and gene therapy with vec- [95–97].
tors encoding GM-CSFRbc has shown efficacy in pre- In vitro studies demonstrated that human MØ stimula-
clinical results [77, 78]. tion with GM-CSF enhances microbicidal response against
Recent clinical data suggest that the expression of GM- M. tuberculosis [98]. GM-CSF KO mice are highly sus-
CSF may enhance the immune response to tumours. The ceptible to mycobacterial infections that produce intra-
first FDA approved vaccine against tumours (Sipuleucel-T; bronchial and intra-alveolar lesions without formation of
ProvengeÒ) is an individually tailored DC-based vaccine granulomas that are usually produced in wild-type mice
that incorporates the use of a key prostate cancer antigen [99]. GM-CSF protects alveolar structure and regulates
and GM-CSF to activate DC ex vivo, which upon DC early recruitment of MØ and DC, which help to control
reconstitution recruits and stimulates T cells to destroy mycobacteria growth through granulomas formation [100].
prostate tumour cells [79]. Another therapy approach has In mice, overexpression of GM-CSF by gene therapy has
been to reduce the elevated levels of transforming growth shown protection for the reactivation of latent tuberculosis
factor (TGF)-b2 that are frequently linked to immunosup- and avoids the transmissibility of hypervirulent M. tuber-
pression in cancer patients by simultaneously administer- culosis [101].
ing a TGF-b2 antisense and GM-CSF plasmid and cancer The administration of GM-CSF to septic patients did not
vaccines destined to enhance autologous tumour cells produce favourable results. GM-CSF mildly increases the
recognition [80]. A third type of is currently in a phase III number of circulating neutrophils in small-for-gestational
clinical trial to evaluate the results of expressing GM-CSF age babies but do not improve neonatal sepsis-free survival
together with IL-12 in allogeneic cancer cells as treatment [102]. Besides, production of GM-CSF negatively corre-
for lung cancer [81, 82]. This kind of therapy may be also lates with survival to septic shock [103]. Extremely high
effective against colorectal cancer, metastatic renal cell doses of GM-CSF may have a negative impact with
carcinoma and pancreatic adenocarcinoma [83–85]. The extensive lesions and production of cytokines and inflam-
experimental administration of an adenovirus encoding matory mediators that failed to concentrate T cells and MØ
GM-CSF together with an adenovirus encoding p16 tumour into sites of infection, MØ accumulation, blindness and
suppressor gene induces a potent antitumour response by severe damages to various tissues [104, 105].
increasing the expression of MHC I molecules and the
cytotoxic activity of CD8 T cells [86]. Moreover, the
administration of therapeutic vaccines with plasmids Prophylactic vaccines based on GM-CSF
encoding GM-CSF and tumour antigens enhances the IgG
antibody response antitumour antigens, as well as the IFN- The adjuvant role of GM-CSF in prophylactic vaccines
c and IL-6 production [87, 88]. Another interesting strat- against infectious diseases has been evaluated in preclinical
egy, still in experimental phase, is the combined applica- trials. Vaccines based on recombinant vectors encoding
tion of armed oncolytic adenovirus expressing IL-12 and mycobacterial antigens or vaccines based on recombinant
GM-CSF with radiotherapy to suppress primary hepato- BCG have shown effectiveness in experimental models of
carcinoma in mice [89]. TB. However, the use of GM-CSF as adjuvant improves
In vitro and in vivo microbicidal activity of neutrophils protection against disseminated infection increasing the
and MØ is enhanced by GM-CSF. GM-CSF also prevents number of APC as well as the production of IL-12, IFN-c,
the immunosuppressant effect of dexamethasone, allowing antibodies and the cytotoxic response [106–111]. Intra-
an effective response against Aspergillus fumigatus hyphae cerebral administration of recombinant rabies virus
[90]. Preincubation of neutrophils with GM-CSF increases encoding GM-CSF prevents the development of experi-
the fungicidal activity against Candida glabrata and His- mental rabies after infection with the wild-type virus [112].
toplasma capsulatum [7, 91, 92]. In despite of this, there A similar DNA vaccine co-expressing GM-CSF against
are no current clinical trials that focus on GM-CSF effect Clostridium botulinum has shown protection [113]. GM-
as an adjuvant for the treatment against invasive fungal CSF is also under evaluation as a new tool for the treatment
diseases. There are also reports about GM-CSF antibacte- for TB, aspergillosis, candida, and influenza [5, 10, 114].
rial activity in immunodeficient patients. GM-CSF enhan- GM-CSF may induce tumour cell death by activation of
ces ex vivo neutrophil bactericidal activity against circulating monocytes. This and its other immunomodulatory

123
Med Oncol (2013) 30:774 Page 11 of 14

properties are currently exploited by means of its adminis- Acknowledgments A.F.C., O.R.E., and M.A.S., are scholarship
tration as adjuvant of vaccines against prostate cancer, and it is holders from the National Council for Science and Technology
(CONACYT), Mexico. Point of view and discussion from León Islas
also under evaluation in clinical trials for treatment for mel- is gratefully acknowledged. The graphic design expertise from Ari-
anoma and haematological malignancies. Nine hundred sixty- adna Méndez is gratefully acknowledged. The authors declare not to
eight studies were found related to ‘‘sargramostim’’ or ‘‘GM- have any conflict of interest related to publishing this article.
CSF’’ in the clinicaltrials.gov page in October 2013. Most
current studies are analysing the usefulness of GM-CSF in
haematological disorders and cancer as adjuvant therapy. References
Additionally, GM-CSF is also currently being evaluated as
adjuvant for prophylactic vaccines against HIV, sepsis, peri- 1. Metcalf D. Hematopoietic cytokines. Blood. 2008;111:485–91.
2. Bradley TR, Metcalf D. The growth of mouse bone marrow cells
odontitis and Crohn’s disease. Table 1 (offered as Supple- in vitro. Aust J Exp Biol Med Sci. 1966;44:287–99.
mental Material) shows some of the most interesting ongoing 3. Sheridan JW, Metcalf D. A low molecular weight factor in lung-
clinical trials and the potential clinical applications of GM- conditioned medium stimulating granulocyte and monocyte
CSF that now motivate pharmaceutical companies to inves- colony formation in vitro. J Cell Physiol. 1973;81:11–23.
4. Burgess AW, Camakaris J, Metcalf D. Purification and proper-
tigate the immunoregulatory and protective effects of this ties of colony-stimulating factor from mouse lung–conditioned
cytokine in a variety of illnesses. medium. J Biol Chem. 1977;252:1998–2003.
5. Hamilton JA. Colony-stimulating factors in inflammation and
autoimmunity. Nat Rev Immunol. 2008;8:533–44.
6. Lane TA, Law P, Maruyama M, et al. Harvesting and enrich-
Conclusion ment of hematopoietic progenitor cells mobilized into the
peripheral blood of normal donors by granulocyte-macrophage
CSF has a long history of clinical use, but their entire colony-stimulating factor (GM-CSF) or G-CSF: potential role in
potential has not yet been fully exploited. They have been allogeneic marrow transplantation. Blood. 1995;85:275–82.
7. Armitage JO. Emerging applications of recombinant human
almost exclusively applied to increase the absolute num- granulocyte-macrophage colony-stimulating factor. Blood.
bers of circulating innate immune effector cells which 1998;92:4491–508.
greatly differ since every CSF has different effect on bone 8. Conti L, Gessani S. GM-CSF in the generation of dendritic cells
marrow cells production. Furthermore, neither maturation, from human blood monocyte precursors: recent advances.
Immunobiology. 2008;213:859–70.
chemotaxis, phagocytosis nor microbicidal functions have 9. Fukuzawa H, Sawada M, Kayahara T, et al. Identification of
been considered when analysing the effect of diverse CSF, GM-CSF in Paneth cells using single-cell RT-PCR. Biochem
which has made researchers to miss the very important Biophys Res Commun. 2003;312:897–902.
value of GM-CSF as immunoregulatory cytokine. It is 10. Hamilton JA, Anderson GP. GM-CSF biology. Growth Factors.
2004;22:225–31.
quite intriguing how little overlap there is in biological 11. Xing Z, Braciak T, Ohkawara Y, et al. Gene transfer for cytokine
actions among lymphoid and haematopoietic regulatory functional studies in the lung: the multifunctional role of GM-CSF
factors. One difference between haematopoietic and lym- in pulmonary inflammation. J Leukoc Biol. 1996;59:481–8.
phoid regulatory factors is the circumscribed site in bone 12. de Groot RP, Coffer PJ, Koenderman L. Regulation of prolif-
eration, differentiation and survival by the IL-3/IL-5/GM-CSF
marrow of their production and action confining its stim- receptor family. Cell Signal. 1998;10:619–28.
ulation to specific cell colony types. This is one of the most 13. Hansen G, Hercus TR, McClure BJ, et al. The structure of the
important differences between G-CSF GM-CSF and GM-CSF receptor complex reveals a distinct mode of cytokine
M-CSF, which should be considered as cytokines that receptor activation. Cell. 2008;134:496–507.
14. Matsuguchi T, Zhao Y, Lilly M, et al. The cytoplasmic domain
affect rather the quality than the quantity of leucocytes in of the granulocyte-macrophage colony-stimulating factor (GM-
circulation. Such functional effect should be first consid- CSF) receptor subunit is essential for both GM-CSF-mediated
ered in the clinical guidelines for therapeutic use. growth and differentiation. J Biol Chem. 1997;272:17450–9.
The inclusion of biological products has changed and 15. McClure BJ, Hercus TR, Cambareri BA, et al. Molecular
assembly of the ternary granulocyte-macrophage colony-stimu-
expanded the possibilities of modern therapy. GM-CSF lating factor receptor complex. Blood. 2003;101:1308–15.
effects on the inflammatory response influence the outcome 16. Sawada M, Itoh Y, Suzumura A, et al. Expression of cytokine
of multiple tissue insults in several systems and organs. receptors in cultured neuronal and glial cells. Neurosci Lett.
Virus-based immune agents such as GM-CSF armed vec- 1993;160:131–4.
17. Carr PD, Gustin SE, Church AP, et al. Structure of the complete
tors are among the early efforts to reach a potent immu- extracellular domain of the common subunit of the human GM-
nostimulation therapy. GM-CSF has a wide range of CSF, IL-3, and IL-5 receptors reveals a novel dimer configura-
lymphoid regulatory actions, linking innate and adaptive tion. Cell. 2001;104:291–300.
immunity, acting as a bridge between haematopoietic and 18. McClure BJ, Hercus TR, Cambareri BA, et al. Molecular
assembly of the ternary granulocyte-macrophage colony-stimu-
lymphoid factors and should be considered first-line treat- lating factor receptor complex. Blood. 2003;101:1308–15.
ment for all kind of illnesses requiring a fully competent 19. Hansen G, Hercus TR, Xu Y, et al. Crystallization and pre-
immune response. liminary X-ray diffraction analysis of the ternary human

123
Page 12 of 14 Med Oncol (2013) 30:774

GM-CSF receptor complex. Acta Crystallogr Sect F Struct Biol macrophage colony-stimulating factor in humans. Kidney Int.
Cryst Commun. 2008;64:711–4. 1996;50:2070–8.
20. Choi JK, Kim KH, Park H, et al. Granulocyte macrophage- 38. Hart PH, Whitty GA, Piccoli DS, et al. Synergistic activation of
colony stimulating factor shows anti-apoptotic activity in neural human monocytes by granulocyte-macrophage colony-stimu-
progenitor cells via JAK/STAT5-Bcl-2 pathway. Apoptosis. lating factor and IFN-gamma. Increased TNF-alpha but not IL-1
2011;16:127–34. activity. J Immunol. 1988;141:1516–21.
21. O’Mahony DS, Pham U, Iyer R, et al. Differential constitutive 39. Hazenberg BP, Van Leeuwen MA, Van Rijswijk MH, et al.
and cytokine-modulated expression of human Toll-like receptors Correction of granulocytopenia in Felty’s syndrome by granu-
in primary neutrophils, monocytes, and macrophages. Int J Med locyte-macrophage colony-stimulating factor. Simultaneous
Sci. 2008;5:1–8. induction of interleukin-6 release and flare-up of the arthritis.
22. Tanimoto A, Murata Y, Wang KY, et al. Monocyte chemoat- Blood. 1989;74:2769–70.
tractant protein-1 expression is enhanced by granulocyte-mac- 40. de Vries EG, Willemse PH, Biesma B, et al. Flare-up of rheu-
rophage colony-stimulating factor via Jak2-Stat5 signalling and matoid arthritis during GM-CSF treatment after chemotherapy.
inhibited by atorvastatin in human monocytic U937 cells. J Biol Lancet. 1991;338:517–8.
Chem. 2008;283:4643–51. 41. Bischof RJ, Zafiropoulos D, Hamilton JA, et al. Exacerbation of
23. Liontos LM, Dissanayake D, Ohashi PS, et al. The Src-like acute inflammatory arthritis by the colony-stimulating factors
adaptor protein regulates GM-CSFR signalling and monocytic CSF-1 and granulocyte macrophage (GM)-CSF: evidence of
dendritic cell maturation. J Immunol. 2011;186:1923–33. macrophage infiltration and local proliferation. Clin Exp
24. Domen J, Weissman IL. Hematopoietic stem cells need two Immunol. 2000;119:361–7.
signals to prevent apoptosis; BCL-2 can provide one of these, 42. Campbell IK, Bendele A, Smith DA, et al. Granulocyte-mac-
Kitl/c-Kit signalling the other. J Exp Med. 2000;192:1707–18. rophage colony stimulating factor exacerbates collagen induced
25. Chen J, Cárcamo JM, Golde DW. The alpha subunit of the arthritis in mice. Ann Rheum Dis. 1997;56:364–8.
granulocyte-macrophage colony-stimulating factor receptor 43. Burmester GR, Weinblatt ME, McInnes IB, et al. Efficacy and
interacts with c-Kit and inhibits c-Kit signalling. J Biol Chem. safety of mavrilimumab in subjects with rheumatoid arthritis.
2006;281:22421–6. Ann Rheum Dis. 2013;72:1445–52.
26. Lilly MB, Zemskova M, Frankel AE, et al. Distinct domains of 44. Mudzinski SP, Christian TP, Guo TL, et al. Expression of HLA-
the human granulocyte-macrophage colony-stimulating factor DR (major histocompatibility complex class II) on neutrophils
receptor alpha subunit mediate activation of Jak/Stat signalling from patients treated with granulocyte-macrophage colony-
and differentiation. Blood. 2001;97:1662–70. stimulating factor for mobilization of stem cells. Blood.
27. Crosier KE, Wong GG, Mathey-Prevot B, et al. A functional 1995;86:2452–3.
isoform of the human granulocyte/macrophage colony-stimu- 45. Fanger NA, Liu C, Guyre PM, et al. Activation of human T cells
lating factor receptor has an unusual cytoplasmic domain. Proc by major histocompatibility complex class II expressing neu-
Natl Acad Sci USA. 1991;88:7744–8. trophils: proliferation in the presence of superantigen, but not
28. Raines MA, Liu L, Quan SG, et al. Identification and molecular tetanus toxoid. Blood. 1997;89:4128–35.
cloning of a soluble human granulocyte-macrophage colony-stim- 46. Herold S, Mayer K, Lohmeyer J. Acute lung injury. How
ulating factor receptor. Proc Natl Acad Sci USA. 1991;88:8203–7. macrophages orchestrate resolution of inflammation and tissue
29. LeukineÒ Prescribing information. Available at http://products. repair. Front Immunol. 2001;2:6.
sanofi.us/Leukine/Leukine.html Accessed on September 2013. 47. Cakarova L, Marsh LM, Wilhelm J, et al. Macrophage tumor
30. Jiang D, Schwarz H. Regulation of granulocyte and macrophage necrosis factor-alpha induces epithelial expression of granulo-
populations of murine bone marrow cells by G-CSF and CD137 cyte-macrophage colony-stimulating factor: impact on alveolar
protein. PLoS One. 2010;5:e15565. epithelial repair. Am J Respir Crit Care Med. 2009;180:521–32.
31. Crawford J, Armitage J, Balducci L, et al. Myeloid growth 48. Krausgruber T, Blazek K, Smallie T, et al. IRF5 promotes
factors. J Natl Compr Canc Netw. 2009;7:64–83. inflammatory macrophage polarization and TH1-TH17 respon-
32. Kelsen JR, Rosh J, Heyman M, et al. Phase I trial of sargram- ses. Nat Immunol. 2011;12:231–8.
ostim in pediatric Crohn’s disease. Inflamm Bowel Dis. 49. Miah MA, Yoon CH, Kim J, et al. CISH is induced during DC
2010;16:1203–8. development and regulates DC-mediated CTL activation. Eur J
33. The FDA Safety Information and Adverse Event Reporting Immunol. 2012;42:58–68.
Program, 2008. Leukine safety information Available at http:// 50. Hornell TM, Beresford GW, Bushey A, et al. Regulation of the
www.fda.gov/safety/medwatch/safetyinformation/safetyalerts class II MHC pathway in primary human monocytes by granu-
forhumanmedicalproducts/ucm090918.htm, Accessed on Octo- locyte-macrophage colony-stimulating factor. J Immunol.
ber 2013. 2003;171:2374–83.
34. Martinez-Moczygemba M, Doan ML, Elidemir O, et al. Pul- 51. McCormick S, Santosuosso M, Zhang XZ, et al. Manipulation of
monary alveolar proteinosis caused by deletion of the GM- dendritic cells for host defence against intracellular infections.
CSFR{alpha} gene in the X chromosome pseudoautosomal Biochem Soc Trans. 2006;34:283–6.
region 1. J Exp Med. 2008;205:2711–6. 52. Moldenhauer LM, Keenihan SN, Hayball JD, et al. GM-CSF is
35. Hamilton JA, Whitty GA, Stanton H, et al. Effects of macro- an essential regulator of T cell activation competence in uterine
phage-colony stimulating factor on human monocytes: induction dendritic cells during early pregnancy in mice. J Immunol.
of expression of urokinase-type plasminogen activator, but not 2010;185:7085–96.
of secreted prostaglandin E2, interleukin-6, interleukin-1, or 53. Hesske L, Vincenzetti C, Heikenwalder M, et al. Induction of
tumour necrosis factor-alpha. J Leukoc Biol. 1993;53:707–14. inhibitory central nervous system-derived and stimulatory
36. Takahashi GW, Andrews DF 3rd, Lilly MB, et al. Effect of blood-derived dendritic cells suggests a dual role for granulo-
granulocyte-macrophage colony-stimulating factor and inter- cyte-macrophage colony-stimulating factor in central nervous
leukin-3 on interleukin-8 production by human neutrophils and system inflammation. Brain. 2010;133(Pt 6):1637–54.
monocytes. Blood. 1993;81:357–64. 54. Sonderegger I, Iezzi G, Maier R, et al. GM-CSF mediates
37. Selgas R, Fernández de Castro M, Jiménez C, Selgas R, et al. autoimmunity by enhancing IL-6-dependent Th17 cell devel-
Immunomodulation of peritoneal macrophages by granulocyte- opment and survival. J Exp Med. 2008;205:2281–94.

123
Med Oncol (2013) 30:774 Page 13 of 14

55. Biondo M, Nasa Z, Marshall A, Toh BH, Alderuccio F. Local 74. Brosbøl-Ravnborg A, Hvas CL, Agnholt J, et al. Toll-like
transgenic expression of granulocyte macrophage-colony stim- receptor-induced granulocyte-macrophage colony-stimulating
ulating factor initiates autoimmunity. J Immunol. factor secretion is impaired in Crohn’s disease by nucleotide
2001;166(3):2090–9. oligomerization domain 2-dependent and -independent path-
56. Kim DH, Sandoval D, Reed J, et al. The role of GM-CSF in ways. Clin Exp Immunol. 2009;155:487–95.
adipose tissue inflammation. Am J Physiol Endocrinol Metab. 75. Korzenik JR, Dieckgraefe BK, Valentine JF, et al. Sargramostim
2008;295:E1038–46. for active Crohn’s disease. N Engl J Med. 2005;352:2193–201.
57. Exley MA, Koziel MJ. To be or not to be NKT: natural killer T 76. Valentine JF, Fedorak RN, Feagan B, et al. Steroid-sparing
cells in the liver. Hepatology. 2004;40:1033–40. properties of sargramostim in patients with corticosteroid-
58. Brochériou I, Maouche S, Durand H, et al. Antagonistic regu- dependent Crohn’s disease: a randomised, double-blind, pla-
lation of macrophage phenotype by M-CSF and GM-CSF: cebo-controlled, phase 2 study. Gut. 2009;58:1354–62.
implication in atherosclerosis. Atherosclerosis. 77. Tazawa R, Trapnell BC, Inoue Y, et al. Inhaled granulocyte/
2011;214:316–24. macrophage-colony stimulating factor as therapy for pulmonary
59. Saitoh T, Kishida H, Tsukada Y, et al. Clinical significance of alveolar proteinosis. Am J Respir Crit Care Med. 2010;181:
increased plasma concentration of macrophage colony-stimu- 1345–54.
lating factor in patients with angina pectoris. J Am Coll Cardiol. 78. Kleff V, Sorg UR, Bury C, et al. Gene therapy of beta(c)-defi-
2000;35:655–65. cient pulmonary alveolar proteinosis (beta(c)-PAP): studies in a
60. Oren H, Erbay AR, Balci M, et al. Role of novel biomarkers of murine in vivo model. Mol Ther. 2008;16:757–64.
inflammation in patients with stable coronary heart disease. 79. Higano CS, Schellhammer PF, Small EJ, et al. Integrated data
Angiology. 2007;58:148–55. from 2 randomized, double-blind, placebo-controlled, phase 3
61. Kellar RS, Lancaster JJ, Thai HM, et al. Antibody to granulo- trials of active cellular immunotherapy with sipuleucel-T in
cyte-macrophage colony-stimulating factor reduces the number advanced prostate cancer. Cancer. 2009;115:3670–9.
of activated tissue macrophages and improves left ventricular 80. Olivares J, Kumar P, Yu Y, et al. Phase I trial of TGF-{beta}2
function following myocardial infarction in a rat coronary-artery antisense GM-CSF gene-modified autologous tumor cell (TAG)
ligation model. J Cardiovasc Pharmacol. 2011;57:568–74. vaccine. Clin Cancer Res. 2011;17:183–92.
62. Sugiyama Y, Yagita Y, Oyama N, et al. Granulocyte colony- 81. Bradbury PA, Shepherd FA. Immunotherapy for lung cancer.
stimulating factor enhances arteriogenesis and ameliorates J Thorac Oncol. 2008;3:S164–70.
cerebral damage in a mouse model of ischemic stroke. Stroke. 82. Holt GE, Disis ML. Immune modulation as a therapeutic strat-
2011;42:770–5. egy for non-small-cell lung cancer. Clin Lung Cancer.
63. Tu J, Karasavvas N, Heaney ML, et al. Molecular character- 2009;9:S13–9.
ization of a granulocyte macrophage-colony-stimulating factor 83. Staff C, Mozaffari F, Haller BK, et al. A Phase I safety study of
receptor alpha subunit-associated protein. GRAP Blood. plasmid DNA immunization targeting carcinoembryonic antigen
2000;96:794–9. in colorectal cancer patients. Vaccine. 2011;29:6817–22.
64. Stösser S, Schweizerhof M, Kuner R. Hematopoietic colony- 84. Garcia JA, Mekhail T, Elson P, et al. Phase I/II trial of subcu-
stimulating factors: new players in tumor-nerve interactions. taneous interleukin-2, granulocyte-macrophage colony-stimu-
J Mol Med. 2011;89:321–9. lating factor and interferon-a in patients with metastatic renal
65. Ding DX, Rivas CI, Heaney ML, et al. The alpha subunit of the cell carcinoma. BJU Int. 2012;109:63–9.
human granulocyte-macrophage colony-stimulating factor 85. Lutz E, Yeo CJ, Lillemoe KD, et al. A lethally irradiated allo-
receptor signals for glucose transport via a phosphorylation- geneic granulocyte-macrophage colony stimulating factor-
independent pathway. Proc Natl Acad Sci USA. 1994;91: secreting tumor vaccine for pancreatic adenocarcinoma: a phase
2537–41. II trial of safety, efficacy, and immune activation. Ann Surg.
66. Vadhan-Raj S, Keating M, LeMaistre A, et al. Effects of 2011;253:328–35.
recombinant human granulocyte-macrophage colony-stimulat- 86. Wang L, Qi X, Sun Y, et al. Adenovirus-mediated combined
ing factor in patients with myelodysplastic syndromes. N Engl J P16 gene and GM-CSF gene therapy for the treatment of
Med. 1987;317:1545–52. established tumor and induction of antitumor immunity. Cancer
67. Schweizerhof M, Stösser S, Kurejova M, et al. Hematopoietic Gene Ther. 2002;9:819–24.
colony-stimulating factors mediate tumor-nerve interactions and 87. Dranoff G, Crawford AD, Sadelain M, et al. Involvement of
bone cancer pain. Nat Med. 2009;15:802–7. granulocyte-macrophage colony-stimulating factor in pulmon-
68. Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor ary homeostasis. Science. 1994;264:713–6.
cells in cancer: recent progress and prospects. Immunol Cell 88. Sun X, Hodge LM, Jones HP, et al. Co-expression of granulo-
Biol. 2013;91:493–502. cyte-macrophage colony-stimulating factor with antigen
69. Zhang Y, Cheng S, Zhang M, et al. High-infiltration of tumor- enhances humoral and tumor immunity after DNA vaccination.
associated macrophages predicts unfavorable clinical outcome Vaccine. 2002;20:1466–74.
for node-negative breast cancer. PLoS One. 2013;8:e76147. 89. Kim W, Seong J, Oh HJ, et al. A novel combination treatment of
70. Sica A, Mantovani A. Macrophage plasticity and polarization: armed oncolytic adenovirus expressing IL-12 and GM-CSF with
in vivo veritas. J Clin Invest. 2012;122:787–95. radiotherapy in murine hepatocarcinoma. J Radiat Res.
71. Hao NB, Lu MH, Fan YH, Cao YL, Zhang ZR, et al. Macro- 2011;52:646–54.
phages in tumor microenvironments and the progression of 90. Roilides E, Blake C, Holmes A, et al. Granulocyte-macrophage
tumors. Clin Dev Immunol. 2012;2012:948098. colony-stimulating factor and interferon-gamma prevent dexa-
72. Egea L, Hirata Y, Kagnoff MF. GM-CSF: a role in immune and methasone-induced immunosuppression of antifungal monocyte
inflammatory reactions in the intestine. Expert Rev Gastroen- activity against Aspergillus fumigatus hyphae. J Med Vet Mycol.
terol Hepatol. 2010;4:723–31. 1996;34:63–9.
73. Hirata Y, Egea L, Dann SM, et al. GM-CSF-facilitated dendritic 91. Kowanko IC, Ferrante A, Harvey DP, et al. Granulocyte-mac-
cell recruitment and survival govern the intestinal mucosal rophage colony-stimulating factor augments neutrophil killing
response to a mouse enteric bacterial pathogen. Cell Host of Torulopsis glabrata and stimulates neutrophil respiratory
Microbe. 2010;7:151–63. burst and degranulation. Clin Exp Immunol. 1991;83:225–30.

123
Page 14 of 14 Med Oncol (2013) 30:774

92. Newman SL, Gootee L. Colony-stimulating factors activate the lungs severely affects the ability of mice to control myco-
human macrophages to inhibit intracellular growth of Histo- bacterium tuberculosis infection. J Leukoc Biol. 2005;77:
plasma capsulatum yeasts. Infect Immun. 1992;60:4593–7. 914–22.
93. Roilides E, Mertins S, Eddy J, et al. Impairment of neutrophil 105. Lang RA, Metcalf D, Cuthbertson RA, et al. Transgenic mice
chemotactic and bactericidal function in children infected with expressing a hemopoietic growth factor gene (GM-CSF)
human immunodeficiency virus type 1 and partial reversal after develop accumulations of macrophages, blindness, and a fatal
in vitro exposure to granulocyte-macrophage colony-stimulating syndrome of tissue damage. Cell. 1987;51:675–86.
factor. J Pediatr. 1990;117:531–40. 106. Zhang X, Divangahi M, Ngai P, et al. Intramuscular immuni-
94. Page AV, Liles WC. Colony-stimulating factors in the preven- zation with a monogenic plasmid DNA tuberculosis vaccine:
tion and management of infectious diseases. Infect Dis Clin enhanced immunogenicity by electroporation and co-expression
North Am. 2011;25:803–17. of GM-CSF transgene. Vaccine. 2007;25:1342–52.
95. Steinwede K, Tempelhof O, Bolte K, et al. Local delivery of 107. Ryan AA, Wozniak TM, Shklovskaya E, et al. Improved pro-
GM-CSF protects mice from lethal pneumococcal pneumonia. tection against disseminated tuberculosis by Mycobacterium
J Immunol. 2011;187:5346–56. bovis bacillus Calmette-Guerin secreting murine GM-CSF is
96. Lu H, Xing Z, Brunham RC. GM-CSF transgene-based adjuvant associated with expansion and activation of APCs. J Immunol.
allows the establishment of protective mucosal immunity fol- 2007;179:18–24.
lowing vaccination with inactivated Chlamydia trachomatis. 108. Nambiar JK, Ryan AA, Kong CU, et al. Modulation of pul-
J Immunol. 2002;169:6324–31. monary DC function by vaccine-encoded GM-CSF enhances
97. Bo L, Wang F, Zhu J, et al. Granulocyte-colony stimulating protective immunity against Mycobacterium tuberculosis
factor (G-CSF) and granulocyte-macrophage colony stimulating infection. Eur J Immunol. 2010;40:153–61.
factor (GM-CSF) for sepsis: a meta-analysis. Crit Care. 2011; 109. Dou J, Tang Q, Yu F, et al. Investigation of immunogenic effect
15:R58. of the BCG priming and Ag85A- GM-CSF boosting in Balb/c
98. Denis M, Ghadirian E. Granulocyte-macrophage colony-stimu- mice model. Immunobiology. 2010;215:133–42.
lating factor restricts growth of tubercle bacilli in human mac- 110. Zhang X, Divangahi M, Ngai P, et al. Intramuscular immuni-
rophages. Immunol Lett. 1990;24:203–6. zation with a monogenic plasmid DNA tuberculosis vaccine:
99. Chroneos ZC, Midde K, Sever-Chroneos Z, et al. Pulmonary enhanced immunogenicity by electroporation and co-expression
surfactant and tuberculosis. Tuberculosis. 2009;89:S10–4. of GM-CSF transgene. Vaccine. 2007;25:1342–52.
100. Szeliga J, Daniel DS, Yang CH, et al. Granulocyte-macrophage 111. Wang J, Zganiacz A, Xing Z. Enhanced immunogenicity of
colony stimulating factor-mediated innate responses in tuber- BCG vaccine by using a viral-based GM-CSF transgene adju-
culosis. Tuberculosis. 2008;88:7–20. vant formulation. Vaccine. 2002;20:2887–98.
101. Francisco-Cruz A, Mata-Espinosa D, Estrada-Parra S, Xing Z, 112. Wang H, Zhang G, Wen Y, et al. Intracerebral administration of
Hernández-Pando R. Immunotherapeutic effects of recombinant recombinant rabies virus expressing GM-CSF prevents the
adenovirus encoding granulocyte-macrophage colony-stimulat- development of rabies after infection with street virus. PLoS
ing factor in experimental pulmonary tuberculosis. Clin Exp One. 2011;6:e25414.
Immunol. 2013;171:283–97. 113. Li N, Yu YZ, Yu WY, et al. Enhancement of the immunogenicity
102. Marlow N, Morris T, Brocklehurst P, et al. A randomised trial of of DNA replicon vaccine of Clostridium botulinum neurotoxin
granulocyte-macrophage colony-stimulating factor for neonatal serotype A by GM-CSF gene adjuvant. Immunopharmacol Im-
sepsis: outcomes at 2 years. Arch Dis Child Fetal Neonatal Ed. munotoxicol. 2011;33:211–9.
2013;98(1):F46–53. 114. Grabstein KH, Urdal DL, Tushinski RJ, et al. Induction of
103. Mera S, Tatulescu D, Cismaru C, et al. Multiplex cytokine macrophage tumoricidal activity by granulocyte-macrophage
profiling in patients with sepsis. APMIS. 2011;119:155–63. colony-stimulating factor. Science. 1986;232:506–8.
104. Gonzalez-Juarrero M, Hattle JM, Izzo A, et al. Disruption of 115. Kaushansky K. Lineage-specific hematopoietic growth factors.
granulocyte macrophage-colony stimulating factor production in N Engl J Med. 2006;354:2034–45.

123

View publication stats

You might also like