granulocyteâmacrophage colony-stimulating factor: not just another haematopoietic growth factor

14
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 factor (GM-CSF) is often used to treat leucopenia. Other haematopoietins may increase the number of circulating leucocytes with higher efficiency, but GM-CSF has addi- tional effects that may be far more relevant than its hae- matopoietic activity. GM-CSF induces differentiation, proliferation and activation of macrophages and dendritic cells which are necessary for the subsequent T helper cell type 1 and cytotoxic T lymphocyte activation. GM-CSF haematopoietic and non-haematopoietic functions have pro-inflammatory and immune regulatory potential to treat a variety of autoimmune diseases and tumours. On the other hand, GM-CSF deficiency leads to various immune dysfunctions and the current utilization of GM-CSF as haematopoietic factor might be an accurate but very incomplete indication for a cytokine with vast clinical potential. Keywords Haematopoietic growth factor Á Granulocyte–macrophage colony-stimulating factor Á Innate immunity Á Adaptive immunity Abbreviations AML Acute myeloid leukaemia AMØ Alveolar macrophage BM Bone marrow CISH Cytokine inducible SH2-domain DC Dendritic cell GRAP GM-CSFRa subunit-associated protein iNKT Invariant natural killer T IjK IjB kinase Macrophage MAPK Mitogen-activated kinase-like protein NFjB Nuclear factor kappa-light-chain-enhancer of activated B cells IkB Nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor b PI3K Phosphatidylinositol 3 kinase PKC Protein kinase C PAP Pulmonary alveolar proteinosis rhGM-CSF Recombinant human GM-CSF rhG-CSF Recombinant human granulocyte colony- stimulating factor STAT Signal transducers and activators of transcription SLAP SRC-like adapter protein TLR Toll-like receptor Introduction Colony-stimulating factors (CSF) are crucial for survival, proliferation, differentiation, maturation and functional activation of haematopoietic cells [1]. There are three types of CSF, namely macrophage colony-stimulating factor (M- CSF) or CSF1, granulocyte–macrophage colony-stimulating Electronic supplementary material The online version of this article (doi:10.1007/s12032-013-0774-6) contains supplementary material, which is available to authorized users. A. Francisco-Cruz Á O. Ramos-Espinosa Á D. Mata-Espinosa Á B. Marquina-Castillo Á J. Barrios-Payan Á R. Hernandez-Pando (&) Department of Pathology, National Institute of Medical Sciences and Nutrition ‘‘Salvador Zubiran’’, Vasco de Quiroga 15, Tlalpan, 14000 Me ´xico City, Mexico e-mail: [email protected] A. Francisco-Cruz Á M. Aguilar-Santelises Department of Immunology, National School of Biological Sciences, National Polytechnic Institute, Me ´xico City, Mexico 123 Med Oncol (2013) 30:774 DOI 10.1007/s12032-013-0774-6

Upload: rogelio

Post on 21-Dec-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

REVIEW ARTICLE

Granulocyte–macrophage colony-stimulating factor: not justanother 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

� Springer Science+Business Media New York 2013

Abstract Granulocyte–macrophage colony-stimulating

factor (GM-CSF) is often used to treat leucopenia. Other

haematopoietins may increase the number of circulating

leucocytes with higher efficiency, but GM-CSF has addi-

tional effects that may be far more relevant than its hae-

matopoietic activity. GM-CSF induces differentiation,

proliferation and activation of macrophages and dendritic

cells which are necessary for the subsequent T helper cell

type 1 and cytotoxic T lymphocyte activation. GM-CSF

haematopoietic and non-haematopoietic functions have

pro-inflammatory and immune regulatory potential to treat

a variety of autoimmune diseases and tumours. On the

other hand, GM-CSF deficiency leads to various immune

dysfunctions and the current utilization of GM-CSF as

haematopoietic factor might be an accurate but very

incomplete indication for a cytokine with vast clinical

potential.

Keywords Haematopoietic growth factor �Granulocyte–macrophage colony-stimulating factor �Innate immunity � Adaptive immunity

Abbreviations

AML Acute myeloid leukaemia

AMØ Alveolar macrophage

BM Bone marrow

CISH Cytokine inducible SH2-domain

DC Dendritic cell

GRAP GM-CSFRa subunit-associated protein

iNKT Invariant natural killer T

IjK IjB kinase

MØ Macrophage

MAPK Mitogen-activated kinase-like protein

NFjB Nuclear factor kappa-light-chain-enhancer of

activated B cells

IkB Nuclear factor of kappa light polypeptide

gene enhancer in B cells inhibitor bPI3K Phosphatidylinositol 3 kinase

PKC Protein kinase C

PAP Pulmonary alveolar proteinosis

rhGM-CSF Recombinant human GM-CSF

rhG-CSF Recombinant human granulocyte colony-

stimulating factor

STAT Signal transducers and activators of

transcription

SLAP SRC-like adapter protein

TLR Toll-like receptor

Introduction

Colony-stimulating factors (CSF) are crucial for survival,

proliferation, differentiation, maturation and functional

activation of haematopoietic cells [1]. There are three types

of CSF, namely macrophage colony-stimulating factor (M-

CSF) or CSF1, granulocyte–macrophage colony-stimulating

Electronic supplementary material The online version of thisarticle (doi:10.1007/s12032-013-0774-6) contains supplementarymaterial, which is available to authorized users.

A. Francisco-Cruz � O. Ramos-Espinosa � D. Mata-Espinosa �B. Marquina-Castillo � J. Barrios-Payan �R. Hernandez-Pando (&)

Department of Pathology, National Institute of Medical Sciences

and Nutrition ‘‘Salvador Zubiran’’, Vasco de Quiroga 15,

Tlalpan, 14000 Mexico City, Mexico

e-mail: [email protected]

A. Francisco-Cruz � M. Aguilar-Santelises

Department of Immunology, National School of Biological

Sciences, National Polytechnic Institute, Mexico City, Mexico

123

Med Oncol (2013) 30:774

DOI 10.1007/s12032-013-0774-6

factor (GM-CSF) or CSF2 and granulocyte colony-stimu-

lating factor (G-CSF) or CSF3 (Fig. 1). CSF were uninten-

tionally discovered when solid-state culture systems allowed

specific precursors of granulocytes and macrophages to

proliferate and form colonies of maturing but non-autono-

mous proliferating cells [1, 2]. The newly found cell product

that needed to be added to stimulate colony formation

received the operational name ‘‘colony-stimulating factor’’

[1, 2].

As part of the haematopoietin family, GM-CSF is a

four-helix packing cytokine (Fig. 2a). The GM-CSF gene

is located in the 5q31 region. Deletion of 5q is associated

with acute myeloid leukaemia (AML), but AML is not

always generated by deletion of 5q. The same chromo-

somal location is associated with other genes encoding

cytokines such as IL-4, IL-5 and IL-13 [2–4]. The stem cell

factor (SCF), leukaemia inhibitory factor, Flt3 ligand

(Flt3L), erythropoietin (EPO), thrombopoietin (TPO) and

IL-3, IL-5, IL-11 also have haematopoietic activity [1].

GM-CSF was first described for its ability to induce

mouse myelopoiesis, in addition to its capacity to stimulate

granulocyte, macrophage, eosinophil, megakaryocyte and

erythroid colony formation. However, there was not a clear

functional distinction between CSF and IL-3 until the

human GM-CSF was sequenced in 1985. The murine GM-

CSF was sequenced in 1994. Cloning techniques also

revealed in 1986 that the hypothetical human ‘‘pluripoie-

tin’’ was actually G-CSF which has since then been used as

the most important haematopoietic factor in humans [2–5].

Deletion of genes encoding for EPO, G-CSF, TPO or

M-CSF produces dramatic reductions in the amount of

cells that are normally stimulated by each one of these

cytokines. However, deletion of the GM-CSF gene only

reduces neutrophils function without significantly affecting

Multipotent stem cell

Primitive progenitor cells

Committed precursor cells

Linage commited cells

Neutrophils, eosinophils, basophilsMonocytes

Self-renewal

SCF TPO

GM-CSF

HSC

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

CMP

GM

MP GP

Fig. 1 GM–CSF relationship

with haematopoiesis. Formation

of blood cells progress from a

haematopoietic stem cell, which

can undergo either self-renewal

or differentiation into a

multilineage committed

progenitor cell named common

myeloid progenitor. This cell

gives rise to a more

differentiated progenitor by

stimulation with GM–CSF

committed to the granulocytes

and macrophages (GM) linage.

Finally, these cells give rise to

uni-lineage committed

progenitors for granulocytes or

monocytes. SCF stem cell

factor, TPO thrombopoietin

[115]

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

123

their number in circulation [1]. Recombinant human GM-

CSF (rhGM-CSF) has been available in USA for thera-

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].

Clinical trials revealed its pro-inflammatory effect and, in

consequence, its potential usefulness as a prophylactic or

adjuvant therapeutic agent for patients at high risk of

infection [7]. In 1996, the Food and Drug Administration

approved GM-CSF for treatment for leucopenia associated

with fungal infections [7]. GM-CSF applications will be

most probably further extended due to its ability to induce

inflammatory responses of differentiated eosinophils,

neutrophils, macrophages (MØ) and dendritic cells (DC)

[1, 5, 6].

GM-CSF expression and target cells

GM-CSF can be detected under physiological conditions at

serum concentrations ranging from 20 to 100 pg/ml [8].

Higher serum levels of GM-CSF require stimulation with

cytokines, antigens, microbial products or inflammatory

agents such as IL-1, TNF-a or lipopolysaccharide (LPS) [8].

In fact, GM-CSF was first purified from the conditioned

medium of lung tissue from mice after LPS administration

Fig. 2 Structure, cellular sources and cellular targets of GM–CSF.

a Type I cytokine or alpha short chain with four packing helix, native

GM–CSF is a monomeric glycosylated protein of 127 aa. rhGM-CSF

is associated with dimeric complexes. Structure obtained from Protein

Data Bank PDB: 2GMF. b GM-CSF production usually requires

stimulation with cytokines, antigens, microbial products or inflam-

matory agents (IL-1, TNF-a or LPS). Both in steady state and induced

conditions GM-CSF may be produced by CD4? T cells (TH1, TH2

and TH17), B lymphocytes, macrophages, keratinocytes, eosinophils,

neutrophils, endothelial cells, CD56hiCD16- NK cells, bone-forming

osteoblasts, mast cells, multipotent mesenchymal stromal cells and

Paneth’s cell. Upon appropriate stimulation, much of the production

and action of GM-CSF occurs locally at the site of inflammation.

c The major target cells and effects of GM-CSF are maturation in DC,

activation, survival and differentiation in granulocytes, steady-state

differentiation in alveolar MØ (AMØ) and invariant natural killer T

(iNKT) cells and proliferation in microglia and pneumocytes type I

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

123

[8]. GM-CSF is produced by a variety of cells, including

stromal cells, Paneth’s cells, MØ, DC, mast cells, endothelial

cells, smooth muscle cells, fibroblasts, chondrocytes, as well

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 has a particularly important role in the crosstalk

between antigen-presenting cells and T cells [8, 9]. Although

the major role for GM-CSF seems to induce DC maturation,

GM-CSF also induces granulocytes activation, microglia

proliferation, steady-state differentiation in invariant natural

killer T (iNKT) cells and survival and differentiation of

alveolar MØ (AMØ; Fig. 2c). Moreover, lymphocytes and

endothelial cells have GM-CSF receptors (GM-CSFR)

which makes them susceptible to GM-CSF stimulation, but

the consequences of such stimulation are not clear [8–11].

GM-CSF knockout (KO) mice do not suffer abnormal-

ities in myelopoiesis, but they do have the characteristic

abnormalities of pulmonary alveolar proteinosis (PAP),

indicating that GM-CSF might have a significant role in

maintenance of the normal lung physiology [5, 10]. GM-

CSF deficient mice also have a deficient iNKT cells dif-

ferentiation during thymic ontogeny since GM-CSF acts at

the early thymic iNKT cell precursor stage, after lineage

commitment and positive selection by regulating the

acquisition of secretory function for immune synapse-

directed release of immunoregulatory cytokines [5, 10].

GM-CSF receptor

The GM-CSFR is a heterodimer formed by a ligand-spe-

cific a subunit (GM-CSFRa or CD116; 60–80 kDa) and a

b subunit (GM-CSFRbc or CD131; 120–140 kDa) shared

with IL-3 and IL-5 receptors [12]. Both subunits are type I

transmembrane glycoprotein structurally characterized by

the presence of cytokine-receptor homology modules,

consisting of two domains of fibronectin type III [13]. GM-

CSFRa binds its ligand with low affinity (KD =

0.2–100 nM), but the expression of GM-CSFRbc increases

GM-CSFRa affinity to KD = 100 pM [13]. Eight confor-

mational variants have been described for GM-CSFRa, but

only two are biologically relevant for its transductional

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

the fact that the complete deletion of its cytoplasmic region

results in lack of cell growth and differentiation. GM-

CSFRbc is constitutively expressed on the cell surface

[14]. Similarly to IL-3R and IL-5R, GM-CSFR is expres-

sed at very low levels on the surface of haematopoietic

cells, with only 100–1,000 molecules per cell [13]. GM-

CSFR is also expressed on granulocytes and macrophages

progenitor cells, mature cells such as monocytes, MØ, DC,

megakaryocytes, neutrophils, plasma cells, T lymphocytes,

vascular endothelial cells, uterine cells, gastrointestinal

tract epithelial cells, astrocytes, oligodendrocytes and

microglia cells [9, 15, 16].

GM-CSF/GM-CSFR interactions

GM-CSF is able to bind GM-CSFRbc in the absence of

GM-CSFRa, but the heterodimerization with both subunits

is required for intracellular signal transduction. GM-CSFR

activation follows general rules regarding receptor dimer-

ization and transphosphorylation of tyrosine residues in the

receptor cytoplasmic domains. GM-CSFR does not have

intrinsic tyrosine kinase activity. Instead, it requires the

association of GM-CSFRbc with JAK-2 for GM-CSFRbc

transphosphorylation, involving two bc chains closely sit-

uated in the cytoplasmic region [17]. Crystallographic

studies have shown that GM-CSFRbc is in fact homodi-

meric, but its cytoplasmic regions are quite separated

(120 A), which makes transphosphorylation difficult to

occur [17–19].

A unique tertiary structure of the GM-CSF/GM-CSFR

complex has been described. It corresponds to a coordi-

nated dodecamer structure, which is necessary for receptor

activation (Fig. 3a). The association between GM-CSF and

GM-CSFR involves three sites of interaction. The first

interaction is between GM-CSF and GM-CSFRa, the sec-

ond is between GM-CSF and two domains of two different

GM-CSFRbc molecules and the third one is a stabilizing

site formed between GM-CSFRa and GM-CSFRbc. These

three combinations determine the formation of a higher-

order dodecamer complex composed by two hexameric

complexes linked by a fourth site of interaction. Antibodies

and mutations directed to the fourth site of interaction

significantly decrease GM-CSF transduction and initiate

the loss of the dodecamer complex [13]. These interactions,

which are only observed in GM-CSFR, explain the trans-

phosphorylation by GM-CSF/GM-CSFR in which JAK-2 is

associated with the beta chain. The dodecamer complex

product relocates closer two GM-CSFRbc, at a distance of

10 A permitting transphosphorylation and the activation of

subsequent signalling pathways [13, 19].

GM-CSF signalling

GM-CSFR transduction is similar to the activation carried

on by the interferon production regulator family. Binding

of GM-CSF with its receptor leads to the activation of

kinases of the JAK family which then phosphorylate signal

transducers and activators of transcription (STAT) that

migrate to the nucleus and bind specific DNA elements

directing the transcription of specific genes related to cell

differentiation. GM-CSF induces cell proliferation mainly

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

123

by activation and signalling of protein kinase C, preventing

cell death by phosphatidylinositol 3 kinase and JAK/

STAT5-Bcl-2 signalling. GM-CSF induces proliferating

and inflammatory responses by mitogen-activated protein

kinases (MAPK) and NFjB activation (Fig. 3B) [14, 20].

GM-CSF increases AMØ and neutrophils function.

Proliferating AMØ increase their own expression of toll-

like receptor 4 (TLR4) and nuclear factor kappa-light-

chain-enhancer of activated B cells (NFjB) by direct GM-

CSFR and IjB kinase (IjK) interaction. GM-CSF regulates

the expression of TLR2 and TLR4 in neutrophils and TLR2

in monocytes. Additionally, GM-CSF induces the release

of IL-12 and TNF-a and enhances the expression of

monocyte chemoattractant protein (MCP)-1 by JAK2-

STAT5 signalling in monocytes [21, 22]. Recently, a spe-

cific signalling pathway has been described for inducing

classic MØ phenotype or M1 and inflammatory DC, which

is described below. SRC-like adapter protein (SLAP)

functions as a negative regulator for DC maturation

induced by GM-CSF. The need for SLAP action suggests a

specific threshold for correct stimulation by GM-CSF for

monocytic DC maturation [23].

GM-CSF binding to GM-CSFRbc triggers most of the

intracellular signals that induce inflammation, cell survival,

differentiation and proliferation in monocytes and granu-

locytes. c-Kit and GM-CSFRa are well-known markers of

haematopoietic cells differentiation. Haematopoietic stem

cells down-regulate c-Kit and up-regulate GM-CSFRawhen they enter to the granulocyte/macrophage lineage

[24, 25]. GM-CSFRa has two isoforms. Stimulation of the

GM-CSFRa-1 isoform induces overexpression of CD86

and F4/80 cell surface proteins, and it may also induce

intense phosphorylation of JAK-2 [26]. GM-CSFRa-2

isoform has a different cytoplasmic domain, but it associ-

ates with GM-CSFRbc in the same manner than GM-

CSFRa-1 does it [27]. There also exist soluble isoforms of

both a-subunits derived from alternative mRNA splicing,

but their functions are not known [27, 28].

GM-CSF clinical applications as haematopoietin

GM-CSF is the growth factor most extensively used as

hemopoietin in the clinical practice. rhGM-CSF is pro-

duced in S. cerevisiae, and it is named sargramostim or

Leukine� (Bayer HealthCare Pharmaceuticals, USA). Its

amino acid sequence is homologous to the endogenous

human GM-CSF, except for a leucine instead of a proline

at the 23rd position and different glycosylated motives. The

degree of glycosylation affects the biological activity,

antigenicity, toxicity and pharmacokinetics. When rhGM-

CSF is produced in E. coli is named molgramostim

(Zenotech Laboratories, India) and when it is produced in

Chinese hamster ovary cells is named regramostim (Lan-

ospharma Laboratories, China) [7]. Sargramostim may help

to prevent infections by enhancing the immune cell func-

tion. It is the only growth factor approved in USA for

treatment of older adults with AML, after induction che-

motherapy, to shorten the time to neutrophil recovery and

to reduce the incidence of life-threatening infections [7,

29]. Sargramostim is also approved in USA for myeloid

reconstitution following allogeneic bone marrow trans-

plantation (BMT), autologous BMT or peripheral blood

stem cell transplantation. Furthermore, sargramostim is

also approved for peripheral blood stem cell mobilization,

BMT failure and engraftment delay [29].

Pharmacokinetics and pharmacodynamics of rhGM-

CSF

When sargramostim is administered to the patients by

intravenous infusion during 2 h, it reaches a mean beta

half-life of approximately 60 min. The peak concentra-

tions of GM-CSF are observed in blood samples

obtained during or immediately after completion of the

sargramostim infusion, while minor concentrations are

still detected in blood 6 h after the beginning of the

infusion [7, 29]. GM-CSF is also detected in serum first

after 15 min of subcutaneous injection of sargramostim

to healthy volunteers. Then, the mean beta half-life is

approximately 162 min, and peak levels are reached after

one to 3 h post-injection and remain detectable for up to

6 h. Parenteral administration of rhGM-CSF alters the

kinetics of myeloid progenitor cells in bone marrow

accelerating the cell-cycle entry and reducing the cell-

cycle time. These effects are reversible once adminis-

tration is discontinued. However, the numbers of mobi-

lized progenitor cells that are released in peripheral

blood by GM-CSF are 10 times lower than those

released by G-CSF [6, 30].

rhGM-CSF and rhG-CSF in leucopenia

rhGM-CSF produces small increases in the number of

peripheral neutrophils and circulating monocytes. It is

currently indicated as second-line treatment for patients

with severe neutropenia [7, 31]. rhGM-CSF also induces a

mild increase in the number of circulating eosinophils and

basophils and enhances the phagocytic function of neu-

trophils. Filgrastim (rhG-CSF) and pegfilgrastim (pegylat-

ed rhG-CSF) are currently approved by FDA for prevention

of chemotherapy-induced neutropenia [1, 7].

The therapeutic application of rhGM-CSF for leuco-

penia could be overestimated. G-CSF recommendations

for neutropenia are supported by various results from

clinical trials and head-to-head comparative studies about

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

123

clinical benefits of G-CSF compared with GM-CSF are

lacking. Moreover, G-CSF has higher myeloproliferative

activity than GM-CSF [30]. CD34? mononuclear cells

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-

tively). These effects are not synergistic when both CSF

are combined [6, 7, 30]. Besides, there are ample differ-

ences in therapeutic doses between G-CSF (5 lg/kg) and

GM-CSF (250 lg/m2), which are considered in the clin-

ical guidelines from the National Comprehensive Cancer

Network (NCCN) and the American Society of Clinical

Oncology (ASCO) recommending rhG-CSF as first-line

therapy for febrile neutropenia [31].

Toxicological profile of rhGM-CSF

Sargramostim is usually well tolerated by healthy subjects,

who have not shown clinical alterations in their clinical

analysis, as compared to placebo-treated individuals [29].

NCCN guidelines recommend sargramostim administration

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

123

to 55 years old and older AML patients 24 h after induc-

tion chemotherapy [31]. Although sargramostim adminis-

tration increases cellularity in haematological, heart and

lung tissue on monkeys, these effects have not been serious

enough to fulfil toxicity standard criteria [29]. However,

sargramostim-treated patients with febrile neutropenia

secondary to chemotherapy, risk to develop fluid retention,

respiratory symptoms due to sequestration of granulocytes

in pulmonary circulation, cardiovascular symptoms, renal

and hepatic dysfunction, as well as adverse reactions

related to bone marrow transplant or peripheral blood

progenitor cell transplant [31]. Paediatric patients suffering

Crohn’s disease have been treated with subcutaneous doses

of 4 or 6 lg/kg sargramostim every day during 8 weeks,

and 90 % of them only had mild injection site reactions,

whereas the resting 10 % required dose reductions due to

elevated absolute neutrophil counts [32]. Syncope is the

worst side effect triggered thus far by sargramostim

administration to human subjects. Since it was attributed to

EDTA that was included in the liquid form, the FDA

prohibited the preparation of Leukine� in its liquid for-

mulation [33].

Non-haematopoietic functions of GM-CSF

GM-CSF has significant non-haematopoietic effects in

various physiological processes other than haematopoiesis.

GM-CSF KO mice do not suffer alterations in the steady

state of haematopoiesis. Instead, GM-CSF KO animals

acquire a particular lung injury phenotype characterized by

accumulation of lipids and surfactant proteins in alveolar

spaces, similar to the human condition described as PAP

[3, 4, 34].

Inflammation

The contribution of GM-CSF to the inflammatory response

has been evaluated in vitro. rhGM-CSF induces longer

survival in monocytes, MØ and neutrophils, augments the

amount of inflammatory mediators released by these cells

and contributes to the elimination of pathogens and

tumours [35, 36]. MØ are more efficiently stimulated by

secondary stimuli such as LPS and IFN-c, when they are

initially stimulated with GM-CSF. Intra-peritoneal admin-

istration of GM-CSF induces a strong recruitment of

human MØ, and both human and murine monocytes show

higher pro-inflammatory response when they are primary

stimulated with GM-CSF and then restimulated with LPS

[37, 38].

One of the most interesting features of GM-CSF was

observed in patients with Felty’s syndrome (rheumatoid

arthritis, splenomegaly and neutropenia) that received

rhGM-CSF and experienced worsening of arthritis [39].

Additionally, patients with rheumatoid arthritis (RA) also

experienced symptoms exacerbation when treated with

rhGM-CSF after chemotherapy [40]. Moreover, the exac-

erbation of RA symptoms may be reproduced with GM-

CSF administration in experimental models [41, 42]. On

the other hand, administration of mavrilimumab, a human

monoclonal antibody targeting GM-CSFRa, induces rapid

and significant responses in RA subjects [43].

Activation and regulation of the immune system

GM-CSF stimulates the expression of adhesion molecules,

IgGFcR and activated complemented receptors on neutro-

phils, enhancing their response to chemotactic factors,

phagocytosis, synthesis of leukotriene B4, arachidonic acid

release and superoxide anion generation. Moreover, GM-

CSF increases survival of neutrophils and induces its

MHC-II expression enabling them to activate T cells

response to superantigens [44, 45].

AMØ orchestrate structural and functional parenchyma

repair processes that are essential for homoeostasis [46].

Different extracellular signals are integrated to shape up

pulmonary MØ phenotypes, during lung inflammation

Fig. 3 Dodecameric complex of GM-CSFR activation and signalling

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-

CSFRa. Interaction with free dimeric forms of GM-CSFRbc chains

makes possible the formation of a hexameric complex formed by two

GM-CSFRbc chains. Each one of these two chains is joined to one

GM-CSFRa chain and carries a GM-CSF molecule (2:2:2). The

dodecameric complex, the highest order complex, is formed by the

lateral aggregation of two hexameric complexes thanks to a fourth site

of interaction that forms a signalling competent structure, diminishing

the distance between bc chains and allowing the transphosphorylation

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

recruited for inducing activation of nuclear factors that up-regulate the

expression of specific genes related to inflammation, survival,

differentiation and cell proliferation. One of the GM-CSFRbc trans-

phosphorylated domains induces the activation of Ras and mitogen-

activated protein kinases (MAPK) with the consequent induction of

c-fos and c-jun and PI3 K/Akt/p21waf-1. The other phosphorylated

domain mediates the activation of Janus kinase 2 (JAK2)/signal

transducer and activator of transcription 5 (STAT5) signalling

pathway. Activated STAT5 migrates into the nucleus after mutual

phosphorylation and dimerization and binds to specific DNA-binding

sites, followed by the expression of STAT5 target genes. Jak kinase–

STAT5 transcription factor pathway induces the expression of

interferon-regulating factor 5 (IRF5). High expression of IRF5 results

in classical MØ activation (M1) or inflammatory MØ induction. GM–

CSF supports differentiation, survival and proliferation in part through

phosphorylated STAT5 (pSTAT5) signalling. In parallel, pSTAT

activity leads to an accumulation of cytoplasmic cytokine inducible

SH2-domain protein (CISH). CISH induces feedback inhibition of

STAT5 activation, triggering the differentiation of type 1 polarized

DC. GM-CSFRa subunit-associated protein (GRAP), a protein that

binds to the cytoplasmic region of the alpha chain, is highly expressed

in neoplastic cells

b

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

123

TNF-a released from activated AMØ induces epithelial

GM-CSF expression, which initiates alveolar epithelial cell

proliferation and helps to restore the alveolar barrier

function [47]. Pneumocytes type II express GM-CSFRaand GM-CSFRbc. Proliferating type II pneumocytes may

then down-regulate GM-CSFR expression during its trans-

differentiation into type I pneumocytes occurring in

response to acute lesion [46, 47].

GM-CSF regulates the inflammatory response by acti-

vation of the JAK kinase–STAT5 transcription factor

pathway, inducing the expression of the interferon-regu-

lating factor 5 (IRF5). A high IRF5 expression by GM-CSF

or IFN-c results in M1 MØ activation. IRF5 acts together

with RelA (NFjB protein) inducing TNF-a gene expres-

sion and IRF5 inhibits the transcription of IL-10 and pro-

motes IFN-c expression [48]. In contrast, when M-CSF

predominates, IRF4 is overexpressed inducing a M2 MØ

phenotype. Several cell types, including fibroblasts, endo-

thelial cells, stromal cells and osteoblasts, constitutively

produce M-CSF. There is a steady-state production of

M-CSF that turns MØ towards the M2 phenotype trough

IRF4, probably balancing the pro-inflammatory microen-

vironment achieved by the expression of IL-12, IL-6 and

IL-23 that are produced by GM-CSF-stimulated MØ [46,

48].

GM-CSF has an important role in the production of BM-

derived type 1 DC. BM-derived DC acquires potent anti-

gen-presenting capacity, representing a crucial reservoir of

professional antigen-presenting cells (APC). In type 1 DC

differentiation, the expression and activation of STAT5 is

gradually enhanced by GM-CSF during the early stage of

DC development. In parallel, pSTAT activity leads to the

accumulation of cytoplasmic cytokine inducible SH2-

domain (CISH) protein, at the later stage [49]. When a

threshold concentration of CISH is reached, CISH inhibits

the GM-CSF-mediated STAT5 phosphorylation, triggering

its differentiation to strong type 1 polarized DC, promoting

MHC class I expression. Thus, CISH can act as a molecular

switch from the DC progenitor stage to immature DC and a

control point for cytotoxic T lymphocytes activation [49].

GM-CSF is a DC-activating cytokine inducing a strong

differentiation of TH1 type cells. GM-CSF enhances the

oxidative metabolism, cytotoxicity and antibody-dependent

phagocytosis [7, 9]. Human monocytes and monocyte-

derived DC treated with GM-CSF exhibited higher

expression of MHC-II molecules, CD80, CD86 and CD40,

increasing the immune response against antimicrobial

agents [50, 51]. However, the endometrium originated

GM-CSF induces tolerance during early pregnancy [52],

GM-CSF directs DC from the central nervous system

(CNS) towards an inhibitory phenotype. In contrast, DC

recruited by GM-CSF from the peripheral circulation into

the CNS tissue has a pro-inflammatory phenotype, which

has been related to the pathogenesis of autoimmune

encephalomyelitis [53].

GM–CSF relationships with disease

The ability of GM-CSF to regulate the immune system has

been related to Felty’s syndrome, RA and various other

autoimmune, cardiovascular and metabolic diseases [43,

54]. Local over expression of GM-CSF in the stomach of

mice leads to autoimmune gastritis [55]. On the other hand,

GM-CSF deficient mice are less prone to develop experi-

mental allergic encephalitis, myocarditis and collagen-

induced arthritis [5, 8, 10]. GM-CSF promotes autoim-

munity by enhancing IL-6-dependent survival of antigen-

specific T CD4? cells. It has been described as the en-

cephalitogenic factor produced by IL-23-driven pathogenic

TH cells. GM-CSF is required for the response to IL-6 and

IL-23 by DC and subsequent TH17 cell generation [54].

Moreover, GM-CSF promotes autoimmunity by enhancing

IL-6-dependent survival of antigen-specific T CD4? cells

[54, 55].

GM-CSF is expressed by adipose tissue, but its role in

there remains unclear. GM-CSF KO mice are obese,

hyperphagic and show larger mesenteric fat adipocytes,

decreased number of MØ, lower transcription of pro-

inflammatory cytokines and enhanced peripheral uptake of

glucose when receive a high-fat diet [56]. iNKT cells

constitute up to 50 % of liver lymphocytes, and the relation

between GM-CSF, liver iNKT cells and metabolism has

not yet been investigated [57]. On the other hand, M-CSF

production in CD68? MØ found in human fatty streaks and

atherosclerotic plaques suggests that the ratio between

M-CSF and GM-CSF is important to determine MØ het-

erogeneity in atherosclerotic lesions [58]. The antagonism

between M-CSF and GM-CSF is also important to define

the functional polarization of M2 and M1 MØ during the

course of diverse inflammatory and metabolic disorders

[59, 60]. MØ migration to areas of myocardial infarction is

associated with tissue damage, whereas the treatment with

anti-GM-CSF antibodies decreases the amount of damaged

tissue after infarction [61]. A model of cerebral ischaemia

also shows that GM-CSF promotes collateral arteries

growth and reduces cerebral ischaemia [62].

GM–CSF relationship with tumour cells

The presence of GM-CSFR on tumour cells makes them

susceptible to GM-CSF stimulation. GM-CSFRa binding

may be particularly important for signal transduction in

tumour cells overexpressing the GM-CSFRa subunit-

associated protein [63]. The interaction between tumour

cells and sensory neurons seems to be part of the patho-

physiology of cancer-induced pain [64, 65]. The intense

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

123

bone pain occurring as side effect in rhGM-CSF-treated

patients that suffer myelodysplastic syndromes may be

caused by a two-way interaction between GM-CSFRabearing tumour cells and neurons [66]. GM-CSFRa is also

expressed on pancreatic nerves from healthy subjects and

in large hypertrophic nerves close to tumours in pancreatic

carcinoma patients [67]. GM-CSFR on nerve fibres may

contribute to tumour-induced pain through the JAK-

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-

erogeneous population of immature granulocytic and

monocytic cells, not expressing cell surface markers

associated with differentiated monocytes, MØ or DC.

MDSC exert an immunosuppressive effect of the immune

response against cancer [68]. Although the suppression

mechanism is not clear, it has been strongly associated with

the local presence of MDSC and M2 MØ in mice and

humans [68, 69]. In humans, MØ can be differentiated

either towards pro-inflammatory M1 MØ or anti-inflam-

matory M2 MØ. M1 MØ express IL-12high, IL- 23high, IL-

10low and have high bactericidal and tumoricidal capacity,

whereas M2 MØ expressing IL-12low, IL-23low, IL-10high

are involved with promotion of tissue remodelling, tumour

growth and immunoregulation [70]. The possibility exists

that tumour cells may be favoured by GM-CSF since they

can be stimulated to in vitro proliferation. However,

tumour cells proliferation may not be the predominant

result, due to the presence of GM-CSF and interaction with

other GM-CSF responder cells. If tumour growth promo-

tion occurs, it may be rather due to M-CSF stimulation,

leading to activation of M2 MØ and other tumour-associ-

ated macrophages that produce anti-inflammatory cyto-

kines which are linked to in vivo growth, migration and

spreading of a variety of cancers [48, 71].

Therapeutic applications of GM-CSF

GM-CSF KO mice are more susceptible than wild-type

mice to several infections and intestinal inflammation [72].

Whereas GM-CSF production is necessary to maintain a

fully functional intestinal innate immunity and to facilitate

recruitment and survival of intestinal DC, lack of GM-CSF

may be related to an immune deficiency associated with

Crohn’s disease [73]. Mutations in the intracellular pattern

recognition receptor NOD2, also known as inflammatory

bowel disease protein 1, have been identified in patients

with Crohn’s disease. These patients produce normal levels

of TNF-a, but they do not produce enough GM-CSF [74].

Multicentre randomized clinical trials have demonstrated

that subcutaneous administration of GM-CSF induces sig-

nificant remission of Crohn’s disease [75, 76]. PAP is

another disease in which GM-CSF production is relevant.

Tumor cells

Inflammatory cells

Sensory nerves

HyperalgesiaNerver emodelling

Release of proinflammatorycytokines

Proliferation

Fig. 4 Tumour–nerve interactions and cancer-induced pain. GM–

CSF may contribute to autocrine and paracrine mechanisms involving

proliferation of tumour cells. GM–CSF sensitizes nerves to mechan-

ical stimuli in vitro and in vivo, potentiates calcitonin gene-related

peptide (CGRP) release and causes sprouting of sensory nerve

endings in the skin mediating the sensitization of nociceptors

following exposure to CSF trough JAK-STAT3 and ERK1/2 signal-

ling. GM–CSF production can lead to recruitment and activation of

inflammatory MØs and monocytes and subsequent release of pro-

inflammatory cytokines such as TNF-a

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

123

PAP is a fatal lung disease characterized by respiratory

failure secondary to a deficient AMØ clearance, abnor-

malities in host defence and accumulation of surfactant

proteins [34]. Autoantibodies against GM-CSF and defi-

cient expression of the GM-CSFRa or bc chain have been

described in patients with PAP [34]. rhGM-CSF inhalation

is beneficial for PAP patients, and gene therapy with vec-

tors encoding GM-CSFRbc has shown efficacy in pre-

clinical results [77, 78].

Recent clinical data suggest that the expression of GM-

CSF may enhance the immune response to tumours. The

first FDA approved vaccine against tumours (Sipuleucel-T;

Provenge�) is an individually tailored DC-based vaccine

that incorporates the use of a key prostate cancer antigen

and GM-CSF to activate DC ex vivo, which upon DC

reconstitution recruits and stimulates T cells to destroy

prostate tumour cells [79]. Another therapy approach has

been to reduce the elevated levels of transforming growth

factor (TGF)-b2 that are frequently linked to immunosup-

pression in cancer patients by simultaneously administer-

ing a TGF-b2 antisense and GM-CSF plasmid and cancer

vaccines destined to enhance autologous tumour cells

recognition [80]. A third type of is currently in a phase III

clinical trial to evaluate the results of expressing GM-CSF

together with IL-12 in allogeneic cancer cells as treatment

for lung cancer [81, 82]. This kind of therapy may be also

effective against colorectal cancer, metastatic renal cell

carcinoma and pancreatic adenocarcinoma [83–85]. The

experimental administration of an adenovirus encoding

GM-CSF together with an adenovirus encoding p16 tumour

suppressor gene induces a potent antitumour response by

increasing the expression of MHC I molecules and the

cytotoxic activity of CD8 T cells [86]. Moreover, the

administration of therapeutic vaccines with plasmids

encoding GM-CSF and tumour antigens enhances the IgG

antibody response antitumour antigens, as well as the IFN-

c and IL-6 production [87, 88]. Another interesting strat-

egy, still in experimental phase, is the combined applica-

tion of armed oncolytic adenovirus expressing IL-12 and

GM-CSF with radiotherapy to suppress primary hepato-

carcinoma in mice [89].

In vitro and in vivo microbicidal activity of neutrophils

and MØ is enhanced by GM-CSF. GM-CSF also prevents

the immunosuppressant effect of dexamethasone, allowing

an effective response against Aspergillus fumigatus hyphae

[90]. Preincubation of neutrophils with GM-CSF increases

the fungicidal activity against Candida glabrata and His-

toplasma capsulatum [7, 91, 92]. In despite of this, there

are no current clinical trials that focus on GM-CSF effect

as an adjuvant for the treatment against invasive fungal

diseases. There are also reports about GM-CSF antibacte-

rial activity in immunodeficient patients. GM-CSF enhan-

ces ex vivo neutrophil bactericidal activity against

S. aureus in children with HIV infection [93]. The use of

GM-CSF in haematopoietic stem cell transplanted patients

reduces the incidence of bacterial infections and decreases

the infection-related hospitalization rates [94]. Local

administration of GM-CSF protects mice against lethal

pneumococcal pneumonia and Chlamydia trachomatis

[95–97].

In vitro studies demonstrated that human MØ stimula-

tion with GM-CSF enhances microbicidal response against

M. tuberculosis [98]. GM-CSF KO mice are highly sus-

ceptible to mycobacterial infections that produce intra-

bronchial and intra-alveolar lesions without formation of

granulomas that are usually produced in wild-type mice

[99]. GM-CSF protects alveolar structure and regulates

early recruitment of MØ and DC, which help to control

mycobacteria growth through granulomas formation [100].

In mice, overexpression of GM-CSF by gene therapy has

shown protection for the reactivation of latent tuberculosis

and avoids the transmissibility of hypervirulent M. tuber-

culosis [101].

The administration of GM-CSF to septic patients did not

produce favourable results. GM-CSF mildly increases the

number of circulating neutrophils in small-for-gestational

age babies but do not improve neonatal sepsis-free survival

[102]. Besides, production of GM-CSF negatively corre-

lates with survival to septic shock [103]. Extremely high

doses of GM-CSF may have a negative impact with

extensive lesions and production of cytokines and inflam-

matory mediators that failed to concentrate T cells and MØ

into sites of infection, MØ accumulation, blindness and

severe damages to various tissues [104, 105].

Prophylactic vaccines based on GM-CSF

The adjuvant role of GM-CSF in prophylactic vaccines

against infectious diseases has been evaluated in preclinical

trials. Vaccines based on recombinant vectors encoding

mycobacterial antigens or vaccines based on recombinant

BCG have shown effectiveness in experimental models of

TB. However, the use of GM-CSF as adjuvant improves

protection against disseminated infection increasing the

number of APC as well as the production of IL-12, IFN-c,

antibodies and the cytotoxic response [106–111]. Intra-

cerebral administration of recombinant rabies virus

encoding GM-CSF prevents the development of experi-

mental rabies after infection with the wild-type virus [112].

A similar DNA vaccine co-expressing GM-CSF against

Clostridium botulinum has shown protection [113]. GM-

CSF is also under evaluation as a new tool for the treatment

for TB, aspergillosis, candida, and influenza [5, 10, 114].

GM-CSF may induce tumour cell death by activation of

circulating monocytes. This and its other immunomodulatory

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

123

properties are currently exploited by means of its adminis-

tration as adjuvant of vaccines against prostate cancer, and it is

also under evaluation in clinical trials for treatment for mel-

anoma and haematological malignancies. Nine hundred sixty-

eight studies were found related to ‘‘sargramostim’’ or ‘‘GM-

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.

Additionally, GM-CSF is also currently being evaluated as

adjuvant for prophylactic vaccines against HIV, sepsis, peri-

odontitis and Crohn’s disease. Table 1 (offered as Supple-

mental Material) shows some of the most interesting ongoing

clinical trials and the potential clinical applications of GM-

CSF that now motivate pharmaceutical companies to inves-

tigate the immunoregulatory and protective effects of this

cytokine in a variety of illnesses.

Conclusion

CSF has a long history of clinical use, but their entire

potential has not yet been fully exploited. They have been

almost exclusively applied to increase the absolute num-

bers of circulating innate immune effector cells which

greatly differ since every CSF has different effect on bone

marrow cells production. Furthermore, neither maturation,

chemotaxis, phagocytosis nor microbicidal functions have

been considered when analysing the effect of diverse CSF,

which has made researchers to miss the very important

value of GM-CSF as immunoregulatory cytokine. It is

quite intriguing how little overlap there is in biological

actions among lymphoid and haematopoietic regulatory

factors. One difference between haematopoietic and lym-

phoid regulatory factors is the circumscribed site in bone

marrow of their production and action confining its stim-

ulation to specific cell colony types. This is one of the most

important differences between G-CSF GM-CSF and

M-CSF, which should be considered as cytokines that

affect rather the quality than the quantity of leucocytes in

circulation. Such functional effect should be first consid-

ered in the clinical guidelines for therapeutic use.

The inclusion of biological products has changed and

expanded the possibilities of modern therapy. GM-CSF

effects on the inflammatory response influence the outcome

of multiple tissue insults in several systems and organs.

Virus-based immune agents such as GM-CSF armed vec-

tors are among the early efforts to reach a potent immu-

nostimulation therapy. GM-CSF has a wide range of

lymphoid regulatory actions, linking innate and adaptive

immunity, acting as a bridge between haematopoietic and

lymphoid factors and should be considered first-line treat-

ment for all kind of illnesses requiring a fully competent

immune response.

Acknowledgments A.F.C., O.R.E., and M.A.S., are scholarship

holders from the National Council for Science and Technology

(CONACYT), Mexico. Point of view and discussion from Leon Islas

is gratefully acknowledged. The graphic design expertise from Ari-

adna Mendez is gratefully acknowledged. The authors declare not to

have any conflict of interest related to publishing this article.

References

1. Metcalf D. Hematopoietic cytokines. Blood. 2008;111:485–91.

2. Bradley TR, Metcalf D. The growth of mouse bone marrow cells

in vitro. Aust J Exp Biol Med Sci. 1966;44:287–99.

3. Sheridan JW, Metcalf D. A low molecular weight factor in lung-

conditioned medium stimulating granulocyte and monocyte

colony formation in vitro. J Cell Physiol. 1973;81:11–23.

4. Burgess AW, Camakaris J, Metcalf D. Purification and proper-

ties of colony-stimulating factor from mouse lung–conditioned

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-

ment of hematopoietic progenitor cells mobilized into the

peripheral blood of normal donors by granulocyte-macrophage

colony-stimulating factor (GM-CSF) or G-CSF: potential role in

allogeneic marrow transplantation. Blood. 1995;85:275–82.

7. Armitage JO. Emerging applications of recombinant human

granulocyte-macrophage colony-stimulating factor. Blood.

1998;92:4491–508.

8. Conti L, Gessani S. GM-CSF in the generation of dendritic cells

from human blood monocyte precursors: recent advances.

Immunobiology. 2008;213:859–70.

9. Fukuzawa H, Sawada M, Kayahara T, et al. Identification of

GM-CSF in Paneth cells using single-cell RT-PCR. Biochem

Biophys Res Commun. 2003;312:897–902.

10. Hamilton JA, Anderson GP. GM-CSF biology. Growth Factors.

2004;22:225–31.

11. Xing Z, Braciak T, Ohkawara Y, et al. Gene transfer for cytokine

functional studies in the lung: the multifunctional role of GM-CSF

in pulmonary inflammation. J Leukoc Biol. 1996;59:481–8.

12. de Groot RP, Coffer PJ, Koenderman L. Regulation of prolif-

eration, differentiation and survival by the IL-3/IL-5/GM-CSF

receptor family. Cell Signal. 1998;10:619–28.

13. Hansen G, Hercus TR, McClure BJ, et al. The structure of the

GM-CSF receptor complex reveals a distinct mode of cytokine

receptor activation. Cell. 2008;134:496–507.

14. Matsuguchi T, Zhao Y, Lilly M, et al. The cytoplasmic domain

of the granulocyte-macrophage colony-stimulating factor (GM-

CSF) receptor subunit is essential for both GM-CSF-mediated

growth and differentiation. J Biol Chem. 1997;272:17450–9.

15. McClure BJ, Hercus TR, Cambareri BA, et al. Molecular

assembly of the ternary granulocyte-macrophage colony-stimu-

lating factor receptor complex. Blood. 2003;101:1308–15.

16. Sawada M, Itoh Y, Suzumura A, et al. Expression of cytokine

receptors in cultured neuronal and glial cells. Neurosci Lett.

1993;160:131–4.

17. Carr PD, Gustin SE, Church AP, et al. Structure of the complete

extracellular domain of the common subunit of the human GM-

CSF, IL-3, and IL-5 receptors reveals a novel dimer configura-

tion. Cell. 2001;104:291–300.

18. McClure BJ, Hercus TR, Cambareri BA, et al. Molecular

assembly of the ternary granulocyte-macrophage colony-stimu-

lating factor receptor complex. Blood. 2003;101:1308–15.

19. Hansen G, Hercus TR, Xu Y, et al. Crystallization and pre-

liminary X-ray diffraction analysis of the ternary human

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

123

GM-CSF receptor complex. Acta Crystallogr Sect F Struct Biol

Cryst Commun. 2008;64:711–4.

20. Choi JK, Kim KH, Park H, et al. Granulocyte macrophage-

colony stimulating factor shows anti-apoptotic activity in neural

progenitor cells via JAK/STAT5-Bcl-2 pathway. Apoptosis.

2011;16:127–34.

21. O’Mahony DS, Pham U, Iyer R, et al. Differential constitutive

and cytokine-modulated expression of human Toll-like receptors

in primary neutrophils, monocytes, and macrophages. Int J Med

Sci. 2008;5:1–8.

22. Tanimoto A, Murata Y, Wang KY, et al. Monocyte chemoat-

tractant protein-1 expression is enhanced by granulocyte-mac-

rophage colony-stimulating factor via Jak2-Stat5 signalling and

inhibited by atorvastatin in human monocytic U937 cells. J Biol

Chem. 2008;283:4643–51.

23. Liontos LM, Dissanayake D, Ohashi PS, et al. The Src-like

adaptor protein regulates GM-CSFR signalling and monocytic

dendritic cell maturation. J Immunol. 2011;186:1923–33.

24. Domen J, Weissman IL. Hematopoietic stem cells need two

signals to prevent apoptosis; BCL-2 can provide one of these,

Kitl/c-Kit signalling the other. J Exp Med. 2000;192:1707–18.

25. Chen J, Carcamo JM, Golde DW. The alpha subunit of the

granulocyte-macrophage colony-stimulating factor receptor

interacts with c-Kit and inhibits c-Kit signalling. J Biol Chem.

2006;281:22421–6.

26. Lilly MB, Zemskova M, Frankel AE, et al. Distinct domains of

the human granulocyte-macrophage colony-stimulating factor

receptor alpha subunit mediate activation of Jak/Stat signalling

and differentiation. Blood. 2001;97:1662–70.

27. Crosier KE, Wong GG, Mathey-Prevot B, et al. A functional

isoform of the human granulocyte/macrophage colony-stimu-

lating factor receptor has an unusual cytoplasmic domain. Proc

Natl Acad Sci USA. 1991;88:7744–8.

28. Raines MA, Liu L, Quan SG, et al. Identification and molecular

cloning of a soluble human granulocyte-macrophage colony-stim-

ulating factor receptor. Proc Natl Acad Sci USA. 1991;88:8203–7.

29. Leukine� Prescribing information. Available at http://products.

sanofi.us/Leukine/Leukine.html Accessed on September 2013.

30. Jiang D, Schwarz H. Regulation of granulocyte and macrophage

populations of murine bone marrow cells by G-CSF and CD137

protein. PLoS One. 2010;5:e15565.

31. Crawford J, Armitage J, Balducci L, et al. Myeloid growth

factors. J Natl Compr Canc Netw. 2009;7:64–83.

32. Kelsen JR, Rosh J, Heyman M, et al. Phase I trial of sargram-

ostim in pediatric Crohn’s disease. Inflamm Bowel Dis.

2010;16:1203–8.

33. The FDA Safety Information and Adverse Event Reporting

Program, 2008. Leukine safety information Available at http://

www.fda.gov/safety/medwatch/safetyinformation/safetyalerts

forhumanmedicalproducts/ucm090918.htm, Accessed on Octo-

ber 2013.

34. Martinez-Moczygemba M, Doan ML, Elidemir O, et al. Pul-

monary alveolar proteinosis caused by deletion of the GM-

CSFR{alpha} gene in the X chromosome pseudoautosomal

region 1. J Exp Med. 2008;205:2711–6.

35. Hamilton JA, Whitty GA, Stanton H, et al. Effects of macro-

phage-colony stimulating factor on human monocytes: induction

of expression of urokinase-type plasminogen activator, but not

of secreted prostaglandin E2, interleukin-6, interleukin-1, or

tumour necrosis factor-alpha. J Leukoc Biol. 1993;53:707–14.

36. Takahashi GW, Andrews DF 3rd, Lilly MB, et al. Effect of

granulocyte-macrophage colony-stimulating factor and inter-

leukin-3 on interleukin-8 production by human neutrophils and

monocytes. Blood. 1993;81:357–64.

37. Selgas R, Fernandez de Castro M, Jimenez C, Selgas R, et al.

Immunomodulation of peritoneal macrophages by granulocyte-

macrophage colony-stimulating factor in humans. Kidney Int.

1996;50:2070–8.

38. Hart PH, Whitty GA, Piccoli DS, et al. Synergistic activation of

human monocytes by granulocyte-macrophage colony-stimu-

lating factor and IFN-gamma. Increased TNF-alpha but not IL-1

activity. J Immunol. 1988;141:1516–21.

39. Hazenberg BP, Van Leeuwen MA, Van Rijswijk MH, et al.

Correction of granulocytopenia in Felty’s syndrome by granu-

locyte-macrophage colony-stimulating factor. Simultaneous

induction of interleukin-6 release and flare-up of the arthritis.

Blood. 1989;74:2769–70.

40. de Vries EG, Willemse PH, Biesma B, et al. Flare-up of rheu-

matoid arthritis during GM-CSF treatment after chemotherapy.

Lancet. 1991;338:517–8.

41. Bischof RJ, Zafiropoulos D, Hamilton JA, et al. Exacerbation of

acute inflammatory arthritis by the colony-stimulating factors

CSF-1 and granulocyte macrophage (GM)-CSF: evidence of

macrophage infiltration and local proliferation. Clin Exp

Immunol. 2000;119:361–7.

42. Campbell IK, Bendele A, Smith DA, et al. Granulocyte-mac-

rophage colony stimulating factor exacerbates collagen induced

arthritis in mice. Ann Rheum Dis. 1997;56:364–8.

43. Burmester GR, Weinblatt ME, McInnes IB, et al. Efficacy and

safety of mavrilimumab in subjects with rheumatoid arthritis.

Ann Rheum Dis. 2013;72:1445–52.

44. Mudzinski SP, Christian TP, Guo TL, et al. Expression of HLA-

DR (major histocompatibility complex class II) on neutrophils

from patients treated with granulocyte-macrophage colony-

stimulating factor for mobilization of stem cells. Blood.

1995;86:2452–3.

45. Fanger NA, Liu C, Guyre PM, et al. Activation of human T cells

by major histocompatibility complex class II expressing neu-

trophils: proliferation in the presence of superantigen, but not

tetanus toxoid. Blood. 1997;89:4128–35.

46. Herold S, Mayer K, Lohmeyer J. Acute lung injury. How

macrophages orchestrate resolution of inflammation and tissue

repair. Front Immunol. 2001;2:6.

47. Cakarova L, Marsh LM, Wilhelm J, et al. Macrophage tumor

necrosis factor-alpha induces epithelial expression of granulo-

cyte-macrophage colony-stimulating factor: impact on alveolar

epithelial repair. Am J Respir Crit Care Med. 2009;180:521–32.

48. Krausgruber T, Blazek K, Smallie T, et al. IRF5 promotes

inflammatory macrophage polarization and TH1-TH17 respon-

ses. Nat Immunol. 2011;12:231–8.

49. Miah MA, Yoon CH, Kim J, et al. CISH is induced during DC

development and regulates DC-mediated CTL activation. Eur J

Immunol. 2012;42:58–68.

50. Hornell TM, Beresford GW, Bushey A, et al. Regulation of the

class II MHC pathway in primary human monocytes by granu-

locyte-macrophage colony-stimulating factor. J Immunol.

2003;171:2374–83.

51. McCormick S, Santosuosso M, Zhang XZ, et al. Manipulation of

dendritic cells for host defence against intracellular infections.

Biochem Soc Trans. 2006;34:283–6.

52. Moldenhauer LM, Keenihan SN, Hayball JD, et al. GM-CSF is

an essential regulator of T cell activation competence in uterine

dendritic cells during early pregnancy in mice. J Immunol.

2010;185:7085–96.

53. Hesske L, Vincenzetti C, Heikenwalder M, et al. Induction of

inhibitory central nervous system-derived and stimulatory

blood-derived dendritic cells suggests a dual role for granulo-

cyte-macrophage colony-stimulating factor in central nervous

system inflammation. Brain. 2010;133(Pt 6):1637–54.

54. Sonderegger I, Iezzi G, Maier R, et al. GM-CSF mediates

autoimmunity by enhancing IL-6-dependent Th17 cell devel-

opment and survival. J Exp Med. 2008;205:2281–94.

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

123

55. Biondo M, Nasa Z, Marshall A, Toh BH, Alderuccio F. Local

transgenic expression of granulocyte macrophage-colony stim-

ulating factor initiates autoimmunity. J Immunol.

2001;166(3):2090–9.

56. Kim DH, Sandoval D, Reed J, et al. The role of GM-CSF in

adipose tissue inflammation. Am J Physiol Endocrinol Metab.

2008;295:E1038–46.

57. Exley MA, Koziel MJ. To be or not to be NKT: natural killer T

cells in the liver. Hepatology. 2004;40:1033–40.

58. Brocheriou I, Maouche S, Durand H, et al. Antagonistic regu-

lation of macrophage phenotype by M-CSF and GM-CSF:

implication in atherosclerosis. Atherosclerosis.

2011;214:316–24.

59. Saitoh T, Kishida H, Tsukada Y, et al. Clinical significance of

increased plasma concentration of macrophage colony-stimu-

lating factor in patients with angina pectoris. J Am Coll Cardiol.

2000;35:655–65.

60. Oren H, Erbay AR, Balci M, et al. Role of novel biomarkers of

inflammation in patients with stable coronary heart disease.

Angiology. 2007;58:148–55.

61. Kellar RS, Lancaster JJ, Thai HM, et al. Antibody to granulo-

cyte-macrophage colony-stimulating factor reduces the number

of activated tissue macrophages and improves left ventricular

function following myocardial infarction in a rat coronary-artery

ligation model. J Cardiovasc Pharmacol. 2011;57:568–74.

62. Sugiyama Y, Yagita Y, Oyama N, et al. Granulocyte colony-

stimulating factor enhances arteriogenesis and ameliorates

cerebral damage in a mouse model of ischemic stroke. Stroke.

2011;42:770–5.

63. Tu J, Karasavvas N, Heaney ML, et al. Molecular character-

ization of a granulocyte macrophage-colony-stimulating factor

receptor alpha subunit-associated protein. GRAP Blood.

2000;96:794–9.

64. Stosser S, Schweizerhof M, Kuner R. Hematopoietic colony-

stimulating factors: new players in tumor-nerve interactions.

J Mol Med. 2011;89:321–9.

65. Ding DX, Rivas CI, Heaney ML, et al. The alpha subunit of the

human granulocyte-macrophage colony-stimulating factor

receptor signals for glucose transport via a phosphorylation-

independent pathway. Proc Natl Acad Sci USA. 1994;91:

2537–41.

66. Vadhan-Raj S, Keating M, LeMaistre A, et al. Effects of

recombinant human granulocyte-macrophage colony-stimulat-

ing factor in patients with myelodysplastic syndromes. N Engl J

Med. 1987;317:1545–52.

67. Schweizerhof M, Stosser S, Kurejova M, et al. Hematopoietic

colony-stimulating factors mediate tumor-nerve interactions and

bone cancer pain. Nat Med. 2009;15:802–7.

68. Khaled YS, Ammori BJ, Elkord E. Myeloid-derived suppressor

cells in cancer: recent progress and prospects. Immunol Cell

Biol. 2013;91:493–502.

69. Zhang Y, Cheng S, Zhang M, et al. High-infiltration of tumor-

associated macrophages predicts unfavorable clinical outcome

for node-negative breast cancer. PLoS One. 2013;8:e76147.

70. Sica A, Mantovani A. Macrophage plasticity and polarization:

in vivo veritas. J Clin Invest. 2012;122:787–95.

71. Hao NB, Lu MH, Fan YH, Cao YL, Zhang ZR, et al. Macro-

phages in tumor microenvironments and the progression of

tumors. Clin Dev Immunol. 2012;2012:948098.

72. Egea L, Hirata Y, Kagnoff MF. GM-CSF: a role in immune and

inflammatory reactions in the intestine. Expert Rev Gastroen-

terol Hepatol. 2010;4:723–31.

73. Hirata Y, Egea L, Dann SM, et al. GM-CSF-facilitated dendritic

cell recruitment and survival govern the intestinal mucosal

response to a mouse enteric bacterial pathogen. Cell Host

Microbe. 2010;7:151–63.

74. Brosbøl-Ravnborg A, Hvas CL, Agnholt J, et al. Toll-like

receptor-induced granulocyte-macrophage colony-stimulating

factor secretion is impaired in Crohn’s disease by nucleotide

oligomerization domain 2-dependent and -independent path-

ways. Clin Exp Immunol. 2009;155:487–95.

75. Korzenik JR, Dieckgraefe BK, Valentine JF, et al. Sargramostim

for active Crohn’s disease. N Engl J Med. 2005;352:2193–201.

76. Valentine JF, Fedorak RN, Feagan B, et al. Steroid-sparing

properties of sargramostim in patients with corticosteroid-

dependent Crohn’s disease: a randomised, double-blind, pla-

cebo-controlled, phase 2 study. Gut. 2009;58:1354–62.

77. Tazawa R, Trapnell BC, Inoue Y, et al. Inhaled granulocyte/

macrophage-colony stimulating factor as therapy for pulmonary

alveolar proteinosis. Am J Respir Crit Care Med. 2010;181:

1345–54.

78. Kleff V, Sorg UR, Bury C, et al. Gene therapy of beta(c)-defi-

cient pulmonary alveolar proteinosis (beta(c)-PAP): studies in a

murine in vivo model. Mol Ther. 2008;16:757–64.

79. Higano CS, Schellhammer PF, Small EJ, et al. Integrated data

from 2 randomized, double-blind, placebo-controlled, phase 3

trials of active cellular immunotherapy with sipuleucel-T in

advanced prostate cancer. Cancer. 2009;115:3670–9.

80. Olivares J, Kumar P, Yu Y, et al. Phase I trial of TGF-{beta}2

antisense GM-CSF gene-modified autologous tumor cell (TAG)

vaccine. Clin Cancer Res. 2011;17:183–92.

81. Bradbury PA, Shepherd FA. Immunotherapy for lung cancer.

J Thorac Oncol. 2008;3:S164–70.

82. Holt GE, Disis ML. Immune modulation as a therapeutic strat-

egy for non-small-cell lung cancer. Clin Lung Cancer.

2009;9:S13–9.

83. Staff C, Mozaffari F, Haller BK, et al. A Phase I safety study of

plasmid DNA immunization targeting carcinoembryonic antigen

in colorectal cancer patients. Vaccine. 2011;29:6817–22.

84. Garcia JA, Mekhail T, Elson P, et al. Phase I/II trial of subcu-

taneous interleukin-2, granulocyte-macrophage colony-stimu-

lating factor and interferon-a in patients with metastatic renal

cell carcinoma. BJU Int. 2012;109:63–9.

85. Lutz E, Yeo CJ, Lillemoe KD, et al. A lethally irradiated allo-

geneic granulocyte-macrophage colony stimulating factor-

secreting tumor vaccine for pancreatic adenocarcinoma: a phase

II trial of safety, efficacy, and immune activation. Ann Surg.

2011;253:328–35.

86. Wang L, Qi X, Sun Y, et al. Adenovirus-mediated combined

P16 gene and GM-CSF gene therapy for the treatment of

established tumor and induction of antitumor immunity. Cancer

Gene Ther. 2002;9:819–24.

87. Dranoff G, Crawford AD, Sadelain M, et al. Involvement of

granulocyte-macrophage colony-stimulating factor in pulmon-

ary homeostasis. Science. 1994;264:713–6.

88. Sun X, Hodge LM, Jones HP, et al. Co-expression of granulo-

cyte-macrophage colony-stimulating factor with antigen

enhances humoral and tumor immunity after DNA vaccination.

Vaccine. 2002;20:1466–74.

89. Kim W, Seong J, Oh HJ, et al. A novel combination treatment of

armed oncolytic adenovirus expressing IL-12 and GM-CSF with

radiotherapy in murine hepatocarcinoma. J Radiat Res.

2011;52:646–54.

90. Roilides E, Blake C, Holmes A, et al. Granulocyte-macrophage

colony-stimulating factor and interferon-gamma prevent dexa-

methasone-induced immunosuppression of antifungal monocyte

activity against Aspergillus fumigatus hyphae. J Med Vet Mycol.

1996;34:63–9.

91. Kowanko IC, Ferrante A, Harvey DP, et al. Granulocyte-mac-

rophage colony-stimulating factor augments neutrophil killing

of Torulopsis glabrata and stimulates neutrophil respiratory

burst and degranulation. Clin Exp Immunol. 1991;83:225–30.

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

123

92. Newman SL, Gootee L. Colony-stimulating factors activate

human macrophages to inhibit intracellular growth of Histo-

plasma capsulatum yeasts. Infect Immun. 1992;60:4593–7.

93. Roilides E, Mertins S, Eddy J, et al. Impairment of neutrophil

chemotactic and bactericidal function in children infected with

human immunodeficiency virus type 1 and partial reversal after

in vitro exposure to granulocyte-macrophage colony-stimulating

factor. J Pediatr. 1990;117:531–40.

94. Page AV, Liles WC. Colony-stimulating factors in the preven-

tion and management of infectious diseases. Infect Dis Clin

North Am. 2011;25:803–17.

95. Steinwede K, Tempelhof O, Bolte K, et al. Local delivery of

GM-CSF protects mice from lethal pneumococcal pneumonia.

J Immunol. 2011;187:5346–56.

96. Lu H, Xing Z, Brunham RC. GM-CSF transgene-based adjuvant

allows the establishment of protective mucosal immunity fol-

lowing vaccination with inactivated Chlamydia trachomatis.

J Immunol. 2002;169:6324–31.

97. Bo L, Wang F, Zhu J, et al. Granulocyte-colony stimulating

factor (G-CSF) and granulocyte-macrophage colony stimulating

factor (GM-CSF) for sepsis: a meta-analysis. Crit Care. 2011;

15:R58.

98. Denis M, Ghadirian E. Granulocyte-macrophage colony-stimu-

lating factor restricts growth of tubercle bacilli in human mac-

rophages. Immunol Lett. 1990;24:203–6.

99. Chroneos ZC, Midde K, Sever-Chroneos Z, et al. Pulmonary

surfactant and tuberculosis. Tuberculosis. 2009;89:S10–4.

100. Szeliga J, Daniel DS, Yang CH, et al. Granulocyte-macrophage

colony stimulating factor-mediated innate responses in tuber-

culosis. Tuberculosis. 2008;88:7–20.

101. Francisco-Cruz A, Mata-Espinosa D, Estrada-Parra S, Xing Z,

Hernandez-Pando R. Immunotherapeutic effects of recombinant

adenovirus encoding granulocyte-macrophage colony-stimulat-

ing factor in experimental pulmonary tuberculosis. Clin Exp

Immunol. 2013;171:283–97.

102. Marlow N, Morris T, Brocklehurst P, et al. A randomised trial of

granulocyte-macrophage colony-stimulating factor for neonatal

sepsis: outcomes at 2 years. Arch Dis Child Fetal Neonatal Ed.

2013;98(1):F46–53.

103. Mera S, Tatulescu D, Cismaru C, et al. Multiplex cytokine

profiling in patients with sepsis. APMIS. 2011;119:155–63.

104. Gonzalez-Juarrero M, Hattle JM, Izzo A, et al. Disruption of

granulocyte macrophage-colony stimulating factor production in

the lungs severely affects the ability of mice to control myco-

bacterium tuberculosis infection. J Leukoc Biol. 2005;77:

914–22.

105. Lang RA, Metcalf D, Cuthbertson RA, et al. Transgenic mice

expressing a hemopoietic growth factor gene (GM-CSF)

develop accumulations of macrophages, blindness, and a fatal

syndrome of tissue damage. Cell. 1987;51:675–86.

106. Zhang X, Divangahi M, Ngai P, et al. Intramuscular immuni-

zation with a monogenic plasmid DNA tuberculosis vaccine:

enhanced immunogenicity by electroporation and co-expression

of GM-CSF transgene. Vaccine. 2007;25:1342–52.

107. Ryan AA, Wozniak TM, Shklovskaya E, et al. Improved pro-

tection against disseminated tuberculosis by Mycobacterium

bovis bacillus Calmette-Guerin secreting murine GM-CSF is

associated with expansion and activation of APCs. J Immunol.

2007;179:18–24.

108. Nambiar JK, Ryan AA, Kong CU, et al. Modulation of pul-

monary DC function by vaccine-encoded GM-CSF enhances

protective immunity against Mycobacterium tuberculosis

infection. Eur J Immunol. 2010;40:153–61.

109. Dou J, Tang Q, Yu F, et al. Investigation of immunogenic effect

of the BCG priming and Ag85A- GM-CSF boosting in Balb/c

mice model. Immunobiology. 2010;215:133–42.

110. Zhang X, Divangahi M, Ngai P, et al. Intramuscular immuni-

zation with a monogenic plasmid DNA tuberculosis vaccine:

enhanced immunogenicity by electroporation and co-expression

of GM-CSF transgene. Vaccine. 2007;25:1342–52.

111. Wang J, Zganiacz A, Xing Z. Enhanced immunogenicity of

BCG vaccine by using a viral-based GM-CSF transgene adju-

vant formulation. Vaccine. 2002;20:2887–98.

112. Wang H, Zhang G, Wen Y, et al. Intracerebral administration of

recombinant rabies virus expressing GM-CSF prevents the

development of rabies after infection with street virus. PLoS

One. 2011;6:e25414.

113. Li N, Yu YZ, Yu WY, et al. Enhancement of the immunogenicity

of DNA replicon vaccine of Clostridium botulinum neurotoxin

serotype A by GM-CSF gene adjuvant. Immunopharmacol Im-

munotoxicol. 2011;33:211–9.

114. Grabstein KH, Urdal DL, Tushinski RJ, et al. Induction of

macrophage tumoricidal activity by granulocyte-macrophage

colony-stimulating factor. Science. 1986;232:506–8.

115. Kaushansky K. Lineage-specific hematopoietic growth factors.

N Engl J Med. 2006;354:2034–45.

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

123