key: cord-0794770-y5qlrci5 authors: Lee, Kevin M C; Achuthan, Adrian A; Hamilton, John A title: GM-CSF: A Promising Target in Inflammation and Autoimmunity date: 2020-10-29 journal: Immunotargets Ther DOI: 10.2147/itt.s262566 sha: 335447ce1b2e1a19eb1a559a35259eda376a1f1b doc_id: 794770 cord_uid: y5qlrci5 The cytokine, granulocyte macrophage-colony stimulating factor (GM-CSF), was firstly identified as being able to induce in vitro the proliferation and differentiation of bone marrow progenitors into granulocytes and macrophages. Much preclinical data have indicated that GM-CSF has a wide range of functions across different tissues in its action on myeloid cells, and GM-CSF deletion/depletion approaches indicate its potential as an important therapeutic target in several inflammatory and autoimmune disorders, for example, rheumatoid arthritis. In this review, we discuss briefly the biology of GM-CSF, raise some current issues and questions pertaining to this biology, summarize the results from preclinical models of a range of inflammatory and autoimmune disorders and list the latest clinical trials evaluating GM-CSF blockade in such disorders. Granulocyte macrophage-colony stimulating factor (GM-CSF, CSF2) was originally defined as a hemopoietic growth factor due to its ability to form colonies of granulocytes and macrophages in vitro by proliferation and differentiation of bone marrow progenitor cells. 1 A number of reports have revealed that GM-CSF and the GM-CSF receptor (GM-CSFR) levels are elevated and correlated with disease severity in many inflammatory/autoimmune diseases, for example, rheumatoid arthritis (RA). 2 In addition, there is much preclinical data providing a strong rationale for the involvement of GM-CSF in such diseases. As a result, GM-CSF and GM-CSFR have both attracted great interest as potential therapeutic targets. This review provides a brief overview of the pleiotropic biology of GM-CSF and outlines some of the most recent preclinical findings and in particular the resultant clinical studies using GM-CSF-or GM-CSFR-targeting monoclonal antibodies (mAbs) in various diseases. It also summarizes some of the contentious issues and outstanding questions pertaining to GM-CSF biology. This review cannot obviously cover all aspect of the broad topic and further background information on GM-CSF biology and targeting can be found in earlier reviews (see, for example, [2] [3] [4] [5] [6] ). latter shared with the interleukin-3 (IL-3) and IL-5 receptors. The activation of GM-CSFR triggers (i) phosphorylation of the GM-CSFRβ subunit, which commonly leads to the binding of signal transducer and activator of transcription 5 (STAT5) thereby initiating Janus Kinase (JAK) 2 signaling [7] [8] [9] and (ii) activation of the MEK/ERK, phosphatidylinositol 3 kinase (PI3K) and NFκB pathways. 10, 11 The hemopoietic-specific transcription factor, interferon regulatory factor 4 (IRF4), is a key signaling molecule for the adoption of dendritic cell (DC)-like properties in GM-CSF -treated precursors, such as monocytes. [12] [13] [14] [15] [16] Recently, it was reported that the GM-CSF stimulation of monocytes/macrophages in vitro leads to the formation of CCL17 via IRF4 as an important pathway, termed the GM-CSF/CCL17 axis (see below). 17 Mechanistically, GM-CSF up-regulates IRF4 expression by enhancing JMJD3 demethylase activity. 17 Additionally, GM-CSF-IRF4 signaling favours the polarization of pro-inflammatory macrophages and increased antigen presentation capability (ie increased MHC class II expression) during in vivo inflammation. 18 In contrast, some literature indicates IRF5, but not IRF4, to be important for GM-CSFinduced macrophage polarization 19,20 and IRF4 has been considered to have an anti-inflammatory role in macrophages (for example, enhanced interleukin (IL)-10 and reduced TNF production). 21-23 While GM-CSF appears to be dispensable for steady state myeloid cell development in vivo, 24 GM-CSF directly regulates the differentiation of liver-derived fetal monocytes into immature alveolar macrophages during embryonic development 25 and also promotes the development of functional alveolar macrophages via PU.1. 25,26 GM-CSF genedeficient mice develop pulmonary alveolar proteinosis, 24, 27 which can also occur in humans due to genetic mutations or endogenous neutralizing antibody (53). 28 This pathology results from compromised alveolar macrophage functions. 29 In addition, GM-CSF deficient mice have been reported to be more susceptible to lung infections. 30,31 Inhaled GM-CSF can protect mice from such infections 30,32 via enhancing macrophage and DC function. 30, 31 These studies suggest that the availability of excess GM-CSF could be beneficial in certain circumstances. A wide range of cells can produce GM-CSF. [2] [3] [4] [5] [6] During inflammation, major sources of GM-CSF are both hematopoietic, for example, T and B cells, and nonhematopoietic, for example, tissue resident cells. GM-CSF has been reported to be produced by T H 1 and T H 17 cells via STAT signaling 33-35 and shown to be crucial for encephalitogenicity. 33 A distinct subset of T cells, known as GM-CSF-producing T H cells, has recently been identified. 35 GM-CSF-producing T H cells express undetectable T-bet, GATA-3, or RORγt and do not express a cytokine signature that other T cell subsets express. 35, 36 In humans, GM-CSF-producing T H cells can be identified as CCR10 + CCR4 + CXCR3 − CCR6 − cells. 36 GM-CSFproducing T H cells have been implicated in autoimmune brain disease as fewer GM-CSF-producing T H cells correlated with less severe experimental autoimmune encephalomyelitis (EAE). 35 The innate lymphoid cell (ILC) family encompasses the classic cytotoxic natural killer (NK) cells and the noncytotoxic ILCs. 37,38 NK cells have been recently reported to produce GM-CSF when infiltrating into joints for the maintenance of inflammatory arthritis. 39 Among the subsets of ILCs, type 3 ILCs have been shown to secrete GM-CSF in intestinal inflammation. [40] [41] [42] In spondyloarthritis, type 3 ILCs are found to be enriched in the inflamed joint and are the predominate source of GM-CSF. 43 A subset of B cells, namely innate response activator (IRA) B cells, resides in nonlymphoid sites, such as the peritoneal and pleural cavities, and provides a first line of defense against infection. 44 In a model of Toll-like receptor (TLR)-induced sepsis, IRA B cells, which produce GM-CSF and also reported to express the GM-CSFR, mediate a GM-CSF-dependent IgM protective mechanism against septic shock. 44-46 Mixed chimeric mice with B cell-restricted GM-CSF deficiency had high bacterial titer morbidity after infection but did not develop alveolar proteinosis, 44 indicating that B cell-derived GM-CSF is dispensable for surfactant clearance by alveolar macrophages. Memory B cells isolated from multiple sclerosis (MS) patients produce high levels of GM-CSF, termed GM-CSF + B cells, and co-culture of these cells with macrophagecolony stimulating factor (M-CSF)-generated, human blood monocyte-derived macrophages initiates proinflammatory responses. 47 Interestingly, dimethyl fumarate ameliorates MS submit your manuscript | www.dovepress.com ImmunoTargets and Therapy 2020:9 226 and depletes GM-CSF-producing B cells in these patients 48,49 consistent with a pathogenic role for these cells. Tissue resident cells can also be a potential source of GM-CSF during inflammation. For example, it has been shown that fibroblast-like synoviocytes are important for the initiation of experimental autoimmune arthritis via their GM-CSF production. 50 GM-CSF has been reported to be expressed by cardiac fibroblasts in models of Kawasaki disease and myocarditis. 51,52 Epithelial cells can produce GM-CSF in response to allergenic stimuli 53,54 and such production can restore alveolar barrier function. 55 In addition, studies also reported that endothelial cells can produce GM-CSF in response to pro-inflammatory cytokine stimulation. 56-58 In vitro, GM-CSF can regulate proliferation and/or activation of myeloid cells, namely monocytes, macrophages, DCs, neutrophils and eosinophils. At sites of inflammation, GM-CSF can be proinflammatory through recruitment of myeloid cells and/or by enhancing their survival and activation. 6, 59 However, it has been reported that prolonged exposure to GM-CSF can lead to the generation of monocyte-derived suppressor cells. 60 In addition to being able to control the development of monocytes and macrophages from bone marrow precursors in vitro, GM-CSF can regulate multiple functions in the differentiated cells, including cell survival, proliferation and maturation, via transcription factors, such as PU. 1 and IRF4. 17, 26, 61 During infection, GM-CSF has been shown to boost macrophage antimicrobial functions, such as enhanced phagocytosis 62 and increased production of reactive oxygen species. 63,64 GM-CSF stimulates monocytes/ macrophages to secrete some pro-inflammatory cytokines (for example, IL-6, IL-23 and CCL17) 17,65-67 and these cells are often hyperinflammatory ("primed") when they encounter a second stimulus (for example, lipopolysaccharide (LPS)). [68] [69] [70] Macrophages are remarkably plastic cells and have been classified into various so-called "polarization states" (for example, M1 vs M2) in different diseased tissues. As regards the expression of certain pro-inflammatory cytokines, GM-CSF has been considered to shift the phenotype of macrophages into a M1-like, pro-inflammatory polarization state; 71 however, such cells have also been considered to have dual M1/M2 characteristics, 53,67 and GM-CSF-activated monocytes have been reported to alleviate experimental colitis. 67 As a result, it has been recommended that the M1/M2 polarization terminology not be applied to GM-CSF action. 2, 17, 61, 72 GM-CSF also promotes the development of migratory CD103 + CD11b + DCs, 40,73 while negatively regulating the development of resident CD8 + DCs. 74 GM-CSF, often in combination with IL-4, is widely used to generate in vitro murine and human DC populations from bone marrow precursors and blood monocytes, respectively. 11, [75] [76] [77] Heterogeneity in GM-CSF-induced bone marrow DCs has been reported, comprising at least two populations, namely the Fms-like tyrosine kinase 3 (FLT-3) + DC or the CD11c + MHCII + CD115 + monocyte-derived DC (MoDC). 78 There has been debate in the literature regarding the role of GM-CSF in the in vivo generation of MoDCs. [79] [80] [81] [82] [83] [84] [85] Some studies have shown GM-CSF to be dispensable for the differentiation of MoDCs; 82 in contrast, it has been demonstrated that NFκB-dependent GM-CSF production in CD4 + T cells is required for the generation of MoDCs. 81, 83, 84 Very recently, to add fuel to this debate GM-CSF has been proposed 86 to differentiate a population of DC3s independently of conventional DCs (cDCs) or monocyte-restricted progenitors. Besides the regulation of DC numbers, there is evidence that GM-CSF also regulates their function, including antigen presentation (as indicated by increased MHCII expression 78 ) and inflammatory responses (as indicated by increased IL-6 and IL-23 secretion 87 ). GM-CSF can enhance the survival, adhesion and trafficking of neutrophils. 88, 89 During infection, GM-CSF also upregulates the antimicrobial functions of neutrophils, such as phagocytosis 90 and formation of extracellular traps. 91 One study reported that the expression of PU.1 in neutrophils of pulmonary alveolar proteinosis patients was normal, indicating that GM-CSF is not involved in steady state neutrophil development. 89 The chemokine, CCL17 (formerly called thymus and activation-regulated chemokine [TARC]) was originally implicated in the preferential attraction of T H 2 cells; 92 however, it can also attract regulatory T cells. 93 Its most recognized receptor is CCR4 94 although the atypical chemokine receptor 2 (ACR2) has also been reported to be a CCL17 receptor. 95 As mentioned above, GM-CSF in vitro upregulates CCL17 formation via an IRF4/JMJD3-dependent mechanism in human and mouse monocytes/macrophages, 17 as well as in inflammation ImmunoTargets and Therapy 2020:9 submit your manuscript | www.dovepress.com DovePress 227 models. 18,80 It is to be noted that GM-CSF is not the only mediator that regulates CCL17 production in these populations; for example, IL-4 also upregulates the CCL17 expression in human monocytes and mouse macrophages via a similar mechanism. 96 This GM-CSF/CCL17 axis has been found to be important in controlling inflammatory arthritic and osteoarthritic pain in pre-clinical models; 17,80,97-99 importantly, this axis appears to be active in humans, since a neutralizing GM-CSFR mAb in RA patients leads to a sustained reduction in circulating CCL17 levels. 100 Preclinical studies suggest that CCL17 may not necessarily be acting as a T cell chemokine in its control of inflammation and its associated pain. 17,80,97-99 Mechanistically, whether CCL17 has a direct effect on neurons for pain induction remains an open question 101 -there are conflicting reports as to whether CCR4, the CCL17 receptor, is expressed on neurons. [101] [102] [103] [104] However, whatever the mechanism, it is clear that neutralizing CCL17 peripherally with a mAb ameliorates inflammatory arthritic 99 and osteoarthritic 97,98 pain. Additionally, other studies have also reported a nonchemotactic role for CCL17, for example, a role(s) in regulating inflammation by restricting regulatory T cell expansion. 105, 106 There are still a number of issues and questions pertaining to GM-CSF biology, which need to be addressed as they have potential implications for the clinical targeting of GM-CSF in inflammation and autoimmunity. It should be borne in mind that this biology may vary depending on the processes and tissues involved in the particular clinical indication in question. Which responding cell type(s) is relevant and whether GM-CSF regulates their number and/or activation/differentiation status are important considerations -the latter issue may have implications for therapeutic delivery and dosing since the extent of GM-CSF neutralization/depletion is likely to be critical. 6 There is still plenty of controversy around the role of GM-CSF in DC development in vivo. [79] [80] [81] [82] [83] [84] [85] It would also be worth knowing how significant is GM-CSF-dependent IRF4 signaling in monocytes/macrophages, including the so-called GM-CSF/CCL17 axis discussed above. Depending on the particular model of inflammation/autoimmunity being studied, the relevant cell type(s) producing GM-CSF varies, again with possible implications for therapeutic strategies. GM-CSF administration systemically can have pro-inflammatory and anti-inflammatory effects -as discussed previously, these responses to exogenous GM-CSF may or may not be predictive of the findings when endogenous (locally acting?) GM-CSF is neutralized/depleted. 2 GM-CSF has been documented for its role in peripheral pain. 17,99,107,108 However, whether in this capacity GM-CSF is acting directly on neurons (nociceptors), including acting centrally, remains unclear as conflicting reports have been published. 101, [109] [110] [111] [112] [113] [114] [115] Additionally, GM-CSF has also been reported to have neuroprotective effects following nerve injury. 116, 117 Further research is obviously needed to clarify how GM-CSF interacts with the nervous system. Early studies measuring cytokines in synovial fluid and blood from patients with RA showed increased GM-CSF levels, as well as increased expression of GM-CSFR, in inflamed synovial tissue. 118, 119 Administration of GM-CSF to RA patients led to disease flares. 120 A genome-wide association study revealed that mutations in CSF2 (the gene that encodes GM-CSF) contribute to genetic susceptibility in RA. 121 Based in part on the priming of blood monocytes with GM-CSF, it was recently suggested that GM-CSF neutralization be considered as a potential therapeutic approach for the treatment of ankylosing spondylitis. 122 The contribution of GM-CSF to the pathogenesis of experimental inflammatory arthritis is well documented in the literature. GM-CSF-deficient mice fail to develop arthritis and associated pain in several inflammatory arthritis models, including collagen-induced arthritis (CIA), antigen-induced arthritis (AIA), zymosan-induced arthritis (ZIA) and K/BxN serum-transfer arthritis (STA). 17,39,108,123 The administration of neutralizing GM-CSF mAbs ameliorated existing disease in these models. 99, 124 Regarding the relevance of GM-CSF to arthritic pain, as mentioned earlier GM-CSF is implicated in regulating inflammatory and arthritic pain via downstream CCL17. 17,98,99 Interestingly, high levels of circulating GM-CSF have been shown to correlate with the responsiveness of RA patients to anti-TNF agents. 125 Consistent with the concept of the GM-CSF/CCL17 axis (see above), RA patients treated with anti-GM-CSFR mAb (mavrilimumab) have reduced circulating CCL17 levels, suggesting that CCL17 could be a biomarker for anti-GM-CSF or anti-GM-CSFR treatment. 100 submit your manuscript | www.dovepress.com ImmunoTargets and Therapy 2020:9 Osteoarthritis Osteoarthritis (OA) was once considered a noninflammatory arthropathy; however, it is now wellrecognized that there can be a significant inflammatory component contributing to OA clinical symptoms, for example, chronic pain. The expression of GM-CSF and its receptor have been found in OA synovial tissue 126, 127 and reported to be negatively correlated with pain. 126 In contrast, in a collagenase-induced, joint instability OA model, GM-CSF-deficient mice were protected from associated pain and osteophyte development. 107 Consistent with this data, neutralizing anti-GM-CSF mAb effectively ameliorated pain in the same model. 97, 107 This pain amelioration was observed when the neutralizing mAb was administered early or late in this model but early administration was needed for it to be effective on joint damage. 97 This data has led to clinical trials in OA for targeting GM-CSF (see below) or CCL17 (https://clinicaltrials.gov/show/ NCT03485365). Synovial inflammation, characterized by macrophage infiltration, is often more prominent in early OA lesions, while advanced OA is more commonly associated with structural changes (for example, cartilage degeneration and/or osteophyte formation). [128] [129] [130] Given that GM-CSF regulates a wide range of macrophage functions (see above), it could be that optimal clinical improvement might be seen in patients with early OA as opposed to patients with advanced OA disease. Multiple sclerosis (MS) is a chronic autoimmune/inflammatory disease of the central nervous system (CNS) and is characterized by demyelination and subsequent axonal degeneration. While it is widely believed that T H 17 cells are the main encephalitogenic population in EAE, 131 the most widely used MS model, it was reported that their key secreted cytokine, IL-17, is dispensable for the development of EAE. 132,133 Instead, it was later shown that GM-CSF secreted by T H 17 cells is the main cytokine contributing to encephalitogenicity 134 via the activation of microglia within the CNS. 135 GM-CSF-activated microglia adopt a M1-like (inflammatory) phenotype 136 and produce highly neurotoxic molecules such as tumor necrosis factor (TNF), IL-1 and IL-6. 137 It has been proposed that GM-CSF promotes the breakdown of the blood brain barrier enabling entry of circulating Ly6C hi monocytes and stimulates the differentiation of monocyte-derived antigen-presenting cells. 138, 139 These differentiated cells share a similar phenotype to macrophages found in active MS lesions. 140, 141 Additional mouse studies have demonstrated that GM-CSF deletion results in fewer monocyte-derived cells in the CNS parenchyma following EAE induction, 142 and GM-CSF administration leads to more cells migrating into the CNS parenchyma. 143 Elevated GM-CSF levels have been reported in the cerebrospinal fluid of patients with active MS. 144, 145 Glatiramer acetate, a FDAapproved drug to treat MS, has been shown to upregulate regulatory T cells and reduce GM-CSF levels in mice with EAE. 146 These reports demonstrate that GM-CSF plays a central role in EAE and indicate that GM-CSF might be a therapeutic target in MS. Inflammatory bowel disease (IBD) is a chronic immunemediated disease affecting the gastrointestinal tract consisting of two main subtypes: Crohn's disease (CD) and ulcerative colitis (UC). 147 Impaired innate immunity plays a critical pathogenic role in IBD. 148 GM-CSF has been identified as a key mediator of chronic inflammation in models of colitis. 41, 149, 150 Other studies using dextran sodium sulfate (DSS)-induced colitis reported that GM-CSF-deficient mice developed more severe colitis; 151, 152 mechanistically, it has been claimed that type 3 ILCderived GM-CSF modulates the macrophage phenotype to prevent intestinal fibrosis. 42 In line with a potential beneficial role of GM-CSF in IBD, GM-CSF administration can improve IBD experimentally 153 and in some patients; 152,154 also, high levels of circulating anti-GM-CSF autoantibodies have been found to correlate with worse CD prognosis. 155, 156 Interstitial Lung Disease Interstitial lung disease (ILD) comprise heterogenous inflammatory lung parenchyma disorders that can lead to alveolitis and ultimately fibrosis. 157 It is also a serious complication associated with systemic rheumatic diseases. 157 Experimentally, the SKG mouse with a mutation in the Zap-70 gene 158,159 develops spontaneous arthritis, ILD and IBD. 160 The lungs of SKG mice develop fibrosis associated with intense infiltrates of GM-CSF + IL-17A + neutrophils, pathological features that are reminiscent of human ILD. 161 GM-CSF blockade reduces these features, including the degree of fibrosis, in SKG mice, with IL-17 blockade being less effective. 162 Interestingly, CCL17-expressing macrophages have been implicated in mediating peritoneal fibrosis. 163 Given these findings, together with the data in lung inflammation models wherein GM-CSF blockade ImmunoTargets and Therapy 2020:9 submit your manuscript | www.dovepress.com DovePress 229 impaired CCL17 expression in alveolar macrophages, 80 in our view further studies examining the role of the GM-CSF /CCL17 axis in lung fibrosis are warranted. Dissecting aortic aneurysm is an important and often lifethreatening condition. It was reported that mice deficient in Kif6, the gene encoding the transcription factor, Krueppel-like factor 6, developed worse aortic aneurysm. 164 In the same study, GM-CSF was identified to be an effector molecule downstream of Krueppel-like factor 6 and the administration of GM-CSF exacerbated aortic aneurysm formation, with GM-CSF antagonism having the opposite effect. 164 In the aortic root GM-CSF induces CD11b + Gr-1 + Ly6C hi inflammatory monocyte accumulation, with anti-GM-CSF mAb administration resulting in reduced inflammation and dilation. 165 These findings suggest that GM-CSF blockade might be an effective therapeutic approach in aortic aneurysm. GM-CSF has been reported to be involved in the T H 2 response in allergic airway inflammation via activation of DCs. 53, 166, 167 In a mouse model of asthma, allergenexposed epithelial cells secrete GM-CSF, which activates DCs and also prolongs eosinophil survival. 53, 168 Administration of a GM-CSF neutralizing mAb also led to reduced allergic hyperresponsiveness. 167, 168 As a result, an anti-GM-CSF mAb has been tested in a Phase II trial for severe asthma (see below). Interestingly, it has been reported that alveolar DC-derived CCL17 is critical for airway inflammation 169, 170 and CCL17 airway expression correlates with asthmatic disease severity. 171, 172 In our view these data warrant a detailed study examining the role of the GM-CSF/CCL17 axis in asthma, with CCL17 being a potential target for treating allergic disease and/or a biomarker for patient selection. Obesity is now widely considered as a low-grade, chronic inflammatory disease that contributes to metabolic dysfunction, ectopic lipid deposition and insulin resistance. 173, 174 With progressive obesity, adipose tissue macrophages (ATMs) have been considered to be a key cell type contributing to metabolic inflammation, insulin resistance and the impairment of adipocyte function. [175] [176] [177] In response to diet-induced obesity (DIO) in mice, elevated GM-CSF levels can be detected in serum, 178 peritoneal fluid, 179 and adipose tissue; 180 moreover, GM-CSF is required for DIOinduced adipose tissue inflammation, as GM-CSF genedeficient mice had reduced number of infiltrating ATMs and crown-like features in adipose tissue, 180 in spite of increased adiposity and body weight. 181, 182 It was also reported that GM-CSF gene-deficient mice exhibited improved metabolic status, namely insulin sensitivity to glucose, compared with their wild-type counterparts, 180 and that GM-CSF-responsive myeloid cells play a key role in this improvement. 181 As a result, GM-CSF has been proposed to be a key mediator whose actions might explain the difference between obese individuals with normal glucose tolerance (metabolically "healthy") and those with type 2 diabetes (metabolically "unhealthy"). 181 In addition to type 2 diabetes, GM-CSF has also been implicated in other obesity-exacerbated diseases, for example, in the obesity-mediated enhancement of breast cancer metastasis. 183 How GM-CSF plays a role during obesityinduced meta-inflammation remains to be explored. In coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), there is a characteristic hyperactive immune response that leads to an overwhelming infiltration of inflammatory myeloid cells (particularly monocytes, macrophages and neutrophils) into the lungs. [184] [185] [186] [187] [188] [189] [190] More recent studies have questioned the validity of referring to the COVID-19 hyperactive immune response as the "cytokine storm" seen in chimeric antigen receptor (CAR) T cell-related cytokine release syndrome (CRS). 191, 192 The COVID-19-related hyperactive immune response resembles a phenotype of secondary haemophagocytic lymphohistiocytosis, often referred to as "macrophage activation syndrome". [184] [185] [186] [187] [188] [189] In COVID-19 patients, increased percentages of GM-CSF-expressing leukocytes have been found in the blood. 193 Inhibition of GM-CSF activity in models of hyperinflammatory conditions that share similar pathology to late stages of COVID-19, such as CAR T cell-related CRS and neurotoxicity, 194 graft versus host disease-associated CRS 195 and inflammatory lung diseases, 162, [196] [197] [198] was shown to be beneficial. The relevance of GM-CSF to COVID-19 and its potential as a therapeutic target have been reviewed recently. 199, 200 submit your manuscript | www.dovepress.com ImmunoTargets and Therapy 2020:9 A number of clinical trials neutralizing GM-CSF (see Table 1 ) or GM-CSFR (see Table 2 ) using mAbs have been/are being carried out. Encouragingly, no serious adverse events have been noted so far, for example infections and compromised lung function, with the data from a long-term open label extension (OLE) study in RA patients being particularly promising in this regard. 100 Otilimab Otilimab (formerly known as MOR-103 and GSK3196165) is an IgG1 mAb, developed by MorphoSys AG, that binds to GM-CSF and prevents its interaction with GM-CSFRα; it is ImmunoTargets and Therapy 2020:9 submit your manuscript | www.dovepress.com DovePress currently being produced by GSK for use in several randomized controlled trials (RCTs). A short-term, dose-escalation phase Ib/IIa trial in randomized RA patients (n=96, NCT01023256) showed improved efficacy in all outcomes (ACR and European League Against Rheumatism (EULAR) response) compared with placebo with no pulmonary function test abnormalities being reported. 201 The subsequent doubleblind, placebo-controlled phase IIa (NCT02799472) and IIb (NCT02504671) trials consistently showed clinical improvement in RA patients receiving otilimab and it was well tolerated. Circulating CCL17 levels declined only in the otilimab group, supporting the existence of the GM-CSF /CCL17 axis in humans. GSK has announced the start of a clinical development program (ContRAst) embracing three Phase III trials aiming to evaluate the efficacy and safety of otilimab in RA patients with inadequate response to i) conventional synthetic/biologic disease modifying anti-rheumatic drugs (DMARDs) (NCT03970837), ii) methotrexate (NCT03980483) and (iii) biologic DMARDs and/or JAK inhibitors (NCT04134728). A long-term safety and efficacy study with otilimab has also commenced (NCT04333147). The results of an exploratory, 12-week, phase IIa study of otilimab in patients with hand OA (n=44, NCT02683785) have been reported and, while not statistically significant in this small study, reduction in pain, accompanied by improvement in functional impairment, was noted. 202 Patients have been recruited to determine the efficacy of otilimab in COVID-19 (NCT04376684). 199 Lenzilumab Lenzilumab (formerly known as KB003) is an IgG1neutralizing anti-GM-CSF mAb and has been tested successfully in a randomized phase II trial in RA (NCT00995449). A Phase I trial using lenzilumab in patients with chronic myelomonocytic leukemia (CMML) has been completed (NCT02546284); 33% of patients showed durable clinical benefit, which appears to be better in a distinct subtype of CMML patients, warranting further studies to identify CMML subtypes more likely to respond. 203 A Phase II, randomized, double-blind, placebocontrolled, 24-week study in asthma patients (n=311; NCT01603277) has been performed; overall, there were no effects on asthma control although there appeared to be improvements in patients with eosinophilic asthma. 204 Recently, lenzilumab was administered to a small cohort of patients (n=12) with COVID-19 pneumonia and found to associate with improved clinical outcome with no mortality observed; 205 a subsequent phase III study has commenced patient recruitment to evaluate efficacy and safety of lenzilumab (NCT04351152). TJM2 is an IgG1-neutralizing anti-GM-CSF mAb. Phase I trials in healthy subjects (NCT03794180) and in patients with severe COVID-19 (NCT04341116) have commenced. Namilumab (formerly known as MT203), an IgG1neutralizing anti-GM-CSF mAb, has been investigated in double-blind, placebo controlled, randomized trials in healthy individuals (NCT02354599) 206 and in RA patients (NCT01317797), 207 which established that namilumab has an acceptable tolerability profile. 206, 207 Phase 1b (NCT01317797) and phase II studies (NCT02379091) in RA demonstrated efficacy, with the latter study further reporting dose-response effects. 207, 208 A phase II trial investigating the efficacy of namilumab in plaque psoriasis was also completed (NCT02129777) with no significant difference being recorded for this end point between placebo-treated and namilumab-treated individuals. 209 Patients are being recruited for a phase IIa trial using namilumab in axial spondyloarthritis (NCT03622658). Gimsilumab (also known as KIN-1901), a fully human IgG1 mAb, has been investigated in a double-blind, placebo controlled, randomized trial in healthy subjects and subjects with ankylosing spondylitis (NCT04205851). Patients are being recruited for a phase II trial in COVID-19 (NCT04351243). Mavrilimumab (CAM-3001) is a humanized IgG4 mAb with high affinity to the GM-CSFRα chain. 210 The efficacy and safety profiles of mavrilimumab have been investigated in a phase I trial in 32 RA patients (NCT00771420) and in the subsequent EARTH clinical development program, including two phase IIa-IIb RCTs (EARTH EXPLORER 1 and EARTH EXPLORER 2). 211, 212 In the phase IIb, placebocontrolled EARTH EXPLORER 1 study (NCT01706926), moderate-to-severe active RA patients (n=236) with ongoing methotrexate treatment, received three different doses of mavrilimumab (150, 100 and 30 mg) . The DAS28-CRP score among these RA patients was significantly decreased submit your manuscript | www.dovepress.com ImmunoTargets and Therapy 2020:9 in all mavrilimumab subgroups compared with placebo, with the optimal response being 150mg mavrilimumab. 212 The EARTH EXPLORER 2 study (NCT01715896), which was a phase II, double-blind, randomized trial, evaluated the benefits of using mavrilimumab (100mg every other week, n=70) in long-standing, active RA patients (mean disease duration 6.7 years, and mean baseline DAS28-ESR 6.5), who had not responded to a conventional synthetic DMARD or TNF inhibitor. 211 This study included a parallel treatment with an anti-TNF mAb (golimumab) (50 mg every 4 weeks, n=68). No statistical difference was seen between RA patients treated with mavrilimumab or golimumab, which could be due to the fact that a suboptimal mavrilimumab dose (100 mg every other week) was used in this trial as opposed to the most effective dose (150mg every other week), confirmed by the EARTH EXPLORER 1 study. 212 Peripheral biomarkers and pathophysiological pathways modulated by mavrilimumab and golimumab were also assessed in the study. While a number of mediators were suppressed by both mAbs, mavrilimumab, but not golimumab, was able to suppress serum levels of CCL17 and CCL22 and to induce sustained differential suppression of peripheral disease markers in anti-TNF inadequate responders. 211 The long-term efficacy and safety profile of mavrilimumab were also explored in an OLE study (NCT01712399). All patients (n=422), who completed the double-blind phase of EARTH EXPLORER 1 and 2 trials (study 1109; NCT01712399), had the opportunity to enter the study and receive mavrilimumab 100 mg every other week plus methotrexate for a 3-year followup period. 100 At week 122, 65.0% and 40.6% patients achieved a DAS28-CRP score of <3.2 and <2.6, respectively, 100 demonstrating a sustained benefit in measures of RA disease outcomes. The overall safely profile of mavrilimumab appears to be promising, particularly regarding pulmonary alveolar proteinosis. In this OLE study, biomarker analyses support the hypothesis that GM-CSF regulates CCL17 and CCL22 as sustained suppression of CCL17 and CCL22 was seen in mavrilimumab-treated patients over a longer follow-up period. New clinical trials are underway to evaluate the benefits of mavrilimumab in patients with giant cell arteritis (NCT03827018) and in COVID-19 patients (NCT04399980 and NCT04397497). 199 It has recently been reported that mavrilimumab treatment was associated with improved clinical outcomes compared with standard care with patients with severe COVID-19. 213 The preclinical rationale for targeting GM-CSF in inflammation/autoimmunity is solid, and the results from early phase clinical trials of GM-CSF or GM-CSFR blockade in RA patients, and possibly asthma and COVID-19, are encouraging. However, careful ongoing evaluation of adverse effects, particularly in the lungs and gut, is clearly paramount but, as mentioned above, it seems so far that the anti-GM-CSF and anti-GM-CSFR mAbs used in clinical trials are without major safety concerns. It is hoped that there are other indications (see Table 1 ) where GM-CSF targeting will turn out to provide potential benefit. The key role of GM-CSF in inflammatory pain was highlighted above. In this connection, the rapid and dramatic effect of mavrilimumab on RA pain has been highlighted 211, 212 and, intriguingly, it has been speculated that the dramatic effects of the JAK1/2 inhibitor, baricitinib, also on RA pain may be due to its inhibition of GM-CSF signaling. 214 Circulating biomarkers, such as CCL17, may aid in the selection of an indication for GM-CSF-based therapeutics and even of patients within such an indication, thus hopefully leading to better clinical outcomes. It is hoped that targeting GM-CSF is successful in patients who are nonresponders to biologics, for example, those targeting other inflammatory mediators, such as TNF and IL-6. The The nature and action of granulocyte-macrophage colony stimulating factors Anti-colony-stimulating factor therapies for inflammatory and autoimmune diseases Colony-stimulating factors in inflammation and autoimmunity CSF in inflammatory diseases GM-CSF: from growth factor to central mediator of tissue inflammation GM-CSF in inflammation The structure of the GM-CSF receptor complex reveals a distinct mode of cytokine receptor activation ImmunoTargets and Therapy 2020:9 submit your manuscript | www Novel GM-CSF signals via IFN-gammaR/IRF-1 and AKT/mTOR license monocytes for suppressor function Defining GM-CSFand macrophage-CSF-dependent macrophage responses by in vitro models Cytokine enhancement of complement-dependent phagocytosis by macrophages: synergy of tumor necrosis factor-alpha and granulocyte-macrophage colony-stimulating factor for phagocytosis of cryptococcus neoformans GM-CSF-deficient mice are susceptible to pulmonary group B streptococcal infection Granulocyte macrophage-colony stimulating factor induced Zn sequestration enhances macrophage superoxide and limits intracellular pathogen survival Role of GM-CSF in the inflammatory cytokine network that regulates neutrophil influx into the colonic mucosa during clostridium difficile infection in mice IL-1beta and TNFalpha promote monocyte viability through the induction of GM-CSF expression by rheumatoid arthritis synovial fibroblasts Reprogramming of monocytes by GM-CSF contributes to regulatory immune functions during intestinal inflammation GM-CSF priming drives bone marrow-derived macrophages to a pro-inflammatory pattern and downmodulates PGE2 in response to TLR2 ligands GM-CSF priming of human monocytes is dependent on ERK1/2 activation Priming effects of GM-CSF, IFN-gamma and TNF-alpha on human neutrophil inflammatory cytokine production Granulocytemacrophage colony-stimulating factor (CSF) and macrophage CSF-dependent macrophage phenotypes display differences in cytokine profiles and transcription factor activities: implications for CSF blockade in inflammation Macrophage activation and polarization: nomenclature and experimental guidelines GM-CSF-dependent, CD103+ dermal dendritic cells play a critical role in Th effector cell differentiation after subcutaneous immunization Polyethylene glycol-modified GM-CSF expands CD11b(high)CD11c(high) CD11c(high) murine dendritic cells in vivo: a comparative analysis with Flt3 ligand Activities of granulocyte-macrophage colony-stimulating factor and interleukin-3 on monocytes ImmunoTargets and Therapy 2020:9 submit your manuscript | www Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor GM-CSF in the generation of dendritic cells from human blood monocyte precursors: recent advances GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c MHCII(+) macrophages and dendritic cells Specific contributions of CSF-1 and GM-CSF to the dynamics of the mononuclear phagocyte system CCL17 in inflammation and pain GM-CSF-responsive monocyte-derived dendritic cells are pivotal in Th17 pathogenesis GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells Synovial CD4+ T-cellderived GM-CSF supports the differentiation of an inflammatory dendritic cell population in rheumatoid arthritis Differentiation of inflammatory dendritic cells is mediated by NF-kappaB1-dependent GM-CSF production in CD4 T cells GM-CSF-dependent inflammatory pathways Transcriptional and functional analysis of CD1c(+) human dendritic cells identifies a CD163(+) subset priming CD8(+)CD103(+) T cells GM-CSF mediates autoimmunity by enhancing IL-6-dependent Th17 cell development and survival Granulocytemacrophage colony-stimulating factor induces neutrophil adhesion to pulmonary vascular endothelium in vivo: role of beta 2 integrins Human GM-CSF autoantibodies and reproduction of pulmonary alveolar proteinosis RNA-seq reveals activation of both common and cytokine-specific pathways following neutrophil priming Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps Compartmentalized production of CCL17 in vivo: strong inducibility in peripheral dendritic cells contrasts selective absence from the spleen Unique chemotactic response profile and specific expression of chemokine receptors CCR4 and CCR8 by CD4(+)CD25(+) regulatory T cells CCR4 and its ligands: from bench to bedside The atypical chemokine receptor ACKR2 drives pulmonary fibrosis by tuning influx of CCR2(+) and CCR5(+) IFNgamma-producing gammadeltaT cells in mice Epigenetic and transcriptional regulation of IL4-induced CCL17 production in human monocytes and murine macrophages Targeting GM-CSF for collagenase-induced osteoarthritis pain and disease in mice CCL17 blockade as a therapy for osteoarthritis pain and disease TNF and granulocyte macrophage-colony stimulating factor interdependence mediates inflammation via CCL17 Mavrilimumab, a fully human granulocyte-macrophage colony-stimulating factor receptor alpha monoclonal antibody: long-term safety and efficacy in patients with rheumatoid arthritis Immune cytokines and their receptors in inflammatory pain Defining the nociceptor transcriptome Chemokines and glycoprotein120 produce pain hypersensitivity by directly exciting primary nociceptive neurons Somatosensory neuron types identified by high-coverage single-cell RNA-sequencing and functional heterogeneity CCL17-expressing dendritic cells drive atherosclerosis by restraining regulatory T cell homeostasis in mice CCL17 promotes intestinal inflammation in mice and counteracts regulatory T cell-mediated protection from colitis Granulocyte-macrophage colony-stimulating factor is a key mediator in experimental osteoarthritis pain and disease development Granulocyte-macrophage colony-stimulating factor is a key mediator in inflammatory and arthritic pain Hematopoietic colony-stimulating factors: new players in tumor-nerve interactions Hematopoietic colony-stimulating factors mediate tumor-nerve interactions and bone cancer pain Distribution of granulocyte-monocyte colony-stimulating factor and its receptor alpha-subunit in the adult human brain with specific reference to Alzheimer's disease Transcriptional mechanisms underlying sensitization of peripheral sensory neurons by granulocyte-/granulocyte-macrophage colony stimulating factors Central inhibition of granulocyte-macrophage colony-stimulating factor is analgesic in experimental neuropathic pain Transcriptional regulation of voltage-gated sodium channels contributes to GM-CSF-induced pain Granulocyte-macrophage colony stimulating factor as an indirect mediator of nociceptor activation and pain A neuroprotective function for the hematopoietic protein granulocyte-macrophage colony stimulating factor (GM-CSF) Granulocyte-macrophage colony stimulating factor exerts protective and immunomodulatory effects in cortical trauma Cytokines in chronic inflammatory arthritis. II. Granulocyte-macrophage colony-stimulating factor in rheumatoid synovial effusions Plasma GM-CSF concentrations in rheumatoid arthritis, systemic lupus erythematosus and spondyloarthropathy Correction of granulocytopenia in Felty's syndrome by granulocyte-macrophage colony-stimulating factor. Simultaneous induction of interleukin-6 release and flare-up of the arthritis Meta-analysis identifies nine new loci associated with rheumatoid arthritis in the Japanese population GM-CSF primes proinflammatory monocyte responses in ankylosing spondylitis Protection from collagen-induced arthritis in granulocyte-macrophage colony-stimulating factor-deficient mice Blockade of collagen-induced arthritis post-onset by antibody to granulocyte-macrophage colony-stimulating factor (GM-CSF): requirement for GM-CSF in the effector phase of disease Response to treatment with TNFalpha inhibitors in rheumatoid arthritis is associated with high levels of GM-CSF and GM-CSF(+) T lymphocytes Expression of granulocyte macrophage-colony stimulating factor and its receptor in the synovium of osteoarthritis patients is negatively correlated with pain Evidence for GM-CSF receptor expression in synovial tissue. An analysis by semi-quantitative polymerase chain reaction on rheumatoid arthritis and osteoarthritis synovial biopsies Unicompartmental and bicompartmental knee osteoarthritis show different patterns of mononuclear cell infiltration and cytokine release in the affected joints Inflammatory cells in patients with endstage knee osteoarthritis: a comparison between the synovium and the infrapatellar fat pad Synovial tissue inflammation in early and late osteoarthritis The pathogenicity of Th17 cells in autoimmune diseases IL-17A and IL-17F do not contribute vitally to autoimmune neuro-inflammation in mice RORgammat drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation The encephalitogenicity of T(H) 17 cells is dependent on IL-1-and IL-23-induced production of the cytokine GM-CSF GM-CSF production by autoreactive T cells is required for the activation of microglial cells and the onset of experimental autoimmune encephalomyelitis T-cell-mediated regulation of neuroinflammation involved in neurodegenerative diseases GM-CSF increases LPS-induced production of proinflammatory mediators via upregulation of TLR4 and CD14 in murine microglia Circulating Ly-6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease GM-CSF promotes migration of human monocytes across the blood brain barrier Macrophages in inflammatory multiple sclerosis lesions have an intermediate activation status Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination GM-CSF promotes chronic disability in experimental autoimmune encephalomyelitis by altering the composition of central nervous system-infiltrating cells, but is dispensable for disease induction Granulocyte macrophage colony-stimulating factor: a new putative therapeutic target in multiple sclerosis Profile of cerebrospinal fluid and serum cytokines in patients with relapsing-remitting multiple sclerosis: a correlation with clinical activity ImmunoTargets and Therapy 2020:9 submit your manuscript | www Markers of activated T lymphocytes and T cell receptor gamma/delta+ in patients with multiple sclerosis Glatiramer acetate increases T-and B -regulatory cells and decreases granulocyte-macrophage colony-stimulating factor (GM-CSF) in an animal model of multiple sclerosis Epidemiology and risk factors for IBD Crohn's disease: an immune deficiency state Dysregulated hematopoietic stem and progenitor cell activity promotes interleukin-23-driven chronic intestinal inflammation Granulocyte macrophage colony-stimulating factor-activated eosinophils promote interleukin-23 driven chronic colitis The role of granulocyte macrophage-colonystimulating factor in acute intestinal inflammation Granulocyte macrophage colony-stimulating factor and the intestinal innate immune cell homeostasis in Crohn's disease Granulocytemacrophage colony-stimulating factor elicits bone marrow-derived cells that promote efficient colonic mucosal healing Treatment of active Crohn's disease with recombinant human granulocyte-macrophage colonystimulating factor Impaired granulocytemacrophage colony-stimulating factor bioactivity accelerates surgical recurrence in ileal Crohn's disease Genetic and transcriptomic variation linked to neutrophil granulocyte-macrophage colonystimulating factor signaling in pediatric crohn's disease Interstitial lung disease in systemic autoimmune rheumatic diseases: a comprehensive review Altered thymic T-cell selection due to a mutation of the ZAP-70 gene causes autoimmune arthritis in mice The SKG mutation in ZAP-70 also confers arthritis susceptibility in C57 black mouse strains Interleukin-23 mediates the intestinal response to microbial beta-1,3-glucan and the development of spondyloarthritis pathology in SKG mice IL-17A(+)GM-CSF(+) neutrophils are the major infiltrating cells in interstitial lung disease in an autoimmune arthritis model GM-CSF but not IL-17 is critical for the development of severe interstitial lung disease in SKG mice Inflammatory macrophages switch to CCL17-expressing phenotype and promote peritoneal fibrosis Granulocyte macrophage colony-stimulating factor is required for aortic dissection/intramural haematoma GM-CSF contributes to aortic aneurysms resulting from SMAD3 deficiency Granulocyte/ macrophage-colony stimulating factor in allergen-induced rhinitis: cellular localization, relation to tissue eosinophilia and influence of topical corticosteroid Intranasal exposure of mice to house dust mite elicits allergic airway inflammation via a GM-CSFmediated mechanism Attenuation of airway hyperresponsiveness in a murine asthma model by neutralization of granulocyte-macrophage colony-stimulating factor (GM-CSF) CCL17 production by dendritic cells is required for NOD1-mediated exacerbation of allergic asthma Blockade of CCR4 in a humanized model of asthma reveals a critical role for DC-derived CCL17 and CCL22 in attracting Th2 cells and inducing airway inflammation Airway epithelial integrin beta4 suppresses allergic inflammation by decreasing CCL17 production Targeting TSLP with shRNA alleviates airway inflammation and decreases epithelial CCL17 in a murine model of asthma Inflammation, metaflammation and immunometabolic disorders Inflammation and metabolic disorders Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance Obesity is associated with macrophage accumulation in adipose tissue Obesity induces a phenotypic switch in adipose tissue macrophage polarization Obesity alters immune and metabolic profiles: new insight from obese-resistant mice on high-fat diet The effect of diet-induced obesity on the inflammatory phenotype of non-adipose-resident macrophages in an in vivo model of gout The role of GM-CSF in adipose tissue inflammation GM-CSF driven myeloid cells in adipose tissue link weight gain and insulin resistance via formation of 2-aminoadipate Granulocyte/macrophage colony-stimulating factor-dependent dendritic cells restrain lean adipose tissue expansion Obesity alters the lung myeloid cell landscape to enhance breast cancer metastasis through IL5 and GM-CSF COVID-19 illness in native and immunosuppressed states: a clinical-therapeutic staging proposal Cytokine release syndrome in severe COVID-19 COVID-19: consider cytokine storm syndromes and immunosuppression Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19 Complex immune dysregulation in COVID-19 patients with severe respiratory failure Targeting potential drivers of COVID-19: neutrophil extracellular traps Immunotherapeutic implications of IL-6 blockade for cytokine storm Is a "cytokine storm" relevant to COVID-19? An inflammatory cytokine signature predicts COVID-19 severity and survival Pathogenic T-cells and inflammatory monocytes incite inflammatory storms in severe COVID-19 patients GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts Graft-versus-host disease, but not graft-versus-leukemia immunity, is mediated by GM-CSFlicensed myeloid cells Lipopolysaccharideinduced lung inflammation is inhibited by neutralization of GM-CSF Neutrophil GM-CSF receptor dynamics in acute lung injury GM-CSF) regulates lung innate immunity to lipopolysaccharide through Akt/Erk activation of NFkappa B and AP-1 in vivo GM-CSF-based treatments in COVID-19: reconciling opposing therapeutic approaches Targeting GM-CSF in COVID-19 pneumonia: rationale and strategies MOR103, a human monoclonal antibody to granulocyte-macrophage colony-stimulating factor, in the treatment of patients with moderate rheumatoid arthritis: results of a phase Ib/IIa randomised, double-blind, placebo-controlled, dose-escalation trial A Phase iia study of anti-GM-CSF antibody GSK3196165 in subjects with inflammatory hand osteoarthritis Phase 1 study of lenzilumab, a recombinant anti-human GM-CSF antibody, for chronic myelomonocytic leukemia Phase 2, randomised placebo-controlled trial to evaluate the efficacy and safety of an anti-GM-CSF antibody (KB003) in patients with inadequately controlled asthma First clinical use of lenzilumab to neutralize GM-CSF in patients with severe COVID-19 pneumonia. medRxiv Randomized, double-blind, placebo-controlled, phase I study of the safety and pharmacokinetics of namilumab in healthy Japanese and Caucasian men Phase 1b randomized, double-blind study of namilumab, an anti-granulocyte macrophage colony-stimulating factor monoclonal antibody, in mild-tomoderate rheumatoid arthritis Efficacy and safety of namilumab, a human monoclonal antibody against granulocyte-macrophage colony-stimulating factor (GM-CSF) ligand in patients with rheumatoid arthritis (RA) with either an inadequate response to background methotrexate therapy or an inadequate response or intolerance to an anti-TNF (tumour necrosis factor) biologic therapy: a randomized, controlled trial Granulocytemacrophage colony-stimulating factor (GM-CSF) as a therapeutic target in psoriasis: randomized, controlled investigation using namilumab, a specific human anti-GM-CSF monoclonal antibody Mavrilimumab, a human monoclonal GM-CSF receptor-alpha antibody for the management of rheumatoid arthritis: a novel approach to therapy A randomized phase IIb study of mavrilimumab and golimumab in rheumatoid arthritis A randomised phase IIb study of mavrilimumab, a novel GM-CSF receptor alpha monoclonal antibody, in the treatment of rheumatoid arthritis ImmunoTargets and Therapy 2020:9 submit your manuscript | www GM-CSF blockade with mavrilimumab in severe COVID-19 pneumonia and systemic hyperinflammation: a single-centre, prospective cohort study Comparison of baricitinib, upadacitinib, and tofacitinib mediated regulation of cytokine signaling in human leukocyte subpopulations ImmunoTargets and Therapy is an international, peer-reviewed open access journal focusing on the immunological basis of diseases, potential targets for immune based therapy and treatment protocols employed to improve patient management. Basic immunology and physiology of the immune system in health, and disease will be also covered. In addition, the journal will focus on the impact of management programs and new therapeutic agents and protocols on patient perspectives such as quality of life, adherence and satisfaction. The manuscript management system is completely online and includes a very quick and fair peer-review system, which is all easy to use. Visit http://www.dovepress.com/testimonials.php to read real quotes from published authors.Submit your manuscript here: http://www.dovepress.com/immunotargets-and-therapy-journal submit your manuscript | www.dovepress.com ImmunoTargets and Therapy 2020:9