Abstract
Macrophages play diverse roles in development, homeostasis, and immunity. Accordingly, the dysfunction of macrophages is involved in the occurrence and progression of various diseases, such as coronavirus disease 2019 and atherosclerosis. The protective or pathogenic effect that macrophages exert in different conditions largely depends on their functional plasticity, which is regulated via signal transduction such as Janus kinase–signal transducer and activator of transcription, Wnt and Notch pathways, stimulated by environmental cues. Over the past few decades, the molecular mechanisms of signaling pathways in macrophages have been gradually elucidated, providing more alternative therapeutic targets for diseases treatment. Here, we provide an overview of the basic physiology of macrophages and expound the regulatory pathways within them. We also address the crucial role macrophages play in the pathogenesis of diseases, including autoimmune, neurodegenerative, metabolic, infectious diseases, and cancer, with a focus on advances in macrophage‐targeted strategies exploring modulation of components and regulators of signaling pathways. Last, we discuss the challenges and possible solutions of macrophage‐targeted therapy in clinical applications. We hope that this comprehensive review will provide directions for further research on therapeutic strategies targeting macrophage signaling pathways, which are promising to improve the efficacy of disease treatment.
Keywords: diseases, macrophage, signaling pathway, therapeutic targets
In this review, the authors introduce the research advances related to macrophage signaling pathways by first providing an overview of the basic physiology of macrophages and expound on the regulatory pathways within them. Then, the pathogenic effect of macrophages and the therapeutic strategies targeting macrophage signaling pathways are addressed in various diseases.

1. INTRODUCTION
Tissue homeostasis refers to the state of dynamic balance of tissue structure and function, the destruction of which leads to the occurrence of diseases. 1 , 2 The maintenance of tissue homeostasis relies on a series of sensors, signals, and effectors. 2 Deviation of the regulatory variable from the normal range can activate sensors, which leads to the generation of signals acting on effectors, thereby correcting the deviation of the regulatory variable. 2 Macrophages are professional sensors due to their wide distribution in almost every organ tissue and their marvelous adaptability to diverse stimuli. 3 , 4
Plasticity is one of the hallmarks of macrophages, which refers to the ability to change their phenotypes, functions, and physiological characteristics in response to various external challenges. 3 , 5 This is one of the underlying reasons why macrophages can exert multiple key effects in organisms. Transcriptional regulation of gene expression underlies macrophage plasticity, and the bridge between stimulation and transcription is a complex network of signaling pathways involving activation and inhibition of signaling cascade responses. 5 , 6 The perception and integration of information by macrophages result in extensive activation and significant crosstalk of signaling pathways, resulting in delicate and subtle responses to environmental changes.
Unfortunately, macrophage plasticity is not only involved in organism protection but also has pathogenic effects. 7 In some conditions, various stimuli lead to abnormal macrophage activation or dysfunction, resulting in a causal association between macrophages and disease states. 8 The identification of molecules and signaling pathways involved in macrophage plasticity and activation provide the basis for macrophage‐centered therapeutic strategies.
In this review, we first outline the latest understanding of the basic physiology of macrophages, including their origins, replenishment, polarization, and physiological functions. Then, we focus on important signaling pathways regulating the functions of macrophages in a receptor‐centered way. On this basis, we discuss the current therapeutic approaches in clinical use or under clinical trials that target key components of these signaling pathways, including ligands, receptors and effectors, in different diseases. Finally, we discuss the challenges and future research directions of macrophage‐targeted therapy.
2. BASIC PHYSIOLOGY AND ROLE OF MACROPHAGES
Macrophages, originally identified because of their phagocytic nature, are a heterogeneous group of immune cells that are widely distributed throughout the organism and play important roles in development and homeostasis maintenance. Furthermore, it is expected that the origins and polarization of macrophages will determine their phenotypes and functions in specific microenvironments. In this section, we will provide an overview of the basic biology of macrophages, including their origins, replenishment, polarization, and physiological functions.
2.1. Origins of macrophages
Although macrophages were initially thought to originate primarily from hematopoietic stem cells (HSCs) and circulating monocytes, 9 accumulating evidence has clarified an embryo‐derived macrophage lineage with macrophage precursors derived from erythro‐myeloid progenitors (EMPs) in yolk sacs and fetal liver. 9 , 10 The EMP‐derived macrophage precursors can settle throughout the embryo and form subpopulations of tissue‐specific macrophages during organogenesis. And these tissue‐specific macrophages can establish stable spatial and functional relationships with specialized tissue cells and are termed “tissue‐resident macrophages (TRMs).” 11 The EMPs then disappear during fetal life, but EMP‐derived TRMs persist and self‐renew in adults, such as microglia, Kupffer cells, and Langerhans cells. 12 In addition, a small proportion of TRMs are gradually replaced or replenished by HSCs‐derived macrophages. For example, alveolar macrophages and osteoclasts are derived from EMPs and HSCs in adults, 11 while the mucosal macrophages in the gut lamina propria are derived from HSCs and circulating monocytes in adults. 13 Furthermore, macrophages functioning in inflammatory or other pathological states are predominantly derived from circulating monocytes. 5 , 14 , 15
2.2. Replenishment of macrophages
Macrophages must maintain or increase their population to better perform functions, and two strategies are at stake. 14 , 16 One is the recruitment of monocytes, and the other is to increase the proliferation of TRMs by enhancing their self‐renewal capacity. 14 , 17
2.2.1. Recruitment of macrophages
Under tissue stress conditions, including inflammation and cancer, circulating monocytes can be recruited to specific sites and differentiate into macrophages with specific functions. This process relies on the participation of chemokines or cytokines. 18 C‐C motif ligand (CCL) 2 and CCL5 specifically attract and activate monocytes and are key players in the regulation of monocyte/macrophage migration and infiltration. 19 , 20 Colony‐stimulating factor‐1 (CSF‐1) and vascular endothelial growth factor (VEGF) have potent chemotactic effects on monocytes and macrophages via CSF‐1 receptor (CSF‐1R) and VEGF‐R1, respectively. 21 , 22 Endothelial monocyte‐activating polypeptide II was found to promote chemotaxis of monocytes and macrophages, with endothelins (ET)‐1 having a chemotactic effect on human monocytes and ET‐2 on macrophages. 19
2.2.2. In situ proliferation of macrophages
Recent evidence points to the self‐renewal and numerical maintenance of macrophages in adult tissues under homeostatic conditions by in situ proliferation of TRMs rather than an influx of hematopoietic progenitors. 23 , 24 It has been suggested that local proliferation of TRMs is more involved in tissue macrophage regeneration after mild injury, whereas monocyte‐derived macrophages are more engaged in severe conditions. 3 , 25 Notably, TRMs gradually lose their self‐renewal ability with aging. 26 However, the mechanisms underlying the local proliferation and self‐renewal ability of TRMs are still being explored.
2.3. Polarization and functional phenotypes of macrophages
Polarization of macrophages is a process of altering phenotypes and function in response to microenvironmental stimuli, 27 which is essential for macrophages to function appropriately in different environments. 28 , 29
M1 and M2 macrophages represent the two main subpopulations of macrophages. 29 M1 macrophages, also called classically activated macrophages, express molecular markers such as CD80, CD86, CD68, major histocompatibility complex class II (MHC‐II), and Toll‐like receptor (TLR) 4, 29 which are activated by lipopolysaccharide (LPS), interferon‐γ (IFN‐γ), or tumor necrosis factor α (TNF‐α). M1 macrophages secrete a variety of proinflammatory cytokines30, such as interleukin (IL)‐1, IL‐6, TNF‐α, nitric oxide (NO), reactive oxygen species (ROS), and downregulate the expression of IL‐12, IL‐23, and IL‐10. 31 M1 macrophages mainly promote Th1 immune responses, possess potent antimicrobial and antitumor effects, and mediate ROS‐induced tissue damage that impairs tissue regeneration and wound healing. 31
To avoid tissue damage and maintain homeostasis, anti‐inflammatory M2 macrophage‐driven regulatory mechanisms suppress chronic inflammatory responses. 30 M2 macrophages, also known as alternatively activated macrophages, express surface markers, including CD206, CD163, CD209, FIZZ1, Ym1/2 and galactose receptor. 29 , 32 M2 macrophages are mainly activated by CSF‐1, IL‐4, IL‐13, and transforming growth factor‐β (TGF‐β) and can secrete IL‐10, arginase 1 (Arg 1) and TGF‐β, thus playing important roles in inflammation inhibition, tissue remodeling, wound healing, angiogenesis, the T helper type 2 (Th2) immune response, and immune regulation. 31 , 32
As the study progressed, it was established that macrophage polarization is on a continuum, and in addition to M1 and M2 macrophages, there exist other phenotypes of macrophages, such as CD169+ and T cell receptor positive (TCR+) macrophages. 33 CD169+ macrophages are a unique subpopulation of macrophages predominantly located in lymphoid organs such as lymph nodes and spleen. They express molecular markers such as CD169, CD206, and vascular cell adhesion molecule‐1 (VCAM‐1). CD169+ macrophages have the ability to release CCL22 and participate in erythropoiesis, immune tolerance, antigen presentation, and immune regulation. 33 , 34 , 35 TCR+ macrophages express TCRαβ or γδ and CD3. They also express molecules necessary for TCR signaling on lymphocytes, such as ZAP70 and Fyn, but the concentrations of these molecules are different when macrophages are stimulated by IL‐4 or IFN‐γ. TCR+ macrophages can release CCL2 and have a high phagocytosis capacity, playing a role in inflammation and infectious diseases. 33
In conclusion, macrophages are heterogeneous populations composed of multiple subpopulations with different phenotypes and functions, each with their own characteristics and functions. In vivo, macrophages can flexibly adopt specific functional phenotypes in response to subtle and continuous variations in the tissue microenvironment and then play an important role in development and homeostasis. 14
2.4. The role of macrophages in development and homeostasis
Macrophages play essential roles in all stages of development. 36 For instance, during embryogenesis, macrophages derived from yolk sac progenitors contribute to tissue structural reconstruction through the elimination of apoptotic cells. 4 , 36 The development and maintenance of the central nervous system (CNS) depend on the precise regulation of microglia. 37 , 38 In addition, macrophages also serve important functions in angiogenesis, 39 , 40 lymph angiogenesis, 41 mammary duct branching and pancreas islet formation, 42 , 43 adipogenesis, myocyte development and growth, and erythropoiesis. 3 , 8
Macrophages utilize their powerful biological functions to play diverse roles in tissue homeostasis maintenance as well. 4 , 7 , 8 Macrophages block the occurrence of tissue inflammation and injury by phagocytic clearance of debris, damaged cells, dead cells and apoptotic cells. 29 , 44 When tissue damage occurs, TRMs can regulate extracellular matrix formation to effectively repair the tissue and thus maintain the stable state of the tissue. 45 , 46 , 47 Macrophages also serve an essential immune function, as they can recognize invasive microbes or tumor cells, exert direct killing effects, 48 and transmit signals to other immune cells to respond. 49 , 50 Moreover, macrophages are involved in the tissue metabolism of calcium, 51 iron, 52 bilirubin, amino acids, and lipids, 53 regulating the stable level of these metabolites required in organs to ensure their normal function. 4
3. SIGNALING PATHWAYS IN MACROPHAGES
The diversity of macrophage functions depends on their plasticity in response to environmental stimuli. In response to diverse microenvironmental signals or under distinct pathophysiological conditions, macrophages can obtain different functional phenotypes through polarization. This process is regulated by various ligand–receptor recognitions through receptor‐mediated signaling transduction and downstream signaling pathways. The integration of different stimulus signals and the crosstalk and balance among signaling pathways ultimately determine the phenotype and function of macrophages. And these ligands, receptors, and signaling pathways have been shown to serve as targets for disease treatment. Thus, we then introduce these important signaling pathways mainly in a receptor‐centered way.
3.1. Pattern recognition receptor (PRR)‐associated signaling pathways
3.1.1. TLRs
TLRs are important PRRs expressed in macrophages 54 that regulate the survival, activation, and function of macrophages by two distinct signaling pathways, the myeloid differentiation primary‐response protein 88 (MyD88)‐dependent pathway and the TLR/IL‐1R (TIR) domain‐containing adaptor protein inducing IFN‐β (TRIF)‐dependent pathway (Figure 1). 54 , 55
FIGURE 1.

Activation of the NF‐κB signaling pathway in macrophages. Activation of NF‐κB in macrophages is mainly associated with pro‐inflammatory cytokines such as TNF‐α, or recognition of microbial products by TLRs. These ligand–receptor interactions converge on IKK activation through diverse upstream signaling pathways. The IKK complex, consisting of IKKα, IKKβ, and IKKγ, can degrade IκB and thereby activate NF‐κB. Activated NF‐κB mediates the expression of a series of pro‐inflammatory cytokine and chemokine genes. In addition, TLR4 can mediate type I interferon expression by activating IRF3 through TRIF, and TNF‐α mediates caspase‐3‐dependent apoptosis by activating FADD through TNFR1. Abbreviations: TLR, Toll‐like receptor; MyD88, myeloid differentiation primary‐response protein 88; TRIF, TIR domain‐containing adaptor protein inducing IFN‐β; IRAK, IL‐1R‐associated kinase; TRAF, tumor‐necrosis factor (TNF)‐receptor‐associated factor; TAK, transforming‐growth‐factor‐β‐activated kinase; TAB, TAK1‐binding protein 1; TBK, TRAF‐family‐member‐associated NF‐κB activator‐binding kinase 1; IKK, inhibitor of nuclear factor‐κB (IκB)‐kinase; SHP2, Src homology 2 (SH2) domain‐containg protein tyrosine phosphatase; RIP1, receptor‐interacting protein 1; IRF3, IFN‐regulatory factor 3; cIAP1/2, cellular inhibitors of apoptosis1/2; NIK, NF‐κB‐inducing kinase; TNF‐α, tumor necrosis factor‐α; TNFR, TNF receptor; TRADD, TNFR1‐associated death domain protein; FADD, Fas‐associated death domain; MAPK, mitogen‐activated protein kinases; IκB, inhibitor of nuclear factor‐κB; NF‐κB, nuclear factor kappa‐B. (Created with BioRender.com).
Upon binding to ligands, some TLRs, such as TLR2, TLR4, and TLR6, recruit MyD88, which binds to the IL‐1R‐associated kinase (IRAK) 4 complex, leading to the autophosphorylation of IRAK1. 54 Phosphorylated IRAK1 binds to TNF‐receptor‐associated factor (TRAF) 6, and then both detach from the receptor complex to form a new complex with TGF‐β‐activated kinase (TAK) 1, TAK1‐binding protein (TAB) 1 and TAB2 on the plasma membrane. Then, IRAK1 is degraded, while the rest of the complex is translocated to the cytoplasm, where TRAF6 interacts with ubiquitin‐conjugating enzyme 13 (UBC13) and UBC E2 variant 1 to activate TAK1. TAK1 activates mitogen‐activated protein kinase (MAPK) and then activator protein‐1 (AP‐1) to induce the transcription of IL‐1, IL‐6, TNF‐α, and other inflammatory factor genes. 56 TAK1 also phosphorylates the nuclear factor‐κB (IκB)‐kinase (IKK) complex to promote the nuclear translocation of nuclear factor kappa‐B (NF‐κB) and the expression of target genes. 56 , 57
TLR3 and TLR4 can also activate the TRIF‐dependent pathway. 54 TLR3/4 recruits TRIF, which binds to TRAF3/6. Then, TRAF3 recruits TBK1 and IKKε, which leads to the phosphorylation and nuclear translocation of IFN regulatory factor (IRF) 3, inducing the expression of type I IFN. 56 TRIF can also activate MAPK and NF‐κB signaling via UBC13/TAK1 by binding to TRAF6 and the serine/threonine kinase receptor‐interacting protein 1 (RIP1). 57 , 58
3.1.2. Nucleotide‐binding oligomerization domain‐like receptors (NLRs)
The NLR family includes 22 members of three subfamilies, NLR family C‐terminal caspase activation and recruitment domain (CARD) containing protein (NLRC), NLR thermal protein domain associated protein (NLRP), and ICE‐protease activating factor (IPAF), among which NLRP3 59 and NLRC3 60 play regulatory roles in macrophage polarization and the inflammatory response.
Upon stimulation by multiple pathogen‐associated molecular patterns or damage‐associated molecular patterns, NLRP3 oligomerizes and recruits apoptosis‐associated speck‐like protein containing a C‐terCARD (ASC) to form ASC speck through its PYD domain. 61 Then, ASC speck and pro‐caspase‐1 form the NLRP3 inflammasome via CARD–CARD interactions, promoting the maturation of caspase‐1 through pro‐caspase‐1 self‐cleaving. 62 Caspase‐1 cleaves pro‐IL‐1 and pro‐IL‐18 to form activated IL‐1β and IL‐18 secreted extracellularly to mediate the inflammatory response. 62 Caspase‐1 also releases the activity of GSDMDNterm by removing the carboxyl terminus of gasdermin D (GSDMD). 63 Then, GSDMDNterm binds to the cell membrane to form membrane pores, leading to cell death and the release of large amounts of proinflammatory factors. 64 This process, known as pyroptosis, is an inflammatory programmed cell death that plays a role in pathogen clearance by macrophages. 64 In addition, NLRP3 promotes NO production by inducible NO synthase (iNOS). 65
In contrast to NLRP3, NLRC3 inhibits macrophage activation and prevents inflammatory factor production by negatively regulating multiple inflammatory pathways. 60 For example, NLRC3 can inhibit the activation of TRAF6 and IRAK1 and promote their degradation, thus inhibiting the activation of NF‐κB signaling. 66 In addition, NLRC3 can disrupt the formation of inflammasomes by blocking the ASC and pro‐caspase‐1 interaction through the CARD domain. 67
3.1.3. Cyclic GMP‐AMP synthase (cGAS)‐stimulator of IFN genes (STING)
The cGAS‐STING signaling pathway regulates macrophage polarization and the production of various cytokines, which play a role in pathogen clearance, the inflammatory response, and antitumor immunity. 68 , 69
As an important PRR, cGAS is a direct sensor of DNA in host cells. cGAS can recognize and bind to cytoplasmic dsDNA to undergo conformational changes and activation and then catalyze the synthesis of cyclic GMP‐AMP (cGAMP) from intracellular adenosine triphosphate (ATP) and guanosine triphosphate (GTP). cGAMP acts as a second messenger to bind to STING, a linker protein anchored in the endoplasmic reticulum (ER), causing conformational changes and activation. 68 In addition, cyclic dinucleotides produced by bacteria 68 , 70 can directly activate STING. Subsequently, activated STING translocates from the ER to the Golgi, where it undergoes palmitoylation and activates TBK1 and IFN‐regulatory factor 3 to induce the strong expression of type I IFNs. 71 , 72 STING can also activate the NF‐κB signaling pathway through molecules such as TRAF6, 73 IKK, 74 TKB1, 75 and signal transducer and activator of transcription 6 (STAT6). 76
3.2. TNFR‐related signaling pathways
TNF‐α plays an important role in regulating macrophage proliferation, apoptosis, and inflammation by activating signaling pathways, including NF‐κB and MAPK, through binding to its receptor TNFR1/2 (Figure 1). 77 TNFR1 recruits TNFR1‐associated death domain protein (TRADD) through its death domain, which is isolated by the silence of death domain in the absence of TNF‐α. 78 TRADD attracts Fas‐associated death domain (FADD), TRAF2, and RIP1. 78 , 79 FADD mediates apoptosis by sequentially activating caspase‐8 and caspase‐3 through FADD‐like IL‐1β‐converting enzyme. TRAF2 binds ubiquitin ligases cellular inhibitors of apoptosis (cIAPs) 1/2, contributing to polyubiquitin chain assembly of diverse signaling molecules, including NF‐κB‐inducing kinase (NIK), RIP1, TRAF2, and cIAP itself. 80 , 81 TRAF2 can also activate IKK, thereby activating NF‐κB and promoting the expression of proinflammatory cytokines and other target genes. 77 In addition, TRAF2 also activates MAPK kinase kinase 1 (MEKK1), a MAP3K that activates c‐Jun kinases (JNKs) and p38 MAPK through MAP2K4/7, regulating macrophage inflammation and survival. 79 , 82 RIP1 not only acts as an adaptor to recruit IKK to TNFR1 by interacting with IKKγ but can also indirectly activate the IKKβ subunit of IKK by activating TAK1 through ubiquitination.
TNFR2 activates not only the classical NF‐κB and MAPK pathways mentioned above but also the alternative p52‐RelB NF‐κB signaling pathway. 83 TNFR2 recruits TRAF3 and cIPA1/2 through TRAF2, and TRAF3 binds to NIK and promotes NIK degradation. 81 cIPA1/2 mediates the ubiquitination and degradation of TRAF2 and TRAF3 to release NIK. 84 , 85 NIK phosphorylates IKKα to promote the phosphorylation and partial degradation of p100 to generate p52, which allows p52 to bind RelB, forming an NF‐κB dimer to mediate the transcription of target genes. 83 , 84 Degradation of TRAF3 also unblocks the TRAF2/6:MAP3K signaling complexes, leading to translocation of the complex from the receptor to the cytoplasm, where it initiates MAPK phosphorylation and cascade activation. 81
In addition, CD40 is also a member of the TNFR superfamily expressed on macrophages. Binding of CD40 to its ligand CD40L activates the NF‐κB and MAPK signaling pathways mainly through TRAFs (TRAF2/3/5/6), inducing stronger macrophage antigen‐presenting ability and the production of proinflammatory cytokines and chemokines. 82 , 86
3.3. IL receptor‐related signaling pathways
3.3.1. IL‐4/IL‐4R
IL‐4 is an important activator of macrophage M2 polarization and initiates the JAK‐STAT (Figure 2) and phosphatidylinositol 3‐kinase (PI3K)/protein kinase B (Akt) signaling pathways by binding to IL‐4R. 32 , 87 There are two types of IL‐4R: type I receptors consisting of the IL‐4R alpha chain (IL‐4Rα) and common gamma chain (γc) and type II receptors consisting of IL‐4Rα and IL‐13Rα1. Regardless of the type of receptor, these receptor subunits all interact with the Janus kinase (JAK) family. The JAK family consists of four isoforms: JAK1, JAK2, JAK3, and TYK2, all of which are nonreceptor tyrosine kinases. IL‐4Rα binds JAK1, while γc and IL‐13Rα1 bind JAK3 and TYK2, respectively. The interaction between IL‐4 and IL‐4R activates the STAT6 through JAK1/3. Then, activated STAT6 dissociates from IL‐4R to form a dimer with another STAT6 molecule through its SH2 domain. 87 , 88 Translocation of this dimer into the nucleus promotes transcription of M2‐related genes such as Mrc1, Arg1, and IL‐10. 88 In addition, IL‐4Rα phosphorylated tyrosine residues can also recruit and phosphorylate the insulin receptor substrate (IRS) family, specifically IRS2. Phosphorylated IRS attracts the p85 subunit of PI3K, leading to the activation of PI3K/Akt/mammalian target of rapamycin (mTOR) signaling. 89 Moreover, IRS interacts with growth factor receptor‐bound protein 2 (GRB2) to activate son of sevenless (SOS), which results in MAPK pathway activation. 90
FIGURE 2.

Activation of the JAK‐STAT signaling pathway in macrophages. The JAK‐STAT axis is one of the important pathways that regulate macrophage polarization and function. Various cytokines such as IFN‐γ, GM‐CSF, IL‐4/13, IL‐6, and IL‐10 can utilize specific combinations of JAK and STAT members to activate the formation of STAT homodimers or heterodimers, thereby affecting gene transcription. STAT homodimers can directly bind to GAS to promote the transcription of M1/M2‐related genes, while heterodimers such as STAT1/STAT2 bind to IRF9 and then to ISRE to promote M1‐related genes transcription. Abbreviations: IFN, interferon; GM‐CSF, granulocyte‐macrophage colony‐stimulating factor; IL, interleukin; JAK, Janus kinase; STAT, signal transducer and activator of transcription; ISRE, IFN‐stimulated response elements; GAS, IFN‐γ‐activated sequences. (Created with BioRender.com).
3.3.2. IL‐6/IL‐6R
The receptor complex formed by IL‐6, IL‐6R, and gp130 activates intracellular signaling cascades, including the JAK1‐STAT3 (Figure 2) and MAPK signaling pathways, to promote M2 macrophage polarization. 91 In addition, IL‐6 signaling enhances the macrophage response to IL‐4. gp130‐bound JAKs, especially JAK1, are activated after receptor complex formation. Then, activated JAK1 phosphorylates different tyrosine residues in the intracellular segment of the receptor and recruits STAT3, which is also phosphorylated by JAK1. STAT3 forms a homodimer and translocates to the nucleus to activate the transcription of M2‐related genes. Phosphorylated receptor tyrosine residues also activate GRB2 to trigger the Ras/Raf/MAPK signaling cascade.
3.3.3. IL‐10/IL‐10R
IL‐10 is a critical anti‐inflammatory cytokine and is involved in promoting M2 polarization of macrophages. IL‐10R consists of two subunits, IL‐10Rα and IL‐10Rβ, which interact with JAK1 and TYK2, respectively. In the presence of IL‐10, IL‐10R activates JAK1 and TYK2 to promote STAT3 phosphorylation. 92 Activated STAT3 dimerizes and translocates to the nucleus to promote M2‐related gene expression and inhibit IL‐6, IL‐8, and TNF‐α production. 92 , 93
3.4. IFN receptor‐related signaling pathways
Recognition of IFN and IFN receptor promotes M1 macrophage polarization by activating the JAK‐STAT signaling pathway (Figure 2). Although type I IFN and IFN‐γ bind to different receptors, these receptors all interact with members of the JAK family. The binding of IFN‐γ causes dimerization and activation of IFN‐γR1 and IFN‐γR2, which leads to autophosphorylation of JAK 1 and JAK2. 94 Activated JAK2 phosphorylates IFN‐γR to form a docking site for STAT1, causing STAT1 recruitment and phosphorylation. Phosphorylated STAT1 dissociates from IFN‐γR and undergoes homodimerization and nuclear translocation to regulate IFN‐stimulated genes (ISGs) expression by binding to IFN‐γ‐activated sequences. 95 In addition, type I IFNs, such as IFN‐α and IFN‐β, promote the formation of not only STAT homodimers but also heterodimers, such as STAT1‐STAT2. STAT1‐STAT2 binds to IRF9 to form a triplet complex called ISG factor 3 (ISGF3). 96 ISGF3 regulates the expression of ISGs by binding to ISRE to promote the M1 polarization of macrophages. 95 , 96
3.5. CSF‐related signaling pathways
3.5.1. CSF‐1/CSF‐1R
Signaling pathways mediated by CSF‐1/CSF‐1R regulate macrophage survival, proliferation, chemotaxis, and differentiation. 21 , 97 The binding of CSF‐1 to its receptor CSF‐1R rapidly triggers CSF‐1R dimerization and phosphorylation of multiple intracellular tyrosine sites, 98 , 99 allowing CSF‐1R to phosphorylate molecules such as PI3K, Src, and GRB2. 100 PI3K is an essential downstream signaling molecule of CSF‐1R (Figure 3). 101 Activated PI3K induces phosphorylation of phosphati‐dylinositol‐4,5‐bisphosphate (PIP2) to form phosphati‐dylinositol‐3,4,5‐triphosphate, which not only recruits and activates Akt through the pleckstrin homology domain but also enhances Akt activation by phosphorylating mammalian target‐of‐rapamycin complex 2 (mTORC2). 102 Akt regulates macrophage proliferation, polarization, and metabolism through various downstream effectors. 102 For example, Akt inhibits forkhead box protein O1 (FoxO1), a transcription factor that promotes the expression of proinflammatory and proliferation‐ and survival‐related genes in M1 macrophages. 103 The phosphorylation of glycogen synthase kinase‐3β (GSK‐3β) by Akt can promote the M2 polarization of macrophages. 104 CCAAT/enhancer binding protein β (C/EBPβ) is involved in macrophage activation, whose expression is controlled by Akt2. 105 mTORC1 is also an important downstream signaling molecule of Akt. 106 mTORC1 regulates the synthesis of various proteins in macrophages by activating S6K and 4EBP1, thus regulating the metabolism and function of macrophages. 102 , 107 In addition, CSF‐1 regulates macrophage adhesion and motility by activating Rho, Rac, and Cdc, which are central regulators of the actin cytoskeleton and adhesion structures, through at least two independent pathways (the Src/Pyk2 and PI3K pathways). 97 In addition, phosphorylation of CSF‐1R Y697 activates GRB2 and suppressor of cytokine signaling 1 (SOCS1), while Y706 phosphorylation is required for complete STAT1 activation. 98 GRB2 is involved in the activation of the Ras/Raf/mitogen‐activated extracellular signal‐regulated kinase (MEK)/extracellular signal‐regulated kinase (ERK) pathway. 21 , 97 SOCS1 is an important negative feedback regulator of signal transduction that inhibits JAK‐STAT1 and NF‐κB pathways activation in macrophages. 108
FIGURE 3.

Activation of PI3K/Akt and Notch signaling pathways in macrophages. When bound to CSF‐1, phosphorylation sites in intracellular segment of CSF‐1R activate PI3K to initiate PI3K/Akt signaling pathway. Akt regulates the proliferation, polarization, and survival of macrophages by activating or inhibiting transcription factors such as FoxO1, GSK‐3β, and C/EBPβ. PTEN and lactic acid are negative regulators of this pathway, while glycine and ROS promoted the signaling response by inhibiting PTEN. The binding of Notch to its ligand Dll4 results in the release of Notch intracellular domain into the nucleus and the formation of transcription activation complex to promote the expression of M1‐related genes in macrophages. In addition, Jagged1‐Notch axis promotes M2‐related genes expression in tumor‐associated macrophages. Abbreviations: CSF‐1, colony‐stimulating factor‐1; CSF‐1R, colony‐stimulating factor‐1 receptor; PI3K, phosphatidylinositol 3‐kinase; SFK, Src family tyrosine kinases; RSK, ribosome protein subunit 6 kinase; PTEN, phosphatase and tensin homolog deleted on chromosome ten; PIP2, phosphati‐dylinositol‐4,5‐bisphosphate; PIP3, phosphati‐dylinositol‐3,4,5‐triphosphate; PKD, protein kinase D; mTORC, mammalian target‐of‐rapamycin complex; TSC, tuberous sclerosis complex; FoxO1, forkhead box protein O1; GSK‐3β, glycogen synthase kinase‐3β; C/EBPβ, CCAAT/enhancer binding protein β; Dll4, Delta‐like 4; NICD, Notch intracellular domain; MAML1, mastermind‐like transcriptional coactivator 1; RBPJ, recombination signal binding protein for immunoglobulin κJ region. (Created with BioRender.com).
3.5.2. GM‐CSF)/GM‐CSF and its receptor (GM‐CSFR)
The interaction between GM‐CSF and its receptor (GM‐CSFR) triggers M1 macrophage polarization and proinflammatory mediator production mainly through the JAK2‐STAT5 signaling pathway. GM‐CSF binds to its receptors GM‐CSFRα and GM‐CSFRβ to form a dodecamer ligand‒receptor complex that promotes STAT5 phosphorylation and homodimerization via JAK2. 109 Then, STAT5 dimers promote the transcription of M1‐related genes and the production of inflammatory mediators such as TNF‐α, IL‐6, IL‐12, and IL‐23. 110 In addition, GM‐CSF also activates the macrophage NF‐κB and PI3K/Akt and Ras/MEK/ERK signaling pathways. 110 , 111
3.6. Growth factor‐related signaling pathways
3.6.1. TGF‐β/TGF‐β receptor (TβR)
TGF‐β regulates macrophage polarization and function through the TβR in Sma‐ and Mad‐related protein (SMAD)‐dependent or SMAD‐independent signal transduction pathways (Figure 4). 112 When TGF‐β drives the formation of a heterotetrameric complex consisting of two TβRI and two TβRII, TβRII phosphorylates the serine or threonine residues of TβRI, thereby phosphorylating SMAD2/3 to initiate the SMAD signaling cascade. 112 Activated SMAD2/3 separate from TβR and then interact with SMAD4, moving into the nucleus to promote the expression of M2‐related genes. In addition, TGF‐β/SMAD6 induces polyubiquitination and degradation of MyD88 by recruiting the E3 ubiquitin ligases Smurf 1/2 to inhibit TLR/NF‐κB. 113 SMAD6 also segregates Pellino‐1, which is required for the IL‐1R/TLR proinflammatory response. 114 TGF‐β/SMAD7 affects TNF signaling by blocking TNF‐induced TAK1 activity. 115 Moreover, TGF‐β can phosphorylate PI3K, 116 ERK, 117 , 118 independent of SMADs, which is mainly achieved by TβR‐induced ubiquitination of TRAF6.
FIGURE 4.

Activation of MAPK and TGF‐β‐SMAD signaling pathways in macrophages. MAPK signaling pathway can be activated by molecules such as TGF‐β and EGF. The MAPK signal cascade consists of three kinds of protein kinases (MAP3K, MAP2K, and MAPK), which transmit upstream signals to downstream response molecules through sequential phosphorylation. Raf and MEKKs belong to MAP3K; MEKs belong to MAP2K; JNK, ERKs and p38 belong to MAPK. AP‐1, a heterodimer composed of c‐Fos and c‐Jun. TGF‐β activates SMAD4 via SMAD2/3 to regulate the expression of M2 polarization‐related genes. Abbreviations: EGF, epidermal growth factor; SHC, Src homolog and collagen homolog; GRB2, growth factor receptor‐bound protein 2; SOS, son of sevenless; MAP3K, mitogen‐activated protein kinase kinase kinase; MEK, mitogen‐activated extracellular signal‐regulated kinase, MAP2K, mitogen‐activated protein kinase kinase; JNK, c‐Jun kinase; ERK, extracellular signal‐regulated kinase; MAPK, mitogen‐activated protein kinase; ATF2, activating transcription factor 2; TGF‐β, transforming growth factor‐β; TβR, TGF‐β receptor; SMAD, Sma‐ and Mad‐related protein; SARA, the SMAD anchor for receptor activation; Smurf2, SMAD‐specific E3 ubiquitin protein ligase 2; RhoA, Ras homolog family member A. (Created with BioRender.com).
3.6.2. Epidermal growth factor (EGF)/EGF and its receptor (EGFR)
EGF plays an important role in macrophage activation through MAPK signaling pathways (Figure 4). 119 , 120 The binding of EGFR causes EGFR dimerization, which leads to C‐terminal tail autophosphorylation of EGFR, and the phosphorylation region acts as an anchor for GRB2. 121 Then, GRB2 recruits and binds to SOS to trigger the Ras/Raf/MAPK signaling pathway. 121 Activation of the MAPK cascade results in the transcription of many cytokine genes. 119 , 120 Phosphorylated EGFR also causes tyrosine phosphorylation of PI3K to activate the PI3K/Akt/mTOR signaling pathway. 121
3.7. G‐protein‐coupled receptor (GPCR) signaling pathway
GPCRs are a large family of receptors that can bind to various ligands, including chemokines, hormones, ions, neurotransmitters, and odorant molecules. 122 , 123 Macrophages express many GPCRs, such as the chemokine receptor Frizzled (Fzd), which plays a key role in regulating the differentiation and function of macrophages. 124 Here, we mainly introduce C‐C chemokine receptor (CCR) type 2 (CCR2)/CCR5‐ and Fzd‐mediated signal transduction in macrophages (the regulation of other GPCRs on macrophages has been detailed in the review 124 ).
The binding of CCL2/CCL5 to its receptor CCR2/CCR5 promotes macrophage M2 polarization through the GPCR signaling pathway. When bound to its ligand, the GPCR undergoes conformational changes that cause the dissociation of G protein into Gα and Gβγ. Then, Gα and Gβγ activate their downstream signaling molecules and generate different intracellular signal transduction pathways (Figure 5). 122 Gα activates adenylyl cyclase to convert ATP to cyclic adenosine monophosphate (cAMP), 125 which activates protein kinase A (PKA). Activated PKA promotes the phosphorylation of cAMP‐responsive element binding protein (CREB), 126 and CREB promotes the expression of anti‐inflammatory cytokines and M2‐related genes. PKA also activates ERK1/2 and STAT3 but inhibits NF‐κB and STAT1. 127 Gα activates phospholipase C (PLC) to catalyze the production of inositol triphosphate (IP3) and diacylglycerol from PIP2 as well. The binding of IP3 to calcium channel receptors on the ER membrane leads to ER calcium efflux, increasing cytoplasmic Ca2+, which activates various protein kinases, including protein kinase C (PKC) and calmodulin‐dependent protein kinase type II (CaMKII), to regulate macrophage activation and function. 124
FIGURE 5.

Activation of GPCR signaling pathway in macrophages. Chemokines such as CCL2/CCL5, and lactic acid, are signaling molecules that bind to GPCRs causing conformational changes that activate the dissociation of G proteins into Gα and Gβγ. Gα activates PKA by promoting cAMP production, inducing CREB phosphorylation and nuclear translocation thereby promoting anti‐inflammatory cytokine production and macrophage M2 polarization. PKA also inhibits STAT1 phosphorylation and promotes STAT3 phosphorylation contributing to M2 macrophage polarization. Wnt combines with its receptor Fzd 1 to activate downstream signal transduction and further regulate macrophage polarization and function. Wnt2b and Wnt3a inhibit the destruction complex formed by APC, Axin, βTrCP and GSK‐3β through Dvl, leading to the accumulation of β‐catenin and its translocation to the nucleus where it binds to TCF/LEF to mediate anti‐inflammatory and M2‐related gene expression. In contrast, Wnt5a binding to Fzd5 activates the phagocytic activity and pro‐inflammatory response of macrophages through Dvl activation of RhoA, Rac1/Cdc42, and PLC. Abbreviations: GPCR, G‐protein‐coupled receptors; AC, adenylate cyclase; cAMP, cyclic adenosine monophosphate; PKA, protein kinase A; CREB, cAMP‐response element binding protein; Wnt, wingless homolog; Fzd, Frizzled; LRP, LDL receptor‐related protein; Dvl, dishevelled; PLC, phospholipase C; DAG, diacylglycerol; IP3, inositol triphosphate; CaMKII, calmodulin‐dependent protein kinase type II; NFAT, nuclear factor of activated T cells; ROCK, Rho‐associated kinase; CK‐1, casein kinase 1; TCF, T‐cell factor; LEF, lymphoid enhancer‐binding factor. (Created with BioRender.com).
Unlike CCR2/CCR5, Fzd1 is a member of another family of GPCRs. When combined with its ligand wingless homolog (Wnt), Fzd1 activates the Wnt/β‐catenin, Wnt/planar cell polarity (PCP), and Wnt/Ca2+ pathways (Figure 5) to regulate macrophage polarization and function, and different Wnt isoforms can contribute to M2 or M1 polarization of macrophages. 128 Some Wnt molecules, such as Wnt2b and Wnt3a, can mediate the anti‐inflammatory response and M2 polarization of macrophages through the Wnt/β‐catenin signaling pathway. 129 , 130 In the absence of Wnt signaling, cytosolic β‐catenin is phosphorylated and then degraded by the destruction complex (DC), which is formed by APC, Axin, β‐transducin repeats‐containing protein (βTrCP), and GSK‐3β. Binding of Wnt3a to Fzd1 blocks the effect of DC on β‐catenin through dishevelled (Dvl), thereby leading to the accumulation of β‐catenin in the cytoplasm and translocation to the nucleus, where it binds to T‐Cell Factor/Lymphoid Enhancer‐Binding Factor to promote the transcription of downstream target genes such as c‐myc, c‐jun, peroxisome proliferator activated receptor (PPAR) δ, and cyclin D1. 128 Moreover, the Wnt/β‐catenin pathway can antagonize the Notch pathway 131 and restrain NF‐κB signaling activation by inhibiting GSK‐3β activity. 129 , 132
The interaction between Wnt5a and Fzd5 activates Wnt/PCP and Wnt/Ca2+, thereby stimulating the phagocytic activity of macrophages and the secretion of proinflammatory cytokines. 129 , 133 Fzd5 activates RhoA and Rac1/Cdc42, which activate Rho‐associated kinase (ROCK) and JNK, respectively, via Dvl. ROCK and JNK translocate into the nucleus to bind to c‐jun and Activating Transcription Factor 2 to regulate gene expression and promote proinflammatory factor production. 128 Dvl also activates PLC, promoting the accumulation of Ca2+ in the cytoplasm, which induces the expression of proinflammatory cytokines by activating CaMKII. 134
3.8. Notch signaling pathway
The Notch signaling pathway is involved in regulating macrophage polarization and function. 135 To date, four NOTCH receptors (Notch1–4) and five NOTCH ligands (Jagged1, Jagged2, Delta‐like [Dll] 1, Dll3, and Dll4) have been identified. 136 Dll4‐Notch can promote the inflammatory response and M1 polarization of macrophages, 137 , 138 while the Jagged1‐Notch axis promotes M2 macropahge polarization. 139 The Notch receptor is composed of Notch extracellular domain, Notch intracellular domain (NICD), and transmembrane domain. 136 When binding to Dll4 on the surface of adjacent cells, Notch proteolysis occurs twice to release NICD into the nucleus, which binds to the nuclear transcription factor CSL. 137 , 140 In the absence of Notch signaling, CSL binds specifically to co‐inhibitory receptors, histone deacetylase, and DNA sequences, forming a CSL‐DNA binding protein complex to silence the expression of downstream genes. 141 When CSL binds to NCID, it separates from repressor molecules and recruits transcription coactivators to form a transcription activation complex, resulting in the promotion of M1‐related gene expression (Figure 3). 140
3.9. Triggering receptor expressed on myeloid cells (TREM) signaling pathway
The TREM is a class of innate immune receptors that plays a key role in regulating the inflammatory response by amplifying or inhibiting TLR‐induced signals. 142 TREM exists in two forms, membrane receptor TREM and soluble TREM (sTREM), and sTREM negatively regulates membrane TREM signaling by competitively neutralizing ligands. 142
TREM‐1 is an activating receptor expressed on monocytes and macrophages that effectively amplifies the inflammatory response by inducing inflammatory mediator secretion and cooperating with PRR‐mediated signaling pathways. 143 Since TREM‐1 does not contain any signaling motif, it binds to DAP12, an adaptor protein with an ITAM to mediate intracellular signaling. 143 The ITAMs of DAP12 undergo tyrosine residue phosphorylation, providing a docking site for SYK. SYK activates multiple signal transduction pathways, including Ras/ERK and NF‐κB, thereby promoting the expression of proinflammatory cytokines. 144 , 145 TREM‐1 also prolongs macrophage survival by upregulating the Egr2‐dependent antiapoptotic protein Bcl‐2 via ERK and increasing mitofusin 1/2, which maintains mitochondrial integrity. 146 Moreover, TREM‐1 enhances the activity of molecules including MyD88, IκBα, and NF‐κB. 147 , 148
TREM‐2 is an immunonegative regulator that inhibits macrophage activation and the inflammatory response. 149 The low‐avidity interaction between TREM‐2 and DAP12 results in incomplete phosphorylation of DAP12, which fails to recruit and activate SYK but leads to preferential activation of the inhibitory phosphatase SHP1, thereby inhibiting TLR‐mediated activation. 144 In addition, DAP12‐activated PI3K/Akt blocks MAP3K, and DAP12‐activated PLCγ may reduce PIP2, a TIRAP docking site that is required for MyD88 recruitment to the TLR signaling complex. 144
3.10. Immunosuppressive receptor‐related signaling pathways
Signal regulatory protein α (SIRPα), a myeloid inhibitory receptor containing immunoreceptor tyrosine‐based inhibitory motifs (ITIMs), inhibits macrophage phagocytosis and proinflammatory cytokine production by interacting with its cell surface ligand CD47. 150 When SIRPα binds CD47, ITIMs of SIRPα are tyrosine phosphorylated by SFK to bind and inhibit the function of SHP1 and SHP2. Then, SHP1 and SHP2 are engaged in various signaling pathways, such as the GRB2/Ras/Raf/MAPK, JAK‐STAT and PI3K pathways. 151 , 152
Similar to SIRPα, LILRB, 153 PD‐1, 154 and Siglec‐10 155 are also important immunosuppressive receptors expressed on macrophages, all of which contain ITIMs. 156 When these receptors bind to their ligands MHC I, programmed death‐ligand 1 (PD‐L1), and CD24, they inhibit the activation of Ras/ERK and NF‐κB through ITIMs, thereby inhibiting the phagocytosis and activation of macrophages. 153 , 154 , 155
3.11. Redox signaling pathways
ROS are composed of free radicals and non‐free radicals formed by partial reduction of oxygen, including superoxide anion, hydrogen peroxide, and hydroxyl radical. 157 One of the most important characteristics of ROS is that they can snatch electrons from others and are extremely susceptible to oxidation reactions with others. ROS are not only important mediators produced by macrophages to clear pathogens and participate in innate immunity but also have the ability to participate in signaling pathways as second messengers to regulate the phenotype and function of macrophages. 158 , 159 ROS promote the proinflammatory response and M1 polarization of macrophages by promoting the NF‐κB and MAPK signaling cascades. ROS induce thioredoxin, an inhibitor of apoptotic signaling regulated kinase (ASK1), to dissociate from the trx‐ASK1 complex, resulting in phosphorylation of ASK1 and activation of downstream p38. 160 ROS also indirectly maintain the activity of this cascade by inactivating JNK‐inhibiting phosphatase. 161 Moreover, ROS can trigger NLRP3 activation to regulate macrophage pyroptosis. 162 In addition, ROS enhance the phosphorylation of IκB to activate NF‐κB 163 and participate in the activation of STAT1 and IRF5. 164 However, ROS are also involved in regulating M2 macrophage polarization under certain circumstances. For instance, in the presence of M‐CSF, ROS are involved in the late activation of ERK signaling required for M2‐like polarization of macrophages in cancer, 165 and in alveolar macrophages, ROS directly oxidize specific cysteine residues of STAT6 to promote transcription of the M2‐related genes Arg 1 and FIZZ1. 166 ROS can also maintain the activation of the PI3K/Akt pathway by inhibiting PTEN. 167
3.12. Lactic acid signaling pathways
Lactate drives macrophage polarization to anti‐inflammatory, pro‐repair phenotypes through regulation of multiple signaling pathways. 168 , 169 First, lactate inhibits proinflammatory signaling through GPCRs. 170 Lactate specifically inhibits the activation of inflammasome and TLR4‐mediated NF‐κB by activating GPR81 on macrophages. 170 Lactic acidosis mediates cAMP‐dependent signaling by activating plasma membrane GPCRs, inducing the expression of M2 genes and the transcriptional repressor cAMP‐responsive element modulator (CREM/ICER). CREM inhibits the expression of inflammatory genes, including TNF and NOS2. 171 , 172 Second, lactate not only stabilizes HIF1α 173 but also decreases ATP6V0d2 expression by activating mTORC1. ATP6V0d2 is involved in the formation of vacuolar proton pumps that promote lysosomal acidification and protein degradation. Reduced expression of ATP6V0D2 leads to a reduction in the degradation of HIF2α by lysosomes, which induces high expression of VEGF and M2 homeostatic genes. 174 Third, lactate inhibits RIG‐I‐MAVS‐mediated type I IFN production by binding to MAVS, thereby suppressing proinflammatory responses. In addition, lactate inhibits the excessive activation of the NLRP3 inflammasome and caspase‐1 to suppress macrophage pyroptosis and the inflammatory response. 175
3.13. Glycinergic signaling pathways
Glycine can shape macrophage polarization and function by regulating signaling pathways such as NF‐κB and PI3K/Akt. 176 First, glycine inhibits the activation of IKK by inhibiting the phosphorylation of IKKα and IKKβ and the degradation of IκB in proinflammatory macrophages, 177 thereby inhibiting NF‐κB activation and M1 polarization. Second, glycine restrains NLRP3 expression and inflammasome assembly in M1 macrophages through upregulation of the Nrf‐2/HO‐1 axis, 178 inhibiting the inflammatory response and pyroptosis. Finally, glycine could block PTEN to upregulate the activation of Akt in microglia, which inhibits the activation of NF‐κB and HIF1α. 179
4. THE ROLE OF MACROPHAGES IN DISEASES AND TARGETED THERAPIES
Since macrophages are important contributors to development, tissue homeostasis, immunity, and repair, macrophage dysfunction is involved in the initiation and development of many diseases, such as autoimmune, neurodegenerative, metabolic, and infectious diseases and cancer. It is becoming increasingly clear that the flexible and precise transformation of the functional phenotypes of macrophages plays important roles in the progression or remission of diseases. Therapies targeting components and regulators of signaling pathways in macrophages hold great potential and are a prospective direction for the treatment of these diseases (Table 1; Figure 6).
TABLE 1.
Drug candidates in clinic/clinical trials for signaling pathway targeted therapy of macrophage in human diseases.
| Therapeutic target | Agent | Mechanism of action | Application in disease | Drug status | Ref. |
|---|---|---|---|---|---|
| Drugs targeting ligand/receptor on macrophage | |||||
| TNF‐α | Infliximab | TNF‐α inhibitor | RA, IBD | Marketed | 362, 363 |
| COVID‐19 | Clinical trial Phase IV | 364 | |||
| Adalimumab | TNF‐α inhibitor | RA, IBD | Marketed | 362, 363 | |
| COVID‐19 | Clinical trial Phase III | 365 | |||
| Golimumab | TNF‐α inhibitor | RA, IBD | Marketed | 362, 363 | |
| Certolizumab pegol | TNF‐α inhibitor | RA, IBD | Marketed | 362, 363 | |
| Etanercept | Soluble recombinant TNF receptor fusion protein | RA | Marketed | 362 | |
| AD | Clinical trial Phase II | 222 | |||
| Tuberculosis | Clinical trial Phase IV | 297 | |||
| IL‐6 | Siltuximab | IL‐6 inhibitor | RA | Marketed | 366 |
| COVID‐19 | Clinical trial Phase III | 367 | |||
| Sirukumab | IL‐6 inhibitor | RA | Clinical trial Phase III | 368 | |
| COVID‐19 | Clinical trial Phase II | 306 | |||
| Olokizumab | IL‐6 inhibitor | RA | Clinical trial Phase III | 369 | |
| COVID‐19 | Clinical trial Phase III | 306 | |||
| Clazakizumab | IL‐6 inhibitor | RA | Clinical trial Phase II | 370 | |
| COVID‐19 | Clinical trial Phase II | 371 | |||
| Tocilizumab | IL‐6R inhibitor | RA | Marketed | 366 | |
| ALS | Clinical trial Phase II | 250 | |||
| AS | Clinical trial Phase III | 286 | |||
| COVID‐19 | Marketed | 372 | |||
| Salirumab | IL‐6R inhibitor | RA | Clinical trial Phase II | 373 | |
| Levilimab | IL‐6R inhibitor | COVID‐19 | Clinical trial Phase III | 374 | |
| IL‐1 | Anakinra | IL‐1R antagonist | RA | Marketed | 261 |
| ALS | Clinical trial Phase II | 252 | |||
| T2DM | Clinical trial Phase IV | 261 | |||
| AS | Clinical trial Phase IV | 286 | |||
| COVID‐19 | Clinical trial Phase III | 375 | |||
| Canakinumab | IL‐1β antagonist | RA | Clinical trial Phase IV | 376 | |
| T2DM | Clinical trial Phase III | 377 | |||
| AS | Clinical trial Phase III | 284, 285 | |||
| COVID‐19 | Clinical trial Phase IV | 378 | |||
| MABp1 | IL‐1α antagonist | T2DM | Clinical trial Phase II | 262 | |
| IL‐12/23 | Ustekinumab | Antagonist of the p40 subunit of IL‐12 and IL‐23 | RA | Clinical trial Phase II | 379 |
| IBD | Marketed | 363 | |||
| Risankizumab | Antagonist of the p19 subunit of IL‐23 | IBD | Marketed | 363 | |
| COVID‐19 | Clinical trial Phase II | 306 | |||
| Brazikumab | Antagonist of the p19 subunit of IL‐23 | IBD | Clinical trial Phase III | 363 | |
| Mirikizumab | Antagonist of the p19 subunit of IL‐23 | IBD | Clinical trial Phase III | 380 | |
| Guselkumab | Antagonist of the p19 subunit of IL‐23 | RA | Clinical trial Phase II | 379 | |
| IBD | Clinical trial Phase III | 381 | |||
| GM‐CSF | KB003 | GM‐CSF inhibitor | RA | Clinical trial Phase II | 190 |
| MOR103 | GM‐CSF inhibitor | RA | Clinical trial Phase II | 382 | |
| MS | Clinical trial Phase I/II | 206 | |||
| MORAb‐022 | GM‐CSF inhibitor | RA | Clinical trial Phase I | 190 | |
| Namilumab | GM‐CSF inhibitor | RA | Clinical trial Phase II | 383 | |
| Gimsilumab | GM‐CSF inhibitor | COVID‐19 | Clinical trial Phase II | 384 | |
| Otilimab | GM‐CSF inhibitor | COVID‐19 | Clinical trial Phase II | 385 | |
| Lenzilumab | GM‐CSF inhibitor | COVID‐19 | Clinical trial Phase III | 386 | |
| TJ003234 | GM‐CSF inhibitor | COVID‐19 | Clinical trial Phase II/III | 306 | |
| Mavrilimumab | GM‐CSFR inhibitor | RA | Clinical trial Phase II | 387 | |
| COVID‐19 | Clinical trial Phase III | 306 | |||
| CSF‐1 | Masitinib | CSF‐1R inhibitor | MS | Clinical trial Phase III | 249 |
| ALS | Clinical trial Phase III | 248 | |||
|
Emactuzumab (RG7155) |
CSF‐1R inhibitor | Enosynovial giant cell tumor | Clinical trial Phase III | 327 | |
|
Pexidartinib (PLX3397) |
CSF‐1R inhibitor | Enosynovial giant cell tumor | Marketed | 329 | |
| CCR/CCL | Cenicriviroc | CCR2/CCR5 inhibitor | NAFLD | Clinical trial Phase III | 271 |
| Leronlimab | CCR5 inhibitor | COVID‐19 | Clinical trial Phase III | 388 | |
| PF‐04136309 | CCR2 inhibitor | Pancreatic cancer | Clinical trial Phase II | 315 | |
| Carlumab | CCL2 inhibitor | Castration‐resistant prostate cancer | Clinical trial Phase II | 316 | |
| Maraviroc | CCR5 inhibitor | Colorectal cancer | Clinical trial Phase I | 317 | |
| Vicriviroc | CCR5 inhibitor | Colorectal Cancer | Clinical trial Phase II | 317 | |
| CXCR/ CXCL | Plerixafor | CXCR4 inhibitor | Glioblastoma | Clinical trial Phase I/II | 321 |
| Motixafortide | CXCR4 inhibitor | Pancreatic ductal adenocarcinoma | Clinical trial Phase II | 389 | |
| TLR | Eritoran | TLR4 antagonist | COVID‐19 | Clinical trial Phase III | 390 |
| PUL‐042 | Synthetic ligands for TLR 2/6/9 | COVID‐19 | Clinical trial Phase II | 306 | |
| Famotidine | Inhibit TLR3 | COVID‐19 | Clinical trial Phase IV | 391 | |
| PPAR | Pioglitazone | PPARγ agonist | AD | Clinical trial Phase III | 392 |
| T2DM | Marketed | 393 | |||
| NAFLD | Clinical trial Phase IV | 394 | |||
| COVID‐19 | Clinical trial Phase IV | 306 | |||
| Rosiglitazone | PPARγ agonist | AD | Clinical trial Phase III | 395 | |
| T2DM | Marketed | 396 | |||
| NAFLD | Clinical trial Phase IV | 394 | |||
| TZDs | PPARγ agonist | T2DM | Marketed | 396 | |
| NAFLD | Clinical trial Phase IV | 394 | |||
| COVID‐19 | Clinical trial Phase IV | 306 | |||
| Saroglitazar | PPARα/PPARγ agonist | NAFLD | Clinical trial Phase IV | 306 | |
| Aleglitazar | PPARα/PPARγ agonist | NAFLD | Clinical trial Phase III | 280 | |
| Elafibranor | PPARα/PPARδ agonist | NAFLD | Clinical trial Phase III | 281 | |
| Lanifibranor | Pan‐PPAR agonist | NAFLD | Clinical trial Phase III | 282 | |
| GLP‐1R | Exenatide | GLP‐1R agonist | T2DM | Marketed | 394, 397 |
| NAFLD | Clinical trial Phase IV | 394 | |||
| Liraglutide | GLP‐1R agonist | T2DM | Marketed | 397 | |
| NAFLD | Clinical trial Phase IV | 394 | |||
| Lixisenatide | GLP‐1R agonist | T2DM | Marketed | 397 | |
|
Extended‐ release exenatide |
GLP‐1R agonist | T2DM | Marketed | 397 | |
| Albiglutide | GLP‐1R agonist | T2DM | Clinical trial Phase IV | 397 | |
| Dulaglutide | GLP‐1R agonist | T2DM | Marketed | 397 | |
| NAFLD | Clinical trial Phase IV | 394 | |||
| Semaglutide | GLP‐1R agonist | T2DM | Marketed | 397 | |
| NAFLD | Clinical trial Phase IV | 394 | |||
| S1PR | Fingolimod | S1PRs (1, 3, 4, 5) agonist | MS | Marketed | 398 |
| ALS | Clinical trial Phase II | 251 | |||
| COVID‐19 | Clinical trial Phase II | 399 | |||
| Ozanimod | S1PRs (1, 5) agonist | COVID‐19 | Marketed | 400 | |
| SIRPα/CD47 | Hu5F9‐G4 | CD47 inhibitor | Non‐Hodgkin lymphoma, colorectal neoplasms | Clinical trial Phase I/II | 335 |
| CC‐90002 | CD47 inhibitor | Acute myeloid leukemia | Clinical trial Phase I | 336 | |
| FXR | Obeticholic acid | FXR agonist | NAFLD | Marketed | 274, 275 |
| Drugs targeting signaling pathway in macrophage | |||||
| FcR pathway | Spebrutinib | BTK inhibitor | RA | Clinical trial Phase II | 192 |
| Fenebrutinib | BTK inhibitor | RA | Clinical trial Phase III | 193 | |
| Tolebrutinib | BTK inhibitor | MS | Clinical trial Phase III | 208 | |
| Evobrutinib | BTK inhibitor | MS | Clinical trial Phase III | 401 | |
| Ibrutinib | BTK inhibitor | COVID‐19 | Clinical trial Phase II | 306 | |
| Acalabrutinib | BTK inhibitor | COVID‐19 | Clinical trial Phase III | 306 | |
| NF‐κB pathway | NSAIDs | Inhibit NF‐κB | RA, IBD, COVID‐19 | Marketed | 306, 363, 402 |
| Corticosteroids | Inhibit NF‐κB and AP‐1 | RA, IBD, MS, COVID‐19 | Marketed | 363, 398, 402, 403 | |
| Cyclophosphamide | Inhibit NF‐κB | RA | Marketed | 402 | |
| Thalidomide | Inhibits the release of TNF‐α and NF‐κB activation | IBD | Clinical trial Phase III | 404 | |
| DMF | Blockade the JAK‐STAT pathway and downregulate NF‐κB | MS | Marketed | 398 | |
| COVID‐19 | Clinical trial Phase IV | 306 | |||
| NP001 | Inhibit the NF‐κB pathway | ALS | Clinical trial Phase II | 405 | |
| Minocycline | Downregulate NF‐κB | ALS | Clinical trial Phase III | 241 | |
| JAK‐STAT pathway | Tofacitinib | JAK1/2/3/TYK2 inhibitor | RA | Marketed | 406 |
| IBD | Marketed | 406 | |||
| COVID‐19 | Clinical trial Phase IV | 407 | |||
| Baricitinib | JAK1/2 inhibitor | RA | Marketed | 406 | |
| COVID‐19 | Clinical trial Phase IV | 408 | |||
| Peficitinib | JAK1/2/3 inhibitor | RA | Clinical trial Phase III | 409 | |
| IBD | Clinical trial Phase II | 410 | |||
| Upadacitinib | JAK1/3 inhibitor | RA | Marketed | 406 | |
| IBD | Marketed | 406 | |||
| COVID‐19 | Clinical trial Phase IV | 411 | |||
| Filgotinib | JAK1 inhibitor | RA | Clinical trial Phase IV | 412 | |
| IBD | Clinical trial Phase III | 413 | |||
| Ritlecitinib | JAK3 inhibitor | RA | Marketed | 406 | |
| IBD | Marketed | 406 | |||
| Izencitinib | Pan‐JAK inhibitor | IBD | Clinical trial Phase III | 406 | |
| Deucravacitinib | TYK2 inhibitor | IBD | Marketed | 406 | |
| Brepocitinib | JAK1/TYK2 inhibitor | IBD | Clinical trial Phase II | 406 | |
| Pacritinib | JAK2 inhibitor | COVID‐19 | Clinical trial Phase II | 414 | |
| Ruxolitinib | JAK1/2 inhibitor | COVID‐19 | Clinical trial Phase III | 415 | |
| Nezulcitinib | Pan‐JAK inhibitor | COVID‐19 | Clinical trial Phase I | 416 | |
| Glatiramer acetate | Inhibit the JAK‐STAT pathway | MS | Marketed | 398 | |
| Laquinimod | Inhibit the JAK‐STAT, Akt and JNK pathway | MS | Marketed | 398 | |
| DMF | Blockade the JAK‐STAT pathway and downregulate NF‐κB | MS | Marketed | 398 | |
| COVID‐19 | Clinical trial Phase IV | 306 | |||
|
PI3K/Akt/ mTOR pathway |
Duvelisib | Inhibit PI3Kδ and PI3Kγ | COVID‐19 | Clinical trial Phase II | 306 |
| T‐cell lymphoma | Marketed | 417 | |||
| Laquinimod | Inhibit the JAK‐STAT, Akt and JNK pathway | MS | Marketed | 398 | |
| RNS60 | Inhibit PI3Kγ and activate PI3Kα/β | ALS | Clinical trial Phase II | 253 | |
| Metformin | Inhibit mTOR and activate the AMPK pathway | T2DM | Marketed | 265 | |
| COVID‐19 | Clinical trial Phase III | 418 | |||
| Rapamycin | mTOR inhibitor | COVID‐19 | Clinical trial Phase II | 306 | |
| RTB101 | mTOR inhibitor | COVID‐19 |
Clinical trial Phase II |
419 | |
| NLRP3 | Colchicine | Inhibit NLRP3 activity | AS |
Clinical trial Phase IV |
288, 289 |
| COVID‐19 |
Clinical trial Phase IV |
420 | |||
| Azithromycin | Inhibit NLRP3 activity | COVID‐19 |
Clinical trial Phase IV |
421 | |
| Melatonin | Inhibit NLRP3 activity | COVID‐19 | Clinical trial Phase II/III | 422 | |
|
Dapansutrile (OLT1177) |
NLRP3 inhibitor | COVID‐19 | Clinical trial Phase II | 306 | |
| MAPK pathway | Losmapimod | Inhibit p38α and p38β | COVID‐19 | Clinical trial Phase III | 306 |
| Azathioprine | Inhibit the JNK phosphorylation | RA | Marketed | 402 | |
| IBD |
Clinical trial Phase IV |
423 | |||
| MS | Marketed | 398 | |||
| Laquinimod | Inhibit the JAK‐STAT, Akt and JNK pathway | MS | Marketed | 398 | |
|
18β‐ glycyrrhetinic acid |
Suppress MAPK pathway | MS | Marketed | 398 | |
| Selonsertib | ASK1 inhibitor | NAFLD | Clinical trial Phase III | 276 | |
| AMPK pathway | Methotrexate | Activate the AMPK pathway | RA | Marketed | 402 |
| COVID‐19 | Clinical trial Phase III | 306 | |||
| Metformin | Inhibit mTOR and activate AMPK pathway | T2DM | Marketed | 265 | |
| COVID‐19 | Clinical trial Phase III | 418 | |||
Abbreviations: AD, Alzheimer's disease; ALS, amyotrophic lateral sclerosis; AS, atherosclerosis; BTK, Bruton's tyrosine kinase; COVID‐19, coronavirus disease 2019; CSF‐1R, colony‐stimulating factor‐1 receptor; CXCL, CXC motif ligand; CXCR, CXC chemokine receptor; DMF, dimethyl fumarate; FcR, Fc receptor; FXR, farnesoid X receptor; GLP‐1R, glucagon‐like peptide 1 receptor; GM‐CSF, granulocyte‐macrophage colony‐stimulating factor; GM‐CSFR, GM‐CSF receptor; IBD, inflammatory bowel disease; IL‐6, interleukin 6; JAK‐STAT, Janus kinase–signal transducer and activator of transcription; MABp1, monoclonal antibody against IL‐1 alpha; MAPK, mitogen‐activated protein kinase; MS, multiple sclerosis; NAFLD, nonalcoholic fatty liver disease; NF‐κB, nuclear factor kappa‐B; NSAIDs, nonsteroidal antiinflammatory drugs; PI3K, phosphatidylinositol 3‐kinase; PD, Parkinson's disease; RA, rheumatoid arthritis; S1PRs, sphingosine 1‐phosphate receptor; T2DM, type 2 diabetes mellitus; TB, tuberculosis; TLR, Toll‐like receptor; TNF‐α, tumor necrosis factor α; TZDs, thiazolidinedione.
FIGURE 6.

Therapeutic targets targeting macrophage in human diseases. Macrophages are widely present in almost all tissues of adult and play important roles in pathological processes. With the gradual in‐depth understanding of molecular mechanisms, more alternative therapeutic targets of macrophage have been proposed and applied in clinic/clinical trials. Abbreviations: COVID‐19, coronavirus disease 2019; NAFLD, nonalcoholic fatty liver disease; RA, rheumatoid arthritis; MS, multiple sclerosis; AD, Alzheimer's disease; ALS, amyotrophic lateral sclerosis; TB, tuberculosis; AS, atherosclerosis; T2DM, type 2 diabetes mellitus; IBD, inflammatory bowel disease. (Created with BioRender.com).
4.1. Macrophages in autoimmune diseases
Autoimmune diseases refer to tissue trauma caused by excessive and persistent inflammation owing to autoantibodies or self‐reactive lymphocytes when immune tolerance is broken. 180 In particular, macrophages have received increasing attention in the occurrence and progression of autoimmune diseases. Here, we focus on the role of macrophages in the pathogenesis of rheumatoid arthritis (RA), inflammatory bowel disease (IBD), and multiple sclerosis (MS) and summarize macrophage‐targeted therapeutics currently available.
4.1.1. RA
RA is characterized by the massive infiltration of inflammatory cells and proinflammatory mediators in the synovial membrane of multiple joints. Notably, macrophages are one of the most numerous cells in the RA synovium, the number of which is positively correlated with the severity of clinical signs. 181 The altered microenvironment in RA affects macrophage survival and proliferation and promotes M1 macrophage polarization through membrane receptor‐mediated signal transduction, including the NF‐κB, JAK‐STAT1, IRF5, 182 , 183 and Notch 184 signaling pathways, thus promoting joint inflammation. In RA patients, macrophages have high mitochondrial activity and produce high levels of ATP and ROS, 185 giving rise to oxidative damage to bone and cartilage. In addition, macrophages display enhanced resistance to apoptosis owing to sustained activation of the NF‐κB, STAT3, and PI3K signaling pathways, possibly related to the pathogenesis of RA. 186
In view of the critical role of macrophages in the pathogenesis of RA, efforts are being made to develop macrophage‐targeted therapeutics. Several types of agents have succeeded in clinical trials for the treatment of RA. Inflammatory cytokine inhibitors targeting TNF‐α, IL‐6, IL‐1, and IL‐12/23 have been developed, among which the most widely used is anti‐TNF agents. These agents promote macrophage polarization toward the M2 phenotype and increase macrophage apoptosis, leading to the resolution of inflammation in RA. 187 , 188 , 189 GM‐CSF inhibitors can decrease M1 polarization and relieve inflammation. Abundant clinical trials have affirmed the safety and efficacy of GM‐CSF or GM‐CSFR inhibitors in RA patients. 190 In addition, five JAK inhibitors have been approved for the clinical treatment of RA and have shown comparable efficacy to TNF inhibitors. 191 Bruton's tyrosine kinase (BTK), a component of Fc receptor (FcR) signaling pathway, is involved in the secretion of NO, TNF‐α, and IL‐1β by macrophages and is a therapeutic target for RA. 192 An irreversible BTK inhibitor spebrutinib and a highly selective noncovalent BTK inhibitor fenebrutinib have been evaluated in phase II trials and demonstrated efficacy and safety in RA. 193
4.1.2. IBD
IBD is a chronic inflammatory disease in the gastrointestinal tract, primarily encompassing Crohn's disease and ulcerative colitis. Patients with IBD have incomplete immune tolerance and thus initiate innate and adaptive immunity in the intestine with the participation of gut microbiota, leading to inflammation that is hyperactive and difficult to self‐limit. Intestinal macrophages play a major role in the establishment and maintenance of gut homeostasis, and dysfunction of intestinal macrophages is proposed to be the basis of chronic inflammation in IBD. 194 As with other inflammatory diseases, intestinal macrophages convert toward the M1 phenotype with the guidance of a variety of environmental signals in IBD, contributing to the overproduction of proinflammatory factors, suppression of anti‐inflammatory cytokines, and delayed clearance of bacteria. 195 , 196
Some of the anti‐TNF agents and anti‐IL‐12/23 agents used in RA are also widely used in IBD, and JAK inhibitors have entered late‐stage clinical trials. The well‐known mechanism for the effect of TNF‐α blockers on IBD is the induced differentiation of macrophages to the M2 phenotype and the secretion of the anti‐inflammatory cytokine IL‐10, which depends on FcR signaling. 197 , 198 In addition to suppressing the proinflammatory effect of several cytokines, JAK inhibitors downregulate the M1 pathway and permit macrophages to shift their balance toward a regulatory phenotype in patients with IBD. 199
In addition to the above agents, some of the first‐line treatments for IBD can have an impact on signaling pathways to alter the proinflammatory profiles of macrophages. For instance, corticosteroids direct monocytes to differentiate toward M2 macrophages by inactivating NF‐κB and AP‐1. 200 , 201 Salicylate abrogates NF‐κB activation predominantly in macrophages, leading to decreased proinflammatory gene expression. 202 The commonly used immunosuppressive drug azathioprine can exert an anti‐inflammatory effect in a mouse macrophage model by inhibiting Rac1 activity, JNK phosphorylation, and iNOS expression. 203
4.1.3. MS
MS is an idiopathic inflammatory demyelinating disease of the CNS that is caused by immune responses to self‐antigens. The role of microglia/macrophages in MS pathogenesis is similar to that of other autoimmune diseases mentioned above, such as oxidative damage and proinflammatory cytokine and chemokine production, resulting in inflammation and destruction in the brain.
Drugs targeting chemokines, especially the CCL2/CCR2 axis, achieve satisfactory efficacy in preclinical studies and animal models; however, they encounter challenges in clinical research. 204 AZD1480, a JAK1/2 inhibitor, deactivates the M1 phenotype and diminishes disease severity in multiple preclinical models of MS, 205 implying the potential possibility of applying JAK‐STAT inhibitors in the clinic. Furthermore, the GM‐CSF antibody MOR103 and the BTK inhibitors tolebrutinib and evobrutinib for the treatment of MS have entered the clinical trial stage. 206 , 207 , 208
Fingolimod (FTY720) is an agonist of the S1P receptor, which skews microglial polarization to the M2 phenotype, decreases the expression of inflammatory cytokines and induces apoptosis by modulating several signaling pathways involving STAT1/3, IRF8, p38, PI3K/Akt, and NF‐κB. 209 , 210 , 211 Glatiramer acetate, laquinimod, and dimethyl fumarate (DMF) can block the JAK‐STAT pathway and have beneficial effects. 212 In addition, laquinimod and DMF can reduce the M1 phenotype of microglia and the secretion of proinflammatory mediators by suppressing the expression of NF‐κB and reducing the activity of the JNK and Akt pathways, respectively. 213 , 214 18β‐glycyrrhetinic acid significantly decreases the proinflammatory profiles of microglia by suppressing the MAPK signaling pathway and promotes remyelination in an experimental autoimmune encephalomyelitis model. 215
4.2. Macrophages in neurodegenerative diseases
Neurodegenerative diseases feature progressive and irreversible loss of selective populations of neurons, mainly AD, PD, and ALS, according to primary clinical features. 216 As the role of microglia/macrophages in disease pathogenesis and progression is gradually being recognized, microglia have the potential to be prime targets for future therapeutic strategies.
4.2.1. Alzheimer's disease (AD)
AD is the most prevalent degenerative disease of the CNS and is characterized by progressive memory loss, behavioral change, and cognitive dysfunction. One of the most characteristic pathological features of AD is the deposition of Aβ and consequently the formation of neurotic plaques, which is considered by many scholars to be the initial factor of neuronal degeneration. 217 Other pathological features of AD are tau protein hyperphosphorylation and abnormal aggregation, leading to the formation of neurofibrillary tangle and the dysfunction of normal neurons and synapses. Notably, activated microglia elevate tau hyperphosphorylation in a manner dependent on CX3CR1 and IL‐1/p38 MAPK 218 and drive tau propagation and toxicity via NF‐κB signaling. 219 The intricate interactions between the two features of AD and neuroinflammation primarily triggered by microglia have become a hot topic in this field of research.
TNF is a common target to suppress inflammation in various diseases, and the same is true in AD. However, whether to use TNF‐α inhibitors for the treatment of AD is a matter of debate both in clinical trials and preclinical research. Pilot studies reported improvement in efficacy variables with treatment with etanercept, a TNF‐alpha inhibitor in patients with AD, 220 , 221 while a double‐blind phase II study observed no statistically significant changes in cognition, behavior, or global function. 222 Meanwhile, although there is evidence that anti‐TNF‐α reduces the levels of amyloid plaques and tau phosphorylation, 223 a study argues against long‐term use of pan‐anti‐TNF‐α inhibitors in AD, owing to the importance of intact TNF‐α receptor signaling for microglial‐mediated uptake of extracellular Aβ peptide pools. 224 Recently, a novel strategy of targeting the TNF pathway that selectively inhibits proinflammatory and neurotoxic pathways mediated by TNFR1 while preserving the neuroprotective pathway mediated by TNFR2 to counteract Aβ pathology has been proposed, 225 which may advance immunotherapy for AD.
Nuclear receptor PPARγ is another promising target. Activated PPARγ in microglia can stimulate NF‐κB (and, to a lesser extent, AP‐1 and STATs) to reduce the production of inflammatory mediators. 226 The results of clinical trials for PPARγ agonists in AD are positive overall. 227 , 228 Notably, in some trials, the efficacy of PPARγ agonists in individuals depends on their ApoE genotype. 229 The basis of this effect remains unexplained.
Moreover, attempts have been made to modulate other targets in microglia, such as p38, TLRs, NF‐κB, NLRP3, TREM‐2, and the CD200‐CD200R axis, 230 , 231 , 232 , 233 , 234 which need to be further investigated and evaluated in clinical trials before being moved from bench to bedside.
4.2.2. Parkinson's disease (PD)
PD is a class of movement disorders characterized by bradykinesia, muscular rigidity, rest tremor, and postural and gait impairment. Its key pathological features include loss of dopaminergic neurons within the substantia nigra of the midbrain, Lewy pathology with α‐synuclein aggregation, and neuroinflammation. 235 As one of the key cellular players in neuroinflammation, microglia bridge the three pathological features during the pathogenesis of PD.
The induction effect of α‐synuclein on neuroinflammation depends in part on the activation of the TLR/NF‐κB pathway in activated microglia, which causes the increased transcription of NLRP3, pro‐IL‐1β, and pro‐IL‐18. In α‐synuclein high expression mouse models, inhibiting the component of the TLR‐NF‐κB‐NLRP3 axis demonstrated a reduction in neuroinflammation, nigral dopaminergic degeneration and loss, and behavioral deficits. 236 Although not yet in clinical trials, many PPAR agonists have been recognized to protect nerve cells from inflammation in preclinical studies of PD. The administration of the PPARγ agonist pioglitazone in in vitro experiments or animals experiencing PD offered a neuroprotective effect with a decrease in NF‐κB and microglial activation and the preservation of dopamine striatal fibers and substantia nigra nerve cells. PPARβ/δ, PPARα, and PPARα/γ agonists can exert similar actions. 237 CX3CL1 is a unique chemokine produced in neurons and signals through its sole receptor CX3CR1 expressed on microglia. Studies have shown that CX3CL1/CX3CR1 signaling is closely related to the disease progression of PD. Increased CX3CL1 signaling decreases neuronal loss from α‐synuclein exposure by modulating microglia to a more neuroprotective and less proinflammatory phenotype, and loss of CX3CR1 on microglia leads to an increase in PD pathology. Administration of CX3CL1 via a virus or osmotic minipumps in animal models of PD can have an effect of reducing neurodegeneration and microglia activation, holding great promise. 238
4.2.3. Amyotrophic lateral sclerosis (ALS)
ALS is a motor neuron disease that is characterized by the progressive loss of both upper and lower motor neurons. It can affect any voluntary muscle, resulting in heterogeneous presentations of motor deficits. The degree of microglial activation in the motor cortex is significantly correlated with the severity of clinical signs in ALS. 239
Minocycline, a widely used broad‐spectrum antibiotic, selectively inhibits microglia by downregulating NF‐κB, that is, it suppresses M1 microglia during the progressive phase but does not affect the function of M2 microglia. 240 Nevertheless, in a phase III randomized trial, minocycline was observed to have even a harmful effect on patients with ALS after continuous administration. 241 In addition, minocycline does not confer long‐term benefit in patients with other neurodegenerative diseases, 242 , 243 indicating that the application of minocycline in neurodegenerative diseases requires additional caution.
Novel treatment approaches are under development for patients with ALS. NP001, a formulation of purified sodium chlorite, converts into taurine chloramine within monocytes/macrophages that induce proinflammatory macrophages to transform to a phagocytic state by inhibiting the NF‐κB pathway. 244 Its safety and early efficacy in ALS have been affirmed by phase I and II clinical trials. 245 , 246 Masitinib, an oral tyrosine kinase inhibitor (TKI), prevents microglial proliferation, migration, and inflammatory mediator production via selectively inhibiting CSF‐1R in the SOD1 mutant rat model of ALS. 247 A phase II trial showed that masitinib can benefit ALS patients, 248 as well as MS patients. 249 Clinical trials for other common targets in macrophage signaling pathways, such as TNF‐α, IL‐6, IL‐1, PI3K/Akt, and sphingosine 1‐phosphate receptor (S1PR), are being conducted in ALS patients, among which the S1PR agonist fingolimod, IL‐6R antagonist tocilizumab, and IL‐1R antagonist anakinra demonstrate good safety and tolerability, 250 , 251 , 252 and a modulator of the PI3K/Akt pathway RNS60 shows a positive effect, 253 whereas the trial of the TNF‐α antagonist thalidomide has failed. 254
4.3. Macrophages in metabolic diseases
Metabolic homeostasis in the human body depends on the coregulation of metabolic organs such as the pancreas, liver, and adipose tissue. Meanwhile, the immune response and metabolic regulation are highly integrated, and their proper function depends on each other. 3 Once this homeostasis is broken, it will trigger a series of chronic metabolic diseases, particularly obesity, type 2 diabetes mellitus (T2DM), nonalcoholic fatty liver disease (NAFLD), and cardiovascular diseases. 255
4.3.1. T2DM
Obesity is the major cause of insulin resistance. However, obese patients do not always develop T2DM. Hyperglycemia occurs only when islet inflammation impairs the function of β cells, preventing them from producing enough insulin to compensate for insulin resistance. Macrophages that present the proinflammatory classically activated M1‐like phenotype dominate in T2DM islet inflammation, 256 and the ratio of M1/M2 is related to the degree of insulin resistance. 257 Circulating factors such as glucose and free fatty acids (FFAs) induce macrophage secretion of cytokines, particularly IL‐1β and TNF‐α, by binding to TLR4 on macrophages and activating the NF‐κB pathway. 258 These cytokines promote β cell apoptosis and attenuate glucose‐stimulated insulin secretion by activating the NF‐κB and JNK pathways and deteriorating ER stress in β cells. 258
Considering the injurious effect of TNF‐α and IL‐1 produced by macrophages on the insulin signaling pathway, insulin resistance and glycemic control can theoretically be improved by therapies targeting these cytokines. In fact, the treatment of neutralizing cytokines is indeed efficacious in diabetic rodent models. 259 , 260 However, clinical trials have been less cheerful with mixed results. 261 , 262 , 263 , 264
PPARγ agonists, metformin, and glucagon‐like peptide‐1 receptor agonists (GLP‐1RAs) are three widely used and effective classes of antidiabetic agents. Their inhibitory effect on macrophages likely helps to explain their glucose‐lowering role. PPARγ agonists used in T2DM include thiazolidinedione, pioglitazone, and rosiglitazone, the modulation mechanism of which has been discussed in the AD section. Metformin has anti‐inflammatory actions by suppressing the activity of the NLRP3 inflammasome and the maturation of IL‐1β in macrophages by activating the AMPK pathway. 265 GLP‐1 is a kind of brain‐gut peptide secreted by ileal endocrine cells. Various types of cells, including macrophages, express the GLP‐1 receptor (GLP‐1R). After treatment with GLP‐1RAs or GLP‐1 in vivo and in vitro, the phenotype of macrophages is modified toward M2, and inflammatory cytokine secretion is inhibited, possibly because the STAT pathways are affected. 266 Enthusiasm for the development of GLP‐1RAs is growing rapidly, and there are already seven clinically approved GLP‐1R agonists currently used for T2DM.
4.3.2. NAFLD
FFAs released from adipocytes, damaged hepatocytes, endotoxins, and translocated bacteria resulting from increased intestinal permeability and changes in gut microbiota are factors that activate Kupffer cells in NAFLD. 267 Activated macrophages promote the development of hepatic steatosis, steatohepatitis, fibrosis, and ultimately hepatic failure through inflammatory cytokines, oxidative stress, and accumulated endotoxins owing to impaired phagocytic activity.
At present, there are a variety of macrophage signaling‐targeted therapeutic approaches for NAFLD treatment that have shown efficacy in disease models. 268 Here, we discuss only those therapeutic approaches that have already been evaluated in human trials owing to space limitations.
Cenicriviroc, a dual CCR2/CCR5 inhibitor, can inhibit monocyte‐derived macrophage recruitment mediated by CCL2 and significantly relieve hepatic steatosis and liver fibrosis in experimental models. 269 Phase II trials substantiated that cenicriviroc was well tolerated and exhibited an antifibrogenic effect in patients with nonalcoholic steatohepatitis 270 , 271 ; however, a phase III trial was recently terminated owing to undesirable results in the interim analysis. 272 Farnesoid X receptor (FXR) is another promising nuclear receptor target. FXR signaling not only is central to metabolic homeostasis but also reduces liver inflammation by skewing macrophages toward the M2 phenotype and downregulating inflammatory cytokines. 273 Its agonist, obeticholic acid, has obtained satisfactory results in patients with NAFLD. 274 , 275 ASK1, a serine/threonine kinase, promotes inflammation and fibrosis partly by activating MAPK pathways in macrophages. A phase II clinical trial demonstrated the efficacy of the ASK1 inhibitor selonsertib in relieving liver fibrosis in NAFLD patients. 276 The two aforementioned classes of agents, GLP1RAs and PPAR agonists, have also been extensively evaluated in NAFLD patients. 277 , 278 Moreover, PPARα, PPARδ, and PPARγ have similar effects on macrophages. Several drugs have been developed to target multiple receptor subtypes rather than one, including the PPARα/PPARγ agonists saroglitazar and aleglitazar, the PPARα/PPARδ agonist elafibranor, and the pan‐PPAR agonist lanifibranor. All of them are being investigated in patients with NAFLD in clinical trials. 279 , 280 , 281 , 282
4.3.3. Atherosclerosis (AS)
AS is the pathological basis of many cardiovascular diseases, such as myocardial infarction (MI), stroke, and cardiovascular death. Macrophages are considered to be the central cellular effectors of AS progression and run throughout all stages of disease development. 283 The polarization of macrophages largely affects the formation and stability of atherosclerotic plaques. M1 macrophages are found to be higher in unstable plaques or near the lipid core within advanced plaques and dominate the infarction and rupture of plaques, whereas M2 macrophages are the opposite. Pro‐inflammatory stimuli existing in the plaque microenvironment induce M1 polarization, breaking the balance between M1 and M2.
Although measures to prevent AS have mainly focused on lipid‐lowering treatment until now, progress has been made in targeting inflammatory pathways. The well‐known CANTOS trial is the first large‐scale, randomized, double‐blind trial in human AS. This trial tested the efficacy of canakinumab, an IL‐1β monoclonal antibody, in 10,061 patients with previous MI on the basis of standard therapy. 284 , 285 This trial confirmed that canakinumab could further reduce the recurrence and total number of adverse cardiovascular events in patients with prior MI independent of lipid‐level lowering, providing direct evidence for the inflammation hypothesis of AS and a basis for the subsequent development of more inflammation‐targeted drugs. Beyond IL‐1β, the application of agents targeting other cytokines, such as TNF, IL‐6, and IFN‐γ, which are related to macrophage signaling, is also being explored in AS, although the results may not be as inspiring as those of IL‐1β. 286
Colchicine, which is used for gouty arthritis and familial Mediterranean fever, has been extended to AS. The mechanism of action of colchicine is to disrupt tubulin and subsequently downregulate multiple inflammatory pathways and modulate innate immunity. Its impact on macrophages is mainly reflected in suppressing the release of various proinflammatory substances, such as ROS, NO, and TNF‐α, and inhibiting the maturation of IL‐1β via the NALP3/caspase‐1 or ROCK/caspase‐1 pathway. 287 For the application of colchicine in AS, there are two large clinical trials, COLCOT and LoDoCo2, and both show a significantly reduced risk of cardiovascular events for patients with a recent MI who received colchicine. 288 , 289 According to the long‐term follow‐up of LoDoCo2, the benefit of colchicine continues to accumulate over time. 290
4.4. Macrophages in infectious diseases
When host defense is challenged by invading infectious microbes, macrophages are recruited and differentiate into a proinflammatory phenotype after engagement and recognition of pathogens. These macrophages are activated through inflammatory signaling pathways and can secrete inflammatory mediators to promote pathogen killing in the early stages of the disease; however, they can cause tissue damage and even organ failure in diseases such as tuberculosis (TB) and coronavirus disease 2019 (COVID‐19).
4.4.1. TB
TB, a worldwide infectious disease that has led to major health issues, is caused by Mycobacterium tuberculosis (Mtb), an intracellular pathogenic bacterium. 291 The innate immune system is first challenged by Mtb infection, in which macrophages can present dual characteristics of eliminating pathogenic Mtb and establishing niches for the persistence and dissemination of Mtb. 291 , 292
Currently, there are two approaches targeting macrophages in the treatment of TB. 293 One is to amplify cellular antimicrobial mechanisms, thereby enhancing the effectiveness of anti‐Mtb chemotherapy. Imatinib, a TKI, can promote acidification and maturation of macrophage phagosomes, thus reducing intracellular Mtb survival. 294 , 295 Gefitinib, an inhibitor of EGFR, induces autophagy, thus restricting Mtb growth and replication in macrophage cytoplasm. 296 The other approach is to reduce inflammation caused by macrophages, such as the administration of TNF blockers, JAK inhibitors, thalidomide analogs, statins, and PPARγ agonists in severe Mtb infections. 293 , 297 , 298 , 299 , 300 However, these immunomodulators might reactivate latent Mtb infection or exacerbate active Mtb infection if not administered concurrently with antimycobacterial chemotherapy. Therefore, further research on drug dose and clinical value is needed.
4.4.2. COVID‐19
COVID‐19 is a contagious viral disease caused by SARS‐CoV‐2 and has become a global health concern due to its worldwide pandemic. Innate immunity functions as the first line of host defense, in which macrophages play a crucial role. 301 While the undeniable antiviral effect of macrophages is widely accepted, hyperinflammation and the subsequent cytokine storm triggered by overactivated macrophages are gaining concern as complications observed in severe COVID‐19 patients. 302
Macrophages are first activated through the TLR4/TLR7‐TRAF6‐NF‐κB pathway by identifying the SARS‐CoV‐2 structure. Meanwhile, the virus enters the macrophage cytoplasm through the ACE2 receptor, activating the NLRP3 inflammasome in macrophages. Chemokines released by infected epithelial cells and cytokines released by activated NK cells and T cells further promote the recruitment and activation of macrophages through the JAK‐STAT pathway. 302 , 303 In mild or moderate COVID‐19 patients, activated macrophages defend against SARS‐CoV‐2 infection by inflammatory cytokine production and suicide by pyroptosis. However, in severe cases, a reduced or delayed type I IFN response leads to increased cytopathic effects and enhanced chemokine release, thus contributing to sustained recruitment of macrophages. The continuous activation of macrophages leads to the overproduction of inflammatory cytokines and cytokine storms that eventually cause multiple organ failure and even death in patients. 301 , 302 , 304
It has been shown that blocking the entry and replication of virus in macrophages, as well as inhibiting the production of inflammatory factors, inflammasome activation and the type I IFN response can reduce the overactive immune inflammatory response. On the one hand, antiviral drugs remdesivir, molnupiravir, and nirmatrelvir‐ritonavir are proven to be effective in preventing hospitalization and death. 305 On the other hand, medications attempting to block inflammatory pathways, such as TLR/NF‐κB, NLRP3, and JAK‐STAT or inflammatory cytokines, including IL‐1β, IFNs and TNF‐α, are currently under clinical trials. 302 , 306 The results of clinical trials that have already been published show that inflammatory cytokine or signaling pathway inhibition therapies indeed improve the clinical outcomes of most patients, principally reflected in more rapid clinical symptom improvement, reduced inflammatory indices, decreased oxygen and assisted ventilation requirements, and a lower risk of mortality.
4.5. Macrophages in cancer
The approach for cancer treatment in recent years has focused on the tumor microenvironment (TME), which includes malignant, endothelial, stromal, and immune cells. Tumor‐associated macrophages (TAMs) are a plastic and heterogeneous cell population that account for the largest fraction of the myeloid infiltrate in most of human solid malignancies. 48 TAMs can arise from both TRMs and circulating monocytes. 307 And TAMs tend to be considered M2‐like macrophages as they have much of the representative properties of M2 macrophages. However, recently developed techniques such as single‐cell RNA sequencing have shown that TAMs rarely display bona fide M1 or M2 phenotypes, and the complexity of these cells cannot easily be resolved with the traditional binary “M1‐M2” system. 5 , 308 , 309 In respect to the function of TAMs, it is generally agreed that M1‐like TAMs exert antitumor effects, 310 while M2‐like TAMs promote the occurrence, metastasis, and angiogenesis of tumor cells, and inhibit the anti‐tumor immune response mediated by T cells. 311 The two phenotypes of TAMs can be converted into each other in response to environmental factors. 312 Therefore, targeting macrophage‐related signaling pathways to promote TAM depletion, inhibit monocyte recruitment, enhance TAM phagocytosis, and reprogram M2‐like TAMs to M1‐like TAMs could be promising therapeutic strategies for enhancing anti‐cancer immunity.
4.5.1. Targeting TAM recruitment
In tumor‐bearing mice, blockade of CCL2/CCR2 signaling inhibits the recruitment of inflammatory monocytes, delays cancer growth and metastasis, reduces postsurgical recurrence, and enhances survival. 313 However, interruption of CCL2 inhibition leads to an unexpected influx of monocytes from the bone marrow, which enhances cancer cell metastases and accelerates death in mouse models of metastatic breast cancer. 314 Further studies are needed to avoid this lethal rebound.
Several clinical trials targeting CCR2/CCL2 are ongoing. For example, a phase 1b trial in patients with borderline resectable and locally advanced pancreatic cancer showed decreased infiltration of TAMs in tumors and increased responsiveness to chemotherapy in patients treated with the small‐molecule CCR2 inhibitor PF‐04136309 combined with folinic acid, fluorouracil, irinotecan, and oxaliplatin (FOLFIRINOX), compared with those receiving FOLFIRINOX alone. 315 However, another phase 2 study of a human monoclonal antibody against CCL2, carlumab (CNTO 888), showed no antitumor activity as a single agent in patients with metastatic castration‐resistant prostate cancer. 316
In mouse models of various cancer types, including colorectal cancer (CRC), gastric cancer, and breast cancer, CCR5 inhibitors, including maraviroc, vicriviroc, and TAK‐779, have demonstrated antitumor effects. 317 In PDXs of malignant phyllodes tumors, CCR5 blockade with maraviroc showed macrophage repolarization with antitumor effects. 318 These antitumor effects were then confirmed in several clinical trials. A phase I trial with the CCR5 antagonist maraviroc in patients with liver metastases of advanced refractory CRC showed mitigation of the tumor‐promoting microenvironment and objective tumor responses (NCT01736813). 319 The combination of maravironic and pembrolizumab also showed beneficial activity and toxicity in mismatch repair‐proficient CRC (NCT03274804). 320
Preclinical studies with intracranial gliomas in both mice and rats clearly demonstrate that the marked increase in macrophage entry into irradiated tumors is mediated by the CXC motif ligand (CXCL) 12‐CXC chemokine receptor (CXCR) 4 pathway and that inhibition of this pathway blocks macrophage entry, thereby improving local control and survival after radiation therapy. 321 A phase I/II study in patients with newly diagnosed glioblastoma using the reversible CXCR4 inhibitor plerixafor after radiation was conducted, and the results showed that the infusion of plerixafor was well tolerated as an adjunct to standard chemoirradiation and that the local control of tumor recurrence was improved. 321
4.5.2. Targeting TAM activation and proliferation
Several small molecules and antibodies of the CSF‐1/CSF‐1R axis have been developed and tested in a substantial array of animal models. For example, in one study, RG7155, a monoclonal antibody that inhibits CSF‐1R activation, caused macrophage cell death in vitro and reduced TAM densities accompanied by an increase in the CD8(+)/CD4(+) T‐cell ratio in vivo. 322 Furthermore, experimental studies have started to assess combinations of CSF‐1R inhibitors with chemotherapy, 323 radiotherapy, 324 or ICB. 307 In mouse models of different cancer types, such as glioblastoma, 324 prostate cancer, 325 and pancreatic tumors, 326 CSF‐1R has been observed to reduce side effects and potentiate the efficacy of radiotherapy. 324 , 325 , 326
In addition, various drugs targeting CSF‐1(R), including small‐molecule inhibitors such as sotuletinib, ARRY‐382, and pexidartinib and antagonistic monoclonal antibodies such as emactuzumab, cabiralizumab, LY3022855, and axatilimab, have been or are being tested in clinical trials. For patients with tenosynovial giant cell tumor, a rare, locally aggressive neoplasm that overexpresses CSF‐1, objective and complete responses were observed during a phase 1 study of emactuzumab, 327 a phase 2 trial of PLX3397, 328 and a randomized phase 3 trial of pexidartinib. 329 Additionally, the effectiveness of the combination therapy in animal trials led to a number of clinical trials combining CSF‐1 or CSF‐1R inhibitors with radiotherapy, ICBs, or chemotherapeutic agents. For example, a phase 1 study of emactuzumab as a single agent or in combination with paclitaxel in patients with advanced solid tumors revealed depletion of immunosuppressive M2‐like macrophages (NCT01494688). 330
4.5.3. Strengthening TAM phagocytosis
Tumor cells overexpress CD47 in many cancer types, disguising them as healthy cells and avoiding phagocytosis. Therefore, strategies targeting the CD47/SIRPα axis to enable tumor cell killing through cellular phagocytosis have emerged as promising cancer immunotherapies. 331
In a variety of immunocompetent mouse tumor models, including ovarian cancer, 332 breast cancer, 333 and pancreatic cancer, 334 treatment with anti‐CD47 antibodies stimulated macrophage phagocytosis in vitro and suppressed tumor growth in vivo.
Clinical trials are underway for both solid and hematological malignancies using anti‐CD47 antibodies. To date, anti‐CD47 antibodies such as Hu5F9‐G4, 335 CC‐90002, 336 and SRF231 337 are under phase 1 clinical trials. Owing to the expression of CD47 on platelets and red blood cells, potential on‐target toxicity, in particular anemia and thrombocytopenia, is seriously considered. As an approach to achieving greater specificity toward tumors, bispecific agents have been generated. For example, a novel affinity‐tuned bispecific antibody targeting CD47 and PD‐L1 to antagonize both innate and adaptive immune checkpoint pathways is under clinical trial (NCT04881045). 338
As mentioned above, Siglec‐10 is an inhibitory I‐type lectin that can be expressed by TAMs and binds the potent anti‐phagocytic signal CD24 that is overexpressed in several solid tumor types. The binding of CD24 to Siglec10 elicits inhibitory signaling that blocks the cytoskeletal rearrangement required for cellular engulfment by macrophages. Clinical data have shown that CD24 expression is significantly higher in triple‐negative breast cancer cells than in normal breast cells. 339 Moreover, stratification of patients by CD24 expression revealed an increased relapse‐free survival for patients with ovarian cancer and an overall survival advantage for patients with breast cancer with lower CD24 expression, and anti‐SIGLEC10 rescued the macrophage capability to limit tumor growth. 340
4.5.4. Targeting M2‐M1 repolarization
The NF‐κB signaling pathway is important in cancer‐related inflammation and malignant progression. Studies have shown that when NF‐κB signaling is inhibited specifically in macrophages in the TME, they switch to M1 and become cytotoxic to tumor cells. 341 , 342 Inhibition of the NF‐κB pathway in liver macrophages by genetic deletion of IKKβ results in a marked reduction in tumor onset and load. 343 A micellar nanodrug effectively functions in M2‐to‐M1 repolarization via M2‐targeted codelivery of IKKβ siRNA and the STAT6 inhibitor AS1517499, which suppresses tumor growth and metastasis. Because the M2‐targeting peptides are hidden in the pH‐sheddable polyethylene glycol (PEG) corona so that active targeting of M2‐like macrophages is triggered only in the acidic TME rather than the neutral‐pH environment in healthy organs, immune side effects are reduced. 344
PI3K, the upstream regulator of Akt, has been shown to mediate M2 macrophage phenotypes. Activation of PI3Kγ signaling in macrophages has been reported to drive TAM immunosuppressive activities in models of lung cancer, 345 pancreatic cancer, 346 and melanoma. 347 The pharmacologic inhibition of PI3Kγ has shown the ability to reprogram TAMs and increase T‐cell recruitment into tumors, resulting in tumor growth inhibition. Accordingly, several PI3K inhibitors are being tested for different cancer indications, such as duvelisib (dual PI3Kδ/γ inhibitor) in a phase 1 trial and preclinical models of T‐cell lymphoma, 348 alpelisib (α‐specific PI3K inhibitor) for patients with epithelial ovarian cancer, 349 and umbralisib (dual PI3Kδ/CK1ε Inhibitor) in patients with relapsed or refractory indolent lymphoma. 350
5. DISCUSSION
Macrophages are widely present in almost all tissues of adult and play important roles in physiological and pathological processes. Although extraordinary efforts have been made to understand the functions and regulating mechanisms of macrophages, our evolving knowledge about the intricate signaling network within and between macrophage and its microenvironment is still not satisfactory yet.
The crosstalk of different signaling pathways and the bidirectional effect of the same signaling pathway leads to the complexity of macrophage regulation. First, the perception and integration of various external stimuli by macrophages lead to the activation of multiple signaling pathways and significant crosstalk of different signaling pathways, while the complicated crosstalk of signaling pathways can cause synergistic or antagonistic effects in macrophage regulation. An example of the synergistic regulation of macrophage inflammatory responses is the crosstalk between PI3K/Akt/mTOR and TLRs/NF‐κB signaling to promote inflammatory responses in Streptococcus uberis infection. 351 In contrast, an example of the antagonism of different signaling pathways in macrophages to maintain macrophage immune homeostasis is that immunosuppressive receptors antagonize the signal transduction mediated by activatory receptors such as FcR, through ITIMs, to inhibit macrophage phagocytosis and cytokine production. 156 Second, the same signaling pathway may have a bidirectional regulatory effect on macrophages, which means that the signaling pathway not only participates in the M1 macrophage polarization but also can promote the M2 polarization of macrophages under certain conditions. This bidirectional regulatory effect of the same signaling pathway on macrophages depends on factors such as different ligand–receptor interactions, intensity and duration of stimulation, phosphorylation sites, and kinase isoforms. For instance, LPS upregulates CCL2 expression through TLR4/MyD88 signaling, which activates JNK and promotes M1 polarization; 352 however, in IL‐4‐activated macrophages, scavenger receptor 1 leads to JNK activation through K63 ubiquitination, which promotes the polarization of macrophages to M2. 353 In this regard, JNK signaling pathway plays a bidirectional role in macrophage polarization.
It is the complexity of the associated regulatory signaling pathways that allows macrophages to exhibit a remarkable degree of functional and phenotypic plasticity and heterogeneity in response to environmental stimuli. Many of these macrophage phenotypes are tissue‐ and environment‐specific and play different or even opposing roles in human diseases. We have outlined various types of antibody‐based drugs and small molecule drugs targeting environmental signals or macrophage receptors and intracellular signaling pathways. These drugs exert therapeutic effects by blocking or activating macrophage‐related signaling pathways and gene transcription to alter the functional or physiological properties of macrophages.
However, with the development of drugs aiming at different molecular targets of macrophages, the next challenge is to effectively and selectively deliver these drugs to specific disease‐related macrophage subpopulations to improve efficacy and reduce off‐target effects. Studies have found that targeting of macrophages by phagocytosis of nanoparticles or liposomes or by antibodies binding to macrophage‐specific receptors can improve the therapeutic efficacy of drugs. 354 , 355 For example, a mannose‐modified nanoparticle is masked by acid‐sensitive PEG to prevente recognition by TRMs in a neutral pH. While in acidic TME, PEG is shed thereby exposing mannose and binding to mannose receptors on TAMs to achieve intratumor TAM targeting. 356 Besides, lactoferrin‐modified liposomes (LF‐lipo) can specifically bind to low‐density lipoprotein receptor‐related protein expressed on activated colonic macrophages for cell‐targeted anti‐inflammatory therapy, showing enhanced therapeutic efficacy in a mouse colitis model. 357 Moreover, bispecific monoclonal antibodies, which may overcome off‐target effect by the increased infinity with disease‐specific targets, have dual localization and therapeutic roles and are able to precisely target macrophages in the specific site. A classic example in cancer is bispecific antibodies targeting CD47 and tumor‐associated antigens such as CD20 358 and PD‐L1 359 that specifically target macrophages in tumor site, promoting phagocytosis of tumor cells by macrophages but sparing the host cells that do not express the tumor antigens such as platelets and red blood cells, showing limited toxicity.
Furthermore, due to the complexity of macrophage regulatory pathways, the efficacy of a single drug is limited, and combination therapy is a promising direction. For instance, CD40 could enhance the phagocytic activity of macrophages recovered by CD47‐SIRPα blockade, and a fusion protein with high affinity to bind CD40 and CD47 performed better than either CD47 blockade or CD40 agonist alone in a mouse CT26 tumor model. 360 Although combination therapy can better treat the disease and reduce toxic or side effects, it should also be noted that there may be antagonism or enhanced toxicity between drugs. For example, drugs aimed at blocking macrophage recruitment or depleting macrophages, such as CCL2 or CSF‐1R inhibitors, may be incompatible with drugs that enhance macrophage antitumor activity like CD40 agonists. 361 In order to design effective combination therapies that fully exploit drug synergies, an insightful understanding of drug targets and resistance mechanisms is required.
To sum up, macrophage‐targeted therapy has been proven to be a promising therapeutic strategy. However, due to the wide distribution of macrophages throughout the organism and the diversity and complexity of regulatory factors, further understanding of the functional and phenotypic heterogeneity of macrophages in specific microenvironment or disease is needed to improve the efficacy of targeted macrophage therapy and reduce off‐targeting. We comprehensively introduce the signal pathways that regulate macrophages and some key regulators in these pathways that can be used as therapeutic targets. The in‐depth understanding of these signal pathways and molecular targets is important for guiding the development of new therapeutic approaches.
AUTHOR CONTRIBUTIONS
M.L., M.J.W., Y.J.W., and H.F.Z. conceived and drafted the manuscript. G.N.Z. edited and revised the manuscript. Q.L.G. supervised and revised the manuscript. All authors read and approved the final manuscript.
CONFLICT OF INTEREST STATEMENT
The authors declare no conflicts of interest.
ETHICS STATEMENT
No ethical approval was required for this study.
ACKNOWLEDGMENTS
This work was supported by grants from the National Key Technology Research and Development Programme of China (2022YFC2704200 and 2022YFC2704205) and National Natural Science Foundation of China (81902933, 81772787 and 82072889). All figures were created with BioRender.
Li M, Wang M, Wen Y, Zhang H, Zhao G‐N, Gao Q. Signaling pathways in macrophages: molecular mechanisms and therapeutic targets. MedComm. 2023;4:e349. 10.1002/mco2.349
Contributor Information
Guang‐Nian Zhao, Email: zhaogn@tjh.tjmu.edu.cn.
Qinglei Gao, Email: qingleigao@hotmail.com.
DATA AVAILABILITY STATEMENT
Not applicable.
REFERENCES
- 1. Kotas ME, Medzhitov R. Homeostasis, inflammation, and disease susceptibility. Cell. 2015;160(5):816‐827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Meizlish ML, Franklin RA, Zhou X, Medzhitov R. Tissue homeostasis and inflammation. Annu Rev Immunol. 2021;39:557‐581. [DOI] [PubMed] [Google Scholar]
- 3. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496(7446):445‐455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Mosser DM, Hamidzadeh K, Goncalves R. Macrophages and the maintenance of homeostasis. Cell Mol Immunol. 2021;18(3):579‐587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Locati M, Curtale G, Mantovani A. Diversity, mechanisms, and significance of macrophage plasticity. Annu Rev Pathol. 2020;15:123‐147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Lawrence T. Coordinated regulation of signaling pathways during macrophage activation. Microbiol Spectr. 2016;4(5): 10.1128/microbiolspec.MCHD-0025-2015 [DOI] [PubMed] [Google Scholar]
- 7. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11(11):723‐737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Lavin Y, Mortha A, Rahman A, Merad M. Regulation of macrophage development and function in peripheral tissues. Nat Rev Immunol. 2015;15(12):731‐744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Ginhoux F, Guilliams M. Tissue‐resident macrophage ontogeny and homeostasis. Immunity. 2016;44(3):439‐449. [DOI] [PubMed] [Google Scholar]
- 10. Epelman S, Lavine KJ, Randolph GJ. Origin and functions of tissue macrophages. Immunity. 2014;41(1):21‐35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Lazarov T, Juarez‐Carreno S, Cox N, Geissmann F. Physiology and diseases of tissue‐resident macrophages. Nature. 2023;618(7966):698‐707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Hoeffel G, Chen J, Lavin Y, et al. C‐Myb(+) erythro‐myeloid progenitor‐derived fetal monocytes give rise to adult tissue‐resident macrophages. Immunity. 2015;42(4):665‐678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Bain CC, Schridde A. Origin, differentiation, and function of intestinal macrophages. Front Immunol. 2018;9:2733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Italiani P, Boraschi D. From monocytes to M1/M2 macrophages: phenotypical vs. functional differentiation. Front Immunol. 2014;5:514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity. 2016;44(3):450‐462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Schulz C, Gomez Perdiguero E, Chorro L, et al. A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science. 2012;336(6077):86‐90. [DOI] [PubMed] [Google Scholar]
- 17. Sieweke MH, Allen JE. Beyond stem cells: self‐renewal of differentiated macrophages. Science. 2013;342(6161):1242974. [DOI] [PubMed] [Google Scholar]
- 18. Mildner A, Marinkovic G, Jung S. Murine monocytes: origins, subsets, fates, and functions. Microbiol Spectr. 2016;4(5): 10.1128/microbiolspec.MHCD-0033-2016 [DOI] [PubMed] [Google Scholar]
- 19. Murdoch C, Giannoudis A, Lewis CE. Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood. 2004;104(8):2224‐2234. [DOI] [PubMed] [Google Scholar]
- 20. Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat Rev Immunol. 2011;11(11):762‐774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Stanley ER, Chitu V. CSF‐1 receptor signaling in myeloid cells. Cold Spring Harb Perspect Biol. 2014;6(6):a021857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Sawano A, Iwai S, Sakurai Y, et al. Flt‐1, vascular endothelial growth factor receptor 1, is a novel cell surface marker for the lineage of monocyte‐macrophages in humans. Blood. 2001;97(3):785‐791. [DOI] [PubMed] [Google Scholar]
- 23. Bruttger J, Karram K, Wortge S, et al. Genetic cell ablation reveals clusters of local self‐renewing microglia in the mammalian central nervous system. Immunity. 2015;43(1):92‐106. [DOI] [PubMed] [Google Scholar]
- 24. Hashimoto D, Chow A, Noizat C, et al. Tissue‐resident macrophages self‐maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity. 2013;38(4):792‐804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Jenkins SJ, Hume DA. Homeostasis in the mononuclear phagocyte system. Trends Immunol. 2014;35(8):358‐367. [DOI] [PubMed] [Google Scholar]
- 26. Molawi K, Wolf Y, Kandalla PK, et al. Progressive replacement of embryo‐derived cardiac macrophages with age. J Exp Med. 2014;211(11):2151‐2158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Ivashkiv LB. Epigenetic regulation of macrophage polarization and function. Trends Immunol. 2013;34(5):216‐223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Sica A, Erreni M, Allavena P, Porta C. Macrophage polarization in pathology. Cell Mol Life Sci. 2015;72(21):4111‐4126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Shapouri‐Moghaddam A, Mohammadian S, Vazini H, et al. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 2018;233(9):6425‐6440. [DOI] [PubMed] [Google Scholar]
- 30. Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM. M‐1/M‐2 macrophages and the Th1/Th2 paradigm. J Immunol. 2000;164(12):6166‐6173. [DOI] [PubMed] [Google Scholar]
- 31. Kadomoto S, Izumi K, Mizokami A. Macrophage polarity and disease control. Int J Mol Sci. 2021;23(1):144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32(5):593‐604. [DOI] [PubMed] [Google Scholar]
- 33. Chavez‐Galan L, Olleros ML, Vesin D, Garcia I. Much more than M1 and M2 macrophages, There are also CD169(+) and TCR(+) macrophages. Front Immunol. 2015;6:263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Grabowska J, Lopez‐Venegas MA, Affandi AJ, den Haan JMM. CD169+ macrophages capture and dendritic cells instruct: the interplay of the gatekeeper and the general of the immune system. Frontiers In Immunology. 2018;9:2472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Martinez‐Pomares L, Gordon S. CD169+ macrophages at the crossroads of antigen presentation. Trends Immunol. 2012;33(2):66‐70. [DOI] [PubMed] [Google Scholar]
- 36. Suzanne M, Steller H. Shaping organisms with apoptosis. Cell Death Differ. 2013;20(5):669‐675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Michell‐Robinson MA, Touil H, Healy LM, et al. Roles of microglia in brain development, tissue maintenance and repair. Brain. 2015;138(Pt 5):1138‐1159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Prinz M, Masuda T, Wheeler MA, Quintana FJ. Microglia and central nervous system‐associated macrophages‐from origin to disease modulation. Annu Rev Immunol. 2021;39:251‐277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Rao S, Lobov IB, Vallance JE, et al. Obligatory participation of macrophages in an angiopoietin 2‐mediated cell death switch. Development. 2007;134(24):4449‐4458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Fantin A, Vieira JM, Gestri G, et al. Tissue macrophages act as cellular chaperones for vascular anastomosis downstream of VEGF‐mediated endothelial tip cell induction. Blood. 2010;116(5):829‐840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Gordon EJ, Rao S, Pollard JW, Nutt SL, Lang RA, Harvey NL. Macrophages define dermal lymphatic vessel calibre during development by regulating lymphatic endothelial cell proliferation. Development. 2010;137(22):3899‐3910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Pollard JW. Trophic macrophages in development and disease. Nat Rev Immunol. 2009;9(4):259‐270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Stefater JA, 3rd , Ren S, Lang RA, Duffield JS. Metchnikoff's policemen: macrophages in development, homeostasis and regeneration. Trends Mol Med. 2011;17(12):743‐752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Gordy C, Pua H, Sempowski GD, He YW. Regulation of steady‐state neutrophil homeostasis by macrophages. Blood. 2011;117(2):618‐629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Arnold L, Henry A, Poron F, et al. Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis. J Exp Med. 2007;204(5):1057‐1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Batoon L, Millard SM, Wullschleger ME, et al. CD169(+) macrophages are critical for osteoblast maintenance and promote intramembranous and endochondral ossification during bone repair. Biomaterials. 2019;196:51‐66. [DOI] [PubMed] [Google Scholar]
- 47. Liu C, Wu C, Yang Q, et al. Macrophages mediate the repair of brain vascular rupture through direct physical adhesion and mechanical traction. Immunity. 2016;44(5):1162‐1176. [DOI] [PubMed] [Google Scholar]
- 48. Kerneur C, Cano CE, Olive D. Major pathways involved in macrophage polarization in cancer. Front Immunol. 2022;13:1026954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Bellora F, Castriconi R, Dondero A, et al. The interaction of human natural killer cells with either unpolarized or polarized macrophages results in different functional outcomes. Proc Natl Acad Sci U S A. 2010;107(50):21659‐21664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Arens R, Schoenberger SP. Plasticity in programming of effector and memory CD8 T‐cell formation. Immunol Rev. 2010;235(1):190‐205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Charles JF, Aliprantis AO. Osteoclasts: more than ‘bone eaters’. Trends Mol Med. 2014;20(8):449‐459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Soares MP, Hamza I. Macrophages and iron metabolism. Immunity. 2016;44(3):492‐504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Kosteli A, Sugaru E, Haemmerle G, et al. Weight loss and lipolysis promote a dynamic immune response in murine adipose tissue. J Clin Invest. 2010;120(10):3466‐3479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Fitzgerald KA, Kagan JC. Toll‐like receptors and the control of immunity. Cell. 2020;180(6):1044‐1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Akira S, Takeda K. Toll‐like receptor signalling. Nat Rev Immunol. 2004;4(7):499‐511. [DOI] [PubMed] [Google Scholar]
- 56. Kawai T, Akira S. TLR signaling. Semin Immunol. 2007;19(1):24‐32. [DOI] [PubMed] [Google Scholar]
- 57. Kawai T, Akira S. Signaling to NF‐kappaB by Toll‐like receptors. Trends Mol Med. 2007;13(11):460‐469. [DOI] [PubMed] [Google Scholar]
- 58. Meylan E, Burns K, Hofmann K, et al. RIP1 is an essential mediator of Toll‐like receptor 3‐induced NF‐kappa B activation. Nat Immunol. 2004;5(5):503‐507. [DOI] [PubMed] [Google Scholar]
- 59. Zhang J, Liu X, Wan C, et al. NLRP3 inflammasome mediates M1 macrophage polarization and IL‐1beta production in inflammatory root resorption. J Clin Periodontol. 2020;47(4):451‐460. [DOI] [PubMed] [Google Scholar]
- 60. Sun D, Xu J, Zhang W, et al. Negative regulator NLRC3: its potential role and regulatory mechanism in immune response and immune‐related diseases. Front Immunol. 2022;13:1012459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Lu A, Magupalli VG, Ruan J, et al. Unified polymerization mechanism for the assembly of ASC‐dependent inflammasomes. Cell. 2014;156(6):1193‐1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Guey B, Bodnar M, Manie SN, Tardivel A, Petrilli V. Caspase‐1 autoproteolysis is differentially required for NLRP1b and NLRP3 inflammasome function. Proc Natl Acad Sci USA. 2014;111(48):17254‐17259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526(7575):660‐665. [DOI] [PubMed] [Google Scholar]
- 64. Liu X, Zhang Z, Ruan J, et al. Inflammasome‐activated gasdermin D causes pyroptosis by forming membrane pores. Nature. 2016;535(7610):153‐158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Davanso MR, Crisma AR, Braga TT, et al. Macrophage inflammatory state in Type 1 diabetes: triggered by NLRP3/iNOS pathway and attenuated by docosahexaenoic acid. Clin Sci (Lond). 2021;135(1):19‐34. [DOI] [PubMed] [Google Scholar]
- 66. Schneider M, Zimmermann AG, Roberts RA, et al. The innate immune sensor NLRC3 attenuates Toll‐like receptor signaling via modification of the signaling adaptor TRAF6 and transcription factor NF‐kappaB. Nat Immunol. 2012;13(9):823‐831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Eren E, Berber M, Ozoren N. NLRC3 protein inhibits inflammation by disrupting NALP3 inflammasome assembly via competition with the adaptor protein ASC for pro‐caspase‐1 binding. J Biol Chem. 2017;292(30):12691‐12701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Motwani M, Pesiridis S, Fitzgerald KA. DNA sensing by the cGAS‐STING pathway in health and disease. Nat Rev Genet. 2019;20(11):657‐674. [DOI] [PubMed] [Google Scholar]
- 69. Li T, Chen ZJ. The cGAS‐cGAMP‐STING pathway connects DNA damage to inflammation, senescence, and cancer. J Exp Med. 2018;215(5):1287‐1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Shu C, Yi G, Watts T, Kao CC, Li P. Structure of STING bound to cyclic di‐GMP reveals the mechanism of cyclic dinucleotide recognition by the immune system. Nat Struct Mol Biol. 2012;19(7):722‐724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Mukai K, Konno H, Akiba T, et al. Activation of STING requires palmitoylation at the Golgi. Nat Commun. 2016;7:11932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Ishikawa H, Ma Z, Barber GN. STING regulates intracellular DNA‐mediated, type I interferon‐dependent innate immunity. Nature. 2009;461(7265):788‐792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Chen X, Chen Y. Ubiquitination of cGAS by TRAF6 regulates anti‐DNA viral innate immune responses. Biochem Biophys Res Commun. 2019;514(3):659‐664. [DOI] [PubMed] [Google Scholar]
- 74. Fang R, Wang C, Jiang Q, et al. NEMO‐IKKbeta are essential for IRF3 and NF‐kappaB activation in the cGAS‐STING pathway. J Immunol. 2017;199(9):3222‐3233. [DOI] [PubMed] [Google Scholar]
- 75. Balka KR, Louis C, Saunders TL, et al. TBK1 and IKKepsilon act redundantly to mediate STING‐induced NF‐kappaB responses in myeloid cells. Cell Rep. 2020;31(1):107492. [DOI] [PubMed] [Google Scholar]
- 76. Chen H, Sun H, You F, et al. Activation of STAT6 by STING is critical for antiviral innate immunity. Cell. 2011;147(2):436‐446. [DOI] [PubMed] [Google Scholar]
- 77. Shi JH, Sun SC. Tumor necrosis factor receptor‐associated factor regulation of nuclear factor kappaB and mitogen‐activated protein kinase pathways. Front Immunol. 2018;9:1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Chen G, Goeddel DV. TNF‐R1 signaling: a beautiful pathway. Science. 2002;296(5573):1634‐1635. [DOI] [PubMed] [Google Scholar]
- 79. van Loo G, Bertrand MJM. Death by TNF: a road to inflammation. Nat Rev Immunol. 2023;23:289‐303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Varfolomeev E, Goncharov T, Maecker H, et al. Cellular inhibitors of apoptosis are global regulators of NF‐kappaB and MAPK activation by members of the TNF family of receptors. Sci Signal. 2012;5(216):ra22. [DOI] [PubMed] [Google Scholar]
- 81. Karin M, Gallagher E. TNFR signaling: ubiquitin‐conjugated TRAFfic signals control stop‐and‐go for MAPK signaling complexes. Immunol Rev. 2009;228(1):225‐240. [DOI] [PubMed] [Google Scholar]
- 82. Aggarwal BB. Signalling pathways of the TNF superfamily: a double‐edged sword. Nat Rev Immunol. 2003;3(9):745‐756. [DOI] [PubMed] [Google Scholar]
- 83. Rauert H, Wicovsky A, Muller N, et al. Membrane tumor necrosis factor (TNF) induces p100 processing via TNF receptor‐2 (TNFR2). J Biol Chem. 2010;285(10):7394‐7404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Sun SC. The noncanonical NF‐kappaB pathway. Immunol Rev. 2012;246(1):125‐140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Li X, Yang Y, Ashwell JD. TNF‐RII and c‐IAP1 mediate ubiquitination and degradation of TRAF2. Nature. 2002;416(6878):345‐347. [DOI] [PubMed] [Google Scholar]
- 86. Suttles J, Stout RD. Macrophage CD40 signaling: a pivotal regulator of disease protection and pathogenesis. Semin Immunol. 2009;21(5):257‐264. [DOI] [PubMed] [Google Scholar]
- 87. Elo LL, Jarvenpaa H, Tuomela S, et al. Genome‐wide profiling of interleukin‐4 and STAT6 transcription factor regulation of human Th2 cell programming. Immunity. 2010;32(6):852‐862. [DOI] [PubMed] [Google Scholar]
- 88. Souza PP, Brechter AB, Reis RI, Costa CA, Lundberg P, Lerner UH. IL‐4 and IL‐13 inhibit IL‐1beta and TNF‐alpha induced kinin B1 and B2 receptors through a STAT6‐dependent mechanism. Br J Pharmacol. 2013;169(2):400‐412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Kelly‐Welch AE, Hanson EM, Boothby MR, Keegan AD. Interleukin‐4 and interleukin‐13 signaling connections maps. Science. 2003;300(5625):1527‐1528. [DOI] [PubMed] [Google Scholar]
- 90. Jimenez‐Garcia L, Herranz S, Luque A, Hortelano S. Critical role of p38 MAPK in IL‐4‐induced alternative activation of peritoneal macrophages. Eur J Immunol. 2015;45(1):273‐286. [DOI] [PubMed] [Google Scholar]
- 91. Wang SW, Sun YM. The IL‐6/JAK/STAT3 pathway: potential therapeutic strategies in treating colorectal cancer (Review). Int J Oncol. 2014;44(4):1032‐1040. [DOI] [PubMed] [Google Scholar]
- 92. Saxton RA, Tsutsumi N, Su LL, et al. Structure‐based decoupling of the pro‐ and anti‐inflammatory functions of interleukin‐10. Science. 2021;371(6535):eabc8433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Jung M, Ma Y, Iyer RP, et al. IL‐10 improves cardiac remodeling after myocardial infarction by stimulating M2 macrophage polarization and fibroblast activation. Basic Res Cardiol. 2017;112(3):33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Hu X, Herrero C, Li WP, et al. Sensitization of IFN‐gamma Jak‐STAT signaling during macrophage activation. Nat Immunol. 2002;3(9):859‐866. [DOI] [PubMed] [Google Scholar]
- 95. Ivashkiv LB, Donlin LT. Regulation of type I interferon responses. Nat Rev Immunol. 2014;14(1):36‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Platanias LC. Mechanisms of type‐I‐ and type‐II‐interferon‐mediated signalling. Nat Rev Immunol. 2005;5(5):375‐386. [DOI] [PubMed] [Google Scholar]
- 97. Pixley FJ, Stanley ER. CSF‐1 regulation of the wandering macrophage: complexity in action. Trends Cell Biol. 2004;14(11):628‐638. [DOI] [PubMed] [Google Scholar]
- 98. Mouchemore KA, Pixley FJ. CSF‐1 signaling in macrophages: pleiotrophy through phosphotyrosine‐based signaling pathways. Crit Rev Clin Lab Sci. 2012;49(2):49‐61. [DOI] [PubMed] [Google Scholar]
- 99. Munoz‐Garcia J, Cochonneau D, Teletchea S, et al. The twin cytokines interleukin‐34 and CSF‐1: masterful conductors of macrophage homeostasis. Theranostics. 2021;11(4):1568‐1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Xiang C, Li H, Tang W. Targeting CSF‐1R represents an effective strategy in modulating inflammatory diseases. Pharmacol Res. 2022;187:106566. [DOI] [PubMed] [Google Scholar]
- 101. Lee AW, States DJ. Both src‐dependent and ‐independent mechanisms mediate phosphatidylinositol 3‐kinase regulation of colony‐stimulating factor 1‐activated mitogen‐activated protein kinases in myeloid progenitors. Mol Cell Biol. 2000;20(18):6779‐6798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Covarrubias AJ, Aksoylar HI, Horng T. Control of macrophage metabolism and activation by mTOR and Akt signaling. Semin Immunol. 2015;27(4):286‐296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Fan W, Morinaga H, Kim JJ, et al. FoxO1 regulates Tlr4 inflammatory pathway signalling in macrophages. EMBO J. 2010;29(24):4223‐4236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Liu S, Lv X, Wei X, et al. TRIB3‒GSK‐3beta interaction promotes lung fibrosis and serves as a potential therapeutic target. Acta Pharm Sin B. 2021;11(10):3105‐3119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Arranz A, Doxaki C, Vergadi E, et al. Akt1 and Akt2 protein kinases differentially contribute to macrophage polarization. Proc Natl Acad Sci USA. 2012;109(24):9517‐9522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Wu X, Wang Y, Chen H, Wang Y, Gu Y. Phosphatase and tensin homologue determine inflammatory status by differentially regulating the expression of Akt1 and Akt2 in macrophage alternative polarization of periodontitis. J Clin Periodontol. 2023;50(2):220‐231. [DOI] [PubMed] [Google Scholar]
- 107. Weichhart T, Hengstschlager M, Linke M. Regulation of innate immune cell function by mTOR. Nat Rev Immunol. 2015;15(10):599‐614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Wilson HM. SOCS proteins in macrophage polarization and function. Front Immunol. 2014;5:357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Lehtonen A, Matikainen S, Miettinen M, Julkunen I. Granulocyte‐macrophage colony‐stimulating factor (GM‐CSF)‐induced STAT5 activation and target‐gene expression during human monocyte/macrophage differentiation. J Leukoc Biol. 2002;71(3):511‐519. [PubMed] [Google Scholar]
- 110. Achuthan AA, Lee KMC, Hamilton JA. Targeting GM‐CSF in inflammatory and autoimmune disorders. Semin Immunol. 2021;54:101523. [DOI] [PubMed] [Google Scholar]
- 111. Hamilton JA. GM‐CSF in inflammation. J Exp Med. 2020;217(1):e20190945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Batlle E, Massague J. Transforming growth factor‐beta signaling in immunity and cancer. Immunity. 2019;50(4):924‐940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Lee YS, Park JS, Kim JH, et al. Smad6‐specific recruitment of Smurf E3 ligases mediates TGF‐beta1‐induced degradation of MyD88 in TLR4 signalling. Nat Commun. 2011;2:460. [DOI] [PubMed] [Google Scholar]
- 114. Choi KC, Lee YS, Lim S, et al. Smad6 negatively regulates interleukin 1‐receptor‐Toll‐like receptor signaling through direct interaction with the adaptor Pellino‐1. Nat Immunol. 2006;7(10):1057‐1065. [DOI] [PubMed] [Google Scholar]
- 115. Hong S, Lim S, Li AG, et al. Smad7 binds to the adaptors TAB2 and TAB3 to block recruitment of the kinase TAK1 to the adaptor TRAF2. Nat Immunol. 2007;8(5):504‐513. [DOI] [PubMed] [Google Scholar]
- 116. Hamidi A, Song J, Thakur N, et al. TGF‐beta promotes PI3K‐AKT signaling and prostate cancer cell migration through the TRAF6‐mediated ubiquitylation of p85alpha. Sci Signal. 2017;10(486):eaal4186. [DOI] [PubMed] [Google Scholar]
- 117. Lee MK, Pardoux C, Hall MC, et al. TGF‐beta activates Erk MAP kinase signalling through direct phosphorylation of ShcA. EMBO J. 2007;26(17):3957‐3967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Yamashita M, Fatyol K, Jin C, Wang X, Liu Z, Zhang YE. TRAF6 mediates Smad‐independent activation of JNK and p38 by TGF‐beta. Mol Cell. 2008;31(6):918‐924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Hardbower DM, Singh K, Asim M, et al. EGFR regulates macrophage activation and function in bacterial infection. J Clin Invest. 2016;126(9):3296‐3312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Zhao G, Liu L, Peek RM, Jr. , et al. Activation of Epidermal growth factor receptor in macrophages mediates feedback inhibition of M2 polarization and gastrointestinal tumor cell growth. J Biol Chem. 2016;291(39):20462‐20472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Tomas A, Futter CE, Eden ER. EGF receptor trafficking: consequences for signaling and cancer. Trends Cell Biol. 2014;24(1):26‐34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Pierce KL, Premont RT, Lefkowitz RJ. Seven‐transmembrane receptors. Nat Rev Mol Cell Biol. 2002;3(9):639‐650. [DOI] [PubMed] [Google Scholar]
- 123. Lefkowitz RJ. Seven transmembrane receptors: something old, something new. Acta Physiol (Oxf). 2007;190(1):9‐19. [DOI] [PubMed] [Google Scholar]
- 124. Wang X, Iyer A, Lyons AB, Korner H, Wei W. Emerging roles for G‐protein coupled receptors in development and activation of macrophages. Front Immunol. 2019;10:2031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Kamenetsky M, Middelhaufe S, Bank EM, Levin LR, Buck J, Steegborn C. Molecular details of cAMP generation in mammalian cells: a tale of two systems. J Mol Biol. 2006;362(4):623‐639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Negreiros‐Lima GL, Lima KM, Moreira IZ, et al. Cyclic AMP regulates key features of macrophages via PKA: recruitment, reprogramming and efferocytosis. Cells. 2020;9(1):128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Kamthong PJ, Wu M. Inhibitor of nuclear factor‐kappaB induction by cAMP antagonizes interleukin‐1‐induced human macrophage‐colony‐stimulating‐factor expression. Biochem J. 2001;356(Pt 2):525‐530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Malsin ES, Kim S, Lam AP, Gottardi CJ. Macrophages as a source and recipient of Wnt signals. Front Immunol. 2019;10:1813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Schaale K, Neumann J, Schneider D, Ehlers S, Reiling N. Wnt signaling in macrophages: augmenting and inhibiting mycobacteria‐induced inflammatory responses. Eur J Cell Biol. 2011;90(6‐7):553‐559. [DOI] [PubMed] [Google Scholar]
- 130. Feng Y, Ren J, Gui Y, et al. Wnt/beta‐catenin‐promoted macrophage alternative activation contributes to kidney fibrosis. J Am Soc Nephrol. 2018;29(1):182‐193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Collu GM, Hidalgo‐Sastre A, Brennan K. Wnt‐Notch signalling crosstalk in development and disease. Cell Mol Life Sci. 2014;71(18):3553‐3567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Du Q, Zhang X, Cardinal J, et al. Wnt/beta‐catenin signaling regulates cytokine‐induced human inducible nitric oxide synthase expression by inhibiting nuclear factor‐kappaB activation in cancer cells. Cancer Res. 2009;69(9):3764‐3771. [DOI] [PubMed] [Google Scholar]
- 133. Maiti G, Naskar D, Sen M. The Wingless homolog Wnt5a stimulates phagocytosis but not bacterial killing. Proc Natl Acad Sci USA. 2012;109(41):16600‐16605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Liu Q, Song J, Pan Y, et al. Wnt5a/CaMKII/ERK/CCL2 axis is required for tumor‐associated macrophages to promote colorectal cancer progression. Int J Biol Sci. 2020;16(6):1023‐1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Radtke F, MacDonald HR, Tacchini‐Cottier F. Regulation of innate and adaptive immunity by Notch. Nat Rev Immunol. 2013;13(6):427‐437. [DOI] [PubMed] [Google Scholar]
- 136. Kopan R, Ilagan MX. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell. 2009;137(2):216‐233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Nakano T, Fukuda D, Koga J, Aikawa M. Delta‐like ligand 4‐Notch signaling in macrophage activation. Arterioscler Thromb Vasc Biol. 2016;36(10):2038‐2047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Pagie S, Gerard N, Charreau B. Notch signaling triggered via the ligand DLL4 impedes M2 macrophage differentiation and promotes their apoptosis. Cell Commun Signal. 2018;16(1):4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Tao S, Chen Q, Lin C, Dong H. Linc00514 promotes breast cancer metastasis and M2 polarization of tumor‐associated macrophages via Jagged1‐mediated notch signaling pathway. J Exp Clin Cancer Res. 2020;39(1):191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Shang Y, Smith S, Hu X. Role of Notch signaling in regulating innate immunity and inflammation in health and disease. Protein Cell. 2016;7(3):159‐174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Chen W, Liu Y, Chen J, et al. The Notch signaling pathway regulates macrophage polarization in liver diseases. Int Immunopharmacol. 2021;99:107938. [DOI] [PubMed] [Google Scholar]
- 142. Ford JW, McVicar DW. TREM and TREM‐like receptors in inflammation and disease. Curr Opin Immunol. 2009;21(1):38‐46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Klesney‐Tait J, Turnbull IR, Colonna M. The TREM receptor family and signal integration. Nat Immunol. 2006;7(12):1266‐1273. [DOI] [PubMed] [Google Scholar]
- 144. Turnbull IR, Colonna M. Activating and inhibitory functions of DAP12. Nat Rev Immunol. 2007;7(2):155‐161. [DOI] [PubMed] [Google Scholar]
- 145. Lo TH, Tseng KY, Tsao WS, et al. TREM‐1 regulates macrophage polarization in ureteral obstruction. Kidney Int. 2014;86(6):1174‐1186. [DOI] [PubMed] [Google Scholar]
- 146. Yuan Z, Syed MA, Panchal D, et al. Triggering receptor expressed on myeloid cells 1 (TREM‐1)‐mediated Bcl‐2 induction prolongs macrophage survival. J Biol Chem. 2014;289(21):15118‐15129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Klesney‐Tait J, Colonna M. Uncovering the TREM‐1‐TLR connection. Am J Physiol Lung Cell Mol Physiol. 2007;293(6):L1374‐1376. [DOI] [PubMed] [Google Scholar]
- 148. Dower K, Ellis DK, Saraf K, Jelinsky SA, Lin LL. Innate immune responses to TREM‐1 activation: overlap, divergence, and positive and negative cross‐talk with bacterial lipopolysaccharide. J Immunol. 2008;180(5):3520‐3534. [DOI] [PubMed] [Google Scholar]
- 149. Turnbull IR, Gilfillan S, Cella M, et al. Cutting edge: TREM‐2 attenuates macrophage activation. J Immunol. 2006;177(6):3520‐3524. [DOI] [PubMed] [Google Scholar]
- 150. Kuo TC, Chen A, Harrabi O, et al. Targeting the myeloid checkpoint receptor SIRPalpha potentiates innate and adaptive immune responses to promote anti‐tumor activity. J Hematol Oncol. 2020;13(1):160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Okazawa H, Motegi S, Ohyama N, et al. Negative regulation of phagocytosis in macrophages by the CD47‐SHPS‐1 system. J Immunol. 2005;174(4):2004‐2011. [DOI] [PubMed] [Google Scholar]
- 152. Shi L, Kidder K, Bian Z, Chiang SKT, Ouellette C, Liu Y. SIRPalpha sequesters SHP‐2 to promote IL‐4 and IL‐13 signaling and the alternative activation of macrophages. Sci Signal. 2021;14(702):eabb3966. [DOI] [PubMed] [Google Scholar]
- 153. van der Touw W, Chen HM, Pan PY, Chen SH. LILRB receptor‐mediated regulation of myeloid cell maturation and function. Cancer Immunol Immunother. 2017;66(8):1079‐1087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Hartley GP, Chow L, Ammons DT, Wheat WH, Dow SW. Programmed cell death ligand 1 (PD‐L1) signaling regulates macrophage proliferation and activation. Cancer Immunol Res. 2018;6(10):1260‐1273. [DOI] [PubMed] [Google Scholar]
- 155. Altevogt P, Sammar M, Huser L, Kristiansen G. Novel insights into the function of CD24: a driving force in cancer. Int J Cancer. 2021;148(3):546‐559. [DOI] [PubMed] [Google Scholar]
- 156. Munitz A. Inhibitory receptors on myeloid cells: new targets for therapy? Pharmacol Ther. 2010;125(1):128‐137. [DOI] [PubMed] [Google Scholar]
- 157. Ray PD, Huang BW, Tsuji Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 2012;24(5):981‐990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Morris G, Gevezova M, Sarafian V, Maes M. Redox regulation of the immune response. Cell Mol Immunol. 2022;19(10):1079‐1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Tan HY, Wang N, Li S, Hong M, Wang X, Feng Y. The reactive oxygen species in macrophage polarization: reflecting its dual role in progression and treatment of human diseases. Oxid Med Cell Longev. 2016;2016:2795090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Tobiume K, Matsuzawa A, Takahashi T, et al. ASK1 is required for sustained activations of JNK/p38 MAP kinases and apoptosis. EMBO Rep. 2001;2(3):222‐228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Kamata H, Honda S, Maeda S, Chang L, Hirata H, Karin M. Reactive oxygen species promote TNFalpha‐induced death and sustained JNK activation by inhibiting MAP kinase phosphatases. Cell. 2005;120(5):649‐661. [DOI] [PubMed] [Google Scholar]
- 162. Liu Y, Sun Y, Kang J, et al. Role of ROS‐induced NLRP3 inflammasome activation in the formation of calcium oxalate nephrolithiasis. Front Immunol. 2022;13:818625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Takada Y, Mukhopadhyay A, Kundu GC, Mahabeleshwar GH, Singh S, Aggarwal BB. Hydrogen peroxide activates NF‐kappa B through tyrosine phosphorylation of I kappa B alpha and serine phosphorylation of p65: evidence for the involvement of I kappa B alpha kinase and Syk protein‐tyrosine kinase. J Biol Chem. 2003;278(26):24233‐24241. [DOI] [PubMed] [Google Scholar]
- 164. Padgett LE, Burg AR, Lei W, Tse HM. Loss of NADPH oxidase‐derived superoxide skews macrophage phenotypes to delay type 1 diabetes. Diabetes. 2015;64(3):937‐946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Zhang Y, Choksi S, Chen K, Pobezinskaya Y, Linnoila I, Liu ZG. ROS play a critical role in the differentiation of alternatively activated macrophages and the occurrence of tumor‐associated macrophages. Cell Res. 2013;23(7):898‐914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. He C, Larson‐Casey JL, Gu L, Ryan AJ, Murthy S, Carter AB. Cu,Zn‐superoxide dismutase‐mediated redox regulation of Jumonji domain containing 3 modulates macrophage polarization and pulmonary fibrosis. Am J Respir Cell Mol Biol. 2016;55(1):58‐71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Lee SR, Yang KS, Kwon J, Lee C, Jeong W, Rhee SG. Reversible inactivation of the tumor suppressor PTEN by H2O2. J Biol Chem. 2002;277(23):20336‐20342. [DOI] [PubMed] [Google Scholar]
- 168. Sun S, Li H, Chen J, Qian Q. Lactic acid: no longer an inert and end‐product of glycolysis. Physiology (Bethesda). 2017;32(6):453‐463. [DOI] [PubMed] [Google Scholar]
- 169. Ivashkiv LB. The hypoxia‐lactate axis tempers inflammation. Nat Rev Immunol. 2020;20(2):85‐86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Yang K, Xu J, Fan M, et al. Lactate suppresses macrophage pro‐inflammatory response to LPS stimulation by inhibition of YAP and NF‐kappaB activation via GPR81‐mediated signaling. Front Immunol. 2020;11:587913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Chen P, Zuo H, Xiong H, et al. Gpr132 sensing of lactate mediates tumor‐macrophage interplay to promote breast cancer metastasis. Proc Natl Acad Sci USA. 2017;114(3):580‐585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Bohn T, Rapp S, Luther N, et al. Tumor immunoevasion via acidosis‐dependent induction of regulatory tumor‐associated macrophages. Nat Immunol. 2018;19(12):1319‐1329. [DOI] [PubMed] [Google Scholar]
- 173. Colegio OR, Chu NQ, Szabo AL, et al. Functional polarization of tumour‐associated macrophages by tumour‐derived lactic acid. Nature. 2014;513(7519):559‐563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Liu N, Luo J, Kuang D, et al. Lactate inhibits ATP6V0d2 expression in tumor‐associated macrophages to promote HIF‐2alpha‐mediated tumor progression. J Clin Invest. 2019;129(2):631‐646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Sun S, Xu X, Liang L, et al. Lactic acid‐producing probiotic Saccharomyces cerevisiae attenuates ulcerative colitis via suppressing macrophage pyroptosis and modulating gut microbiota. Front Immunol. 2021;12:777665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Gan Z, Zhang M, Xie D, et al. Glycinergic signaling in macrophages and its application in macrophage‐associated diseases. Front Immunol. 2021;12:762564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Contreras‐Nunez E, Blancas‐Flores G, Cruz M, et al. Participation of the IKK‐alpha/beta complex in the inhibition of the TNF‐alpha/NF‐kappaB pathway by glycine: possible involvement of a membrane receptor specific to adipocytes. Biomed Pharmacother. 2018;102:120‐131. [DOI] [PubMed] [Google Scholar]
- 178. Zhang Y, Ma X, Jiang D, et al. Glycine attenuates lipopolysaccharide‐induced acute lung injury by regulating NLRP3 inflammasome and NRF2 signaling. Nutrients. 2020;12(3):611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Liu R, Liao XY, Pan MX, et al. Glycine exhibits neuroprotective effects in ischemic stroke in rats through the inhibition of M1 microglial polarization via the NF‐kappaB p65/Hif‐1alpha signaling pathway. J Immunol. 2019;202(6):1704‐1714. [DOI] [PubMed] [Google Scholar]
- 180. Wang L, Wang FS, Gershwin ME. Human autoimmune diseases: a comprehensive update. J Intern Med. 2015;278(4):369‐395. [DOI] [PubMed] [Google Scholar]
- 181. Haringman JJ, Gerlag DM, Zwinderman AH, et al. Synovial tissue macrophages: a sensitive biomarker for response to treatment in patients with rheumatoid arthritis. Ann Rheum Dis. 2005;64(6):834‐838. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Draijer C, Boorsma CE, Robbe P, et al. Human asthma is characterized by more IRF5+ M1 and CD206+ M2 macrophages and less IL‐10+ M2‐like macrophages around airways compared with healthy airways. J Allergy Clin Immunol. 2017;140(1):280‐283.e283. [DOI] [PubMed] [Google Scholar]
- 183. van der Vorst EPC, Theodorou K, Wu Y, et al. High‐density lipoproteins exert pro‐inflammatory effects on macrophages via passive cholesterol depletion and PKC‐NF‐kappaB/STAT1‐IRF1 signaling. Cell Metab. 2017;25(1):197‐207. [DOI] [PubMed] [Google Scholar]
- 184. Keewan E, Naser SA. The role of Notch signaling in macrophages during inflammation and infection: implication in rheumatoid arthritis? Cells. 2020;9(1):111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Zeisbrich M, Yanes RE, Zhang H, et al. Hypermetabolic macrophages in rheumatoid arthritis and coronary artery disease due to glycogen synthase kinase 3b inactivation. Ann Rheum Dis. 2018;77(7):1053‐1062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Yang CA, Chiang BL. Inflammasomes and human autoimmunity: a comprehensive review. J Autoimmun. 2015;61:1‐8. [DOI] [PubMed] [Google Scholar]
- 187. Narazaki M, Tanaka T, Kishimoto T. The role and therapeutic targeting of IL‐6 in rheumatoid arthritis. Expert Rev Clin Immunol. 2017;13(6):535‐551. [DOI] [PubMed] [Google Scholar]
- 188. Dinarello CA. Interleukin‐1 in the pathogenesis and treatment of inflammatory diseases. Blood. 2011;117(14):3720‐3732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Degboe Y, Rauwel B, Baron M, et al. Polarization of rheumatoid macrophages by TNF targeting through an IL‐10/STAT3 mechanism. Front Immunol. 2019;10:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Zhao J, Guo S, Schrodi SJ, He D. Molecular and cellular heterogeneity in rheumatoid arthritis: mechanisms and clinical implications. Front Immunol. 2021;12:790122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Harrington R, Al Nokhatha SA, Conway R. JAK inhibitors in rheumatoid arthritis: an evidence‐based review on the emerging clinical data. J Inflamm Res. 2020;13:519‐531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Mukhopadhyay S, Mohanty M, Mangla A, et al. Macrophage effector functions controlled by Bruton's tyrosine kinase are more crucial than the cytokine balance of T cell responses for microfilarial clearance. J Immunol. 2002;168(6):2914‐2921. [DOI] [PubMed] [Google Scholar]
- 193. Cohen S, Tuckwell K, Katsumoto TR, et al. Fenebrutinib versus Placebo or adalimumab in rheumatoid arthritis: a randomized, double‐blind, phase II trial (ANDES study). Arthritis Rheumatol. 2020;72(9):1435‐1446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. de Souza HS, Fiocchi C. Immunopathogenesis of IBD: current state of the art. Nat Rev Gastroenterol Hepatol. 2016;13(1):13‐27. [DOI] [PubMed] [Google Scholar]
- 195. Na YR, Stakenborg M, Seok SH, Matteoli G. Macrophages in intestinal inflammation and resolution: a potential therapeutic target in IBD. Nat Rev Gastroenterol Hepatol. 2019;16(9):531‐543. [DOI] [PubMed] [Google Scholar]
- 196. Zhu W, Yu J, Nie Y, et al. Disequilibrium of M1 and M2 macrophages correlates with the development of experimental inflammatory bowel diseases. Immunol Invest. 2014;43(7):638‐652. [DOI] [PubMed] [Google Scholar]
- 197. Bloemendaal FM, Levin AD, Wildenberg ME, et al. Anti‐tumor necrosis factor with a glyco‐engineered Fc‐region has increased efficacy in mice with colitis. Gastroenterology. 2017;153(5):1351‐1362 e1354. [DOI] [PubMed] [Google Scholar]
- 198. Koelink PJ, Bloemendaal FM, Li B, et al. Anti‐TNF therapy in IBD exerts its therapeutic effect through macrophage IL‐10 signalling. Gut. 2020;69(6):1053‐1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Cordes F, Lenker E, Spille LJ, et al. Tofacitinib reprograms human monocytes of IBD patients and healthy controls toward a more regulatory phenotype. Inflamm Bowel Dis. 2020;26(3):391‐406. [DOI] [PubMed] [Google Scholar]
- 200. Oakley RH, Cidlowski JA. The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol. 2013;132(5):1033‐1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Ehrchen J, Steinmuller L, Barczyk K, et al. Glucocorticoids induce differentiation of a specifically activated, anti‐inflammatory subtype of human monocytes. Blood. 2007;109(3):1265‐1274. [DOI] [PubMed] [Google Scholar]
- 202. Bantel H, Berg C, Vieth M, Stolte M, Kruis W, Schulze‐Osthoff K. Mesalazine inhibits activation of transcription factor NF‐kappaB in inflamed mucosa of patients with ulcerative colitis. Am J Gastroenterol. 2000;95(12):3452‐3457. [DOI] [PubMed] [Google Scholar]
- 203. Marinkovic G, Hamers AA, de Vries CJ, de Waard V. 6‐Mercaptopurine reduces macrophage activation and gut epithelium proliferation through inhibition of GTPase Rac1. Inflamm Bowel Dis. 2014;20(9):1487‐1495. [DOI] [PubMed] [Google Scholar]
- 204. Fantuzzi L, Tagliamonte M, Gauzzi MC, Lopalco L. Dual CCR5/CCR2 targeting: opportunities for the cure of complex disorders. Cell Mol Life Sci. 2019;76(24):4869‐4886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Liu Y, Holdbrooks AT, De Sarno P, et al. Therapeutic efficacy of suppressing the Jak/STAT pathway in multiple models of experimental autoimmune encephalomyelitis. J Immunol. 2014;192(1):59‐72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Constantinescu CS, Asher A, Fryze W, et al. Randomized phase 1b trial of MOR103, a human antibody to GM‐CSF, in multiple sclerosis. Neurol Neuroimmunol Neuroinflamm. 2015;2(4):e117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Montalban X, Arnold DL, Weber MS, et al. Placebo‐Controlled trial of an oral BTK inhibitor in multiple sclerosis. N Engl J Med. 2019;380(25):2406‐2417. [DOI] [PubMed] [Google Scholar]
- 208. Reich DS, Arnold DL, Vermersch P, et al. Safety and efficacy of tolebrutinib, an oral brain‐penetrant BTK inhibitor, in relapsing multiple sclerosis: a phase 2b, randomised, double‐blind, placebo‐controlled trial. Lancet Neurol. 2021;20(9):729‐738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Qin C, Fan WH, Liu Q, et al. Fingolimod protects against ischemic white matter damage by modulating microglia toward M2 polarization via STAT3 pathway. Stroke. 2017;48(12):3336‐3346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210. Cipriani R, Chara JC, Rodriguez‐Antiguedad A, Matute C. FTY720 attenuates excitotoxicity and neuroinflammation. J Neuroinflammation. 2015;12:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Yao S, Li L, Sun X, et al. FTY720 inhibits MPP(+)‐induced microglial activation by affecting NLRP3 inflammasome activation. J Neuroimmune Pharmacol. 2019;14(3):478‐492. [DOI] [PubMed] [Google Scholar]
- 212. Benveniste EN, Liu Y, McFarland BC, Qin H. Involvement of the Janus Kinase/Signal Transducer and Activator of Transcription signaling pathway in multiple sclerosis and the animal model of experimental autoimmune encephalomyelitis. J Interferon Cytokine Res. 2014;34(8):577‐588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213. Mishra MK, Wang J, Keough MB, et al. Laquinimod reduces neuroaxonal injury through inhibiting microglial activation. Ann Clin Transl Neurol. 2014;1(6):409‐422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Peng H, Li H, Sheehy A, Cullen P, Allaire N, Scannevin RH. Dimethyl fumarate alters microglia phenotype and protects neurons against proinflammatory toxic microenvironments. J Neuroimmunol. 2016;299:35‐44. [DOI] [PubMed] [Google Scholar]
- 215. Zhou J, Cai W, Jin M, et al. 18Beta‐glycyrrhetinic acid suppresses experimental autoimmune encephalomyelitis through inhibition of microglia activation and promotion of remyelination. Sci Rep. 2015;5:13713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216. Dugger BN, Dickson DW. Pathology of neurodegenerative diseases. Cold Spring Harb Perspect Biol. 2017;9(7):a028035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Li N, Deng M, Hu G, Li N, Yuan H, Zhou Y. New insights into microglial mechanisms of memory impairment in Alzheimer's disease. Biomolecules. 2022;12(11):1722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Bhaskar K, Konerth M, Kokiko‐Cochran ON, Cardona A, Ransohoff RM, Lamb BT. Regulation of tau pathology by the microglial fractalkine receptor. Neuron. 2010;68(1):19‐31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Wang C, Fan L, Khawaja RR, et al. Microglial NF‐kappaB drives tau spreading and toxicity in a mouse model of tauopathy. Nat Commun. 2022;13(1):1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Tobinick E, Gross H, Weinberger A, Cohen H. TNF‐alpha modulation for treatment of Alzheimer's disease: a 6‐month pilot study. MedGenMed. 2006;8(2):25. [PMC free article] [PubMed] [Google Scholar]
- 221. Tobinick EL, Gross H. Rapid improvement in verbal fluency and aphasia following perispinal etanercept in Alzheimer's disease. BMC Neurol. 2008;8:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Butchart J, Brook L, Hopkins V, et al. Etanercept in Alzheimer disease: a randomized, placebo‐controlled, double‐blind, phase 2 trial. Neurology. 2015;84(21):2161‐2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Shi JQ, Shen W, Chen J, et al. Anti‐TNF‐alpha reduces amyloid plaques and tau phosphorylation and induces CD11c‐positive dendritic‐like cell in the APP/PS1 transgenic mouse brains. Brain Res. 2011;1368:239‐247. [DOI] [PubMed] [Google Scholar]
- 224. Montgomery SL, Mastrangelo MA, Habib D, et al. Ablation of TNF‐RI/RII expression in Alzheimer's disease mice leads to an unexpected enhancement of pathology: implications for chronic pan‐TNF‐alpha suppressive therapeutic strategies in the brain. Am J Pathol. 2011;179(4):2053‐2070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Steeland S, Gorle N, Vandendriessche C, et al. Counteracting the effects of TNF receptor‐1 has therapeutic potential in Alzheimer's disease. EMBO Mol Med. 2018;10(4):e8300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226. Daynes RA, Jones DC. Emerging roles of PPARs in inflammation and immunity. Nat Rev Immunol. 2002;2(10):748‐759. [DOI] [PubMed] [Google Scholar]
- 227. Watson GS, Cholerton BA, Reger MA, et al. Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study. Am J Geriatr Psychiatry. 2005;13(11):950‐958. [DOI] [PubMed] [Google Scholar]
- 228. Sato T, Hanyu H, Hirao K, Kanetaka H, Sakurai H, Iwamoto T. Efficacy of PPAR‐gamma agonist pioglitazone in mild Alzheimer disease. Neurobiol Aging. 2011;32(9):1626‐1633. [DOI] [PubMed] [Google Scholar]
- 229. Risner ME, Saunders AM, Altman JF, et al. Efficacy of rosiglitazone in a genetically defined population with mild‐to‐moderate Alzheimer's disease. Pharmacogenomics J. 2006;6(4):246‐254. [DOI] [PubMed] [Google Scholar]
- 230. Munoz L, Ammit AJ. Targeting p38 MAPK pathway for the treatment of Alzheimer's disease. Neuropharmacology. 2010;58(3):561‐568. [DOI] [PubMed] [Google Scholar]
- 231. Thawkar BS, Kaur G. Inhibitors of NF‐kappaB and P2×7/NLRP3/Caspase 1 pathway in microglia: novel therapeutic opportunities in neuroinflammation induced early‐stage Alzheimer's disease. J Neuroimmunol. 2019;326:62‐74. [DOI] [PubMed] [Google Scholar]
- 232. Nizami S, Hall‐Roberts H, Warrier S, Cowley SA, Di Daniel E. Microglial inflammation and phagocytosis in Alzheimer's disease: potential therapeutic targets. Br J Pharmacol. 2019;176(18):3515‐3532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Manich G, Recasens M, Valente T, Almolda B, Gonzalez B, Castellano B. Role of the CD200‐CD200R axis during homeostasis and neuroinflammation. Neuroscience. 2019;405:118‐136. [DOI] [PubMed] [Google Scholar]
- 234. McDonald CL, Hennessy E, Rubio‐Araiz A, et al. Inhibiting TLR2 activation attenuates amyloid accumulation and glial activation in a mouse model of Alzheimer's disease. Brain Behav Immun. 2016;58:191‐200. [DOI] [PubMed] [Google Scholar]
- 235. Kalia LV, Lang AE. Parkinson's disease. Lancet. 2015;386(9996):896‐912. [DOI] [PubMed] [Google Scholar]
- 236. Li Y, Xia Y, Yin S, et al. Targeting microglial α‐synuclein/TLRs/NF‐kappaB/NLRP3 inflammasome axis in Parkinson's disease. Frontiers In Immunology. 2021;12:719807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Behl T, Madaan P, Sehgal A, et al. Elucidating the neuroprotective role of PPARs in Parkinson's disease: a neoteric and prospective target. International Journal of Molecular Sciences. 2021;22(18):10161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Subbarayan MS, Joly‐Amado A, Bickford PC, Nash KR. CX3CL1/CX3CR1 signaling targets for the treatment of neurodegenerative diseases. Pharmacology & Therapeutics. 2022;231:107989. [DOI] [PubMed] [Google Scholar]
- 239. Turner MR, Cagnin A, Turkheimer FE, et al. Evidence of widespread cerebral microglial activation in amyotrophic lateral sclerosis: an [11C](R)‐PK11195 positron emission tomography study. Neurobiol Dis. 2004;15(3):601‐609. [DOI] [PubMed] [Google Scholar]
- 240. Kobayashi K, Imagama S, Ohgomori T, et al. Minocycline selectively inhibits M1 polarization of microglia. Cell Death Dis. 2013;4(3):e525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Gordon PH, Moore DH, Miller RG, et al. Efficacy of minocycline in patients with amyotrophic lateral sclerosis: a phase III randomised trial. Lancet Neurol. 2007;6(12):1045‐1053. [DOI] [PubMed] [Google Scholar]
- 242. Howard R, Zubko O, Bradley R, et al. Minocycline at 2 different dosages vs placebo for patients with mild Alzheimer disease: a randomized clinical trial. JAMA Neurol. 2020;77(2):164‐174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243. Dodel R, Spottke A, Gerhard A, et al. Minocycline 1‐year therapy in multiple‐system‐atrophy: effect on clinical symptoms and [(11)C] (R)‐PK11195 PET (MEMSA‐trial). Mov Disord. 2010;25(1):97‐107. [DOI] [PubMed] [Google Scholar]
- 244. Joo K, Lee Y, Choi D, et al. An anti‐inflammatory mechanism of taurine conjugated 5‐aminosalicylic acid against experimental colitis: taurine chloramine potentiates inhibitory effect of 5‐aminosalicylic acid on IL‐1beta‐mediated NFkappaB activation. Eur J Pharmacol. 2009;618(1‐3):91‐97. [DOI] [PubMed] [Google Scholar]
- 245. Miller RG, Zhang R, Block G, et al. NP001 regulation of macrophage activation markers in ALS: a phase I clinical and biomarker study. Amyotroph Lateral Scler Frontotemporal Degener. 2014;15(7‐8):601‐609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Miller RG, Block G, Katz JS, et al. Randomized phase 2 trial of NP001‐a novel immune regulator: safety and early efficacy in ALS. Neurol Neuroimmunol Neuroinflamm. 2015;2(3):e100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Trias E, Ibarburu S, Barreto‐Nunez R, et al. Post‐paralysis tyrosine kinase inhibition with masitinib abrogates neuroinflammation and slows disease progression in inherited amyotrophic lateral sclerosis. J Neuroinflammation. 2016;13(1):177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Mora JS, Genge A, Chio A, et al. Masitinib as an add‐on therapy to riluzole in patients with amyotrophic lateral sclerosis: a randomized clinical trial. Amyotroph Lateral Scler Frontotemporal Degener. 2020;21(1‐2):5‐14. [DOI] [PubMed] [Google Scholar]
- 249. Vermersch P, Brieva‐Ruiz L, Fox RJ, et al. Efficacy and safety of masitinib in progressive forms of multiple sclerosis: a randomized, phase 3, clinical trial. Neurol Neuroimmunol Neuroinflamm. 2022;9(3):e1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Milligan C, Atassi N, Babu S, et al. Tocilizumab is safe and tolerable and reduces C‐reactive protein concentrations in the plasma and cerebrospinal fluid of ALS patients. Muscle Nerve. 2021;64(3):309‐320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Berry JD, Paganoni S, Atassi N, et al. Phase IIa trial of fingolimod for amyotrophic lateral sclerosis demonstrates acceptable acute safety and tolerability. Muscle Nerve. 2017;56(6):1077‐1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Maier A, Deigendesch N, Muller K, et al. Interleukin‐1 antagonist Anakinra in amyotrophic lateral sclerosis—a pilot study. PLoS One. 2015;10(10):e0139684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Beghi E, Pupillo E, Bianchi E, et al. Effect of RNS60 in amyotrophic lateral sclerosis: a phase II multicentre, randomized, double‐blind, placebo‐controlled trial. Eur J Neurol. 2023;30(1):69‐86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Stommel EW, Cohen JA, Fadul CE, et al. Efficacy of thalidomide for the treatment of amyotrophic lateral sclerosis: a phase II open label clinical trial. Amyotroph Lateral Scler. 2009;10(5‐6):393‐404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860‐867. [DOI] [PubMed] [Google Scholar]
- 256. Boni‐Schnetzler M, Meier DT. Islet inflammation in type 2 diabetes. Semin Immunopathol. 2019;41(4):501‐513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Fujisaka S. The role of adipose tissue M1/M2 macrophages in type 2 diabetes mellitus. Diabetol Int. 2021;12(1):74‐79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Ying W, Fu W, Lee YS, Olefsky JM. The role of macrophages in obesity‐associated islet inflammation and beta‐cell abnormalities. Nat Rev Endocrinol. 2020;16(2):81‐90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor‐alpha: direct role in obesity‐linked insulin resistance. Science. 1993;259(5091):87‐91. [DOI] [PubMed] [Google Scholar]
- 260. Osborn O, Brownell SE, Sanchez‐Alavez M, Salomon D, Gram H, Bartfai T. Treatment with an Interleukin 1 beta antibody improves glycemic control in diet‐induced obesity. Cytokine. 2008;44(1):141‐148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Ruscitti P, Masedu F, Alvaro S, et al. Anti‐interleukin‐1 treatment in patients with rheumatoid arthritis and type 2 diabetes (TRACK): a multicentre, open‐label, randomised controlled trial. PLoS Med. 2019;16(9):e1002901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Timper K, Seelig E, Tsakiris DA, Donath MY. Safety, pharmacokinetics, and preliminary efficacy of a specific anti‐IL‐1alpha therapeutic antibody (MABp1) in patients with type 2 diabetes mellitus. J Diabetes Complications. 2015;29(7):955‐960. [DOI] [PubMed] [Google Scholar]
- 263. Howard C, Noe A, Skerjanec A, et al. Safety and tolerability of canakinumab, an IL‐1beta inhibitor, in type 2 diabetes mellitus patients: a pooled analysis of three randomised double‐blind studies. Cardiovasc Diabetol. 2014;13:94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264. Rissanen A, Howard CP, Botha J, Thuren T, Global I. Effect of anti‐IL‐1beta antibody (canakinumab) on insulin secretion rates in impaired glucose tolerance or type 2 diabetes: results of a randomized, placebo‐controlled trial. Diabetes Obes Metab. 2012;14(12):1088‐1096. [DOI] [PubMed] [Google Scholar]
- 265. Lee HM, Kim JJ, Kim HJ, Shong M, Ku BJ, Jo EK. Upregulated NLRP3 inflammasome activation in patients with type 2 diabetes. Diabetes. 2013;62(1):194‐204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266. Bendotti G, Montefusco L, Lunati ME, et al. The anti‐inflammatory and immunological properties of GLP‐1 Receptor Agonists. Pharmacol Res. 2022;182:106320. [DOI] [PubMed] [Google Scholar]
- 267. Kazankov K, Jorgensen SMD, Thomsen KL, et al. The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol. 2019;16(3):145‐159. [DOI] [PubMed] [Google Scholar]
- 268. Singanayagam A, Triantafyllou E. Macrophages in chronic liver failure: diversity, plasticity and therapeutic targeting. Front Immunol. 2021;12:661182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Krenkel O, Puengel T, Govaere O, et al. Therapeutic inhibition of inflammatory monocyte recruitment reduces steatohepatitis and liver fibrosis. Hepatology. 2018;67(4):1270‐1283. [DOI] [PubMed] [Google Scholar]
- 270. Friedman SL, Ratziu V, Harrison SA, et al. A randomized, placebo‐controlled trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis. Hepatology. 2018;67(5):1754‐1767. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Ratziu V, Sanyal A, Harrison SA, et al. Cenicriviroc treatment for adults with nonalcoholic steatohepatitis and fibrosis: final analysis of the phase 2b CENTAUR study. Hepatology. 2020;72(3):892‐905. [DOI] [PubMed] [Google Scholar]
- 272. Vuppalanchi R, Noureddin M, Alkhouri N, Sanyal AJ. Therapeutic pipeline in nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol. 2021;18(6):373‐392. [DOI] [PubMed] [Google Scholar]
- 273. Peiseler M, Schwabe R, Hampe J, Kubes P, Heikenwalder M, Tacke F. Immune mechanisms linking metabolic injury to inflammation and fibrosis in fatty liver disease ‐ novel insights into cellular communication circuits. J Hepatol. 2022;77(4):1136‐1160. [DOI] [PubMed] [Google Scholar]
- 274. Younossi ZM, Ratziu V, Loomba R, et al. Obeticholic acid for the treatment of non‐alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo‐controlled phase 3 trial. Lancet. 2019;394(10215):2184‐2196. [DOI] [PubMed] [Google Scholar]
- 275. Younossi ZM, Stepanova M, Nader F, et al. Obeticholic acid impact on quality of life in patients with nonalcoholic steatohepatitis: REGENERATE 18‐month interim analysis. Clin Gastroenterol Hepatol. 2022;20(9):2050‐2058.e2012. [DOI] [PubMed] [Google Scholar]
- 276. Loomba R, Lawitz E, Mantry PS, et al. The ASK1 inhibitor selonsertib in patients with nonalcoholic steatohepatitis: a randomized, phase 2 trial. Hepatology. 2018;67(2):549‐559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277. Armstrong MJ, Gaunt P, Aithal GP, et al. Liraglutide safety and efficacy in patients with non‐alcoholic steatohepatitis (LEAN): a multicentre, double‐blind, randomised, placebo‐controlled phase 2 study. Lancet. 2016;387(10019):679‐690. [DOI] [PubMed] [Google Scholar]
- 278. Newsome PN, Buchholtz K, Cusi K, et al. A placebo‐controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. N Engl J Med. 2021;384(12):1113‐1124. [DOI] [PubMed] [Google Scholar]
- 279. Siddiqui MS, Idowu MO, Parmar D, et al. A phase 2 double blinded, randomized controlled trial of saroglitazar in patients with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol. 2021;19(12):2670‐2672. [DOI] [PubMed] [Google Scholar]
- 280. Grobbee EJ, de Jong VD, Schrieks IC, et al. Improvement of non‐invasive tests of liver steatosis and fibrosis as indicators for non‐alcoholic fatty liver disease in type 2 diabetes mellitus patients with elevated cardiovascular risk profile using the PPAR‐alpha/gamma agonist aleglitazar. PLoS One. 2022;17(11):e0277706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281. Ratziu V, Harrison SA, Francque S, et al. Elafibranor, an agonist of the peroxisome proliferator‐activated receptor‐alpha and ‐delta, induces resolution of nonalcoholic steatohepatitis without fibrosis worsening. Gastroenterology. 2016;150(5):1147‐1159 e1145. [DOI] [PubMed] [Google Scholar]
- 282. Francque SM, Bedossa P, Ratziu V, et al. A randomized, controlled trial of the pan‐PPAR agonist lanifibranor in NASH. N Engl J Med. 2021;385(17):1547‐1558. [DOI] [PubMed] [Google Scholar]
- 283. Packard RR, Lichtman AH, Libby P. Innate and adaptive immunity in atherosclerosis. Semin Immunopathol. 2009;31(1):5‐22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284. Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119‐1131. [DOI] [PubMed] [Google Scholar]
- 285. Everett BM, MacFadyen JG, Thuren T, Libby P, Glynn RJ, Ridker PM. Inhibition of interleukin‐1beta and reduction in atherothrombotic cardiovascular events in the CANTOS trial. J Am Coll Cardiol. 2020;76(14):1660‐1670. [DOI] [PubMed] [Google Scholar]
- 286. Poznyak AV, Bharadwaj D, Prasad G, Grechko AV, Sazonova MA, Orekhov AN. Anti‐inflammatory therapy for atherosclerosis: focusing on cytokines. Int J Mol Sci. 2021;22(13):7061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Leung YY, Yao Hui LL, Kraus VB. Colchicine—update on mechanisms of action and therapeutic uses. Semin Arthritis Rheum. 2015;45(3):341‐350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Tardif JC, Kouz S, Waters DD, et al. Efficacy and safety of low‐dose colchicine after myocardial infarction. N Engl J Med. 2019;381(26):2497‐2505. [DOI] [PubMed] [Google Scholar]
- 289. Nidorf SM, Fiolet ATL, Mosterd A, et al. Colchicine in patients with chronic coronary disease. N Engl J Med. 2020;383(19):1838‐1847. [DOI] [PubMed] [Google Scholar]
- 290. Opstal TSJ, van Broekhoven A, Fiolet ATL, et al. Long‐term efficacy of colchicine in patients with chronic coronary disease: insights from LoDoCo2. Circulation. 2022;145(8):626‐628. [DOI] [PubMed] [Google Scholar]
- 291. Liu CH, Liu H, Ge B. Innate immunity in tuberculosis: host defense vs pathogen evasion. Cell Mol Immunol. 2017;14(12):963‐975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. Ahmad F, Rani A, Alam A, et al. Macrophage: a cell with many faces and functions in tuberculosis. Front Immunol. 2022;13:747799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Wallis RS, Hafner R. Advancing host‐directed therapy for tuberculosis. Nat Rev Immunol. 2015;15(4):255‐263. [DOI] [PubMed] [Google Scholar]
- 294. Bruns H, Stegelmann F, Fabri M, et al. Abelson tyrosine kinase controls phagosomal acidification required for killing of Mycobacterium tuberculosis in human macrophages. J Immunol. 2012;189(8):4069‐4078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295. Napier RJ, Rafi W, Cheruvu M, et al. Imatinib‐sensitive tyrosine kinases regulate mycobacterial pathogenesis and represent therapeutic targets against tuberculosis. Cell Host Microbe. 2011;10(5):475‐485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296. Stanley SA, Barczak AK, Silvis MR, et al. Identification of host‐targeted small molecules that restrict intracellular Mycobacterium tuberculosis growth. PLoS Pathog. 2014;10(2):e1003946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297. Wallis RS, Kyambadde P, Johnson JL, et al. A study of the safety, immunology, virology, and microbiology of adjunctive etanercept in HIV‐1‐associated tuberculosis. AIDS. 2004;18(2):257‐264. [DOI] [PubMed] [Google Scholar]
- 298. Wallis RS, van Vuuren C, Potgieter S. Adalimumab treatment of life‐threatening tuberculosis. Clin Infect Dis. 2009;48(10):1429‐1432. [DOI] [PubMed] [Google Scholar]
- 299. Parihar SP, Guler R, Khutlang R, et al. Statin therapy reduces the Mycobacterium tuberculosis burden in human macrophages and in mice by enhancing autophagy and phagosome maturation. J Infect Dis. 2014;209(5):754‐763. [DOI] [PubMed] [Google Scholar]
- 300. Skerry C, Pinn ML, Bruiners N, Pine R, Gennaro ML, Karakousis PC. Simvastatin increases the in vivo activity of the first‐line tuberculosis regimen. J Antimicrob Chemother. 2014;69(9):2453‐2457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301. Knoll R, Schultze JL, Schulte‐Schrepping J. Monocytes and macrophages in COVID‐19. Front Immunol. 2021;12:720109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302. Merad M, Martin JC. Pathological inflammation in patients with COVID‐19: a key role for monocytes and macrophages. Nat Rev Immunol. 2020;20(6):355‐362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303. Yang L, Xie X, Tu Z, Fu J, Xu D, Zhou Y. The signal pathways and treatment of cytokine storm in COVID‐19. Signal Transduct Target Ther. 2021;6(1):255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304. Borczuk AC, Yantiss RK. The pathogenesis of coronavirus‐19 disease. J Biomed Sci. 2022;29(1):87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305. Saravolatz LD, Depcinski S, Sharma M. Molnupiravir and nirmatrelvir‐ritonavir: oral coronavirus disease 2019 antiviral drugs. Clin Infect Dis. 2023;76(1):165‐171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306. Jiang Y, Zhao T, Zhou X, Xiang Y, Gutierrez‐Castrellon P, Ma X. Inflammatory pathways in COVID‐19: mechanism and therapeutic interventions. MedComm. 2022;3(3):e154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307. DeNardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol. 2019;19(6):369‐382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell. 2010;141(1):39‐51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309. Yang M, McKay D, Pollard JW, Lewis CE. Diverse functions of macrophages in different tumor microenvironments. Cancer Res. 2018;78(19):5492‐5503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310. Bruns H, Buttner M, Fabri M, et al. Vitamin D‐dependent induction of cathelicidin in human macrophages results in cytotoxicity against high‐grade B cell lymphoma. Sci Transl Med. 2015;7(282):282ra247. [DOI] [PubMed] [Google Scholar]
- 311. Mantovani A, Sica A. Macrophages, innate immunity and cancer: balance, tolerance, and diversity. Curr Opin Immunol. 2010;22(2):231‐237. [DOI] [PubMed] [Google Scholar]
- 312. Gabrilovich DI, Ostrand‐Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12(4):253‐268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 313. Li X, Yao W, Yuan Y, et al. Targeting of tumour‐infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut. 2017;66(1):157‐167. [DOI] [PubMed] [Google Scholar]
- 314. Bonapace L, Coissieux MM, Wyckoff J, et al. Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature. 2014;515(7525):130‐133. [DOI] [PubMed] [Google Scholar]
- 315. Nywening TM, Wang‐Gillam A, Sanford DE, et al. Targeting tumour‐associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: a single‐centre, open‐label, dose‐finding, non‐randomised, phase 1b trial. Lancet Oncol. 2016;17(5):651‐662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316. Pienta KJ, Machiels JP, Schrijvers D, et al. Phase 2 study of carlumab (CNTO 888), a human monoclonal antibody against CC‐chemokine ligand 2 (CCL2), in metastatic castration‐resistant prostate cancer. Invest New Drugs. 2013;31(3):760‐768. [DOI] [PubMed] [Google Scholar]
- 317. Aldinucci D, Borghese C, Casagrande N. The CCL5/CCR5 axis in cancer progression. Cancers (Basel). 2020;12(7):1765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318. Nie Y, Huang H, Guo M, et al. Breast phyllodes tumors recruit and repolarize tumor‐associated macrophages via secreting CCL5 to promote malignant progression, which can be inhibited by CCR5 inhibition therapy. Clin Cancer Res. 2019;25(13):3873‐3886. [DOI] [PubMed] [Google Scholar]
- 319. Halama N, Zoernig I, Berthel A, et al. Tumoral immune cell exploitation in colorectal cancer metastases can be targeted effectively by anti‐CCR5 therapy in cancer patients. Cancer Cell. 2016;29(4):587‐601. [DOI] [PubMed] [Google Scholar]
- 320. Haag GM, Springfeld C, Grun B, et al. Pembrolizumab and maraviroc in refractory mismatch repair proficient/microsatellite‐stable metastatic colorectal cancer—the PICCASSO phase I trial. Eur J Cancer. 2022;167:112‐122. [DOI] [PubMed] [Google Scholar]
- 321. Thomas RP, Nagpal S, Iv M, et al. Macrophage Exclusion after Radiation Therapy (MERT): a first in human phase I/II trial using a CXCR4 inhibitor in glioblastoma. Clin Cancer Res. 2019;25(23):6948‐6957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322. Ries CH, Cannarile MA, Hoves S, et al. Targeting tumor‐associated macrophages with anti‐CSF‐1R antibody reveals a strategy for cancer therapy. Cancer Cell. 2014;25(6):846‐859. [DOI] [PubMed] [Google Scholar]
- 323. DeNardo DG, Brennan DJ, Rexhepaj E, et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 2011;1(1):54‐67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324. Stafford JH, Hirai T, Deng L, et al. Colony stimulating factor 1 receptor inhibition delays recurrence of glioblastoma after radiation by altering myeloid cell recruitment and polarization. Neuro Oncol. 2016;18(6):797‐806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325. Xu J, Escamilla J, Mok S, et al. CSF1R signaling blockade stanches tumor‐infiltrating myeloid cells and improves the efficacy of radiotherapy in prostate cancer. Cancer Res. 2013;73(9):2782‐2794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 326. Seifert L, Werba G, Tiwari S, et al. Radiation therapy induces macrophages to suppress T‐cell responses against pancreatic tumors in mice. Gastroenterology. 2016;150(7):1659‐1672 e1655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327. Cassier PA, Italiano A, Gomez‐Roca CA, et al. CSF1R inhibition with emactuzumab in locally advanced diffuse‐type tenosynovial giant cell tumours of the soft tissue: a dose‐escalation and dose‐expansion phase 1 study. Lancet Oncol. 2015;16(8):949‐956. [DOI] [PubMed] [Google Scholar]
- 328. Tap WD, Wainberg ZA, Anthony SP, et al. Structure‐guided blockade of CSF1R kinase in tenosynovial giant‐cell tumor. N Engl J Med. 2015;373(5):428‐437. [DOI] [PubMed] [Google Scholar]
- 329. Tap WD, Gelderblom H, Palmerini E, et al. Pexidartinib versus placebo for advanced tenosynovial giant cell tumour (ENLIVEN): a randomised phase 3 trial. Lancet. 2019;394(10197):478‐487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330. Gomez‐Roca CA, Italiano A, Le Tourneau C, et al. Phase I study of emactuzumab single agent or in combination with paclitaxel in patients with advanced/metastatic solid tumors reveals depletion of immunosuppressive M2‐like macrophages. Ann Oncol. 2019;30(8):1381‐1392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331. Murata Y, Kotani T, Ohnishi H, Matozaki T. The CD47‐SIRPalpha signalling system: its physiological roles and therapeutic application. J Biochem. 2014;155(6):335‐344. [DOI] [PubMed] [Google Scholar]
- 332. Tian L, Xu B, Teng KY, et al. Targeting Fc receptor‐mediated effects and the “don't eat me” signal with an oncolytic virus expressing an anti‐CD47 Antibody to treat metastatic ovarian cancer. Clin Cancer Res. 2022;28(1):201‐214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 333. McCracken MN, Cha AC, Weissman IL. Molecular pathways: activating T cells after cancer cell phagocytosis from blockade of CD47 “don't eat me” signals. Clin Cancer Res. 2015;21(16):3597‐3601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 334. Pan Y, Lu F, Fei Q, et al. Single‐cell RNA sequencing reveals compartmental remodeling of tumor‐infiltrating immune cells induced by anti‐CD47 targeting in pancreatic cancer. J Hematol Oncol. 2019;12(1):124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335. Advani R, Flinn I, Popplewell L, et al. CD47 blockade by Hu5F9‐G4 and rituximab in non‐Hodgkin's lymphoma. N Engl J Med. 2018;379(18):1711‐1721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336. Zeidan AM, DeAngelo DJ, Palmer J, et al. Phase 1 study of anti‐CD47 monoclonal antibody CC‐90002 in patients with relapsed/refractory acute myeloid leukemia and high‐risk myelodysplastic syndromes. Ann Hematol. 2022;101(3):557‐569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 337. Peluso MO, Adam A, Armet CM, et al. The fully human anti‐CD47 antibody SRF231 exerts dual‐mechanism antitumor activity via engagement of the activating receptor CD32a. J Immunother Cancer. 2020;8(1):e000413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 338. Chen SH, Dominik PK, Stanfield J, et al. Dual checkpoint blockade of CD47 and PD‐L1 using an affinity‐tuned bispecific antibody maximizes antitumor immunity. J Immunother Cancer. 2021;9(10):e003464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339. Hou L, Pu L, Chen Y, et al. Targeted intervention of NF2‐YAP signaling axis in CD24‐overexpressing cells contributes to encouraging therapeutic effects in TNBC. ACS Nano. 2022;16(4):5807‐5819. [DOI] [PubMed] [Google Scholar]
- 340. Barkal AA, Brewer RE, Markovic M, et al. CD24 signalling through macrophage Siglec‐10 is a target for cancer immunotherapy. Nature. 2019;572(7769):392‐396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 341. Zhang M, Liu ZZ, Aoshima K, et al. CECR2 drives breast cancer metastasis by promoting NF‐κB signaling and macrophage‐mediated immune suppression. Sci Transl Med. 2022;14(630):eabf5473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 342. Tan Y, Sun R, Liu L, et al. Tumor suppressor DRD2 facilitates M1 macrophages and restricts NF‐κB signaling to trigger pyroptosis in breast cancer. Theranostics. 2021;11(11):5214‐5231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 343. Maeda S, Kamata H, Luo JL, Leffert H, Karin M. IKKbeta couples hepatocyte death to cytokine‐driven compensatory proliferation that promotes chemical hepatocarcinogenesis. Cell. 2005;121(7):977‐990. [DOI] [PubMed] [Google Scholar]
- 344. Xiao H, Guo Y, Li B, et al. M2‐like tumor‐associated macrophage‐targeted codelivery of STAT6 inhibitor and IKKbeta siRNA induces M2‐to‐M1 repolarization for cancer immunotherapy with low immune side effects. ACS Cent Sci. 2020;6(7):1208‐1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 345. Joshi S, Liu KX, Zulcic M, et al. Macrophage Syk‐PI3Kgamma inhibits antitumor immunity: SRX3207, a novel dual Syk‐PI3K inhibitory chemotype relieves tumor immunosuppression. Mol Cancer Ther. 2020;19(3):755‐764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 346. Hobbs GA, Baker NM, Miermont AM, et al. Atypical KRAS(G12R) mutant is impaired in PI3K signaling and macropinocytosis in pancreatic cancer. Cancer Discov. 2020;10(1):104‐123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 347. De Henau O, Rausch M, Winkler D, et al. Overcoming resistance to checkpoint blockade therapy by targeting PI3Kgamma in myeloid cells. Nature. 2016;539(7629):443‐447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 348. Flinn IW, Miller CB, Ardeshna KM, et al. DYNAMO: a phase II study of duvelisib (IPI‐145) in patients with refractory indolent non‐Hodgkin lymphoma. J Clin Oncol. 2019;37(11):912‐922. [DOI] [PubMed] [Google Scholar]
- 349. Konstantinopoulos PA, Barry WT, Birrer M, et al. Olaparib and alpha‐specific PI3K inhibitor alpelisib for patients with epithelial ovarian cancer: a dose‐escalation and dose‐expansion phase 1b trial. Lancet Oncol. 2019;20(4):570‐580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 350. Fowler NH, Samaniego F, Jurczak W, et al. Umbralisib, a dual PI3Kdelta/CK1epsilon inhibitor in patients with relapsed or refractory indolent lymphoma. J Clin Oncol. 2021;39(15):1609‐1618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351. Li B, Xi P, Wang Z, et al. PI3K/Akt/mTOR signaling pathway participates in Streptococcus uberis‐induced inflammation in mammary epithelial cells in concert with the classical TLRs/NF‐kB pathway. Vet Microbiol. 2018;227:103‐111. [DOI] [PubMed] [Google Scholar]
- 352. Akhter N, Hasan A, Shenouda S, et al. TLR4/MyD88 ‐mediated CCL2 production by lipopolysaccharide (endotoxin): implications for metabolic inflammation. J Diabetes Metab Disord. 2018;17(1):77‐84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 353. Zhou D, Huang C, Lin Z, et al. Macrophage polarization and function with emphasis on the evolving roles of coordinated regulation of cellular signaling pathways. Cell Signal. 2014;26(2):192‐197. [DOI] [PubMed] [Google Scholar]
- 354. Bart VMT, Pickering RJ, Taylor PR, Ipseiz N. Macrophage reprogramming for therapy. Immunology. 2021;163(2):128‐144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 355. Zhao C, Pang X, Yang Z, Wang S, Deng H, Chen X. Nanomaterials targeting tumor associated macrophages for cancer immunotherapy. J Control Release. 2022;341:272‐284. [DOI] [PubMed] [Google Scholar]
- 356. Zhu S, Niu M, O'Mary H, Cui Z. Targeting of tumor‐associated macrophages made possible by PEG‐sheddable, mannose‐modified nanoparticles. Mol Pharm. 2013;10(9):3525‐3530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 357. Zhao Y, Yang Y, Zhang J, et al. Lactoferrin‐mediated macrophage targeting delivery and patchouli alcohol‐based therapeutic strategy for inflammatory bowel diseases. Acta Pharmaceutica Sinica B. 2020;10(10):1966‐1976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 358. van Bommel PE, He Y, Schepel I, et al. CD20‐selective inhibition of CD47‐SIRPalpha “don't eat me” signaling with a bispecific antibody‐derivative enhances the anticancer activity of daratumumab, alemtuzumab and obinutuzumab. Oncoimmunology. 2018;7(2):e1386361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 359. Wang Y, Ni H, Zhou S, et al. Tumor‐selective blockade of CD47 signaling with a CD47/PD‐L1 bispecific antibody for enhanced anti‐tumor activity and limited toxicity. Cancer Immunol Immunother. 2021;70(2):365‐376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 360. de Silva S, Fromm G, Shuptrine CW, et al. CD40 enhances type i interferon responses downstream of CD47 blockade, bridging innate and adaptive immunity. Cancer Immunol Res. 2020;8(2):230‐245. [DOI] [PubMed] [Google Scholar]
- 361. Long KB, Gladney WL, Tooker GM, Graham K, Fraietta JA, Beatty GL. IFNγ and CCL2 cooperate to redirect tumor‐infiltrating monocytes to degrade fibrosis and enhance chemotherapy efficacy in pancreatic carcinoma. Cancer Discovery. 2016;6(4):400‐413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 362. Radner H, Aletaha D. Anti‐TNF in rheumatoid arthritis: an overview. Wien Med Wochenschr. 2015;165(1‐2):3‐9. [DOI] [PubMed] [Google Scholar]
- 363. Lamb CA, Kennedy NA, Raine T, et al. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut. 2019;68(Suppl 3):s1‐s106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 364. Velez MP, McCarthy MW. Infliximab as a potential treatment for COVID‐19. Expert Rev Anti Infect Ther. 2023;21(1):1‐5. [DOI] [PubMed] [Google Scholar]
- 365. Fakharian A, Barati S, Mirenayat M, et al. Evaluation of adalimumab effects in managing severe cases of COVID‐19: a randomized controlled trial. Int Immunopharmacol. 2021;99:107961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 366. Srivastava S, Rasool M. Underpinning IL‐6 biology and emphasizing selective JAK blockade as the potential alternate therapeutic intervention for rheumatoid arthritis. Life Sci. 2022;298:120516. [DOI] [PubMed] [Google Scholar]
- 367. Declercq J, Van Damme KFA, De Leeuw E, et al. Effect of anti‐interleukin drugs in patients with COVID‐19 and signs of cytokine release syndrome (COV‐AID): a factorial, randomised, controlled trial. Lancet Respir Med. 2021;9(12):1427‐1438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 368. Takeuchi T, Thorne C, Karpouzas G, et al. Sirukumab for rheumatoid arthritis: the phase III SIRROUND‐D study. Ann Rheum Dis. 2017;76(12):2001‐2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 369. Smolen JS, Feist E, Fatenejad S, et al. Olokizumab versus placebo or adalimumab in rheumatoid arthritis. N Engl J Med. 2022;387(8):715‐726. [DOI] [PubMed] [Google Scholar]
- 370. Weinblatt ME, Mease P, Mysler E, et al. The efficacy and safety of subcutaneous clazakizumab in patients with moderate‐to‐severe rheumatoid arthritis and an inadequate response to methotrexate: results from a multinational, phase IIb, randomized, double‐blind, placebo/active‐controlled, dose‐ranging study. Arthritis Rheumatol. 2015;67(10):2591‐2600. [DOI] [PubMed] [Google Scholar]
- 371. Lonze BE, Spiegler P, Wesson RN, et al. A randomized double‐blinded placebo controlled trial of clazakizumab for the treatment of COVID‐19 pneumonia with hyperinflammation. Crit Care Med. 2022;50(9):1348‐1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 372. Trøseid M, Arribas JR, Assoumou L, et al. Efficacy and safety of baricitinib in hospitalized adults with severe or critical COVID‐19 (Bari‐SolidAct): a randomised, double‐blind, placebo‐controlled phase 3 trial. Crit Care. 2023;27(1):9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 373. Fleischmann R, Genovese MC, Maslova K, Leher H, Praestgaard A, Burmester GR. Long‐term safety and efficacy of sarilumab over 5 years in patients with rheumatoid arthritis refractory to TNF inhibitors. Rheumatology (Oxford, England). 2021;60(11):4991‐5001. [DOI] [PubMed] [Google Scholar]
- 374. Lomakin NV, Bakirov BA, Protsenko DN, et al. The efficacy and safety of levilimab in severely ill COVID‐19 patients not requiring mechanical ventilation: results of a multicenter randomized double‐blind placebo‐controlled phase III CORONA clinical study. Inflamm Res. 2021;70(10‐12):1233‐1246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 375. Fanlo P, Gracia‐Tello BDC, Fonseca Aizpuru E, et al. Efficacy and safety of anakinra plus standard of care for patients with severe COVID‐19: a randomized phase 2/3 clinical trial. JAMA Netw Open. 2023;6(4):e237243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 376. Alten R, Gomez‐Reino J, Durez P, et al. Efficacy and safety of the human anti‐IL‐1β monoclonal antibody canakinumab in rheumatoid arthritis: results of a 12‐week, phase II, dose‐finding study. BMC Musculoskelet Disord. 2011;12:153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 377. Everett BM, Donath MY, Pradhan AD, et al. Anti‐inflammatory therapy with canakinumab for the prevention and management of diabetes. J Am Coll Cardiol. 2018;71(21):2392‐2401. [DOI] [PubMed] [Google Scholar]
- 378. Caricchio R, Abbate A, Gordeev I, et al. Effect of canakinumab vs placebo on survival without invasive mechanical ventilation in patients hospitalized with severe COVID‐19: a randomized clinical trial. JAMA. 2021;326(3):230‐239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 379. Smolen JS, Agarwal SK, Ilivanova E, et al. A randomised phase II study evaluating the efficacy and safety of subcutaneously administered ustekinumab and guselkumab in patients with active rheumatoid arthritis despite treatment with methotrexate. Ann Rheum Dis. 2017;76(5):831‐839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 380. Sands BE, Peyrin‐Biroulet L, Kierkus J, et al. Efficacy and safety of mirikizumab in a randomized phase 2 study of patients with Crohn's disease. Gastroenterology. 2022;162(2):495‐508. [DOI] [PubMed] [Google Scholar]
- 381. Feagan BG, Sands BE, Sandborn WJ, et al. Guselkumab plus golimumab combination therapy versus guselkumab or golimumab monotherapy in patients with ulcerative colitis (VEGA): a randomised, double‐blind, controlled, phase 2, proof‐of‐concept trial. Lancet Gastroenterol Hepatol. 2023;8(4):307‐320. [DOI] [PubMed] [Google Scholar]
- 382. Behrens F, Tak PP, Østergaard M, et al. 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. Ann Rheum Dis. 2015;74(6):1058‐1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 383. Taylor PC, Saurigny D, Vencovsky J, et al. 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. Arthritis Res Ther. 2019;21(1):101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 384. Criner GJ, Lang FM, Gottlieb RL, et al. Anti‐granulocyte‐macrophage colony‐stimulating factor monoclonal antibody gimsilumab for COVID‐19 pneumonia: a randomized, double‐blind, placebo‐controlled trial. Am J Respir Crit Care Med. 2022;205(11):1290‐1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 385. Patel J, Bass D, Beishuizen A, et al. A randomised trial of anti‐GM‐CSF otilimab in severe COVID‐19 pneumonia (OSCAR). Eur Respir J. 2023;61(2):2101870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 386. Temesgen Z, Burger CD, Baker J, et al. Lenzilumab in hospitalised patients with COVID‐19 pneumonia (LIVE‐AIR): a phase 3, randomised, placebo‐controlled trial. Lancet Respir Med. 2022;10(3):237‐246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 387. Weinblatt ME, McInnes IB, Kremer JM, et al. A randomized phase iib study of mavrilimumab and golimumab in rheumatoid arthritis. Arthritis Rheumatol. 2018;70(1):49‐59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 388. Gaylis NB, Ritter A, Kelly SA, et al. Reduced Cell surface levels of C‐C chemokine receptor 5 and immunosuppression in long coronavirus disease 2019 syndrome. Clin Infec Dis. 2022;75(7):1232‐1234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 389. Bockorny B, Semenisty V, Macarulla T, et al. BL‐8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial. Nat Med. 2020;26(6):878‐885. [DOI] [PubMed] [Google Scholar]
- 390. Tsukahara K. Contribution to development of remedies for COVID‐19: focusing on eritoran. Nihon Yakurigaku Zasshi. 2022;157(1):38‐40. [DOI] [PubMed] [Google Scholar]
- 391. Brennan CM, Nadella S, Zhao X, et al. Oral famotidine versus placebo in non‐hospitalised patients with COVID‐19: a randomised, double‐blind, data‐intense, phase 2 clinical trial. Gut. 2022;71(5):879‐888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 392. Burns DK, Alexander RC, Welsh‐Bohmer KA, et al. Safety and efficacy of pioglitazone for the delay of cognitive impairment in people at risk of Alzheimer's disease (TOMMORROW): a prognostic biomarker study and a phase 3, randomised, double‐blind, placebo‐controlled trial. Lancet Neurol. 2021;20(7):537‐547. [DOI] [PubMed] [Google Scholar]
- 393. Cusi K, Orsak B, Bril F, et al. Long‐term pioglitazone treatment for patients with nonalcoholic steatohepatitis and prediabetes or type 2 diabetes mellitus: a randomized trial. Ann Intern Med. 2016;165(5):305‐315. [DOI] [PubMed] [Google Scholar]
- 394. Mantovani A, Byrne CD, Targher G. Efficacy of peroxisome proliferator‐activated receptor agonists, glucagon‐like peptide‐1 receptor agonists, or sodium‐glucose cotransporter‐2 inhibitors for treatment of non‐alcoholic fatty liver disease: a systematic review. Lancet Gastroenterol Hepatol. 2022;7(4):367‐378. [DOI] [PubMed] [Google Scholar]
- 395. O'Bryant SE, Zhang F, Petersen M, Johnson L, Hall J, Rissman RA. A precision medicine approach to treating Alzheimer's disease using rosiglitazone therapy: a biomarker analysis of the REFLECT trials. J Alzheimers Dis. 2021;81(2):557‐568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 396. Gross B, Pawlak M, Lefebvre P, Staels B. PPARs in obesity‐induced T2DM, dyslipidaemia and NAFLD. Nat Rev Endocrinol. 2017;13(1):36‐49. [DOI] [PubMed] [Google Scholar]
- 397. Heppner KM, Perez‐Tilve D. GLP‐1 based therapeutics: simultaneously combating T2DM and obesity. Front Neurosci. 2015;9:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 398. De Kleijn KMA, Martens GJM. Molecular effects of FDA‐approved multiple sclerosis drugs on glial cells and neurons of the central nervous system. Int J Mol Sci. 2020;21(12):4229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 399. Teymouri S, Pourbayram Kaleybar S, Hejazian SS, et al. The effect of Fingolimod on patients with moderate to severe COVID‐19. Pharmacol Res Perspect. 2023;11(1):e01039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 400. Samimagham HR, Hassani Azad M, Haddad M, Arabi M, Hooshyar D, KazemiJahromi M. The Efficacy of Famotidine in improvement of outcomes in Hospitalized COVID‐19 Patients: a structured summary of a study protocol for a randomised controlled trial. Trials. 2020;21(1):848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 401. Montalban X, Wallace D, Genovese MC, et al. Characterisation of the safety profile of evobrutinib in over 1000 patients from phase II clinical trials in multiple sclerosis, rheumatoid arthritis and systemic lupus erythematosus: an integrated safety analysis. J Neurol Neurosurg Psychiatry. 2023;94(1):1‐9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 402. Aletaha D, Smolen JS. Diagnosis and management of rheumatoid arthritis: a review. JAMA. 2018;320(13):1360‐1372. [DOI] [PubMed] [Google Scholar]
- 403. Higher dose corticosteroids in patients admitted to hospital with COVID‐19 who are hypoxic but not requiring ventilatory support (RECOVERY): a randomised, controlled, open‐label, platform trial. Lancet (London, England). 2023;401(10387):1499‐1507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 404. Lazzerini M, Martelossi S, Magazzù G, et al. Effect of thalidomide on clinical remission in children and adolescents with refractory Crohn disease: a randomized clinical trial. JAMA. 2013;310(20):2164‐2173. [DOI] [PubMed] [Google Scholar]
- 405. Miller RG, Zhang R, Bracci PM, et al. Phase 2B randomized controlled trial of NP001 in amyotrophic lateral sclerosis: pre‐specified and post hoc analyses. Muscle Nerve. 2022;66(1):39‐49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 406. Tanaka Y, Luo Y, O'Shea JJ, Nakayamada S. Janus kinase‐targeting therapies in rheumatology: a mechanisms‐based approach. Nat Rev Rheumatol. 2022;18(3):133‐145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 407. Guimarães PO, Quirk D, Furtado RH, et al. Tofacitinib in patients hospitalized with Covid‐19 pneumonia. N Engl J Med. 2021;385(5):406‐415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 408. Kalil AC, Patterson TF, Mehta AK, et al. Baricitinib plus remdesivir for hospitalized adults with Covid‐19. N Engl J Med. 2021;384(9):795‐807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 409. Takeuchi T, Tanaka Y, Tanaka S, et al. Efficacy and safety of peficitinib (ASP015K) in patients with rheumatoid arthritis and an inadequate response to methotrexate: results of a phase III randomised, double‐blind, placebo‐controlled trial (RAJ4) in Japan. Ann Rheum Dis. 2019;78(10):1305‐1319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 410. Sands BE, Sandborn WJ, Feagan BG, et al. Peficitinib, an Oral Janus Kinase inhibitor, in moderate‐to‐severe ulcerative colitis: results from a randomised, phase 2 study. J Crohns Colitis. 2018;12(10):1158‐1169. [DOI] [PubMed] [Google Scholar]
- 411. Silverberg JI, de Bruin‐Weller M, Bieber T, et al. Upadacitinib plus topical corticosteroids in atopic dermatitis: week 52 AD Up study results. J Allergy Clin Immunol. 2022;149(3):977‐987. [DOI] [PubMed] [Google Scholar]
- 412. Combe B, Kivitz A, Tanaka Y, et al. Filgotinib versus placebo or adalimumab in patients with rheumatoid arthritis and inadequate response to methotrexate: a phase III randomised clinical trial. Ann Rheum Dis. 2021;80(7):848‐858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 413. Feagan BG, Danese S, Loftus EV, et al. Filgotinib as induction and maintenance therapy for ulcerative colitis (SELECTION): a phase 2b/3 double‐blind, randomised, placebo‐controlled trial. Lancet (London, England). 2021;397(10292):2372‐2384. [DOI] [PubMed] [Google Scholar]
- 414. Cafardi J, Miller C, Terebelo H, et al. Efficacy and safety of pacritinib vs placebo for patients with severe COVID‐19: a phase 2 randomized clinical trial. JAMA Netw Open. 2022;5(12):e2242918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 415. Rein L, Calero K, Shah R, et al. Randomized Phase 3 trial of ruxolitinib for COVID‐19‐associated acute respiratory distress syndrome. Crit Care Med. 2022;50(12):1701‐1713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 416. Belperio J, Nguyen T, Lombardi DA, et al. Efficacy and safety of an inhaled pan‐Janus kinase inhibitor, nezulcitinib, in hospitalised patients with COVID‐19: results from a phase 2 clinical trial. BMJ Open Respir Res. 2023;10(1):e001627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 417. Broccoli A, Argnani L, Zinzani PL. Peripheral T‐cell lymphomas: focusing on novel agents in relapsed and refractory disease. Cancer Treat Rev. 2017;60:120‐129. [DOI] [PubMed] [Google Scholar]
- 418. Bramante CT, Huling JD, Tignanelli CJ, et al. Randomized trial of metformin, ivermectin, and fluvoxamine for Covid‐19. N Engl J Med. 2022;387(7):599‐610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 419. Mannick JB, Teo G, Bernardo P, et al. Targeting the biology of ageing with mTOR inhibitors to improve immune function in older adults: phase 2b and phase 3 randomised trials. Lancet Healthy Longev. 2021;2(5):e250‐e262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 420. Eikelboom JW, Jolly SS, Belley‐Cote EP, et al. Colchicine and the combination of rivaroxaban and aspirin in patients hospitalised with COVID‐19 (ACT): an open‐label, factorial, randomised, controlled trial. Lancet Respir Med. 2022;10(12):1169‐1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 421. Tsalik EL, Rouphael NG, Sadikot RT, et al. Efficacy and safety of azithromycin versus placebo to treat lower respiratory tract infections associated with low procalcitonin: a randomised, placebo‐controlled, double‐blind, non‐inferiority trial. Lancet Infect Dis. 2023;23(4):484‐495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 422. Mousavi SA, Heydari K, Mehravaran H, et al. Melatonin effects on sleep quality and outcomes of COVID‐19 patients: an open‐label, randomized, controlled trial. J Med Virol. 2022;94(1):263‐271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 423. Colombel JF, Sandborn WJ, Reinisch W, et al. Infliximab, azathioprine, or combination therapy for Crohn's disease. N Engl J Med. 2010;362(15):1383‐1395. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.
