Abstract
Background:
Skin fibrosis, characterized by excessive fibroblast proliferation and extracellular matrix deposition in the dermis, is the histopathologic hallmark of dermatologic diseases such as systemic sclerosis, hypertrophic scars, and keloids. Effective anti-scarring therapeutics remain an unmet need, underscoring the complex pathophysiologic mechanisms of skin fibrosis. The Th2 cytokines interleukin (IL)-4 and IL-13 have been implicated as key mediators in the pathogenesis of fibroproliferative disorders.
Objective:
To summarize the current understanding of the role of the IL-4/IL-13 axis in wound healing and skin fibrosis.
Methods:
A literature search identified research studies investigating the roles of IL-4 and IL-13 in fibrotic skin diseases.
Results:
While transforming growth factor-beta has long been regarded as the main driver of fibrotic processes, research into the cellular and molecular biology of wound healing has revealed other pathways that promote scar tissue formation. IL-4 and IL-13 are important mediators of skin fibrosis, supported by evidence from in vitro data, animal models of fibrosis, and clinical studies. Overactive signaling of the IL-4/IL-13 axis contributes to the initiation and perpetuation of fibrotic skin diseases.
Conclusion:
Further insights into the IL-4/IL-13 axis may reveal potential targets for the development of novel therapies that prevent or treat fibrotic skin diseases.
Keywords: cytokines, interleukin-4, interleukin-13, skin fibrosis, scarring
Introduction
Tissue fibrosis is a leading cause of morbidity and mortality worldwide, with an estimated 45% of all deaths in the United States being attributed to fibroproliferative disorders (FPDs) [36, 145]. Fibrosis represents an exaggerated wound healing response following tissue damage, which may be triggered by a variety of stimuli such as infection, autoimmune reaction, and mechanical injury [143, 147]. Characterized by excessive fibroblast proliferation and collagen-rich extracellular matrix (ECM) deposition, fibrosis can affect any organ, leading to permanent scarring and impaired organ function [143]. Skin fibrosis is the histopathologic hallmark of a wide spectrum of dermatologic diseases, including systemic sclerosis (SSc; scleroderma), chronic graft-versus-host disease (GVHD), nephrogenic fibrosing dermopathy, scleromyxedema, and restrictive dermopathy [76, 134]. Localized skin fibrosis can also develop as a sequela of dermal injury (e.g., burns, surgery, trauma, infection, radiation), manifested clinically as hypertrophic scars or keloids [66, 130].
Skin fibrosis is a significant global health problem affecting over 100 million persons per year in the developed world [8, 17]. Cutaneous scars have a profoundly negative impact on patients’ quality of life due to associated pain and pruritus, functional impairment, cosmetic disfigurement, and psychosocial distress [8, 11, 15]. There is no universal consensus on optimal scar management despite high demand for modalities that prevent, reduce, or remove scars, as evidenced by an estimated $12 billion annual market for scar treatment in the US [121, 130]. Current anti-scarring therapeutics have limited clinical efficacy and varying levels of research evidence to support their use, making scar treatment a major unmet need [38, 133]. While significant advances have been made in our understanding of wound healing biology, the exact pathophysiologic mechanisms underlying fibrosis remain unclear.
Research on cutaneous scarring has focused on identifying new therapeutic targets, such as profibrotic cytokines, to develop novel pharmacologic agents that may facilitate optimal or scarless wound healing [42, 79, 136]. Cytokines are signaling molecules of the immune system that regulate fundamental biological processes such as host defense, inflammation, cell growth, angiogenesis, and tissue repair [103]. The T-helper type 2 (Th2) cytokines interleukin (IL)-4 and IL-13 have been implicated in the pathogenesis of FPDs such as pulmonary fibrosis, cirrhosis, myocardial fibrosis, progressive kidney disease, pathological cutaneous scars, and SSc [36, 143].
SSc is an autoimmune connective tissue disorder characterized by chronic inflammation, small vessel vasculopathy, and progressive fibrosis of the skin and internal organs [97]. Since FPDs share common pathomechanistic pathways, SSc is often used as the prototypical fibrotic disease to study the cellular and molecular mechanisms underlying fibrosis [1, 135]. The most thoroughly investigated animal model of fibrosis is the spontaneously occurring tight-skin (TSK/+) mutant mouse, which exhibits a SSc-like syndrome with clinical features including diffuse cutaneous hyperplasia, myocardial hypertrophy, and pulmonary emphysema [10, 26]. The TSK/+ mouse model has been instrumental in revealing the profibrotic effects of IL-4 and IL-13, and will be referred to throughout this review [76].
Herein, we discuss how dysregulated wound healing and Th2-associated immune responses contribute to the pathogenesis of fibrosis. We then review the biological characteristics of IL-4 and IL-13, their signaling pathways, and their pathogenic roles in skin fibrosis as elucidated by laboratory and clinical investigations. This review article serves to highlight the current evidence and therapeutic potential of targeting the IL-4/IL-13 axis for skin fibrosis, as novel biologic agents that inhibit IL-4/IL-13 and their downstream mediators have been recently approved or are currently under development with promising results for the treatment of various allergic and immunologic diseases [57, 115].
Cutaneous Wound Healing
Wound healing is a complex and dynamic process that involves highly regulated pathways to restore tissue architecture and function after injury. Physiologic cutaneous wound healing is characterized by three distinct yet overlapping phases: inflammation (immediate to 1–3 days), proliferation (4 days to 3 weeks), and remodeling (3 weeks to over a year) [42, 105].
The inflammatory phase aims to contain the injury, form a fibrin hemostatic plug, remove tissue debris, and prevent infection [105]. The proliferative phase is characterized by the migration of reparative cells into the provisional wound matrix, angiogenesis, re-epithelialization, formation of granulation tissue, and progressive restoration of tissue function [61, 62]. The predominant cells in granulation tissue are fibroblasts, which produce ECM proteins such as type I collagen, type III collagen, and fibronectin, forming a structural framework for cell adhesion and differentiation [105, 125]. In response to growth factors and cytokines, fibroblasts proliferate and transform into myofibroblasts, which express alpha smooth actin (α-SMA) and are key players of normal tissue repair and fibrosis [25, 143]. Compared to fibroblasts, myofibroblasts have increased activity of ECM biosynthesis and enhanced contractility to induce wound closure [24, 149]. During the remodeling phase, the ECM is reorganized to provide more strength and elasticity to connective tissue, the orientation of collagen fibrils becomes more uniform, and the tensile strength of the wound increases [10, 79].
Normally, the ECM is in a constant flux of remodeling by resident quiescent fibroblasts, whereby synthesis and degradation of ECM components are closely regulated to maintain a homeostatic balance [10, 147]. The perturbation of any wound healing phase can shift this balance to a pathological state of “over repair” or “over healing,” characterized by excessive collagen deposition and failure to restore tissue function, resulting in fibrosis [42, 147]. While scar tissue formation is an expected outcome of wound healing after skin injury, it can range from minimally visible scarring to severe thickening and tightening of the skin [55]. In fibrosis, myofibroblasts drive excessive collagen production in a dysregulated manner and resist the induction of apoptosis [76]. Therefore, it is important to understand the cytokine networks that influence fibroblast activity in normal and pathological wound healing.
Type 2 Immunity and Fibrosis
The immune system plays an essential regulatory role in balancing wound repair and tissue regeneration following insults such as infection and mechanical injury [62, 144]. Type 1 (Th1-mediated) and type 2 (Th2-mediated) immune responses involve opposing effector functions and are each characterized by a distinct repertoire of cytokines (Figure 1) [75]. Pro-inflammatory Th1 cytokines (interferon-gamma, tumor necrosis factor-beta, IL-2 and IL-12) drive cellular immunity and phagocyte-dependent inflammation, which are critical for host defense against intracellular pathogens [36, 103]. In contrast, anti-inflammatory Th2 cytokines (IL-4, IL-5, IL-9, IL-10, and IL-13) evoke strong humoral responses and promote barrier defenses, involved in protection against extracellular parasites [2, 36]. The type 2 immune response is classically viewed as a counterbalance for the tissue damage and inflammation caused by type 1 immunity [34, 73].
Figure 1.

Type 1 immunity, characterized by pro-inflammatory Th1 cytokines, promotes tissue-damaging inflammation as a defense against intracellular pathogens. Type 2 immunity, characterized by anti-inflammatory Th2 cytokines, helps to counter Th1-mediated inflammation and regulates tissue repair following injury. Normally, type 1 and type 2 immune responses are tightly regulated to maintain homeostasis. Over-activation of either immune response may lead to various disease states. For example, disproportionately increased activity of the Th2 cytokines IL-4 and IL-13 contributes to the pathogenesis of fibroproliferative disorders. IFN-γ, interferon-gamma; IL, interleukin; TNF-β, tumor necrosis factor-beta
In addition to suppressing type 1-driven inflammation, type 2 immunity is closely involved in many aspects of wound healing through the production of mediators that facilitate tissue repair and regeneration [2, 34, 36]. Th2 cytokines upregulate genes known to be involved in the mechanisms of wound healing and fibrosis [51]. When Th2 cytokine responses become overactive, chronic, or dysregulated, they may drive overzealous repair mechanisms that trigger disease states such as allergy and fibrosis (Figure 1) [34, 36, 131, 144]. Specifically, IL-4 and IL-13 have been identified as potent mediators that are critical to the induction and perpetuation of atopic diseases and FPDs [33, 144].
IL-4 and IL-13 in Skin Fibrosis and Scarring
IL-4 and IL-13 have similar and distinct biological characteristics
The IL-4/IL-13 axis has been studied extensively as a potential therapeutic target in Th2-driven diseases such as asthma, atopic dermatitis, and cancer [80, 144]. More recently, IL-4 and IL-13 have gained attention for their important roles in tissue repair and fibrosis. Persistent activation of IL-4 and IL-13 signaling leads to abnormal collagen homeostasis and has been implicated in the pathogenesis of FPDs [36, 100, 145].
IL-4 and IL-13 are short α-helix bundle secreted glycoproteins with approximately 30% homology, encoded by a cytokine gene cluster on chromosome 5q [10, 82]. They are mainly produced by Th2-polarized T cells but are also secreted by natural killer T cells, activated mast cells, eosinophils, basophils, macrophages, and dendritic cells [40, 53]. IL-4 and IL-13 elicit many similar biological responses since they share a common receptor chain, IL-4 receptor alpha (IL-4Rα), and the Janus kinase/signal transducer and activator of transcription protein 6 (JAK/STAT6) signaling pathway [145]. In cutaneous wound healing, IL-4 and IL-13 promote fibroblast chemotaxis and proliferation, myofibroblast differentiation, and production of collagen and ECM macromolecules [14, 118, 145]. Although IL-4 and IL-13 share many effector functions, mechanistic studies conducted with knockout mice and neutralizing antibodies have revealed their distinct and non-redundant roles in the regulation of tissue repair [21, 143, 146].
IL-4 and IL-13 activate the IL-4Rα/STAT6 pathway to promote fibrosis
IL-4 and IL-13 can influence almost all cell types as their surface receptors are widely expressed [82, 141]. IL-4 and IL-13 signal through receptor heterodimers composed of three possible subunits: IL-4Rα, the common gamma chain (γc), and IL-13Rα1 (Figure 2) [40, 82]. IL-4 and IL-13 both share the type I receptor complex composed of IL-4Rα and γc. IL-4 also binds to the type II receptor composed of IL-4Rα and IL-13Rα1. IL-13 binds with much higher affinity to the IL-13Rα2 subunit, which is traditionally considered a “decoy” receptor as it has a short cytoplasmic tail with no recognizable signaling motifs [16, 20]. However, Fichtner-Feigl et al. found that IL-13 can activate IL-13Rα2 to induce transforming growth factor-beta (TGF-β) production and ultimately fibrosis [29, 77].
Figure 2.

IL-4 and IL-13 share the type I receptor complex composed of the IL-4Rα chain and the common gamma chain (γc). IL-4 also binds to the type II receptor complex composed of IL-4Rα and the IL-13Rα1 chain. IL-13 binds with high affinity to the IL-13Rα2 subunit that is traditionally considered a “decoy” monomeric receptor, but has been shown to participate in signal transduction.
The binding of IL-4 and IL-13 to their cognate receptors results in JAK activation, with type I receptors activating JAK1/JAK3 and type II receptors activating JAK1 and TYK2/JAK2 [53]. Activated JAKs initiate several intracellular signaling cascades by phosphorylating specific tyrosine residues in the cytoplasmic domain of the IL-4Rα subunit [21, 82]. The phosphorylated tyrosine residues act as docking sites for signaling molecules such as STAT6, which forms homodimers and translocates to the nucleus, activating IL-4- and IL-13-responsive genes (Figure 3) [21, 82]. In the context of fibrosis, IL-4 and IL-13 upregulate the promoter activity and transcription of profibrotic genes such as type I collagen and TGF-β, the prototypical profibrotic cytokine [10, 58]. Thus, overactivity of IL-4 and IL-13 leads to stimulation of profibrotic pathways that drive excessive fibroblast proliferation and ECM deposition.
Figure 3.
IL-4 and IL-13 binding to their cognate cell surface receptors activates the cytoplasmic domain of IL-4Rα and associated JAK1, creating docking sites for signaling molecules such as STAT6. Phosphorylated STAT6 monomers form homodimers and translocate to the nucleus, leading to the activation of target genes involved in fibrosis.
The IL-4Rα/STAT6 signaling pathway has been investigated as a potential therapeutic target to block the progression of FPDs [71, 139]. Alternatively activated (M2) macrophages induced by IL-4Rα signaling has been proposed as a means by which IL-4 and IL-13 mediate skin fibrosis [145, 147]. IL-4Rα-dependent M2 macrophages are found in abundance during chronic Th2 cytokine responses, and during skin repair, they direct wound resolution via collagen fibril assembly and cross-linking [59, 74, 148]. Prolonged infiltration of M2 macrophages has been observed in hypertrophic scars, and the depletion of these macrophages during the subacute phase of wound healing allows normal scarring in mice with human skin transplants [62, 149]. In injured mouse skin, the IL-4Rα signaling pathway is critical for effective wound repair, and overstimulation of IL-4Rα can result in exuberant scar formation [59]. In addition, intact IL-4Rα signaling is required to induce a rapid elevation of proteins associated with Th2-mediated tissue repair in a surgical mouse model [2, 74]. IL-4Rα is more highly expressed in TSK/+ murine fibroblasts and SSc dermal fibroblasts compared with normal controls, resulting in stronger activation of STAT6 upon cytokine stimulation and correlating with increased collagen gene activity [84, 85]. Targeted deletion of the IL-4Rα gene in TSK/+ mice ameliorates the key fibrotic characteristics of the TSK syndrome including dermal thickening, further supporting IL-4Rα’s critical role in the pathogenesis of skin fibrosis [83].
IL-4 and IL-13 exert profibrotic effects through TGF-β, a central mediator of fibrosis
There is strong evidence to suggest that the profibrotic activities of IL-4 and IL-13 are mediated in great extent by TGF-β, a potent profibrotic cytokine and key driver of ECM formation and tissue remodeling [10, 65, 143, 145, 146]. TGF-β is consistently overexpressed in virtually all fibrotic diseases, with production levels correlating with the progression of fibrosis [76, 138, 147]. IL-4 and IL-13 may act through TGF-β to trigger fibrosis by directly activating TGF-β production or stimulating pathways promote TGF-β signaling [143, 147]. The canonical SMAD signaling pathway plays a crucial role in governing TGF-β-induced fibrosis, with downstream targets including connective tissue growth factor, α-SMA, collagens, and matrix metalloproteinases (MMPs), and tissue inhibitors of metalloproteinases (TIMPs) (Figure 4) [49, 64, 87]. Tissue fibrosis is reduced in Smad3-deficient mice, confirming the importance of the TGF-β/SMAD pathway in certain fibrotic processes [143]. The profibrotic effects of TGF-β include the stimulation of fibroblast chemotaxis, differentiation, proliferation, and ECM synthesis and deposition [14, 72]. TGF-β also acts through Smad-independent signal transduction pathways to promote fibrosis, the mechanisms of which are covered in other reviews [49, 64].
Figure 4.

The profibrotic actions of TGF-β are positively regulated by Th2 cytokines IL-4 and IL-13. TGB-β can induce fibrosis via the canonical SMAD signaling pathway. The binding of TGF-β ligands to their cognate receptors activates a cascade of SMAD proteins, which form a complex and translocate to the nucleus to transcribe specific genes. The expression of fibrosis-associated genes, such as collagen and fibronectin, results in ECM deposition, fibroblast proliferation, and myofibroblast differentiation. While these biological processes constitute physiologic wound healing, overactive TGF-β/SMAD signaling can lead to tissue fibrosis.
α-SMA, alpha smooth muscle actin; CTGF, connective tissue growth factor; ECM, extracellular matrix; MMPs, matrix metalloproteinases; TGF-β, transforming growth factor-beta; TIMPs, tissue inhibitors of metalloproteinases
Fibroblasts isolated from hypertrophic scars and keloids overexpress proteins involved in TGF-β signal transduction, and hypertrophic scar fibroblasts have been shown to produce approximately twice as much TGF-β1 as do normal skin fibroblasts [103, 105]. Blockade of TGF-β signaling has been shown to reduce ECM deposition and improve scar appearance in rat wounds, prevent the overexpression of collagen genes in SSc fibroblasts, and prevent skin fibrosis in a sclerodermatous GVHD mouse model [54, 81, 122, 123].
IL-4 is a potent mediator of skin fibrosis and scarring
Several studies have shown that IL-4 is a major profibrotic cytokine that stimulates collagen synthesis by fibroblasts. In response to IL-4 in vitro, human dermal fibroblasts synthesize a dose-dependent increase in pre-collagen mRNAs, resulting in elevated levels of types I and III collagen and fibronectin [37, 104]. This stimulatory effect is neutralized specifically by anti-IL-4 antibodies [104]. Significantly higher levels of IL-4 are detected in skin biopsy specimens, dermal fibroblast cultures, and sera of SSc patients compared to controls [47, 90, 117]. Similar stimulatory effects of IL-4 on collagen synthesis have been observed in human dermal fibroblasts isolated from SSc patients and normal counterparts, as well as in normal and TSK/+ murine fibroblasts [28, 83, 117]. TSK/+ fibroblasts are hyperresponsive to IL-4, displaying increased collagen promoter region activity, collagen gene transcription, and type I collagen protein synthesis compared to controls [84]. IL-4 was also found to promote biosynthesis of collagen proteins by increased stability and transcription of the collagen mRNA in SSc fibroblasts in culture [67].
Significant positive correlations between IL-4 activity and fibrosis have also been demonstrated in animal models. Salmon-Ehr et al. showed that IL-4 is strongly expressed during the early phases of normal wound healing in mouse skin and decreases after wound closure [118]. Topical application of exogenous IL-4 on acute cutaneous wounds in mice induces a significant increase in the formation of fibrotic tissue, whereas the administration of IL-4 antisense oligonucleotides significantly delays wound healing [118]. These results implicate IL-4 production in normal wound healing. Overexpression of IL-4 in transgenic mice results in fibroblast proliferation and increased focal deposition of collagen in the dermis [27]. Conversely, inhibition of IL-4 responses via administration of anti-IL-4 monoclonal antibodies reduces dermal collagen deposition, preventing progression of cutaneous fibrosis in murine models of SSc and chronic skin allograft rejection [88, 99].
The central role of IL-4 in skin fibrosis has also been demonstrated by knockout mouse studies. Neutralizing anti-IL-4 antibodies given to young TSK/+ mice normalizes dermal collagen content and prevents dermal fibrosis [99]. Furthermore, the development of skin fibrosis is completely abrogated in TSK/+ mice with null mutation of either the IL-4 or Stat6 genes [98]. McGaha et al. demonstrated that targeted deletion of the IL-4Rα gene in TSK/+ mice prevents the development of dermal thickening and lowers the level of dermal hydroxyproline content to be similar to that of control mice, highlighting the importance of IL-4 for the development of cutaneous fibrosis [83]. Interestingly, disruption of the IL-4 gene not only rescues mice with the embryonic lethal homozygous TSK/TSK mutation from death, but also prevents the hallmark cutaneous hyperplasia of the TSK syndrome and substantially decreases TGF-β production by fibroblasts in vitro [60].
Altogether, these data show that IL-4 is a key profibrotic agent with important roles in mediating ECM synthesis to maintain tissue integrity. While IL-4 promotes collagen production to maintain normal physiologic processes, its dysregulation can result in abnormal wound healing and fibrotic disease.
IL-13 is a potent mediator of skin fibrosis and scarring
As part of physiologic wound healing, IL-13 directly activates fibroblast proliferation and differentiation, and also induces gene expression of type I collagen and other critical fibrosis-associated proteins such as α-SMA, the hallmark of myofibroblasts [53]. IL-13 has been shown to upregulate total collagen generation in fibroblasts of normal human skin and keloids, with a more rapid and greater magnitude of collagen response to IL-13 stimulation observed in keloid-derived fibroblasts [100]. A study using cDNA microarray analysis to identify IL-13 target genes showed that IL-13 mediates the transcriptional activation of the α2(I) collagen gene in human dermal fibroblasts in a dose-dependent manner [56]. The STAT6 signaling pathway was essential for this induction, as the addition of STAT6 antisense nucleotide resulted in a near complete inhibition of IL-13 effect on α2(I) collagen gene and type I collagen protein expression [56]. Furthermore, IL-13 specifically induces the gene expression of procollagen 3α1, which has been described as an early marker of active fibrosis and a poor clinical prognostic indicator in FPDs [100]. In addition to stimulating dermal collagen deposition, IL-13 also exerts profibrotic effects by hindering collagen degradation through inhibition of MMPs and upregulation of TIMPs in fibroblasts [100]. MMPs are zinc-dependent proteinases that cleave collagen and other ECM components, whereas TIMPs block the activity of MMPs [150]. Therefore, IL-13 has pleiotropic effects on fibrotic processes.
IL-13 has been identified as the dominant effector cytokine of fibrosis in several FPD models [14, 145]. Serum levels of IL-13 are significantly elevated in patients with SSc and localized scleroderma compared with healthy controls, and these levels positively correlate with the extent of skin fibrosis and levels of inflammatory biomarkers [32, 47, 48]. Furthermore, IL-13-producing circulating CD8+ T cells isolated from SSc patients express skin-homing receptors and induce a profibrotic phenotype in normal dermal fibroblasts in vitro, which is inhibited by anti-IL-13 antibody [30, 31]. Interestingly, in the sclerodermatous GVHD murine model, genetic deficiency of either IL-13 or IL-4Rα conferred complete protection from fibrotic disease based on histological and clinical evidence, suggesting that IL-13 signaling is essential in this model of fibrosis [41].
The factors that regulate IL-13 expression in fibrotic disease remain incompletely understood. However, emerging evidence implicates IL-33 as an upstream inducer of IL-13 in the immune system [41]. IL-33 is constitutively expressed in the skin and its increased expression in activated dermal fibroblasts is correlated with skin fibrotic disorders [109]. Subcutaneous injection of IL-33 in mice is sufficient to drive the development of cutaneous fibrosis, with skin samples showing significantly elevated expression of IL-4 and IL-13 [109]. This response is attenuated IL-13 knockout mice [109]. In SSc patients, higher circulating levels of IL-33 correlate with the extent of skin fibrosis and microvascular damage [97].
Altogether, the research studies discussed in this review support the crucial roles of Th2 cytokines in regulating tissue remodeling and ECM homeostasis. While IL-4 and IL-13 exert important wound healing mechanisms in response to tissue damage or type 1 immune-mediated inflammation, their dysregulation can lead to overzealous tissue repair and ultimately fibrotic disease.
Future Directions
There is a large unmet need for innovative therapeutic strategies to prevent and treat skin fibrosis and pathological scars [89]. Despite the substantial societal and economic burden of skin fibrosis, there is no “gold standard” for scar management and current treatments have limited clinical efficacy [39, 133]. The translation of research evidence into clinical practice and therapeutic applications has lagged, underscoring the complexity of fibrotic disease mechanisms.
Understanding the regulation and mechanisms of cytokine action has led to the development of new treatments that have the promise of slowing the progression of disease and potentially altering the natural history of disease. Based on observations in animal models of fibrosis, pathologic scar formation may be mitigated via targeted modulation of profibrotic cytokine responses. Therefore, therapies that inhibit the IL-4/IL-13 axis may be a potential strategy to limit skin fibrosis.
While murine studies have provided significant insights into the pathogenesis of cutaneous scarring, there is no ideal model that accurately recapitulates all human features of dermal fibrosis [4, 9]. As such, the translation of findings from preclinical experiments into therapeutics that attenuate the fibrotic response in humans may not be linear.
Biologic agents targeted against IL-4 and/or IL-13 are already approved or under clinical investigation for therapeutic use in various Th2-driven diseases (Table 1) [7, 63, 78, 80, 129, 137]. Dupilumab, a fully humanized monoclonal antibody against IL-4Rα, is currently the only approved biologic that targets the IL-4/IL-13 axis and is approved for use in the treatment of atopic dermatitis and asthma [112, 124]. In addition, there are two biologic agents (SAR156597 and QBX258) in clinical development that bind and neutralize both IL-4 and IL-13, thereby preventing signaling effects of the entire IL-4/IL-13 axis [68]. Ongoing clinical trials are assessing the efficacy of SAR156597 and QBX258 in treating diffuse cutaneous SSc and lymphedema, respectively [86, 119]. Since FPDs share common mechanistic pathways, these drugs may potentially be repurposed for the treatment of fibrotic skin diseases.
Table 1.
Summary of biologic agents that target the IL-4/IL-13 axis for the treatment of Th2-driven diseases
| Target | Drug name (alternative) | Developers | Molecule | Condition | Developmental status |
|---|---|---|---|---|---|
| IL-4 | Altrakincept (Nuvance) | Amgen | Recombinant human soluble IL-4Rα | Asthma | Halted after Phase III [13, 129] |
| Pascolizumab (SB 240683) | GlaxoSmithKline | Humanized mAb | Asthma | Terminated in Phase I/II [46, 78] | |
| VAK694 | Novartis | Human mAb | AR | Completed Phase II [19] | |
| IL-4Rα | AIR-645 | Altair Therapeutics | Antisense IL-4Rα | Asthma | Completed Phase II [3, 18] |
| AMG-317 | Amgen | Human mAb | Asthma | Completed Phase II [22, 80] | |
| Dupilumab (Dupixent) | Regeneron; Sanofi | Human mAb | AD AR Asthma EoE Nasal polyposis |
FDA approved [124, 126] Undergoing Phase II [110] FDA approved [107] Undergoing Phase III [111] Completed Phase III [6, 120] |
|
| MEDI 9314 | AstraZeneca | Human mAb | AD | Halted after Phase I [5, 63] | |
| Pitrakinra (Aerovant, Aeroderm) | Aerovance | Recombinant human protein of IL-4 mutein | Asthma AD |
Completed Phase II [140] Completed Phase II [80] |
|
| IL-13 | Anrukinzuma b (IMA-638) | Wyeth (Pfizer) | Humanized mAb | Asthma UC |
Halted after Phase II [35, 106] Halted after Phase II [52, 113] |
| Dectrekumab (QAX576) | Novartis | Human mAb | AR Asthma CD EoE IPF Keloids PF due to SSc |
Completed Phase II [91] Halted after Phase II [68] Completed Phase II [92] Completed Phase II [116] Terminated in Phase II [93] Terminated in Phase II [94] Terminated in Phase II [95, 97] |
|
| GSK679586 | GlaxoSmithKline | Humanized mAb | Asthma | Completed Phase II [12] | |
| IMA-026 | Wyeth (Pfizer) | Humanized mAb | Asthma | Completed Phase I/II [35] | |
| Lebrikizumab | Roche; Dermira | Humanized mAb | AD Asthma IPF |
Completed Phase II [127] Completed III [44, 45] Completed Phase II [128] |
|
| Tralokinumab (CAT-354) | MedImmune (AstraZeneca); LEO Pharma |
Human mAb | AD Alopecia areata Asthma IPF UC |
Undergoing Phase III [69, 70, 142] Completed Phase II [43, 114] Completed Phase III [101] Terminated in Phase II [102] Completed Phase II [23] |
|
| RPC4046 (ABT-308) | AbbVie; Receptos (Celgene) | Humanized mAb | Asthma EoE |
Completed Phase I [132] Completed Phase II [50] |
|
| IL-4 and IL-13 | QBX258 (VAK694 + dectrekumab) | Novartis | Combination of two human mAbs | Asthma BCRL |
Completed Phase II [96] Completed pilot study [86] |
| SAR156597 (ACT14604) | Sanofi | Humanized bispecific mAb | IPF Skin fibrosis in SSc |
Completed Phase II [108] Undergoing Phase II [119] |
AD, atopic dermatitis; AR, allergic rhinitis; BCRL, breast cancer related lymphedema; CD, Crohn’s disease; EoE, eosinophilic esophagitis; FDA, Food and Drug Administration; IL, interleukin; IL-4Rα, interleukin-4 receptor α-subunit; IPF, idiopathic pulmonary fibrosis; mAb, monoclonal antibody; PF, pulmonary fibrosis; SSc, systemic sclerosis; UC, ulcerative colitis
Conclusions
Skin fibrosis is the result of an exaggerated wound healing response and underlies the pathogenesis of a wide spectrum of cutaneous diseases such as SSc and pathologic cutaneous scars. Despite the socioeconomic burden of skin fibrosis, effective and durable scar treatment remains a significant unmet need in clinical medicine. Research on cellular and molecular wound healing biology has revealed key molecular players that may be targeted for pharmacologic intervention in the treatment of fibrosis. IL-4 and IL-13 are immunoregulatory Th2 cytokines that mediate important profibrotic effects, including fibroblast proliferation and collagen synthesis. Biologic agents that target the IL-4/IL-13 axis may provide a promising therapeutic modality for patients with skin fibrosis and are worthy of further investigation.
Acknowledgments
Funding: Research reported in this publication was supported by the National Institute of General Medical Sciences of the National Institutes of Health under Award No. K23GM117309.
Footnotes
Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.
Disclosure: The authors have no relevant conflicts of interest to disclose.
References
- 1.Allanore Y, Matucci-Cerinic M, Distler O (2016) Treatment of systemic sclerosis: is there any hope for the future? RMD Open 2:e000260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Allen JE, Wynn TA (2011) Evolution of Th2 Immunity: A Rapid Repair Response to Tissue Destructive Pathogens. PLoS Pathog 7:5–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. [10 Nov 2018];Altair Therapeutics Study Evaluating the Effects of AIR645 on Allergen-Induced Airway Responses in Subjects With Mild Atopic Asthma. https://clinicaltrials.gov/ct2/show/NCT00941577. Accessed.
- 4.Artlett CM (2014) Animal models of systemic sclerosis: Their utility and limitations. Open Access Rheumatol Res Rev 6:65–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.AstraZeneca (2018) Clinical trials appendix Q1 2018 Results update https://www.astrazeneca.com/content/dam/az/PDF/2018/Q1-2018/Q12018Clinicaltrialsappendix.pdf. Accessed 10 Nov 2018
- 6.Bachert C, Mannent L, Naclerio RM, et al. (2016) Effect of Subcutaneous Dupilumab on Nasal Polyp Burden in Patients With Chronic Sinusitis and Nasal Polyposis. JAMA 315:469. [DOI] [PubMed] [Google Scholar]
- 7.Bagnasco D, Ferrando M, Varricchi G, et al. (2016) A Critical Evaluation of Anti-IL-13 and Anti-IL-4 Strategies in Severe Asthma. Int Arch Allergy Immunol 170:122–131. [DOI] [PubMed] [Google Scholar]
- 8.Bayat A, McGrouther D, Ferguson M (2003) Skin scarring. BMJ 326:88–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Beyer C, Schett G, Distler O, Distler JHW (2010) Animal models of systemic sclerosis: Prospects and limitations. Arthritis Rheum 62:2831–2844. [DOI] [PubMed] [Google Scholar]
- 10.Bhogal RK, Stoica CM, McGaha TL, Bona CA (2005) Molecular aspects of regulation of collagen gene expression in fibrosis. J Clin Immunol 25:592–603. [DOI] [PubMed] [Google Scholar]
- 11.Bock O, Schmid-Ott G, Malewski P, Mrowietz U (2006) Quality of life of patients with keloid and hypertrophic scarring. Arch Dermatol Res 297:433–438. [DOI] [PubMed] [Google Scholar]
- 12.De Boever EH, Ashman C, Cahn AP, et al. (2014) Efficacy and safety of an anti–IL-13 mAb in patients with severe asthma: A randomized trial. J Allergy Clin Immunol 133:989–996.e4. [DOI] [PubMed] [Google Scholar]
- 13.Borish LC, Nelson HS, Lanz MJ, et al. (1999) Interleukin-4 receptor in moderate atopic asthma. A phase I/II randomized, placebo-controlled trial. Am J Respir Crit Care Med 160:1816–23. [DOI] [PubMed] [Google Scholar]
- 14.Borthwick LA, Wynn TA, Fisher AJ (2013) Cytokine mediated tissue fibrosis. Biochim Biophys Acta - Mol Basis Dis 1832:1049–1060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Brown BC, McKenna SP, Siddhi K, et al. (2008) The hidden cost of skin scars: quality of life after skin scarring. J Plast Reconstr Aesthetic Surg 61:1049–1058. [DOI] [PubMed] [Google Scholar]
- 16.Brunner SM, Schiechl G, Kesselring R, et al. (2013) IL-13 signaling via IL-13Rα2 triggers TGF-β1-dependent allograft fibrosis. Transplant Res 2:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bush JA, McGrouther DA, Young VL, et al. (2011) Recommendations on clinical proof of efficacy for potential scar prevention and reduction therapies. Wound Repair Regen 19:s32–s37. [DOI] [PubMed] [Google Scholar]
- 18.Catley MC (2010) Asthma & COPD--IQPC’s Second Conference. IDrugs 13:601–4. [PubMed] [Google Scholar]
- 19.Chaker AM, Shamji MH, Dumitru FA, et al. (2016) Short-term subcutaneous grass pollen immunotherapy under the umbrella of anti–IL-4: A randomized controlled trial. J Allergy Clin Immunol 137:452–461.e9. [DOI] [PubMed] [Google Scholar]
- 20.Chiaramonte MG, Mentink-Kane M, Jacobson BA, et al. (2003) Regulation and function of the interleukin 13 receptor alpha 2 during a T helper cell type 2-dominant immune response. J Exp Med 197:687–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Chomarat P, Banchereau J (1998) Interleukin-4 and lnterleukin-13: Their Similarities and Discrepancies. Int Rev Immunol 17:1–52. [DOI] [PubMed] [Google Scholar]
- 22.Corren J, Busse W, Meltzer EO, et al. (2010) A randomized, controlled, phase 2 study of AMG 317, an IL-4Ralpha antagonist, in patients with asthma. Am J Respir Crit Care Med 181:788–96. [DOI] [PubMed] [Google Scholar]
- 23.Danese S, Rudziński J, Brandt W, et al. (2015) Tralokinumab for moderate-to-severe UC: a randomised, double-blind, placebo-controlled, phase IIa study. Gut 64:243–249. [DOI] [PubMed] [Google Scholar]
- 24.Darby IA, Zakuan N, Billet F, Desmoulière A (2016) The myofibroblast, a key cell in normal and pathological tissue repair. Cell Mol Life Sci 73:1145–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Darby IA, Laverdet B, Bonté F, Desmoulière A (2014) Fibroblasts and myofibroblasts in wound healing. Clin Cosmet Investig Dermatol 7:301–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Denton CP, Abraham DJ (2004) Transgenic analysis of scleroderma: understanding key pathogenic events in vivo. Autoimmun Rev 3:285–293. [DOI] [PubMed] [Google Scholar]
- 27.Elbe-Bürger A, Egyed A, Olt S, et al. (2002) Overexpression of IL-4 alters the homeostasis in the skin. J Invest Dermatol 118:767–778. [DOI] [PubMed] [Google Scholar]
- 28.Fertin C, Nicolas JF, Gillery P, et al. (1991) Interleukin-4 stimulates collagen synthesis by normal and scleroderma fibroblasts in dermal equivalents. Cell Mol Biol 37:823–829. [PubMed] [Google Scholar]
- 29.Fichtner-Feigl S, Strober W, Kawakami K, et al. (2006) IL-13 signaling through the IL-13α2 receptor is involved in induction of TGF-β1 production and fibrosis. Nat Med 12:99–106. [DOI] [PubMed] [Google Scholar]
- 30.Fuschiotti P (2011) Role of IL-13 in systemic sclerosis. Cytokine 56:544–549. [DOI] [PubMed] [Google Scholar]
- 31.Fuschiotti P, Larregina AT, Ho J, et al. (2013) Interleukin-13-producing CD8+ T cells mediate dermal fibrosis in patients with systemic sclerosis. Arthritis Rheum 65:236–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Fuschiotti P, Medsger TAJ, Morel PA (2009) Effector CD8+ T cells in systemic sclerosis patients produce abnormally high levels of interleukin-13 associated with increased skin fibrosis. Arthritis Rheum 60:1119–1128. [DOI] [PubMed] [Google Scholar]
- 33.Gandhi NA, Pirozzi G, Graham NMH (2017) Commonality of the IL-4/IL-13 pathway in atopic diseases. Expert Rev Clin Immunol 13:425–437. [DOI] [PubMed] [Google Scholar]
- 34.Gause WC, Wynn TA, Allen JE (2013) Type 2 immunity and wound healing: Evolutionary refinement of adaptive immunity by helminths. Nat Rev Immunol 13:607–614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Gauvreau GM, Boulet L-P, Cockcroft DW, et al. (2011) Effects of Interleukin-13 Blockade on Allergen-induced Airway Responses in Mild Atopic Asthma. Am J Respir Crit Care Med 183:1007–1014. [DOI] [PubMed] [Google Scholar]
- 36.Gieseck RL, Wilson MS, Wynn TA (2018) Type 2 immunity in tissue repair and fibrosis. Nat Rev Immunol 18:62–76. [DOI] [PubMed] [Google Scholar]
- 37.Gillery P, Fertin C, Nicolas JF, et al. (1992) Interleukin-4 stimulates collagen gene expression in human fibroblast monolayer cultures. Potential role in fibrosis. FEBS Lett 302:231–234. [DOI] [PubMed] [Google Scholar]
- 38.Gold MH, Berman B, Clementoni MT, et al. (2014) Updated international clinical recommendations on scar management: Part 1 - Evaluating the evidence. Dermatologic Surg 40:817–824. [DOI] [PubMed] [Google Scholar]
- 39.Gold MH, McGuire M, Mustoe TA, et al. (2014) Updated international clinical recommendations on scar management: Part 2 - Algorithms for scar prevention and treatment. Dermatologic Surg 40:825–831. [DOI] [PubMed] [Google Scholar]
- 40.Greenblatt MB, Aliprantis AO (2013) The immune pathogenesis of scleroderma: context is everything. Curr Rheumatol Rep 15:297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Greenblatt MB, Sargent JL, Farina G, et al. (2012) Interspecies comparison of human and murine scleroderma reveals IL-13 and CCL2 as disease subset-specific targets. Am J Pathol 180:1080–1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Gurtner GC, Werner S, Barrandon Y, Longaker MT (2008) Wound repair and regeneration. Nature 453:314–321. [DOI] [PubMed] [Google Scholar]
- 43.Guttman E. [10 Nov 2018];A Pilot Study of Tralokinumab in Subjects With Moderate to Severe Alopecia Areata. https://clinicaltrials.gov/ct2/show/NCT02684097. Accessed.
- 44.Hanania NA, Korenblat P, Chapman KR, et al. (2016) Efficacy and safety of lebrikizumab in patients with uncontrolled asthma (LAVOLTA I and LAVOLTA II): replicate, phase 3, randomised, double-blind, placebo-controlled trials. Lancet Respir Med 4:781–796. [DOI] [PubMed] [Google Scholar]
- 45.Hanania NA, Noonan M, Corren J, et al. (2015) Lebrikizumab in moderate-to-severe asthma: pooled data from two randomised placebo-controlled studies. Thorax 70:748–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hart TK, Blackburn MN, Brigham-Burke M, et al. (2002) Preclinical efficacy and safety of pascolizumab (SB 240683): a humanized anti-interleukin-4 antibody with therapeutic potential in asthma. Clin Exp Immunol 130:93–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hasegawa M, Fujimoto M, Kikuchi K, Takehara K (1997) Elevated serum levels of interleukin 4 (IL-4), IL-10, and IL-13 in patients with systemic sclerosis. J Rheumatol 24:328–32. [PubMed] [Google Scholar]
- 48.Hasegawa M, Sato S, Nagaoka T, et al. (2003) Serum levels of tumor necrosis factor and interleukin-13 are elevated in patients with localized scleroderma. Dermatology 207:141–7. [DOI] [PubMed] [Google Scholar]
- 49.He W, Dai C (2015) Key Fibrogenic Signaling. Curr Pathobiol Rep 3:183–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hirano I, Collins MH, Assouline-Dayan Y, et al. (2018) RPC4046, a Monoclonal Antibody Against IL13, Reduces Histologic and Endoscopic Activity in Patients With Eosinophilic Esophagitis. Gastroenterology Epub ahead of print [DOI] [PubMed]
- 51.Hoffmann KF, McCarty TC, Segal DH, et al. (2001) Disease fingerprinting with cDNA microarrays reveals distinct gene expression profiles in lethal type 1 and type 2 cytokine-mediated inflammatory reactions. FASEB J Off Publ Fed Am Soc Exp Biol 15:2545–2547. [DOI] [PubMed] [Google Scholar]
- 52.Hua F, Ribbing J, Reinisch W, et al. (2015) A pharmacokinetic comparison of anrukinzumab, an anti-IL-13 monoclonal antibody, among healthy volunteers, asthma and ulcerative colitis patients. Br J Clin Pharmacol 80:101–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Huang X-L, Wang Y-J, Yan J-W, et al. (2015) Role of anti-inflammatory cytokines IL-4 and IL-13 in systemic sclerosis. Inflamm Res 64:151–159. [DOI] [PubMed] [Google Scholar]
- 54.Ihn H, Yamane K, Kubo M, Tamaki K (2001) Blockade of endogenous transforming growth factor beta signaling prevents up-regulated collagen synthesis in scleroderma fibroblasts: association with increased expression of transforming growth factor beta receptors. Arthritis Rheum 44:474–80. [DOI] [PubMed] [Google Scholar]
- 55.Jagdeo J, Shumaker PR (2017) Traumatic Scarring. JAMA Dermatology 153:364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Jinnin M, Ihn H, Yamane K, Tamaki K (2004) Interleukin-13 Stimulates the Transcription of the Human 2(I) Collagen Gene in Human Dermal Fibroblasts. J Biol Chem 279:41783–41791. [DOI] [PubMed] [Google Scholar]
- 57.Kaufman BP, Alexis AF (2018) Biologics and Small Molecule Agents in Allergic and Immunologic Skin Diseases. Curr Allergy Asthma Rep 18:55. [DOI] [PubMed] [Google Scholar]
- 58.Kaviratne M, Hesse M, Leusink M, et al. (2004) IL-13 activates a mechanism of tissue fibrosis that is completely TGF-beta independent. J Immunol 173:4020–4029. [DOI] [PubMed] [Google Scholar]
- 59.Knipper JA, Willenborg S, Brinckmann J, et al. (2015) Interleukin-4 Receptor α Signaling in Myeloid Cells Controls Collagen Fibril Assembly in Skin Repair. Immunity 43:803–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kodera T, McGaha TL, Phelps R, et al. (2002) Disrupting the IL-4 gene rescues mice homozygous for the tight-skin mutation from embryonic death and diminishes TGF-beta production by fibroblasts. Proc Natl Acad Sci U S A 99:3800–3805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Landén NX, Li D, Ståhle M (2016) Transition from inflammation to proliferation: a critical step during wound healing. Cell Mol Life Sci 73:3861–3885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Laurent P, Jolivel V, Manicki P, et al. (2017) Immune-mediated repair: A matter of plasticity. Front Immunol 8:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Lawrence MG, Steinke JW, Borish L (2018) Cytokine-targeting biologics for allergic diseases. Ann Allergy, Asthma Immunol 120:376–381. [DOI] [PubMed] [Google Scholar]
- 64.Leask A, Abraham DJ (2004) TGF-β signaling and the fibrotic response. FASEB J 18:816–827. [DOI] [PubMed] [Google Scholar]
- 65.Lee CG, Homer RJ, Zhu Z, et al. (2001) Interleukin-13 induces tissue fibrosis by selectively stimulating and activating transforming growth factor β1. J Exp Med 194:809–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lee H, Jang Y (2018) Recent Understandings of Biology, Prophylaxis and Treatment Strategies for Hypertrophic Scars and Keloids. Int J Mol Sci 19:711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lee KS, Ro YJ, Ryoo YW, et al. (1996) Regulation of interleukin-4 on collagen gene expression by systemic sclerosis fibroblasts in culture. J Dermatol Sci 12:110–117. [DOI] [PubMed] [Google Scholar]
- 68.Legrand F, Klion AD (2015) Biologic Therapies Targeting Eosinophils: Current Status and Future Prospects. J Allergy Clin Immunol Pract 3:167–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. [10 Nov 2018];LEO Pharma Tralokinumab Monotherapy for Moderate to Severe Atopic Dermatitis - ECZTRA 1 (ECZema TRAlokinumab Trial no. 1) (ECZTRA 1) https://clinicaltrials.gov/ct2/show/NCT03131648. Accessed.
- 70. [10 Nov 2018];LEO Pharma Long-term Extension Trial in Subjects With Atopic Dermatitis Who Participated in Previous Tralokinumab Trials - ECZTEND. https://clinicaltrials.gov/ct2/show/NCT03587805. Accessed.
- 71.Liang H, Zhang Z, Yan J, et al. (2017) The IL-4 receptor α has a critical role in bone marrow–derived fibroblast activation and renal fibrosis. Kidney Int 92:1433–1443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lichtman MK, Otero-Vinas M, Falanga V (2016) Transforming growth factor beta (TGF-β) isoforms in wound healing and fibrosis. Wound Repair Regen 24:215–222. [DOI] [PubMed] [Google Scholar]
- 73.Lloyd CM, Snelgrove RJ (2018) Type 2 immunity: Expanding our view. Sci Immunol 3:1604. [DOI] [PubMed] [Google Scholar]
- 74.Loke P, Gallagher I, Nair MG, et al. (2007) Alternative Activation Is an Innate Response to Injury That Requires CD4+ T Cells to be Sustained during Chronic Infection. J Immunol 179:3926–3936. [DOI] [PubMed] [Google Scholar]
- 75.Lucey DR, Clerici M, Shearer GM (1996) Type 1, and Type 2 cytokine dysregulation in human infectious, neoplastic, and inflammatory diseases. Clin Microbiol Rev 9:532–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Luzina IG, Atamas SP (2008) Fibrotic Skin Diseases. In: Gaspari A, Tyring S (eds) Clinical and Basic Immunodermatology Springer London, London, pp 721–737. [Google Scholar]
- 77.MacDonald TT (2006) Decoy receptor springs to life and eases fibrosis. Nat. Med 12:13–14 [DOI] [PubMed] [Google Scholar]
- 78.Maes T, Joos GF, Brusselle GG (2012) Targeting interleukin-4 in asthma: Lost in translation? Am J Respir Cell Mol Biol 47:261–270. [DOI] [PubMed] [Google Scholar]
- 79.Marshall CD, Hu MS, Leavitt T, et al. (2018) Cutaneous Scarring: Basic Science, Current Treatments, and Future Directions. Adv Wound Care 7:29–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.May RD, Fung M (2015) Strategies targeting the IL-4/IL-13 axes in disease. Cytokine 75:89–116. [DOI] [PubMed] [Google Scholar]
- 81.McCormick LL, Zhang Y, Tootell E, Gilliam AC (1999) Anti-TGF-beta treatment prevents skin and lung fibrosis in murine sclerodermatous graft-versus-host disease: a model for human scleroderma. J Immunol 163:5693–5699. [PubMed] [Google Scholar]
- 82.McCormick SM, Heller NM (2015) Commentary: IL-4 and IL-13 receptors and signaling. Cytokine 75:38–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.McGaha T, Saito S, Phelps RG, et al. (2001) Lack of skin fibrosis in tight skin (TSK) mice with targeted mutation in the interleukin-4R alpha and transforming growth factor-beta genes. J Invest Dermatol 116:136–143. [DOI] [PubMed] [Google Scholar]
- 84.McGaha TL, Le M, Kodera T, et al. (2003) Molecular mechanisms of interleukin-4-induced up-regulation of type I collagen gene expression in murine fibroblasts. Arthritis Rheum 48:2275–2284. [DOI] [PubMed] [Google Scholar]
- 85.McGaha TL, Bona CA (2002) Role of profibrogenic cytokines secreted by T cells in fibrotic processes in scleroderma. Autoimmun Rev 1:174–181. [DOI] [PubMed] [Google Scholar]
- 86.Memorial Sloan Kettering Cancer Center. Immunotherapy for the Treatment of Breast Cancer Related Upper Extremity Lymphedema (BCRL) https://clinicaltrials.gov/ct2/show/NCT02494206. Accessed 10 Nov 2018
- 87.Meng XM, Nikolic-Paterson DJ, Lan HY (2016) TGF-β: The master regulator of fibrosis. Nat Rev Nephrol 12:325–338. [DOI] [PubMed] [Google Scholar]
- 88.Le Moine A, Flamand V, Demoor FX, et al. (1999) Critical roles for IL-4, IL-5, and eosinophils in chronic skin allograft rejection. J Clin Invest 103:1659–1667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Mokos ZB, Jović A, Grgurević L, et al. (2017) Current Therapeutic Approach to Hypertrophic Scars. Front Med 4:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Needleman BW, Wigley FM, Stair RW (1992) Interleukin-1, interleukin-2, interleukin-4, interleukin-6, tumor necrosis factor alpha, and interferon-gamma levels in sera from patients with scleroderma. Arthritis Rheum 35:67–72. [DOI] [PubMed] [Google Scholar]
- 91.Nicholson GC, Kariyawasam HH, Tan AJ, et al. (2011) The effects of an anti–IL-13 mAb on cytokine levels and nasal symptoms following nasal allergen challenge. J Allergy Clin Immunol 128:800–807.e9. [DOI] [PubMed] [Google Scholar]
- 92.Novartis Pharmaceuticals. A Phase II Efficacy Study in Fistulizing Crohn’s Disease Patients https://clinicaltrials.gov/ct2/show/NCT01355614. Accessed 10 Nov 2018
- 93.Novartis Pharmaceuticals. Safety and Efficacy of QAX576 in Patients With Idiopathic Pulmonary Fibrosis (IPF) https://clinicaltrials.gov/ct2/show/NCT01266135. Accessed 10 Nov 2018
- 94.Novartis Pharmaceuticals. Efficacy 2 Part Study of Identification of Keloid Biomarkers and Effect of QAX576 on Keloid Recurrence https://clinicaltrials.gov/ct2/show/NCT00987545. Accessed 10 Nov 2018
- 95.Novartis Pharmaceuticals. QAX576 in Patients With Pulmonary Fibrosis Secondary to Systemic Sclerosis https://clinicaltrials.gov/ct2/show/NCT00581997. Accessed 10 Nov 2018
- 96.Novartis Pharmaceuticals. A Study to Establish the Efficacy of QBX258 in Patients With Moderate to Severe Asthma https://clinicaltrials.gov/ct2/show/NCT01479595. Accessed 10 Nov 2018
- 97.O’Reilly S (2013) Role of interleukin-13 in fibrosis, particularly systemic sclerosis. BioFactors 39:593–596. [DOI] [PubMed] [Google Scholar]
- 98.Ong CJ, Ip S, Teh SJ, et al. (1999) A role for T helper 2 cells in mediating skin fibrosis in tight-skin mice. Cell Immunol 196:60–68. [DOI] [PubMed] [Google Scholar]
- 99.Ong C, Wong C, Roberts CR, et al. (1998) Anti-IL-4 treatment prevents dermal collagen deposition in the tight-skin mouse model of scleroderma. Eur J Immunol 28:2619–2629. [DOI] [PubMed] [Google Scholar]
- 100.Oriente A, Fedarko NS, Pacocha SE, et al. (2000) Interleukin-13 modulates collagen homeostasis in human skin and keloid fibroblasts. J Pharmacol Exp Ther 292:988–994. [PubMed] [Google Scholar]
- 101.Panettieri RA, Sjöbring U, Péterffy A, et al. (2018) Tralokinumab for severe, uncontrolled asthma (STRATOS 1 and STRATOS 2): two randomised, double-blind, placebo-controlled, phase 3 clinical trials. Lancet Respir Med 6:511–525. [DOI] [PubMed] [Google Scholar]
- 102.Parker JM, Glaspole IN, Lancaster LH, et al. (2018) A Phase 2 Randomized Controlled Study of Tralokinumab in Subjects with Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 197:94–103. [DOI] [PubMed] [Google Scholar]
- 103.Perez OA, Berman B (2008) Cytokines and Chemokines. In: Gaspari A, Tyring S (eds) Clinical and Basic Immunodermatology Springer London, London, pp 3–16 [Google Scholar]
- 104.Postlethwaite A, Holness M a, Katai H, Raghow R (1992) Human fibroblasts synthesize elevated levels of extracellular matrix proteins in response to interleukin 4. J Clin Invest 90:1479–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Profyris C, Tziotzios C, Do Vale I (2012) Cutaneous scarring: Pathophysiology, molecular mechanisms, and scar reduction therapeutics Part I. The molecular basis of scar formation. J Am Acad Dermatol 66:1–10. [DOI] [PubMed] [Google Scholar]
- 106.Quirce S, Bobolea I, Domínguez-Ortega J, Barranco P (2014) Future Biologic Therapies in Asthma. Arch Bronconeumol 50:355–361. [DOI] [PubMed] [Google Scholar]
- 107.Rabe KF, Nair P, Brusselle G, et al. (2018) Efficacy and Safety of Dupilumab in Glucocorticoid-Dependent Severe Asthma. N Engl J Med 378:2475–2485. [DOI] [PubMed] [Google Scholar]
- 108.Raghu G, Richeldi L, Crestani B, et al. (2018) SAR156597 in idiopathic pulmonary fibrosis: a phase 2, placebo-controlled study (DRI11772). Eur Respir J Epub ahead of print [DOI] [PubMed]
- 109.Rankin AL, Mumm JB, Murphy E, et al. (2010) IL-33 Induces IL-13-Dependent Cutaneous Fibrosis. J Immunol 184:1526–1535. [DOI] [PubMed] [Google Scholar]
- 110.Regeneron Pharmaceuticals. Dupilumab As An Adjunct For Subcutaneous Grass Immunotherapy https://clinicaltrials.gov/ct2/show/NCT03558997. Accessed 10 Nov 2018
- 111.Regeneron Pharmaceuticals. Study to Determine the Efficacy and Safety of Dupilumab in Adult and Adolescent Patients With Eosinophilic Esophagitis (EoE) https://clinicaltrials.gov/ct2/show/NCT03633617. Accessed 10 Nov 2018
- 112. [10 Nov 2018];Regeneron Pharmaceuticals FDA Approves Asthma Indication for Dupixent® (dupilumab) https://investor.regeneron.com/news-releases/news-release-details/fda-approves-asthma-indication-dupixentr-dupilumab. Accessed.
- 113.Reinisch W, Panés J, Khurana S, et al. (2015) Anrukinzumab, an anti-interleukin 13 monoclonal antibody, in active UC: efficacy and safety from a phase IIa randomised multicentre study. Gut 64:894–900. [DOI] [PubMed] [Google Scholar]
- 114.Renert-Yuval Y, Guttman-Yassky E (2017) The Changing Landscape of Alopecia Areata: The Therapeutic Paradigm. Adv Ther 34:1594–1609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Roesner LM, Zeitvogel J, Heratizadeh A (2019) Common and different roles of IL-4 and IL-13 in skin allergy and clinical implications. Curr Opin Allergy Clin Immunol 19:319–327. [DOI] [PubMed] [Google Scholar]
- 116.Rothenberg ME, Wen T, Greenberg A, et al. (2015) Intravenous anti-IL-13 mAb QAX576 for the treatment of eosinophilic esophagitis. J Allergy Clin Immunol 135:500–7. [DOI] [PubMed] [Google Scholar]
- 117.Salmon-Ehr V, Serpier H, Nawrocki B, et al. (1996) Expression of interleukin-4 in scleroderma skin specimens and scleroderma fibroblast cultures. Potential role in fibrosis. Arch Dermatol 132:802–806. [PubMed] [Google Scholar]
- 118.Salmon-Ehr V, Ramont L, Godeau G, et al. (2000) Implication of interleukin-4 in wound healing. Lab Investig 80:1337–1343. [DOI] [PubMed] [Google Scholar]
- 119.Sanofi. Effectiveness and Safety of SAR156597 in Treating Diffuse Systemic Sclerosis https://clinicaltrials.gov/ct2/show/NCT02921971. Accessed 10 Nov 2018
- 120.Sanofi. A Controlled Clinical Study of Dupilumab in Patients With Nasal Polyps (SINUS-24) https://clinicaltrials.gov/ct2/show/NCT02912468. Accessed 10 Nov 2018
- 121.Sen CK, Gordillo GM, Roy S, et al. (2010) Human skin wounds: a major and snowballing threat to public health and the economy. Wound Repair Regen 17:763–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Shah M, Foreman DM, Ferguson MW (1994) Neutralising antibody to TGF-beta 1,2 reduces cutaneous scarring in adult rodents. J Cell Sci 107:1137–1157. [DOI] [PubMed] [Google Scholar]
- 123.Shah M, Foreman DM, Ferguson MW (1995) Neutralisation of TGF-beta 1 and TGF-beta 2 or exogenous addition of TGF-beta 3 to cutaneous rat wounds reduces scarring. J Cell Sci 108:985–1002. [DOI] [PubMed] [Google Scholar]
- 124.Shirley M (2017) Dupilumab: First Global Approval. Drugs 77:1115–1121. [DOI] [PubMed] [Google Scholar]
- 125.Sidgwick GP, Bayat A (2012) Extracellular matrix molecules implicated in hypertrophic and keloid scarring. J Eur Acad Dermatology Venereol 26:141–152. [DOI] [PubMed] [Google Scholar]
- 126.Simpson EL, Bieber T, Guttman-Yassky E, et al. (2016) Two Phase 3 Trials of Dupilumab versus Placebo in Atopic Dermatitis. N Engl J Med 375:2335–2348. [DOI] [PubMed] [Google Scholar]
- 127.Simpson EL, Flohr C, Eichenfield LF, et al. (2018) Efficacy and safety of lebrikizumab (an anti-IL-13 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis inadequately controlled by topical corticosteroids: A randomized, placebo-controlled phase II trial (TREBLE). J Am Acad Dermatol 78:863–871.e11. [DOI] [PubMed] [Google Scholar]
- 128.Swigris J, Ogura T, Scholand M, et al. The RIFF Study (Cohort A): A Phase II, randomized, double-blind, placebo-controlled trial of lebrikizumab as monotherapy in patients with idiopathic pulmonary fibrosis (abstract A6167). American Thoracic Society 2018 International Conference, San Diego, CA, USA, 2018 [Google Scholar]
- 129.Tan H-TT, Sugita K, Akdis CA (2016) Novel Biologicals for the Treatment of Allergic Diseases and Asthma. Curr Allergy Asthma Rep 16:70. [DOI] [PubMed] [Google Scholar]
- 130.Trace AP, Enos CW, Mantel A, Harvey VM (2016) Keloids and Hypertrophic Scars: A Spectrum of Clinical Challenges. Am J Clin Dermatol 17:201–223. [DOI] [PubMed] [Google Scholar]
- 131.Tredget EE, Yang L, Delehanty M, et al. (2006) Polarized Th2 cytokine production in patients with hypertrophic scar following thermal injury. J Interf Cytokine Res 26:179–189. [DOI] [PubMed] [Google Scholar]
- 132.Tripp CS, Cuff C, Campbell AL, et al. (2017) RPC4046, A Novel Anti-interleukin-13 Antibody, Blocks IL-13 Binding to IL-13 α1 and α2 Receptors: A Randomized, Double-Blind, Placebo-Controlled, Dose-Escalation First-in-Human Study. Adv Ther 34:1364–1381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Tziotzios C, Profyris C, Sterling J (2012) Cutaneous scarring: Pathophysiology, molecular mechanisms, and scar reduction therapeutics: Part II. Strategies to reduce scar formation after dermatologic procedures. J Am Acad Dermatol 66:13–24. [DOI] [PubMed] [Google Scholar]
- 134.Uitto J (1990) Fibrotic Skin Diseases. Arch Dermatol 126:661. [DOI] [PubMed] [Google Scholar]
- 135.Varga J, Abraham D (2007) Systemic sclerosis: a prototypic multisystem fibrotic disorder. J Clin Invest 117:557–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Walmsley GG, Maan ZN, Wong VW, et al. (2015) Scarless wound healing: Chasing the holy grail. Plast Reconstr Surg 135:907–917. [DOI] [PubMed] [Google Scholar]
- 137.Walsh GM (2018) Recent developments in the use of biologics targeting IL-5, IL-4, or IL-13 in severe refractory asthma. Expert Rev Respir Med 12:957–963. [DOI] [PubMed] [Google Scholar]
- 138.Walton KL, Johnson KE, Harrison CA (2017) Targeting TGF-β Mediated SMAD Signaling for the Prevention of Fibrosis. Front Pharmacol 8:461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Weng S-Y, Wang X, Vijayan S, et al. (2018) IL-4 Receptor Alpha Signaling through Macrophages Differentially Regulates Liver Fibrosis Progression and Reversal. EBioMedicine 29:92–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Wenzel S, Wilbraham D, Fuller R, et al. (2007) Effect of an interleukin-4 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies. Lancet (London, England) 370:1422–31. [DOI] [PubMed] [Google Scholar]
- 141.Wills-Karp M, Finkelman FD (2008) Untangling the complex web of IL-4-and IL-13-mediated signaling pathways. Sci Signal 1:1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Wollenberg A, Howell MD, Guttman-Yassky E, et al. (2018) Treatment of atopic dermatitis with tralokinumab, an anti–IL-13 mAb. J Allergy Clin Immunol Epub ahead of print [DOI] [PubMed]
- 143.Wynn TA (2009) Cellular and molecular mechanisms of fibrosis. J Pathol 214:199–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Wynn TA (2015) Type 2 cytokines: Mechanisms and therapeutic strategies. Nat Rev Immunol 15:271–282. [DOI] [PubMed] [Google Scholar]
- 145.Wynn TA (2004) Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol 4:583–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Wynn TA (2003) IL-13 effector functions. Annu Rev Immunol 21:425–456. [DOI] [PubMed] [Google Scholar]
- 147.Wynn TA, Ramalingam TR (2012) Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 18:1028–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Wynn T, Barron L (2010) Macrophages: Master Regulators of Inflammation and Fibrosis. Semin Liver Dis 30:245–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Zhu Z, Ding J, Ma Z, et al. (2016) Systemic depletion of macrophages in the subacute phase of wound healing reduces hypertrophic scar formation. Wound Repair Regen 24:644–656. [DOI] [PubMed] [Google Scholar]
- 150.Zhu Z, Ding J, Shankowsky HA, Tredget EE (2013) The molecular mechanism of hypertrophic scar. J Cell Commun Signal 7:239–252. [DOI] [PMC free article] [PubMed] [Google Scholar]

