Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2423 2023-11-06 01:48:39 |
2 layout Meta information modification 2423 2023-11-06 02:49:58 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zhang, X.; Wu, X.; Li, D. Immunotherapy of Keloids. Encyclopedia. Available online: https://encyclopedia.pub/entry/51164 (accessed on 31 July 2024).
Zhang X, Wu X, Li D. Immunotherapy of Keloids. Encyclopedia. Available at: https://encyclopedia.pub/entry/51164. Accessed July 31, 2024.
Zhang, Xiya, Xinfeng Wu, Dongqing Li. "Immunotherapy of Keloids" Encyclopedia, https://encyclopedia.pub/entry/51164 (accessed July 31, 2024).
Zhang, X., Wu, X., & Li, D. (2023, November 06). Immunotherapy of Keloids. In Encyclopedia. https://encyclopedia.pub/entry/51164
Zhang, Xiya, et al. "Immunotherapy of Keloids." Encyclopedia. Web. 06 November, 2023.
Immunotherapy of Keloids
Edit

Keloids are benign fibroproliferative tumors originating from abnormal wound healing. Many factors can cause keloids, including trauma, surgery, burns, vaccination, acne, and folliculitis, which can be summarized as dermal injury and irritation, in general. However, superficial injuries that do not reach the reticular dermis will not cause keloids, suggesting that keloids result from injury to this skin layer and subsequent abnormal wound healing.

keloid keloid pathogenesis keloid microenvironment immune cells

1. Introduction

Keloids are benign fibroproliferative tumors originating from abnormal wound healing [1]. Many factors can cause keloids, including trauma, surgery, burns, vaccination, acne, and folliculitis, which can be summarized as dermal injury and irritation, in general. However, superficial injuries that do not reach the reticular dermis will not cause keloids, suggesting that keloids result from injury to this skin layer and subsequent abnormal wound healing [2]. Keloids and hypertrophic scars are two types of commonly recognized pathological scars. Keloids have the following three characteristics that distinguish them from hypertrophic scars. First, keloid lesions extend beyond the boundaries of the original lesion and continue to grow from year to year, with little spontaneous regression [1]. Second, fibroblasts from hypertrophic scars and keloids differ. Although hypertrophic scar fibroblasts show a slight increase in basal collagen synthesis, they react normally to growth factors. However, keloidal fibroblasts produce high amounts of collagens, elastin, fibronectin, and proteoglycan and respond abnormally to stimulation. Third, the recurrence rate of keloids is very high [1][3]. Surgical removal of keloids alone has been reported to recur in 70% to 100% of patients and usually results in more intense collagen accumulation and greater lesion formation [4]. Even with surgical resection combined with radiotherapy, the recurrence rate is still high (22%) [5].
The inflammatory, proliferative, and remodeling phases represent three different, although chronologically overlapping, stages of normal wound healing [6]. Keloids are often thought to result from a prolonged proliferative phase and delayed remodeling phase. During normal wound healing, immune cells are recruited during the inflammatory phase to defend against invading microorganisms. Fibroblasts are subsequently stimulated to produce extracellular matrix (ECM) upon which the injured site can be repaired and reshaped [7]. The excess ECM is further digested, and immature collagen III is eventually replaced by mature collagen I [8]. If any steps in this process are disrupted, keloids can develop, which are characterized by abnormal activation of fibroblasts and immune cell infiltration.

2. Immunotherapy of Keloids

Current treatments for keloids include surgical resection, radiotherapy, injections, cryotherapy, laser, radiofrequency ablation, and various drug therapies. However, none of the drugs are keloid-specific [9]. In addition, few current treatments can completely prevent recurrence. The recurrence rate is 70% to 100% for surgical resection alone [4] and 22% for surgical resection plus radiotherapy [5]. Therefore, more effective treatment methods need to be developed. The potential targets for immunotherapy in keloids are summarized here (Table 1).
Table 1. Summary of potential immunotherapies for keloids.

2.1. TGF-β

As the central cellular effector of fibrotic responses, TGF-β is produced and secreted by inflammatory cells, particularly macrophages, as well as fibroblasts and platelets [1]. Types 1, 2, and 3 are the three subtypes of TGF-β. The levels of type 3 TGF-β are not increased in keloids, but those of types 1 and 2, which activate fibroblasts and are involved in fibrosis and inflammation, are increased [20]. In keloids, TGFβ-1 has been associated with increased collagen and fibronectin synthesis by fibroblasts. The addition of TGFβ-1 to keloidal fibroblasts leads to increased synthesis of procollagen RNA [21]. TGF-β is involved in both suppressive and inflammatory immune responses. Under steady-state conditions, TGF-β regulates thymic T-cell selection and maintains homeostasis of the naïve T-cell pool. TGF-β inhibits cytotoxic T-lymphocyte (CTL), Th1, and Th2 cell differentiation while promoting peripheral (p)Treg, Th17, Th9, and Tfh cell generation and T-cell tissue residence in response to immune challenges. Similarly, TGF-β controls the proliferation, survival, activation, and differentiation of B cells, as well as the development and functions of innate cells, including natural killer (NK) cells, macrophages, dendritic cells, and granulocytes [22]. TGF-β promotes keloid formation by regulating fibroblasts and macrophages. TGF-β works in tandem with IL17A to stimulate IL-6 and CCL2 production in fibroblasts, which are macrophage chemokines and offer a way for macrophages to be recruited to the keloid microenvironment [23]. Considering macrophages are an important source of TGF-β production, the recruited macrophages produce more TGF-β and further promote collagen synthesis in the keloid microenvironment. In addition, by co-culturing fibroblasts with Tregs, Chen et al. [8] found that Tregs promoted collagen expression, which was more pronounced in keloids than in non-keloid controls, and that the process required TGF-β and anti-CD3/CD28 stimulation. Therefore, targeting TGF-β-related receptors or pathways is a promising method for the treatment of keloids. Current therapies targeting TGF-β/Smad signaling in fibroblasts can be divided into genetic, cellular, and pharmacological therapies [24]. Several investigational agents that target the TGF-β pathway have entered clinical trials. Clinical trials of the TGF-β neutralizing antibody fresolimumab (GC1008) are underway [10].
Fresolimumab is a pan-TGF neutralizing antibody that targets all three TGF-β isoforms and is being tested in clinical trials for a variety of fibrotic and cancer disorders [10]. Phase 1 clinical studies have been conducted in patients with focal segmental glomerulosclerosis (FSGS), idiopathic pulmonary fibrosis (IPF), advanced malignant melanoma, and renal cell carcinoma (RCC). The administration of fresolimumab was well tolerated in three separate human phase 1 clinical trials, and no treatment-related serious adverse effects were reported [25]. In fibrotic diseases, Rice et al. found that fresolimumab treatment reduced biomarkers and improved clinical symptoms in patients with systemic sclerosis (SSc) [11]. The expression of several TGF-β and collagen-related genes, including CTGF, SERPINE1, and COL10A1, declined after fresolimumab treatment compared with baseline [11]. In addition, fresolimumab treatment was associated with a rapid, dramatic decline in fibroblast infiltration of the deep dermis [11]. Since fibroblasts are the main pathogenic cells of keloids, fresolimumab may be a novel therapeutic strategy for keloids.
However, persistent inhibition of TGF-β function may lead to side effects because TGF-β is involved in a variety of biological processes [10]. It has been shown that systemic reduction in TGF-β activity can be antifibrotic but also blocks its anti-inflammatory function, leading to exacerbation of inflammation [26]. Adverse effects also include bleeding episodes, such as gastrointestinal bleeding, gingival bleeding, or epistaxis [11]. Local injection of TGF-β antibodies or inhibitors may be an alternative treatment for keloids. Fibronectin extra domain A (Fn-EDA) is a component of the ECM and is specifically expressed in the fibrotic region. McGaraughty et al. designed a dual-specific antibody targeting Fn-EDA to deliver TGF-β antibody to fibrotic kidney lesion sites [12]. The molecule partially targets Fn-EDA and partially neutralizes TGF-β using dual variable domain Ig (DVD-Ig) technology. Systemic injection of bispecific antibodies led to notably higher levels of each molecule in obstructed kidneys than in non-obstructed kidneys, ipsilateral kidneys of sham animals, and other tissues. Since Fn-EDA is also enriched in keloids [1], the bispecific molecule targeting Fn-EDA and TGF-β can also be tried in the treatment of keloids. Bispecific antibody treatment may be more effective and have fewer side effects than the anti-TGF-β antibody alone.

2.2. NKG2A/CD94

CD8+ T cells can significantly inhibit fibroblast activity and proliferation [27]. Through scRNA-seq, Xu et al. [28] found that the downregulation of CD8+ CTLs is a feature in the peripheral blood and keloid lesions. In addition, scRNA-seq also showed specific upregulation of the NKG2A/CD94 complex, which may contribute to the reduction in CTLs within the tissue boundaries of keloid lesions. Therefore, it can be inferred that NKG2A/CD94 indirectly promotes the activity and proliferation of fibroblasts in keloids, thereby exacerbating keloid progression.
Monalizumab is a novel checkpoint inhibitor targeting the NKG2A/CD94 complex in tumor immunotherapy [13]. Monalizumab can enhance NK cell activity against various tumor cells and rescue CD8+ T-cell function in combination with programmed-cell-death axis (PDx) blockade [13]. Monalizumab enhances tumor immunity by blocking inhibitory NKG2A receptors to promote CD8+ T-cell effector function [13]. Given that CD8+ T cells inhibit fibroblast proliferation and activity, monalizumab is a promising agent for reducing excessive ECM in keloids by restoring the inhibitory effect of CD8+ T cells on fibroblasts. Furthermore, a prominent advantage of monalizumab is that blocking NKG2A has little toxicity and, in particular, produces no signs of autoimmunity [13]. The most common adverse events of monalizumab were fatigue (17%), pyrexia (13%), and headache (10%). Other rare adverse events were interstitial lung disease, colitis, and hypophosphatemia [13]. In the future, clinical trials of monalizumab in the treatment of keloids may lead to surprising discoveries.

2.3. IL-4/IL-13 Pathway

Th2 cells and their cytokines play an important role in the pathogenesis of keloids. Wu et al. [29] found that markers of Th2, including IL4R, CCR5, CCL11, and TNFSF4/OX40L, were significantly upregulated in both keloid lesional and non-lesional skin compared to normal skin. IL-4 and IL-13 not only promote ECM deposition, inflammation, and pruritus of keloids but also induce the transformation of monocytes into M2 macrophages [30], thus playing a pivotal role in the pathogenesis of keloids. IL-4 and IL-13 can also directly stimulate nerve fibers via IL-4 receptors to induce pruritus. Moreover, TGF-β induces the production of IL-31, a Th2-related cytokine that is elevated in skin wound tissue and is significantly correlated with pruritus intensity [31]. This evidence suggests that Th2 cells and their cytokines, especially IL-4 and IL-13, contribute to keloid development, and the Th2 axis may become a breakout area of research in keloid pruritus treatments.
Dupilumab, which was approved for the treatment of moderate-to-severe atopic dermatitis (AD), inhibits type 2 inflammation via the IL-4/IL-13 pathway, also known as the Th2 axis [32]. Interestingly, AD is an independent risk factor for the development of keloids [33]. The higher risk of keloid formation in AD patients suggests an association between keloids and the Th2 axis. Diaz et al. [14] reported a new use of dupilumab in the treatment of keloids. In a patient with both severe AD and keloids, after 7 months of treatment with a 300 mg dupilumab subcutaneous injection every 2 weeks, AD improved significantly, with a 50% reduction in the total size of the fibrotic plaque, shrinking of the large keloid, flattening of the surrounding margins, and full removal of the smaller keloid. Furthermore, dupilumab relieved pruritus in a phase 3 study of patients with moderate-to-severe AD, indicating that it plays a role in blocking signals from itch neurons [15] and may also improve pruritus of keloids. Injection site reactions are the most common adverse event associated with dupilumab. Other relatively rare adverse events include ocular complications (e.g., dry eyes, conjunctivitis, blepharitis, keratitis, and pruritus), head and neck dermatitis, onset of psoriatic lesions, progression of cutaneous T-cell lymphoma, alopecia areata, hypereosinophilia, and arthritis. Most of these adverse effects can be controlled during continued treatment with dupilumab, but some (such as severe conjunctivitis) may result in discontinuation of treatment [16][17]. Above all, dupilumab, which targets the IL-4/IL-13 pathway, may provide a better treatment choice for keloid patients, especially those with concurrent AD, and is worthy of further large-scale clinical trials.

2.4. TSLP

Thymic stromal lymphopoietin (TSLP) is an epithelial-cell-derived cytokine that directly stimulates Th2 cytokine production, leading to Th2 inflammation [34], and collagen production, leading to skin fibrosis [18][35][36]. Studies have shown that IL-13 induces skin fibrosis in AD via TSLP [35]. In IL-13 transgenic mice, TSLP neutralization or genetic deletion of TSLPR led to a considerable decrease in fibrocytes and cutaneous fibrosis [35]. Given that TSLP expression is also greatly enhanced in keloid lesions, extending to uninvolved skin [29], it is meaningful to investigate whether TSLP also has a role in promoting fibrosis in keloids. Tezepelumab is a human monoclonal antibody that blocks TSLP, which is also implicated in the pathogenesis of asthma by driving Th2 inflammation of the airway. Clinically, tezepelumab is used to treat severe, uncontrolled asthma in adults and adolescents [18]. The most common adverse events are nasopharyngitis, upper respiratory tract infection, headache, and asthma (which was more frequently observed in the placebo group than in the tezepelumab group) [18]. Tezepelumab can decrease the enhanced immune cells in keloid tissues. Although there have been no clinical trials of tezepelumab in keloids, it is worth assessing its effectiveness as a treatment of keloids that may produce similar effects to dupilumab.

2.5. IL-6

IL-6, secreted by M1 macrophages and mast cells [37][38], promotes chemotaxis, residency, and activation of macrophages [39]. Under stimulation of IL-6 and adenosine receptors, macrophages can generate a large amount of IL-10, TGF-β, and VEGF [39]. IL-6 has been shown to induce collagen and α-smooth muscle actin (α-SMA) expression in dermal fibroblasts [19]. Previous research has shown that IL-6 expression is increased in keloid fibroblasts [40]. Inhibiting IL-6 or IL-6 receptor α (IL-6Rα) in keloid fibroblasts revealed a dose-dependent decrease in collagen type I α 2 and fibronectin 1 mRNAs [41]. Furthermore, the mRNA and protein expression levels of gp130 and several downstream targets in IL-6 signaling (JAK1, STAT3, RAF1, and ELK1) were upregulated in keloid fibroblasts versus normal fibroblasts [41]. These findings suggest that IL-6 signaling may play an important role in keloid pathogenesis and provide clues for strategies targeting IL-6 for keloid therapy and prevention. Immunotherapies that target IL-6 signaling, such as tocilizumab, sarilumab, and siltuximab [19], should be further investigated in keloids.
Tocilizumab blocks the IL-6 receptor (IL-6R) and is approved in more than 100 countries worldwide for the treatment of rheumatoid arthritis (RA) and juvenile idiopathic arthritis (JIA) [19]. In fibrotic diseases, the 2018 ACR conference reported the results of a phase III randomized controlled trial of tocilizumab in systemic sclerosis (SSc) treatment [19], in which 210 patients with SSc received 162 mg subcutaneous tocilizumab treatment. Although the primary endpoint was not met at the end of the experiment, the forced vital capacity of patients taking tocilizumab was superior to that of patients taking a placebo, indicating that tocilizumab significantly inhibited the worsening of respiratory disorders due to SSc-induced pulmonary fibrosis. On this basis, tocilizumab is also a valuable candidate for skin fibrosis treatment. In terms of adverse events, the most common adverse events associated with long-term use (96 weeks) were infections, such as pneumonia, infectious tenosynovitis, sepsis, and otitis media [42]. A decrease in neutrophils is often observed during treatment with tocilizumab in RA patients; however, neutropenia is not due to the myelotoxic effects of tocilizumab but rather to changes in the localization of circulating neutrophils [19].

References

  1. Andrews, J.P.; Marttala, J.; Macarak, E.; Rosenbloom, J.; Uitto, J. Keloids: The paradigm of skin fibrosis-Pathomechanisms and treatment. Matrix Biol. 2016, 51, 37–46.
  2. Ogawa, R. Keloid and Hypertrophic Scars Are the Result of Chronic Inflammation in the Reticular Dermis. Int. J. Mol. Sci. 2017, 18, 606.
  3. Robles, D.T.; Moore, E.; Draznin, M.; Berg, D. Keloids: Pathophysiology and management. Dermatol. Online J. 2007, 13, 9.
  4. Gauglitz, G.G.; Korting, H.C.; Pavicic, T.; Ruzicka, T.; Jeschke, M.G. Hypertrophic scarring and keloids: Pathomechanisms and current and emerging treatment strategies. Mol. Med. 2011, 17, 113–125.
  5. Miles, O.J.; Zhou, J.; Paleri, S.; Fua, T.; Ramakrishnan, A. Chest keloids: Effect of surgical excision and adjuvant radiotherapy on recurrence, a systematic review and meta-analysis. ANZ J. Surg. 2021, 91, 1104–1109.
  6. Werner, S.; Grose, R. Regulation of wound healing by growth factors and cytokines. Physiol. Rev. 2003, 83, 835–870.
  7. Gauglitz, G.G. Management of keloids and hypertrophic scars: Current and emerging options. Clin. Cosmet. Investig. Dermatol. 2013, 6, 103–114.
  8. Chen, Y.; Jin, Q.; Fu, X.; Qiao, J.; Niu, F. Connection between T regulatory cell enrichment and collagen deposition in keloid. Exp. Cell Res. 2019, 383, 111549.
  9. Limmer, E.E.; Glass, D.A. A Review of Current Keloid Management: Mainstay Monotherapies and Emerging Approaches. Dermatol. Ther. 2020, 10, 931–948.
  10. Teicher, B.A. TGFbeta-Directed Therapeutics: 2020. Pharmacol. Ther. 2021, 217, 107666.
  11. Rice, L.M.; Padilla, C.M.; McLaughlin, S.R.; Mathes, A.; Ziemek, J.; Goummih, S.; Nakerakanti, S.; York, M.; Farina, G.; Whitfield, M.L.; et al. Fresolimumab treatment decreases biomarkers and improves clinical symptoms in systemic sclerosis patients. J. Clin. Investig. 2015, 125, 2795–2807.
  12. McGaraughty, S.; Davis-Taber, R.A.; Zhu, C.Z.; Cole, T.B.; Nikkel, A.L.; Chhaya, M.; Doyle, K.J.; Olson, L.M.; Preston, G.M.; Grinnell, C.M.; et al. Targeting Anti-TGF-beta Therapy to Fibrotic Kidneys with a Dual Specificity Antibody Approach. J. Am. Soc. Nephrol. 2017, 28, 3616–3626.
  13. Andre, P.; Denis, C.; Soulas, C.; Bourbon-Caillet, C.; Lopez, J.; Arnoux, T.; Blery, M.; Bonnafous, C.; Gauthier, L.; Morel, A.; et al. Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells. Cell 2018, 175, 1731–1743.e13.
  14. Diaz, A.; Tan, K.; He, H.; Xu, H.; Cueto, I.; Pavel, A.B.; Krueger, J.G.; Guttman-Yassky, E. Keloid lesions show increased IL-4/IL-13 signaling and respond to Th2-targeting dupilumab therapy. J. Eur. Acad. Dermatol. Venereol. 2020, 34, e161–e164.
  15. Simpson, E.L.; Bieber, T.; Guttman-Yassky, E.; Beck, L.A.; Blauvelt, A.; Cork, M.J.; Silverberg, J.I.; Deleuran, M.; Kataoka, Y.; Lacour, J.P.; et al. Two Phase 3 Trials of Dupilumab versus Placebo in Atopic Dermatitis. N. Engl. J. Med. 2016, 375, 2335–2348.
  16. Kychygina, A.; Cassagne, M.; Tauber, M.; Galiacy, S.; Paul, C.; Fournie, P.; Simon, M. Dupilumab-Associated Adverse Events During Treatment of Allergic Diseases. Clin. Rev. Allergy Immunol. 2022, 62, 519–533.
  17. Narla, S.; Silverberg, J.I.; Simpson, E.L. Management of inadequate response and adverse effects to dupilumab in atopic dermatitis. J. Am. Acad. Dermatol. 2022, 86, 628–636.
  18. Menzies-Gow, A.; Corren, J.; Bourdin, A.; Chupp, G.; Israel, E.; Wechsler, M.E.; Brightling, C.E.; Griffiths, J.M.; Hellqvist, A.; Bowen, K.; et al. Tezepelumab in Adults and Adolescents with Severe, Uncontrolled Asthma. N. Engl. J. Med. 2021, 384, 1800–1809.
  19. Kang, S.; Tanaka, T.; Narazaki, M.; Kishimoto, T. Targeting Interleukin-6 Signaling in Clinic. Immunity 2019, 50, 1007–1023.
  20. Lee, T.Y.; Chin, G.S.; Kim, W.J.; Chau, D.; Gittes, G.K.; Longaker, M.T. Expression of transforming growth factor beta 1, 2, and 3 proteins in keloids. Ann. Plast. Surg. 1999, 43, 179–184.
  21. Shaffer, J.J.; Taylor, S.C.; Cook-Bolden, F. Keloidal scars: A review with a critical look at therapeutic options. J. Am. Acad. Dermatol. 2002, 46 (Suppl. 2), S63–S97.
  22. Sanjabi, S.; Oh, S.A.; Li, M.O. Regulation of the Immune Response by TGF-beta: From Conception to Autoimmunity and Infection. Cold Spring Harb. Perspect. Biol. 2017, 9, a022236.
  23. Dufour, A.M.; Alvarez, M.; Russo, B.; Chizzolini, C. Interleukin-6 and Type-I Collagen Production by Systemic Sclerosis Fibroblasts Are Differentially Regulated by Interleukin-17A in the Presence of Transforming Growth Factor-Beta 1. Front. Immunol. 2018, 9, 1865.
  24. Zhang, T.; Wang, X.F.; Wang, Z.C.; Lou, D.; Fang, Q.Q.; Hu, Y.Y.; Zhao, W.Y.; Zhang, L.Y.; Wu, L.H.; Tan, W.Q. Current potential therapeutic strategies targeting the TGF-beta/Smad signaling pathway to attenuate keloid and hypertrophic scar formation. Biomed. Pharmacother. 2020, 129, 110287.
  25. Lonning, S.; Mannick, J.; McPherson, J.M. Antibody targeting of TGF-beta in cancer patients. Curr. Pharm. Biotechnol. 2011, 12, 2176–2189.
  26. Isaka, Y. Targeting TGF-beta Signaling in Kidney Fibrosis. Int. J. Mol. Sci. 2018, 19, 2532.
  27. Shan, M.; Liu, H.; Hao, Y.; Song, K.; Feng, C.; Wang, Y. The Role of CD28 and CD8(+) T Cells in Keloid Development. Int. J. Mol. Sci. 2022, 23, 8862.
  28. Xu, H.; Zhu, Z.; Hu, J.; Sun, J.; Wo, Y.; Wang, X.; Zou, H.; Li, B.; Zhang, Y. Downregulated cytotoxic CD8(+) T-cell identifies with the NKG2A-soluble HLA-E axis as a predictive biomarker and potential therapeutic target in keloids. Cell Mol. Immunol. 2022, 19, 527–539.
  29. Wu, J.; Del Duca, E.; Espino, M.; Gontzes, A.; Cueto, I.; Zhang, N.; Estrada, Y.D.; Pavel, A.B.; Krueger, J.G.; Guttman-Yassky, E. RNA Sequencing Keloid Transcriptome Associates Keloids With Th2, Th1, Th17/Th22, and JAK3-Skewing. Front. Immunol. 2020, 11, 597741.
  30. Zhu, J. T Helper Cell Differentiation, Heterogeneity, and Plasticity. Cold Spring Harb. Perspect. Biol. 2018, 10, a030338.
  31. Hawash, A.A.; Ingrasci, G.; Nouri, K.; Yosipovitch, G. Pruritus in Keloid Scars: Mechanisms and Treatments. Acta Derm. Venereol. 2021, 101, adv00582.
  32. Guttman-Yassky, E.; Bissonnette, R.; Ungar, B.; Suarez-Farinas, M.; Ardeleanu, M.; Esaki, H.; Suprun, M.; Estrada, Y.; Xu, H.; Peng, X.; et al. Dupilumab progressively improves systemic and cutaneous abnormalities in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2019, 143, 155–172.
  33. Lu, Y.Y.; Lu, C.C.; Yu, W.W.; Zhang, L.; Wang, Q.R.; Zhang, C.L.; Wu, C.H. Keloid risk in patients with atopic dermatitis: A nationwide retrospective cohort study in Taiwan. BMJ Open 2018, 8, e022865.
  34. He, R.; Oyoshi, M.K.; Garibyan, L.; Kumar, L.; Ziegler, S.F.; Geha, R.S. TSLP acts on infiltrating effector T cells to drive allergic skin inflammation. Proc. Natl. Acad. Sci. USA 2008, 105, 11875–11880.
  35. Oh, M.H.; Oh, S.Y.; Yu, J.; Myers, A.C.; Leonard, W.J.; Liu, Y.J.; Zhu, Z.; Zheng, T. IL-13 induces skin fibrosis in atopic dermatitis by thymic stromal lymphopoietin. J. Immunol. 2011, 186, 7232–7242.
  36. Simpson, E.L.; Parnes, J.R.; She, D.; Crouch, S.; Rees, W.; Mo, M.; van der Merwe, R. Tezepelumab, an anti-thymic stromal lymphopoietin monoclonal antibody, in the treatment of moderate to severe atopic dermatitis: A randomized phase 2a clinical trial. J. Am. Acad. Dermatol. 2019, 80, 1013–1021.
  37. Yu, Y.; Wu, H.; Zhang, Q.; Ogawa, R.; Fu, S. Emerging insights into the immunological aspects of keloids. J. Dermatol. 2021, 48, 1817–1826.
  38. Hesketh, M.; Sahin, K.B.; West, Z.E.; Murray, R.Z. Macrophage Phenotypes Regulate Scar Formation and Chronic Wound Healing. Int. J. Mol. Sci. 2017, 18, 1545.
  39. Zhou, X.; Franklin, R.A.; Adler, M.; Jacox, J.B.; Bailis, W.; Shyer, J.A.; Flavell, R.A.; Mayo, A.; Alon, U.; Medzhitov, R. Circuit Design Features of a Stable Two-Cell System. Cell 2018, 172, 744–757.e17.
  40. Xue, H.; McCauley, R.L.; Zhang, W. Elevated interleukin-6 expression in keloid fibroblasts. J. Surg. Res. 2000, 89, 74–77.
  41. Ghazizadeh, M.; Tosa, M.; Shimizu, H.; Hyakusoku, H.; Kawanami, O. Functional implications of the IL-6 signaling pathway in keloid pathogenesis. J. Investig. Dermatol. 2007, 127, 98–105.
  42. Khanna, D.; Lin, C.J.F.; Furst, D.E.; Wagner, B.; Zucchetto, M.; Raghu, G.; Martinez, F.J.; Goldin, J.; Siegel, J.; Denton, C.P. Long-Term Safety and Efficacy of Tocilizumab in Early Systemic Sclerosis-Interstitial Lung Disease: Open-Label Extension of a Phase 3 Randomized Controlled Trial. Am. J. Respir. Crit. Care Med. 2022, 205, 674–684.
More
Information
Subjects: Dermatology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 314
Revisions: 2 times (View History)
Update Date: 06 Nov 2023
1000/1000
Video Production Service