Immunotherapy of Keloids: Comparison
Please note this is a comparison between Version 2 by Camila Xu and Version 1 by Dongqing Li.

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][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 [47][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.

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
Video Production Service