Next Article in Journal
The Role of the Endometrial Microbiota in Endometrial Cancer: A Systematic Review of the Literature
Previous Article in Journal
Assessing Cardiovascular Risk in Geriatric Patients Without Atherosclerotic Cardiovascular Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

What Is New in Morphea—Narrative Review on Molecular Aspects and New Targeted Therapies

by
Tomasz Stein
1,*,
Paulina Cieplewicz-Guźla
1,
Katarzyna Iżykowska
2,
Monika Pieniawska
2,
Ryszard Żaba
1,
Aleksandra Dańczak-Pazdrowska
1 and
Adriana Polańska
1
1
Department of Dermatology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
2
Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(23), 7134; https://doi.org/10.3390/jcm13237134
Submission received: 19 September 2024 / Revised: 15 November 2024 / Accepted: 17 November 2024 / Published: 25 November 2024
(This article belongs to the Section Immunology)

Abstract

:
Morphea, also known as localized scleroderma, is an autoimmune chronic connective tissue disease. It is characterized by excessive collagen deposition in the dermis and/or subcutaneous tissue. The etiopathogenesis of this disease is not fully understood, with endothelial cell damage, immunological disorders, extracellular matrix disorders and factors such as infection, trauma and other autoimmune diseases being considered. As medicine advances, there is increasing evidence that genetic factors play a significant role in disease risk and progression. In addition to environmental factors and genetic predisposition, epigenetic factors may be potential triggers for morphea. Epigenetics studies changes that affect gene expression without altering the DNA sequence, such as microRNAs, long non-coding RNAs or DNA methylation. Understanding the pathogenesis of this disease is key to identifying potential new treatments. There are anecdotal reports of good therapeutic effects following the use of biological drugs such as tocilizumab, a humanized IgG monoclonal antibody; abatacept, a recombinant soluble fusion protein; JAK inhibitors, such as tofacitinib and baricitinib; and a drug used successfully in cancer treatment, imatinib, a tyrosine kinase receptor inhibitor. In this article, we aim to review up-to-date knowledge on the pathogenesis of morphea, with particular emphasis on genetic and epigenetic factors. In addition, we present the new options of morphea treatment based on several case series treated with new drugs that are potential targets for the development of therapies for this disease.

1. Introduction

Morphea, also known as localized scleroderma, is a chronic autoimmune connective tissue disease with an incompletely understood etiopathogenesis. It is characterized by excessive collagen deposition in the dermis and/or subcutaneous tissue [1]. The disease may also affect deeper tissues such as adipose tissue, fascia, muscle and bone [2]. The clinical picture usually shows an erythematous inflammatory patch and waxy yellow foci of induration [3]. The exact cause that initiates the disease process is still unknown. Certain stimuli such as drugs/infection or trauma lead to dysregulation of angiogenesis and the immune system in genetically predisposed individuals [4]. In particular, T lymphocyte activation and the release of interferon-γ-related cytokines are involved, resulting in the activation of inflammatory and profibrotic pathways leading to excessive collagen production [5,6].
Morphea is a rare autoimmune connective tissue disease, and its incidence appears to be underestimated. The only population-based study conducted by Peterson et al. in Olmsted County, Minnesota, USA, between 1960 and 1993 suggests that the incidence of morphea is approximately 0.4–2.7 cases/100,000 [7]. The disease is 2.6–6 times more common in women than in men [8]. Based on retrospective studies, morphea appears to be more common in Caucasians [9,10]. The incidence is similar in children and adults. There are two peaks of morphea incidence, the first between 2 and 14 years of age and the second in adults between 40 and 50 years of age [11].
Several forms of morphea have been described, each with different clinical manifestations and degrees of connective tissue involvement. In 2009, Kreuter et al. proposed a division into five main clinical types of morphea: limited, generalized, linear (which includes the linear limb variant, en coup de sabre and hemifacial atrophy), deep and mixed [12]. This classification is supported by the European Dermatological Forum [13] and the Polish Dermatological Society [14]. In adults, the most common clinical form of morphea is the plaque-like variety (morphea en plaques, MEP) [15], while in children it is the linear variety [16].
Systemic sclerosis, which is a distinct entity, should be considered in the broad differential diagnosis. Features suggestive of systemic sclerosis include a mask-like face, microstomia, telangiectasias, Raynaud’s sign, sclerodactyly, finger ulcers, the presence of antibodies (anti-Scl-70 and anti-centromere) and the involvement of internal organs [3,17].
Very similar clinically to morphea is lichen sclerosus. Morphea may coexist with lichen sclerosus (LS), especially with its extragenital form. Coexistence of lichen sclerosus and morphea in one lesion has been described clinically, histopathologically and dermoscopically [18]. Kreuteur et al. observed the coexistence of lichen sclerosus and morphea on histopathologic examination in 5.7% of morphea patients [19].
LS similar to morphea is a chronic, inflammatory dermatosis with an incompletely explained pathogenesis with a predilection for the anogenital area. Genetic, autoimmune, environmental, infectious and hormonal factors are believed to play a role. The presence of inflammation caused by chronic irritation, trauma and moisture in the genital area is also important [20]. The incidence is underestimated; it is assumed that in both sexes it may be 0.1% to 0.3% [21]. LS can occur at any age and in both sexes, with the ratio of women to men being as high as 10:1 [22]. In women, there are two peaks of LS incidence—the first before puberty, the second in the peri- and postmenopausal period, which is associated with low estrogen levels in these periods. However, it is currently believed that LS can occur in women of any age [23,24]. In contrast, in men, two peaks of occurrence are observed—in the third and sixth decades of life [25].
LS occurs in genital and extragenital forms, with 85% of patients having lesions in the genital mucosa [26].
In women, lesions are most often located in the prepuce of the clitoris, labia minora and majora, perineum and perianal area. These are hyperkeratotic, waxy, ivory-colored lumps that merge with each other and are accompanied by intense itching [20]. The most important complication of untreated lesions is scarring, leading to narrowing of the vaginal opening, pain and dyspareunia [24]. In prepubertal girls, the clinical picture is similar to that in adult patients, but erosions, petechiae and fissures are more frequently observed [24].
LS in males is usually located on the foreskin and glans penis, and anus involvement is rare. The occurrence of porcelain-white papules leads to phimosis, which is observed in almost 100% of boys with LS. Additionally, scarring can lead to narrowing of the external urethral opening and, as a consequence, dysuria and sexual dysfunction [27].
Skin lesions in the extragenital area are rarely observed, occurring mainly in women. They are usually asymptomatic, ivory-colored lumps located in the submammary region, neck, arms, inner thighs, wrists, and upper back in places of pressure or trauma [26].
LS is diagnosed based on the typical clinical picture; a biopsy of skin lesions for histopathological examination is performed in doubtful situations or when malignant changes are suspected [22].
The differential diagnosis of morphea is presented in Table 1.
Imaging methods can be used to monitor morphea. Ultrasonography is useful for assessing the extent and depth of the disease process and enables measurement of the thickness of the dermis and adipose tissue [28]. High-frequency ultrasonography (HF-USG) can be used to monitor the dynamics of the disease process. In the sclerosis phase, an increase in echogenicity and thickening of the skin is observed. In the inflammatory phase, an echogenicity decrease is observed [28,29]. Salgueiro et al. [30] observed an ultrasound symptom called the “sun sign”. This is a hyperechoic halo surrounding the superficial subcutaneous veins of the limbs in cross-section. There are interesting anecdotal data on the use of several other noninvasive methods like infrared thermography (IRT, skin temperature), laser Doppler imaging (LDI, skin blood flow) and multispectral imaging (MSI, oxygenation) in morphea, although their use requires further research [31].
Depending on the clinical form of morphea, its activity or severity, the depth and extent of the lesions and the degree of tissue damage, local and systemic treatments are used in therapy [4]. Decisions must take into account the chronic and recurrent nature of the disease and the presence of atrophy and extracutaneous complications, which can lead to significant disability [14] (Figure 1).

2. Pathogenesis

Morphea is a chronic disease whose pathogenesis is not fully understood. Several factors may play a role in the development of the disease, including genetic, epigenetic or environmental factors [2]. Damage to the endothelial cells is thought to be of primary importance in the pathogenesis of the disease and may initiate the disease process [32]. The following insults are mentioned: trauma, infection, radiation or drugs (β-blockers, bleomycin, bromocriptine, D-penicillamine). They cause microvascular damage and secondary activation of T lymphocytes [13]. These, in turn, release cytokines and growth factors that play a key role in the fibrotic process. Studies have confirmed that transforming growth factor β (TGF-β), interleukin IL-1β, IL-4, IL-6, IL-10, IL-27, interferon gamma (INF-γ) and, more recently, IL-17A are essential for inflammation and fibrosis [33,34,35,36,37,38]. Ultimately, activation of these pro-inflammatory and fibrosis-inducing mediators leads to excessive collagen production and reduced production of metalloproteinases responsible for collagen degradation [13].
In Figure 2, we show the pathogenesis of morphea.

2.1. Endothelial Cell Damage

The most likely earliest phenomenon underlying the pathogenesis of morphea is thought to be endothelial cell activation resulting from endothelial damage [39]. Endothelial cell activation, together with an increase in endothelin-1 production and a decrease in prostacyclin production, leads to vasoconstriction. Other factors contributing to vasculopathy include an imbalance between pro- and anti-angiogenic mediators, the presence of fibroblast-derived anti-angiogenic factors, and reduced levels of erythropoiesis-associated factor (Friend leukemia integration factor 1, Fli1), which is responsible for regulating immune function and collagen production. Damaged endothelial cells release cytokines that increase the production of adhesion molecules such as intracellular adhesion molecule (ICAM)-1, vascular cell adhesion molecule (VCAM)-1, E-selectin, and P-selectin. Increased levels of adhesion molecules, in turn, lead to increased recruitment of T lymphocytes, which then release cytokines that stimulate the fibrotic process, including IL-4, IL-6, and TGF-β [4]. Damage to the endothelial cells leads to obliteration of the lumen of small blood vessels and release of cellular antigens, which may be the initial trigger for inflammatory and autoimmune responses. The vascular theory in the pathogenesis of morphea is supported by the histopathological picture of the lesional skin. In the inflammatory phase of morphea, endothelial cells are swollen and activated, and there is thickening of the vessel walls surrounded by an inflammatory infiltrate and active fibroblasts. In the late phase of morphea, basement membrane proliferation and a decrease in vascular density are observed [40].

2.2. Immune Dysfunction

An important component in the pathogenesis of morphea is the activation of the immune system. Increased production of adhesion molecules such as ICAM-1, VCAM-1, E-selectin and P-selectin leads to increased recruitment of inflammatory cells, including T lymphocytes, monocytes and other immune cells [41]. Studies using flow cytometry in morphea patients (adults and children) have shown that the immune response is dominated by CD4+ helper T cells (Th) and there is a decrease in regulatory T cells (Treg) [42,43,44]. Histopathological examination of skin lesions taken during the early inflammatory phase of morphea has also shown activation of predominantly T lymphocytes, which may indicate their involvement in tissue damage and fibrosis induction [40]. Studies suggest the existence of an imbalance in the Th lymphocyte population, with a predominance of Th2 over Th1 lymphocytes, which also favors fibrotic processes [44]. Ihn et al., analyzing sera from patients with morphea and SSc, showed elevated levels of CD30 (Th2 lymphocyte surface antigen), which may also indicate that the immune response is associated with increased Th2 lymphocyte activity [5]. It has been postulated that the early, active phase of morphea is dominated by Th1 and Th17 lymphocyte responses. Th1 lymphocytes produce IFN-γ and IL-2, which are stimulated by IL-12 [45]. Cytokines such as interferon gamma-induced protein 10 (IP-10), monocyte chemotactic protein-1 (MCP-1), IL-17a, IL-12p70, granulocyte-macrophage colony-stimulating factor (GM-CSF), platelet-derived growth factor-beta, platelet-derived growth factor-bb (PDGF-bb), IFN-α2 and IFN-γ, measured in peripheral blood mononuclear cells (PBMCs), were increased in morphea patients compared to controls [46]. IL-12 stimulates Th1 lymphocytes to produce IFN-γ, which induces the expression of IP-10 [46]. IP-10 is produced by fibroblasts and endothelial cells, among others. IP-10 is thought to be a chemokine associated with the stimulation of a pro-inflammatory, Th1-dependent response and is positively correlated with disease activity. It has been suggested that this protein may be a future serological marker of disease activity [46]. Th17 lymphocytes produce IL-17A, IL-21, IL-22 [44]. Dańczak-Pazdrowska et al. demonstrated increased IL-17A gene expression in PBMCs from morphea patients compared to controls and a negative correlation between disease duration and IL-17A gene expression [34]. O’Brien et al. also demonstrated increased IL-17A gene expression in PBMC and skin of morphea patients and showed a positive correlation between increased IL-17A gene expression and shorter disease duration [47]. Torok et al. conducted a study in a group of 69 children diagnosed with morphea and showed significantly higher plasma levels of IP-10 and IL-17A and other cytokines—MCP-1, IL-12p70, GM-CSF, PDGF-bb, IFN-α2 and IFN-γ—in morphea patients compared to controls. In summary, the results of this study suggest that a pro-inflammatory response dependent on Th1 (IFN-γ) and Th17 (IL-17A) lymphocytes is more predominant in morphea than in SSc. Increased levels of these cytokines correlated with shorter disease duration. In addition, IL-4, IL-5 and IL-13 secreted by Th2 lymphocytes were not significantly increased [46]. It has been postulated that a Th2 lymphocyte-dependent inflammatory response predominates in the sclerosing phase of morphea [41]. Th2 lymphocytes produce IL-4, IL-5, IL-6, IL-10 and IL-13, which correlate with fibrotic processes [46]. IL-4 induces the production of TGF-β, which not only stimulates collagen production but also induces the differentiation of fibroblasts into smooth muscle cell myofibroblasts. The accumulation of myofibroblasts enhances the fibrotic process by increasing the stiffness of the extracellular matrix [48]. In addition, TGF-β stimulates the production of type I collagen, type III collagen and other extracellular matrix proteins by fibroblasts [49]. In addition, it has been suggested that the distribution of CD34+ skin dendritic cells (DCs) is altered during the sclerosing phase. CD34 expression in the stroma was significantly lower in patients with morphea than in healthy controls [50]. Some studies have reported significant numbers of plasmocytoid DCs in skin lesions in the deeper layers of the skin, around blood vessels and around collagen fibers in the subcutaneous tissue. They are thought to play an important role in both the development of immune tolerance mechanisms and the activation of autoreactive T cells [51].

2.3. Disorders of the Extracellular Matrix

The main regulator of the fibrosis process is TGF-β [40]. An increase in its concentration causes an increase in the expression of collagen types I, III, VI, X, fibronectin and proteoglycans [6,52]. It also causes a decrease in the production of proteases and an increase in their inhibitors [6]. Extracellular matrix metalloproteinases (MMPs) are also inactivated. Tomimura et al. showed that anti-MMP antibodies were present in patients with morphea and SSc, but not in healthy controls [53]. The role of insulin-like growth factor (IGF) in the pathogenesis of morphea has also been highlighted [4]. Fawzi et al. showed increased IGF gene expression in the skin of morphea patients compared to controls [54]. IGF increases collagen production, recruits fibroblasts and activates excessive extracellular matrix production [54].

2.4. Genetics Factors

Understanding the genetic basis of morphea is critical to uncovering the mechanisms underlying this disease and identifying potential treatment targets. Although most cases of morphea are sporadic, reports of familial aggregation suggest a potential hereditary component [55]. This means that genetic factors may contribute to susceptibility and play a significant role in the development and progression of morphea.

2.4.1. HLA Involvement

Human leukocyte antigen (HLA) has been implicated as the most promising target in the pathogenesis of morphea, suggesting a genetic predisposition to the disease [2]. HLA molecules are cell surface glycoproteins that primarily function to present both endogenous and exogenous antigens to T lymphocytes, which facilitate their recognition and response, thereby regulating the immune system [56]. Several studies have identified specific HLA alleles that are more common in people with morphea compared to the healthy population. Papara et. al. summarized a comprehensive correlation of the most significant associations within HLA-DRB1*04:04 and HLA-B*37, emphasizing that the alleles associated with morphea were different from those associated with systemic sclerosis (SSc), indicating that morphea is immunogenetically distinct from SSc [2]. Moreover, the HLA-class II—HLA-DQB1*02:01, HLA-DRB1*03:01 and HLA-DQA1*03:00, and HLA-class I—HLA-C*08 and HLA-C*15—have also been implicated in morphea patients [57]. The presence of HLA alleles might affect the disease’s severity and progression, possibly via pathways involving autoimmunity and sustained inflammatory processes. Moreover, HLA alleles in morphea patients were described to be strongly associated with multiple sclerosis (MS) and autoimmune thyroid disease (AITD) [2]. As mentioned above, HLA-DRB1*04:04 has also been associated with an increased risk of RA in cohorts of patients with morphea, supporting the hypothesis that autoimmune aberrations are involved in the pathogenesis of the condition [57]. Another study investigating the gene expression profiles of juvenile morphea identified significant upregulation of several HLA class II genes, including HLA-DQA1, HLA-DQB1, and HLA-DRB1, which were highly correlated with both inflammation and fibrosis scores [58]. These HLA alleles were also identified in a case-control study comparing the peripheral blood of patients with morphea with a healthy control group, indicating susceptibility to morphea [58]. Additional HLA testing in juvenile SSc supports the notion that HLA-DRB1 and HLA-DQA1 are associated with greater susceptibility to scleroderma [58]. The overlap between morphea and SSc, together with the correlation of these HLA genes with inflammation and collagen deposition in the current study, highlights the interconnection of inflammation and fibrosis in morphea, further characterizing morphea as a fibrotic disease driven by inflammatory processes [58].

2.4.2. Cutaneous Mosaicism

Recent studies have highlighted the involvement of cutaneous mosaicism as a possible factor in morphea’s pathogenesis [59]. Mosaicism refers to the presence of genetically distinct cell populations in an individual resulting from postzygotic mutations [60]. Two subtypes of mosaicism can be distinguished: genomic (caused by de novo mutation, altering the DNA sequence) or epigenomic (change in gene expression not correlated with DNA sequence alterations) [61]. This phenomenon suggests that somatic mutations occurring in embryogenesis may contribute to the development of morphea lesions. Studies have identified mosaic mutations in genes associated with pathways involved in skin development and immune system regulation [61,62,63,64], which potentially may have an impact on the localized scleroderma phenotype, and the presence of mosaicism highlights the possibly complex genetic landscape of morphea—mainly the linear subtype, which follows Blaschko lines during epidermal development, determining the linear distribution of fibrosis processes [65,66].

2.4.3. Transcriptome Alternations

Morphea transcriptome analysis revealed a predominance of IFN-γ-driven T helper 1 immune dysregulation, with relatively few identified fibrotic pathways [67]. In particular, gene expression profiles in morphea-affected skin were found to be consistent with an inflammatory subset of systemic sclerosis (SSc), clearly distinct from the fibroproliferative subset of SSc [67]. Additionally, the not-affected skin of patients with diagnosed morphea did not have the pathological gene expression signatures observed in not-affected SSc skin samples. Further examination of the IFN-γ-mediated chemokines CXCL9 and CXCL10 revealed increased transcription in the skin but not in the blood serum [67]. Despite increased transcriptional activity in the skin, serum CXCL9 levels were elevated and associated with active, widespread skin involvement [67]. These findings suggest that morphea is primarily a skin-directed disorder characterized by dysregulation of the T helper 1 immune system, which contrasts with the fibrotic features and systemic transcriptional changes observed in SSc [67]. A different study, comparing pediatric morphea gene signatures with healthy controls, indicated a distinct inflammatory response gene signature (IRGS) expression pattern, emphasizing genes related to IFNα, IFNγ, and TNFα [68]. Gene set enrichment analysis (GSEA) revealed that this IRGS, which includes interferon-induced chemokines such as CXCL9, CXCL10, CXCL11, and IFNγ, was more pronounced in LS patients showing more inflammatory changes [68]. Moreover, elevated levels of CXCL9 and CXCL10 were detected in the serum of patients with morphea, consistent with increased transcription and expression of CXCL9 in skin lesions, and this increase has been linked to inflammation and disease activity [47,67].

2.5. Epigenetics Factors

Epigenetic factors are considered to be triggers of the morphea, next to environmental factors and genetic predisposition [59]. Epigenetics investigates changes that influence gene expression without altering the DNA sequence, like DNA methylation, post-translational modification of histones, microRNAs (miRNAs), and long non-coding RNAs (lnRNAs) [41].

2.5.1. DNA Methylation

Abnormal DNA methylation resulting in altered gene expression has been shown in many autoimmune disorders, including systemic scleroderma, where both PBMCs and dermal fibroblasts were investigated [69]. Not many studies have been performed on morphea patients. The first and only genome-wide DNA methylation study in PBMCs isolated from a cohort of juvenile localized scleroderma (jLS) was performed by Coit et al. [70]. The methylome of jLS patients was compared not only to healthy individuals but also to juvenile systemic sclerosis patients (jSSc), and the majority of differentially methylated sites and genes were unique to either jSSc or jLS, suggesting differences in epigenetic patterns and inflammatory and cell signaling pathways in both diseases. In general, inflammatory pathways were enriched in genes deferentially methylated in jSSc, including STAT3, NF-kB, and IL-15 pathways, while the HIPPO signaling pathway, which regulates cell proliferation and contact activities, was enriched in jLS [70]. In both jSSc and jLS patients compared to controls, consistent hypermethylation in CpG sites within FGFR2 was identified. FGFR2 encodes the fibroblast growth factor receptor 2 protein that has broad biological functions including promoting cell proliferation, survival, migration, and adhesion. Overall, the analysis showed that PBMNc in jSSc and jLS are unique at the epigenetic level and that the epigenic pattern of jSSc appears to be more inflammatory compared to jLS [70].

2.5.2. MicroRNA

MicroRNAs (miRNAs) are small non-coding RNAs that usually bind to complementary sequences in the 3′ untranslated regions (UTR) of mRNAs, which results in inhibiting their translation into protein. They are involved in the regulation of a variety of cellular processes like cell differentiation, proliferation, apoptosis, and immune response [71]. Although the studies on miRNAs in morphea are rather limited, many recent reports have shown that miRNAs are involved in the regulation of processes that are related to this disorder, like fibrosis, including alterations in the TGF-β signaling cascade (miRNA-18, miRNA-20, miRNA-21, miRNA-23b, miRNA-29, miRNA-140-5p, miRNA-146a, miRNA-206), fibroblast proliferation and differentiation (miRNA-21, miRNA-31, miRNA-146a, miRNA-200), and extracellular matrix synthesis and deposition (miRNA-let-7a, miRNA-7, miRNA-26a, miRNA-29, miRNA-129-5p, miRNA-133a, miRNA-133b, miRNA-150, miRNA-196a) [71,72].
Morphea is characterized by skin fibrosis due to excessive deposition of type I collagen, and studies have shown that several miRNAs are regulators of this collagen expression [73,74,75]. One of them, miR-7, was significantly downregulated in skin, dermal fibroblast, and serum of LSc patients compared with healthy controls [73]. Upon specific inhibition of miR-7 in cultured normal dermal fibroblasts, the upregulation of α2(I) collagen protein was observed. Similar studies were performed for miRNA-196a and let-7a [74,75], which are putative regulators of α1(I) and α2(I) components of type I collagen [71]. The analysis showed that the level of miRNA-196a was markedly decreased in LSc skin tissue and serum [75], and blocking the miR-196a with the specific inhibitor in normal cultures of human dermal fibroblasts led to the upregulation of type I collagen. Let-7a expression was also downregulated not only in the skin of LS patients but also in SSc; however, in terms of serum concentration, the level was significantly decreased especially in LSc patients [74]. The inhibition or overexpression of let-7a in human or mouse skin fibroblasts significantly affected the type I collagen protein expression. All those studies proved that downregulation of miR-7, let-7a, and miR-196a and subsequent overexpression of type 1 collagen in dermal fibroblasts may play a key role in the pathogenesis of LSc [73,74,75].
One of the upregulated miRNAs in patients with morphea and systemic scleroderma was miR-155 [76]. The expression was elevated in skin tissue but also in the experimental skin fibrosis mouse models. MiR-155−/− mice showed less fibrosis in the dermis when challenged with bleomycin in terms of skin thickness and collagen content [76]. Both local and systemic miR-155 silencing in vivo remarkably attenuated bleomycin-induced dermal fibrosis, while miR-155 silencing in primary skin fibroblasts inhibited collagen production. Molecular analysis in vitro showed that Wnt/β-catenin and Akt signaling, pathways necessary for fibrosis development, are regulated by miR-155 via direct targeting of CK1α (casein kinase 1α) and SHIP-1 (inositol phosphatase-1) [76]. This extensive analysis of miR-155 showed this miRNA as an important epigenetic player in scleroderma and a potential treatment target.
Another upregulated miRNA in conditions marked by skin fibrosis, including morphea, was the miR-483-5p [77]. Although the functional validation was performed on systemic sclerosis (SS), it proved that miR-483-5p modulates the expression of fibrosis-related genes involved in myofibroblast transition (αSMA, SM22A) and collagen production (Col4A1, Col4A2, Col1A2, Fli-1) in fibroblasts and endothelial cells. This study proposed that miR-483-5p serves as a new molecular marker for the early stages of the disease and contributes to the establishment of the fibrotic phase [77].

2.5.3. Circulating MicroRNAs

Circulating microRNAs (c-miRNAs), which are present in almost all biological fluids, are promising and sensitive biomarkers for various diseases [78]. Their signatures could be used in the clinic to assess the patient state and treatment efficiency. In morphea patients, the serum levels of six miRNAs—miRNA-181b-5p, miRNA-223-3p, miRNA-21-5p, let 7i-5p, miRNA-29a-3p and miRNA-210-3p—were significantly increased compared to healthy individuals [79]. All of those miRNAs were either previously linked to morphea, SS, or other autoimmune disorders or were related to fibrosis [79]. The authors performed a series of correlation analyses between miRNA expression and the severity and clinical symptoms of morphea. The analysis revealed that, in the female morphea patients, some significant correlations between several miRNAs’ serum levels and the severity and other clinical symptoms of morphea were identified; for example, the increased level of let-7i co-existed with less severe morphea, while the miRNA-21 serum level was inversely correlated with inflammation [79]. The diagnostic and prognostic value of circulating miRNAs is worth further investigation.

2.5.4. Long Non-Coding RNAs

Long non-coding RNAs (lncRNAs) are groups of RNA transcripts longer than 200 nucleotides that regulate gene expression at transcriptional and post-transcriptional levels [80]. The lncRNA profile of inflammatory cells was investigated in skin tissue samples from pediatric morphea and fibroblasts co-cultured with CD4+ T lymphocytes. The analysis revealed 11 candidate lncRNAs; among them, the MIR100HG that might be involved in the fibrosis, via the miR-29a-3p/Tab1/TGF-β1 axis, and ZEB1-AS1 via the ZEB1-AS1/miR-141-3p axis, respectively [80]. The involvement of the TGF-β receptor signaling pathway was also confirmed in the study by GO analysis based on the lncRNA expression profiling.

2.6. Other Factors in the Pathogenesis of Morphea

Infectious factors include the occurrence of morphea-like lesions following infections with measles, varicella, Epstein–Barr, hepatitis B and C viruses [81,82,83,84]. Other possible aetiological factors include infection with Borrelia burgdorferi (B. burgdorferi), the role of which remains controversial and debated. The association between morphea and Lyme disease was first suggested by Aberer et al. in 1987 [85]. Since then, numerous reports using both serological methods and PCR detection of B. burgdorferi DNA in a skin lesion have failed to confirm the association between morphea and B. burgdorferi infection [86,87,88,89]. Weide et al. report that late skin lesions in Lyme disease may be associated with pseudosclerotic changes, but these cannot be considered as changes in the course of morphea [90]. The association of morphea with other autoimmune diseases has been described in the medical literature for years. According to Leitenberger et al., 30% of adult patients with morphea have a coexisting autoimmune disease [10], a higher frequency than in the healthy population. The most common autoimmune diseases are plaque psoriasis, systemic lupus erythematosus, multiple sclerosis and vitiligo [8]. However, recent studies have not shown an increased frequency of positive antithyroid antibodies in adult patients with morphea [91], in contrast to the pediatric population where coexistence with autoimmune thyroiditis is significant [92]. Patients with the generalized form of morphea are much more likely to have co-existing autoimmune diseases than patients with other forms of morphea [8]. The medical literature also describes a more frequent co-occurrence of morphea with lichen sclerosus of the anogenital area, with some data suggesting a frequency of 38% [93]. Since 1990, cases of morphea-like changes have been described in patients following radiotherapy, mainly in patients with a history of breast cancer [94]. The pathogenesis of this phenomenon remains unclear. Data to date suggest that ionizing radiation activates fibroblasts to increase secretion of cytokines with profibrotic effects (IL-4, IL-5, TGF-β) [95]. There are also reports in the medical literature of cases of skin fibrosis associated with chemotherapy, following the use of drugs such as docetaxel, paclitaxel, bleomycin, peplomycin, fluoropyridines, gemcitabine, doxorubicin and cyclophosphamide [96] and associated with the use of checkpoint inhibitors [97]. Trauma has also been mentioned as a possible factor in the pathogenesis of morphea. Some authors postulate the occurrence of the Koebnerization phenomenon as an important factor in the pathogenesis of morphea [98,99]. The cause of this reaction is still unknown, but it is thought that cytokines, stress proteins, adhesion molecules and/or autoantigens are released into the skin under the influence of mechanical trauma [99]. There have also been reports of morphea as a reaction to tattooing, with skin lesions limited to the tattooed area [100].

3. Treatment

The treatment of morphea remains a challenge for clinicians because of the poor clinical response to treatment. The main therapeutic options, depending on the type of morphea, include topical steroids, topical calcineurin inhibitors, imiquimod, and in more advanced cases methotrexate, systemic steroids, mycophenolate mofetil, and phototherapy (PUVA, UVA1, UVB) [41].
Treatment decisions should consider the subtype of morphea, the disease’s activity, and the depth of the skin lesions. The earlier treatment starts, the less damage, such as limb deformity, will occur. Topical corticosteroids are the first-line treatment for patients with superficial peripheral lesions. Treatment is prolonged, sometimes requiring intermittent therapy or alternative therapy such as topical tacrolimus [2].
The first-line treatment for extensive or deeply infiltrating lesions is ultraviolet A (UVA1 phototherapy, 340–400 nm), which has greater tissue penetration than UVB [101].
For very active, widespread lesions, systemic treatment with corticosteroids (intravenous or oral) or methotrexate is required. The combination of systemic corticosteroids and methotrexate has the best effect in this group of patients and is the first-line treatment [41].
Mycophenolate mofetil may be an alternative to methotrexate but is rarely used in dermatological practice. Surgical treatment may be considered in patients with the en coup de sabre type [41].
In pediatric patients, similar to adults, treatment depends on the clinical form of morphea, activity, severity or extent of the disease process [102]. In the case of limited skin involvement, the treatment of choice is the external use of topical agents (e.g., calcineurin inhibitors or calcipotriol [103]). Local application of glucocorticosteroids is effective in the case of active lesions, but in the pediatric population their use should be limited due to the risk of atrophy [104]. Phototherapy—UVA-1 or narrowband UVB—is recommended as second-line treatment in children over 12 years of age [13]. When extensive tissue involvement occurs (linear, deep, generalized form), rapid implementation of general treatment is required. The first-line treatment is methotrexate in monotherapy or in combination with oral or intravenous glucocorticosteroids. In the absence of improvement or intolerance to the above-mentioned treatment, the use of mycophenolate mofetil is recommended [102].
In addition to the methods mentioned, we also have surgical procedures. According to European dermatological recommendations, surgical procedures are indicated mainly in the linear form of limbs to correct differences in limb length [13]. Surgical interventions can also be considered in the case of cosmetic defects in the form of en coup de sabre or Parry–Romberg syndrome. To reduce the risk of disease reactivation, surgery should only be performed in the inactive form of morphea. There are no recommendations specifying the appropriate duration of such treatment; it is recommended that the disease be inactive for 3–5 years [105]. Recent studies indicate that in the case of significant esthetic or functional defects in the course of morphea, autologous fat transplantation can be considered [2]. It has also been shown to have anti-inflammatory and antifibrotic effects by reducing the expression of TGF-b1 and collagen type III [106,107]. Due to individual case reports, further research on this treatment method is needed.
As our understanding of the pathogenesis of this disease has progressed and genetic and autoimmune processes have been identified, we will focus on new therapeutic options using biological drugs, antifibrinolytic and small molecule agents.

3.1. Biological Drugs and Small Molecules

IL-6 plays a crucial role in the pathogenesis of morphea. It stimulates collagen production by fibroblasts and the differentiation of CD4+ cells into pathogenic Th17 cells, making it a pro-inflammatory and profibrotic interleukin [108].
Kreuter et al. [109] showed that IL-6 levels are elevated in the skin and serum of patients with morphea.
Tocilizumab is a humanized monoclonal antibody approved by the FDA for treating rheumatoid arthritis, giant cell arteritis, polyarticular juvenile idiopathic arthritis, systemic juvenile idiopathic arthritis and COVID-19 pneumonia [110].
In a randomized trial of subcutaneous tocilizumab in patients with systemic scleroderma, one effect was a reduction in skin thickness compared with placebo [111].
Based on this, it was hypothesized that the drug might play a role in patients with refractory morphea. The drug has shown promising results in several case series.
Lythgoe et al. [112] used tocilizumab in five pediatric patients with linear morphea. These were patients with an active phase of the disease, despite escalation to maximum tolerated doses of methotrexate and one other disease-modifying drug or biological drug.
The dosing regimen was 8 mg/kg for children weighing 30 kg or more and 12 mg/kg for children weighing less than 30 kg, administered as 0-, 2- and 4-week courses followed by 4-week intervals. Patients received the drug intravenously and were treated for 12 to 25 months. All patients tolerated the drug without serious adverse effects. Two of five patients required steroids at the start of tocilizumab therapy, and one patient resumed methotrexate and mycophenolate mofetil. All patients showed improvement in Physician Global Assessment (PGA) disease activity scores at 6 months. However, changes in the modified Localized Skin Severity Index (mLoSSI) were not statistically significant. The skin damage that occurred before tocilizumab treatment is largely irreversible, so future studies should focus on the pediatric population in the early stages of the disease. The authors suggest that intravenous administration of tocilizumab is safe [112].
Zhang et al. [113] described the case of a 6-year-old female patient treated with tocilizumab. The patient had a severe form of morphea, pansclerotic scleroderma. Before tocilizumab, she was treated with methotrexate, prednisone and mycophenolate mofetil. Due to disease progression and progressive fibrosis, mycophenolate mofetil was discontinued and intravenous tocilizumab 300 mg every four weeks was started, while methotrexate was continued at 25 mg weekly. Within a few months, there was significant improvement. The mLoSSI improved from 22 to 6 and the PGA-A from 30 to 17. Pansclerotic scleroderma is an extremely rare subtype of morphea characterized by an aggressive course and generalized thickening of the skin that can lead to joint deformity and, consequently, a significant reduction in joint mobility. The combination of tocilizumab and methotrexate, and therefore the efficacy of this therapy, requires further study in a larger group of patients.
Martini et al. [114] described the cases of two pediatric patients with pansclerotic scleroderma who, after the failure of disease-modifying drugs, were treated with tocilizumab, which reduced disease activity and halted disease progression. These authors suggest that tocilizumab should be used in the early stages of the disease to prevent tissue damage. The descriptions above were related to pediatric patients.
Lonowski et al. [115] presented three adult women between 41 and 69 years; one woman had linear morphea (en coup de sabre) and two women had generalized morphea. The drug was administered intravenously or subcutaneously according to patient preference (subcutaneously at 162 mg/week or intravenously at 8 mg/kg/month). One patient with linear morphea was also treated with mycophenolate mofetil, and one patient with generalized scleroderma was treated with intravenous immunoglobulin. None of the patients required steroids at baseline or during tocilizumab therapy.
Improvement of scleroderma was observed in all patients. CR (Complete remission) was achieved in two patients and partial improvement in three patients. The drug was well tolerated.
Another potential target is the JAK/STAT pathway. In vitro and mouse studies have shown that JAK inhibitors can effectively reduce or even block TGF-β-induced skin fibrosis [116].
The JAK-STAT pathway is thought to mediate the promotion of fibrosis by TGF- β, IL-6 and other factors, as well as play a role in signaling in the fibrotic process [117].
Tang et al. [118] described the use of tofacitinib, a JAK1 and JAK3 inhibitor, in two pediatric patients with limited morphea. The 6-year-old girl was initially treated with glycyrrhizin, topical tacrolimus and UVA1. The patient’s parents did not agree to treatment with methotrexate and mycophenolate mofetil, so the patient was started on tofacitinib at an initial dose of 2.5 mg 2 × day for 4 months, followed by 2.5 mg every other day for a further 2 months. The drug was well tolerated. After 6 months, the mLoSSI improved from 6 to 1 and LosDi (Localized scleroderma damage index) from 7 to 2. In the second patient, a 13-year-old boy diagnosed with linear morphea, hydroxychloroquine, topical tacrolimus and UVA1 were used; his parents refused methotrexate and corticosteroids. Due to disease progression, tofacitinib was added to the treatment regimen—5 mg 2 × day for 2 months and then, due to the rapid clinical response, the dose was reduced to 5 mg per day for a further 3 months. After 5 months, the mLoSSI improved from 7 to 1 and the LosDi from 7 to 2. In both cases, the skin sclerosis resolved, which was confirmed by histopathology and immunofluorescence, where an upregulation of collagen synthesis was shown to be the basis of the treatment.
Baricitinib, a JAK1 and JAK2 inhibitor, has shown a positive effect in a patient with generalized morphea [119].
Another drug, abatacept, is a fusion protein consisting of the extracellular domain of human cytotoxic T-lymphocyte-associated antigen-4 (CLTA-4) fused to a modified Fc fragment of human immunoglobulin G1 (IgG1). It is produced by recombinant DNA technology in Chinese hamster ovary cells [120]. The drug selectively modulates a key costimulatory signal required for full activation of T cells expressing CD28. Abatacept has a more substantial effect on the response of T lymphocytes that are not in contact with any stimulus than on the response of memory T lymphocytes. In vitro studies show that the drug weakens T lymphocytes by reducing their proliferation rate and ability to produce cytokines [120]. It reduces the production of interferon-gamma, interleukin-2 and TNFα by T lymphocytes, and reduces the plasma concentration of interleukin-6, a product of activated macrophages; thus, the drug may reduce skin fibrosis [120].
Stausbøl-Grøn et al. [121] described two patients with deep morphea treated with abatacept, one of whom showed significantly improved skin and mobility. Both were able to reduce their steroid doses.
Fage et al. [122] presented 13 patients with a refractory, severe form of limited scleroderma. Patients were treated with abatacept with a mean time between disease severity assessments of 16 months, a relatively short interval. Improvement and reduction in lesion size and improvement in the mLoSSI were observed during treatment.
Li et al. [123] retrospectively evaluated the safety and efficacy of abatacept in patients with refractory limited morphea. Patients were followed for 12 to 24 months, with clinical assessments at 6-month intervals, including skin and PGA-A measurements. There was a cohort of 18 patients with a mean age of 13.4 years, most of whom had linear scleroderma with musculoskeletal involvement. Patients had been unsuccessfully treated with corticosteroids, methotrexate and/or mycophenolate mofetil. Abatacept was added to maintenance treatment with a disease-modifying drug; 13 patients were also receiving steroids at the start of abatacept treatment. After just 6 months, there was an improvement in the skin and a reduction in PGA-A. In most patients (80%), there was a significant improvement in skin and musculoskeletal activity at 12 months and every 6 months up to 24 months of follow-up. However, 16.7% of patients discontinued treatment due to adverse effects.
Abatacept proved to be an effective treatment, but the authors conclude that prospective studies are needed.

3.2. Antifibrinolytic Agents

The pathophysiology of morphea is based in part on abnormal signaling via the platelet-derived growth factor (PDGF) and TNFβ, which appears to be one of the key processes in the inflammatory response in morphea and systemic scleroderma [124].
A tyrosine kinase inhibitor (Imatinib) is approved for the treatment of chronic myelogenous leukemia and gastrointestinal stromal tumors. This drug interferes with both of these signaling pathways by blocking the activity of c-Kit, c-Abl and PDGF receptors [124].
In addition, Akmetshina et al. [125] concluded that imatinib may cause fibrosis regression by inhibiting collagen synthesis, leading to increased matrix degradation; hence its anti-fibrinolytic effect.
Coelho-Macias et al. [126] described the case of a 50-year-old patient with generalized scleroderma, diagnosed 10 years before the start of imatinib treatment; in addition, the patient developed multiple ulcers on his extremities during the course of the disease. For this reason, immunosuppressive treatment was not attempted, and the patient was only treated with pentoxifylline. The patient was started on imatinib for 12 months (3 months at 200 mg/day and 9 months at 300 mg/day). Clinical improvement was observed during treatment, with normal ulcer healing and reduction in inflammatory lesions, as confirmed by biopsy and ultrasound.
The limitations of the above trials is the small group size and the lack of a control group. These drugs are not without side effects, but no serious adverse events have been reported in the cases mentioned.
We have included all of the above medicines in Table 2.

4. Discussion

The key challenges for clinicians treating patients with morphea are early diagnosis and the earliest possible initiation of treatment to minimize future damage, such as cosmetic sequelae or limb deformity.
The identified immunological, genetic or epigenetic factors offer the opportunity to identify new therapeutic targets for the treatment of morphea subtypes that are resistant to standard therapies.
Small molecule biologics, used successfully in the treatment of rheumatological diseases, for example, could be an alternative therapy in morphea.
However, randomized, double-blind, placebo-controlled clinical trials are needed to provide answers about the efficacy and, above all, the safety of the new drugs and the possibility of treating the disease.

Author Contributions

Conceptualization, A.D.-P.; writing—original draft preparation, T.S., P.C.-G., K.I., M.P. and A.P.; visualization, T.S.; writing—review and editing, A.P. and R.Ż. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. Writing assistance was not utilized in the production of this manuscript.

References

  1. Chiriac, A.; Brzezinski, P.; Chiriac, A.E.; Coroș, M.F.; Moldovan, C.; Podoleanu, C.; Stolnicu, S. The Value of Ultrasonography in the Diagnosis and Monitoring of Localized Morphea—Case Report. J. Interdiscip. Med. 2016, 1, 193–196. [Google Scholar] [CrossRef]
  2. Papara, C.; De Luca, D.A.; Bieber, K.; Vorobyev, A.; Ludwig, R.J. Morphea: The 2023 update. Front. Med. 2023, 10, 1108623. [Google Scholar] [CrossRef] [PubMed]
  3. Ferreli, C.; Gasparini, G.; Parodi, A.; Cozzani, E.; Rongioletti, F.; Atzori, L. Cutaneous Manifestations of Scleroderma and Scleroderma-Like Disorders: A Comprehensive Review. Clin. Rev. Allergy Immunol. 2017, 53, 306–336. [Google Scholar] [CrossRef]
  4. Fett, N.; Werth, V.P. Update on morphea: Part I. Epidemiology, clinical presentation, and pathogenesis. J. Am. Acad. Dermatol. 2011, 64, 217–228. [Google Scholar] [CrossRef]
  5. Ihn, H.; Yazawa, N.; Kubo, M.; Yamane, K.; Sato, S.; Fujimoto, M.; Kikuchi, K.; Soma, Y.; Tamaki, K. Circulating levels of soluble CD30 are increased in patients with localized scleroderma and correlated with serological and clinical features of the disease. J. Rheumatol. 2000, 27, 698–702. [Google Scholar] [PubMed]
  6. Yamamoto, T. Chemokines and chemokine receptors in scleroderma. Int. Arch. Allergy Immunol. 2006, 140, 345–356. [Google Scholar] [CrossRef]
  7. Peterson, L.S.; Nelson, A.M.; Su, W.P.; Mason, T.; O’Fallon, W.M.; E Gabriel, S. The epidemiology of morphea (localized scleroderma) in Olmsted County 1960–1993. J. Rheumatol. 1997, 24, 73–80. [Google Scholar]
  8. Kreuter, A.; Krieg, T.; Worm, M.; Wenzel, J.; Moinzadeh, P.; Kuhn, A.; Aberer, E.; Scharffetter-Kochanek, K.; Horneff, G.; Reil, E.; et al. German guidelines for the diagnosis and therapy of localized scleroderma. J. Dtsch. Dermatol. Ges. 2016, 14, 199–216. [Google Scholar] [CrossRef]
  9. Christen-Zaech, S.; Hakim, M.D.; Afsar, F.S.; Paller, A.S. Pediatric morphea (localized scleroderma): Review of 136 patients. J. Am. Acad. Dermatol. 2008, 59, 385–396. [Google Scholar] [CrossRef]
  10. Leitenberger, J.J.; Cayce, R.L.; Haley, R.W.; Adams-Huet, B.; Bergstresser, P.R.; Jacobe, H.T. Distinct autoimmune syndromes in morphea: A review of 245 adult and pediatric cases. Arch. Dermatol. 2009, 145, 545–550. [Google Scholar] [CrossRef]
  11. Fett, N. Scleroderma: Nomenclature, etiology, pathogenesis, prognosis, and treatments: Facts and controversies. Clin. Dermatol. 2013, 31, 432–437. [Google Scholar] [CrossRef] [PubMed]
  12. Kreuter, A.; Krieg, T.; Worm, M.; Wenzel, J.; Gambichler, T.; Kuhn, A.; Aberer, E.; Scharffetter-Kochanek, K.; Hunzelmann, N. AWMF Guideline no. 013/066. Diagnosis and therapy of circumscribed scleroderma. J. Dtsch. Dermatol. Ges. 2009, 7 (Suppl. S6), S1–S14. [Google Scholar] [CrossRef] [PubMed]
  13. Knobler, R.; Moinzadeh, P.; Hunzelmann, N.; Kreuter, A.; Cozzio, A.; Mouthon, L.; Cutolo, M.; Rongioletti, F.; Denton, C.; Rudnicka, L.; et al. European Dermatology Forum S1-guideline on the diagnosis and treatment of sclerosing diseases of the skin, Part 1: Localized scleroderma, systemic sclerosis and overlap syndromes. J. Eur. Acad. Dermatol. Venereol. 2017, 31, 1401–1424. [Google Scholar] [CrossRef] [PubMed]
  14. Krasowska, D.; Rudnicka, L.; Dańczak-Pazdrowska, A.; Chodorowska, G.; Woźniacka, A.; Lis-Święty, A.; Czuwara, J.; Maj, J.; Majewski, S.; Sysa-Jędrzejowska, A.; et al. Localized scleroderma (morphea). Diagnostic and therapeutic recommendations of the Polish Dermatological Society. Dermatol. Rev. 2019, 106, 333–353. [Google Scholar] [CrossRef]
  15. Marzano, A.V.; Menni, S.; Parodi, A.; Borghi, A.; Fuligni, A.; Fabbri, P.; Caputo, R. Localized scleroderma in adults and children. Clinical and laboratory investigations on 239 cases. Eur. J. Dermatol. 2003, 13, 171–176. [Google Scholar]
  16. Marsol, I.B. Update on the classification and treatment of localized scleroderma. Actas Dermosifiliogr. 2013, 104, 654–666. [Google Scholar] [CrossRef]
  17. Dańczak-Pazdrowska, A.; Cieplewicz, P.; Żaba, R.; Adamski, Z.; Polańska, A. Controversy around the morphea. Postep. Dermatol. Allergol. 2021, 38, 716–720. [Google Scholar] [CrossRef]
  18. Sharma, P.; Khullar, G.; Nagia, S.; Sharma, S. Clinical, histopathological and dermatoscopic overlap of lichen sclerosus and morphea in the same lesion. Australas. J. Dermatol. 2023, 64, e292–e294. [Google Scholar] [CrossRef]
  19. Kreuter, A.; Wischnewski, J.; Terras, S.; Altmeyer, P.; Stücker, M.; Gambichler, T. Coexistence of lichen sclerosus and morphea: A retrospective analysis of 472 patients with localized scleroderma from a German tertiary referral center. J. Am. Acad. Dermatol. 2012, 67, 1157–1162. [Google Scholar] [CrossRef]
  20. Papini, M.; Russo, A.; Simonetti, O.; Borghi, A.; Corazza, M.; Piaserico, S.; Feliciani, C.; Calzavara-Pinton, P. Diagnosis and management of cutaneous and anogenital lichen sclerosus: Recommendations from the Italian Society of Dermatology (SIDeMaST). Ital. J. Dermatol. Venereol. 2021, 156, 519–533. [Google Scholar] [CrossRef]
  21. Wallace, H.J. Lichen sclerosus et atrophicus. Trans. St. Johns Hosp. Dermatol. Soc. 1971, 57, 9–30. [Google Scholar] [PubMed]
  22. Kirtschig, G.; Becker, K.; Günthert, A.; Jasaitiene, D.; Cooper, S.; Chi, C.; Kreuter, A.; Rall, K.; Aberer, W.; Riechardt, S.; et al. Evidence-based (S3) Guideline on (anogenital) Lichen sclerosus. J. Eur. Acad. Dermatol. Venereol. 2015, 29, e1–e43. [Google Scholar] [CrossRef] [PubMed]
  23. Singh, N.; Ghatage, P. Etiology, Clinical Features, and Diagnosis of Vulvar Lichen Sclerosus: A Scoping Review. Obstet. Gynecol. Int. 2020, 2020, 7480754. [Google Scholar] [CrossRef] [PubMed]
  24. Krapf, J.M.M.; Smith, A.B.; Cigna, S.T.; Goldstein, A.T. Presenting Symptoms and Diagnosis of Vulvar Lichen Sclerosus in Premenopausal Women: A Cross-Sectional Study. J. Low. Genit. Tract Dis. 2022, 26, 271–275. [Google Scholar] [CrossRef]
  25. Kizer, W.S.; Prarie, T.; Morey, A.F. Balanitis xerotica obliterans: Epidemiologic distribution in an equal access health care system. South. Med. J. 2003, 96, 9–11. [Google Scholar] [CrossRef]
  26. Powell, J.; Wojnarowska, F. Childhood vulvar lichen sclerosus: An increasingly common problem. J. Am. Acad. Dermatol. 2001, 44, 803–806. [Google Scholar] [CrossRef]
  27. Becker, K. Lichen sclerosus in boys. Dtsch. Ärzteblatt Int. 2011, 108, 53–58. [Google Scholar] [CrossRef]
  28. Wortsman, X.; Wortsman, J.; Sazunic, I.; Carreño, L. Activity assessment in morphea using color Doppler ultrasound. J. Am. Acad. Dermatol. 2011, 65, 942–948. [Google Scholar] [CrossRef]
  29. Polańska, A.; Dańczak-Pazdrowska, A.; Silny, W.; Sadowska, A.; Jenerowicz, D.; Osmola-Mańkowska, A.; Olek-Hrab, K. High-frequency ultrasonography in monitoring the effects of treatment of selected dermatoses. Post. Dermatol. Alergol. 2011, 28, 255–260. [Google Scholar]
  30. Salgueiro, C.; Baeza, J.; Moll, C.; Alfaro-Sepúlveda, D.; Mellado, G.; Villaseca-Hernández, M.; Vera-Kellet, C.; Wortsman, X. Sun Sign in the Ultrasound Diagnosis and Activity Assessment of Morphea. J. Ultrasound Med. 2022, 41, 1817–1824. [Google Scholar] [CrossRef]
  31. Pain, C.E.; Murray, A.; Dinsdale, G.; Marsden, A.; Manning, J.; Riley, P.; Leone, V.; Amin, T.; Zulian, F.; Herrick, A.L. Non-invasive imaging and clinical skin scores in juvenile localized scleroderma. Rheumatology 2024, 63, 1332–1340. [Google Scholar] [CrossRef] [PubMed]
  32. Narbutt, J.; Hołdrowicz, A.; Lesiak, A. Morphea—Selected local treatment methods and their effectiveness. Reumatologia 2017, 55, 305–313. [Google Scholar] [CrossRef] [PubMed]
  33. Dańczak-Pazdrowska, A.; Kowalczyk, M.J.; Szramka-Pawlak, B.; Gornowicz-Porowska, J.; Szewczyk, A.; Silny, W.; Olewicz-Gawlik, A.; Molińska-Glura, M.; Żaba, R.; Hrycaj, P. Clinical immunology Interleukin 1β in morphea. Cent. Eur. J. Immunol. 2012, 3, 247–252. [Google Scholar] [CrossRef]
  34. Dańczak-Pazdrowska, A.; Kowalczyk, M.; Szramka-Pawlak, B.; Gornowicz-Porowska, J.; Szewczyk, A.; Silny, W.; Olewicz-Gawlik, A.; Molińska-Glura, M.; Żaba, R.; Hrycaj, P. Interleukin-17A and interleukin-23 in morphea. Arch. Med. Sci. 2012, 8, 1089–1095. [Google Scholar] [CrossRef]
  35. Gu, Y.S.; Kong, J.; Cheema, G.S.; Keen, C.L.; Wick, G.; Gershwin, M.E. The immunobiology of systemic sclerosis. Semin. Arthritis Rheum. 2008, 38, 132–160. [Google Scholar] [CrossRef] [PubMed]
  36. Schoenherr, C.; Weiskirchen, R.; Haan, S. Interleukin-27 acts on hepatic stellate cells and induces signal transducer and activator of transcription 1-dependent responses. Cell Commun. Signal. 2010, 8, 19. [Google Scholar] [CrossRef]
  37. Shabgah, A.G.; Fattahi, E.; Shahneh, F.Z. Interleukin-17 in human inflammatory diseases. Postep. Dermatol. Alergol. 2014, 31, 256–261. [Google Scholar] [CrossRef] [PubMed]
  38. Shimozato, O.; Sato, A.; Kawamura, K.; Chiyo, M.; Ma, G.; Li, Q.; Tagawa, M. The secreted form of p28 subunit of interleukin (IL)-27 inhibits biological functions of IL-27 and suppresses anti-allogeneic immune responses. Immunology 2009, 128, e816–e825. [Google Scholar] [CrossRef]
  39. Kahaleh, M.B.; Sherer, G.K.; LeRoy, E.C. Endothelial injury in scleroderma. J. Exp. Med. 1979, 149, 1326–1335. [Google Scholar] [CrossRef]
  40. Gupta, R.A.; Fiorentino, D. Localized scleroderma and systemic sclerosis: Is there a connection? Best Pract. Res. Clin. Rheumatol. 2007, 21, 1025–1036. [Google Scholar] [CrossRef]
  41. Mertens, J.S.; Seyger, M.M.B.; Thurlings, R.M.; Radstake, T.R.D.J.; De Jong, E.M.G.J. Morphea and Eosinophilic Fasciitis: An Update. Am. J. Clin. Dermatol. 2017, 18, 491–512. [Google Scholar] [CrossRef] [PubMed]
  42. Inoshita, T.; Whiteside, T.L.; Rodnan, G.P.; Taylor, F.H. Abnormalities of T lymphocyte subsets in patients with progressive systemic sclerosis (PSS, scleroderma). J. Lab. Clin. Med. 1981, 97, 264–277. [Google Scholar] [PubMed]
  43. Keystone, E.; Lau, C.; Gladman, D.; Wilkinson, S.; Lee, P.; Shore, A. Immunoregulatory T cell subpopulations in patients with scleroderma using monoclonal antibodies. Clin. Exp. Immunol. 1982, 48, 443–488. [Google Scholar] [PubMed]
  44. Mirizio, E.; Marathi, A.; Hershey, N.; Ross, C.; Schollaert, K.; Salgado, C.; Reyes-Mugica, M.; Torok, K.S. Identifying the Signature Immune Phenotypes Present in Pediatric Localized Scleroderma. J. Investig. Dermatol. 2019, 139, 715–718. [Google Scholar] [CrossRef]
  45. Carol, M.A. Immunology of systemic sclerosis. Front. Biosci. 2005, 10, 1707–1719. [Google Scholar] [CrossRef]
  46. Torok, K.S.; Kurzinski, K.; Kelsey, C.; Yabes, J.; Magee, K.; Vallejo, A.N.; Medsger, T.; Feghali-Bostwick, C.A. Peripheral blood cytokine and chemokine profiles in juvenile localized scleroderma: T-helper cell-associated cytokine profiles. Semin. Arthritis Rheum. 2015, 45, 284–293. [Google Scholar] [CrossRef]
  47. O’brien, J.C.; Rainwater, Y.B.; Malviya, N.; Cyrus, N.; Auer-Hackenberg, L.; Hynan, L.S.; Hosler, G.A.; Jacobe, H.T. Transcriptional and Cytokine Profiles Identify CXCL9 as a Biomarker of Disease Activity in Morphea. J. Investig. Dermatol. 2017, 137, 1663–1670. [Google Scholar] [CrossRef]
  48. Rongioletti, F.; Ferreli, C.; Atzori, L.; Bottoni, U.; Soda, G. Scleroderma with an update about clinico-pathological correlation. Ital. J. Dermatol. Venereol. 2018, 153, 208–215. [Google Scholar] [CrossRef]
  49. Fertin, C.; Nicolas, J.F.; Gillery, P.; Kalis, B.; Banchereau, J.; Maquart, F.X. Interleukin-4 stimulates collagen synthesis by normal and scleroderma fibroblasts in dermal equivalents. Cell Mol. Biol. 1991, 37, 823–829. [Google Scholar]
  50. Lee, J.S.; Park, H.; Yoon, H.; Chung, J.H.; Cho, S. CD34 stromal expression is inversely proportional to smooth muscle actin expression and extent of morphea. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 2208–2216. [Google Scholar] [CrossRef]
  51. Osmola-Mańkowska, A.; Teresiak-Mikołajczak, E.; Dańczak-Pazdrowska, A.; Kowalczyk, M.; Żaba, R.; Adamski, Z. The role of dendritic cells and regulatory T cells in the pathogenesis of morphea. Cent. Eur. J. Immunol. 2015, 40, 103–108. [Google Scholar] [CrossRef] [PubMed]
  52. Asano, Y.; Ihn, H.; Jinnin, M.; Mimura, Y.; Tamaki, K. Involvement of alphavbeta5 integrin in the establishment of autocrine tgf-β signaling in dermal fibroblasts derived from localized scleroderma. J. Investig. Dermatol. 2006, 126, 1761–1769. [Google Scholar] [CrossRef] [PubMed]
  53. Tomimura, S.; Ogawa, F.; Iwata, Y.; Komura, K.; Hara, T.; Muroi, E.; Takenaka, M.; Shimizu, K.; Hasegawa, M.; Fujimoto, M.; et al. Autoantibodies against matrix metalloproteinase-1 in patients with localized scleroderma. J. Dermatol. Sci. 2008, 52, 47–54. [Google Scholar] [CrossRef] [PubMed]
  54. Fawzi, M.; Tawfik, S.; Eissa, A.; El-Komy, M.; Abdel-Halim, M.; Shaker, O. Expression of insulin-like growth factor-I in lesional and non-lesional skin of patients with morphoea. Br. J. Dermatol. 2008, 159, 86–90. [Google Scholar] [CrossRef] [PubMed]
  55. Yurtsever, I.; Łukomska, M.; Sobolewski, P.; Szymańska, E.; Owczarek, W.; Walecka, I. Familial localized scleroderma with paediatric onset: A review. Postep. Dermatol. Allergol. 2021, 38, 193–197. [Google Scholar] [CrossRef]
  56. Medhasi, S.; Chantratita, N. Human Leukocyte Antigen (HLA) System: Genetics and Association with Bacterial and Viral Infections. J. Immunol. Res. 2022, 2022, 9710376. [Google Scholar] [CrossRef]
  57. Jacobe, H.; Ahn, C.; Arnett, F.C.; Reveille, J.D. Major histocompatibility complex class I and class II alleles may confer susceptibility to or protection against morphea: Findings from the Morphea in Adults and Children cohort. Arthritis Rheumatol. 2014, 66, 3170–3177. [Google Scholar] [CrossRef]
  58. Schutt, C.; Mirizio, E.; Salgado, C.; Reyes-Mugica, M.; Wang, X.; Chen, W.; Grunwaldt, L.; Schollaert, K.L.; Torok, K.S. Transcriptomic Evaluation of Juvenile Localized Scleroderma Skin with Histologic and Clinical Correlation. Arthritis Rheumatol. 2021, 73, 1921–1930. [Google Scholar] [CrossRef]
  59. Saracino, A.; Denton, C.; Orteu, C. The molecular pathogenesis of morphoea: From genetics to future treatment targets. Br. J. Dermatol. 2017, 177, 34–46. [Google Scholar] [CrossRef]
  60. Thorpe, J.; Osei-Owusu, I.A.; Avigdor, B.E.; Tupler, R.; Pevsner, J. Mosaicism in Human Health and Disease. Annu. Rev. Genet. 2020, 54, 487–510. [Google Scholar] [CrossRef]
  61. Molho-Pessach, V.; Schaffer, J.V. Blaschko lines and other patterns of cutaneous mosaicism. Clin. Dermatol. 2011, 29, 205–225. [Google Scholar] [CrossRef] [PubMed]
  62. Kouzak, S.S.; Mendes, M.S.T.; Costa, I.M.C. Cutaneous mosaicisms: Concepts, patterns and classifications. An. Bras. Dermatol. 2013, 88, 507–517. [Google Scholar] [CrossRef] [PubMed]
  63. Aluri, J.; Cooper, M.A. Genetic Mosaicism as a Cause of Inborn Errors of Immunity. J. Clin. Immunol. 2021, 41, 718–728. [Google Scholar] [CrossRef]
  64. Lee, J.S.; Park, H.-S.; Cho, S.; Yoon, H.-S. Concurrence of Circumscribed Morphea and Segmental Vitiligo: A Case Report. Ann. Dermatol. 2018, 30, 708–711. [Google Scholar] [CrossRef]
  65. Pedowska, M.; Ptasiewicz, M.; Szumiło, J.; Chałas, R. Morphological and clinical picture of the morphea in the oral cavity. Folia Morphol. 2022, 81, 544–550. [Google Scholar] [CrossRef]
  66. Weibel, L.; Harper, J. Linear morphoea follows Blaschko’s lines. Br. J. Dermatol. 2008, 159, 175–181. [Google Scholar] [CrossRef]
  67. Chen, H.W.; Zhu, J.L.; Martyanov, V.; Tsoi, L.C.; Johnson, M.E.; Barber, G.; Popovich, D.; O’brien, J.C.; Coias, J.; Cyrus, N.; et al. Gene Expression Signatures in Inflammatory and Sclerotic Morphea Skin and Sera Distinguish Morphea from Systemic Sclerosis. J. Investig. Dermatol. 2023, 143, 1886–1895.e10. [Google Scholar] [CrossRef] [PubMed]
  68. Mirizio, E.; Liu, C.; Yan, Q.; Waltermire, J.; Mandel, R.; Schollaert, K.L.; Konnikova, L.; Wang, X.; Chen, W.; Torok, K.S. Genetic Signatures from RNA Sequencing of Pediatric Localized Scleroderma Skin. Front. Pediatr. 2021, 9, 669116. [Google Scholar] [CrossRef]
  69. Wu, H.; Chen, Y.; Zhu, H.; Zhao, M.; Lu, Q. The Pathogenic Role of Dysregulated Epigenetic Modifications in Autoimmune Diseases. Front. Immunol. 2019, 10, 2305. [Google Scholar] [CrossRef]
  70. Coit, P.; Schollaert, K.L.; Mirizio, E.M.; Torok, K.S.; Sawalha, A.H. DNA methylation patterns in juvenile systemic sclerosis and localized scleroderma. Clin. Immunol. 2021, 228, 108756. [Google Scholar] [CrossRef]
  71. Wolska-Gawron, K.; Bartosińska, J.; Krasowska, D. MicroRNA in localized scleroderma: A review of literature. Arch. Dermatol. Res. 2020, 312, 317–324. [Google Scholar] [CrossRef] [PubMed]
  72. Babalola, O.; Mamalis, A.; Lev-Tov, H.; Jagdeo, J. The role of microRNAs in skin fibrosis. Arch. Dermatol. Res. 2013, 305, 763–776. [Google Scholar] [CrossRef]
  73. Etoh, M.; Jinnin, M.; Makino, K.; Yamane, K.; Nakayama, W.; Aoi, J.; Honda, N.; Kajihara, I.; Makino, T.; Fukushima, S.; et al. microRNA-7 down-regulation mediates excessive collagen expression in localized scleroderma. Arch. Dermatol. Res. 2013, 305, 9–15. [Google Scholar] [CrossRef] [PubMed]
  74. Makino, K.; Jinnin, M.; Hirano, A.; Yamane, K.; Eto, M.; Kusano, T.; Honda, N.; Kajihara, I.; Makino, T.; Sakai, K.; et al. The downregulation of microRNA let-7a contributes to the excessive expression of type I collagen in systemic and localized scleroderma. J. Immunol. 2013, 190, 3905–3915. [Google Scholar] [CrossRef]
  75. Makino, T.; Jinnin, M.; Etoh, M.; Yamane, K.; Kajihara, I.; Makino, K.; Ichihara, A.; Igata, T.; Sakai, K.; Fukushima, S.; et al. Down-regulation of microRNA-196a in the sera and involved skin of localized scleroderma patients. Eur. J. Dermatol. 2014, 24, 470–476. [Google Scholar] [CrossRef] [PubMed]
  76. Yan, Q.; Chen, J.; Li, W.; Bao, C.; Fu, Q. Correction: Corrigendum: Targeting miR-155 to Treat Experimental Scleroderma. Sci. Rep. 2016, 6, 21941. [Google Scholar] [CrossRef] [PubMed]
  77. Chouri, E.; Servaas, N.H.; Bekker, C.P.J.; Affandi, A.J.; Cossu, M.; Hillen, M.R.; Angiolilli, C.; Mertens, J.S.; van den Hoogen, L.L.; Silva-Cardoso, S.; et al. Serum microRNA screening and functional studies reveal miR-483-5p as a potential driver of fibrosis in systemic sclerosis. J. Autoimmun. 2018, 89, 162–170. [Google Scholar] [CrossRef]
  78. Pozniak, T.; Shcharbin, D.; Bryszewska, M. Circulating microRNAs in Medicine. Int. J. Mol. Sci. 2022, 23, 3996. [Google Scholar] [CrossRef]
  79. Wolska-Gawron, K.; Bartosińska, J.; Rusek, M.; Kowal, M.; Raczkiewicz, D.; Krasowska, D. Circulating miRNA-181b-5p, miRNA-223-3p, miRNA-210-3p, let 7i-5p, miRNA-21-5p and miRNA-29a-3p in patients with localized scleroderma as potential biomarkers. Sci. Rep. 2020, 10, 20218. [Google Scholar] [CrossRef]
  80. Gong, Y.; Liu, H.; Li, G.; Sun, L. Identification of lncRNA expression profiles in pediatric localized scleroderma. J. Cosmet. Dermatol. 2022, 21, 6422–6427. [Google Scholar] [CrossRef]
  81. Longo, F.; Saletta, S.; Lepore, L.; Pennesi, M. Localized scleroderma after infection with Epstein-Barr virus. Clin. Exp. Rheumatol. 1993, 11, 681–683. [Google Scholar] [PubMed]
  82. Mihas, A.A.; Abou-Assi, S.G.; Heuman, D.M. Cutae morphea associated with chronic hepatitis C. J. Hepatol. 2003, 39, 458–459. [Google Scholar] [CrossRef] [PubMed]
  83. Soylu, S.; Gül, Ü.; Gönül, M.; Klç, A.; Çakmak, S.K.; Demiriz, M. An uncommon presentation of the co-existence of morphea and vitiligo in a patient with chronic hepatitis B virus infection: Is there a possible association with autoimmunity? Am. J. Clin. Dermatol. 2009, 10, 336–338. [Google Scholar] [CrossRef] [PubMed]
  84. Spirer, Z.; Ilie, B.; Pick, I.A.; Yaron, M. Localized scleroderma following varicella in a three-year-old girl with IgA deficiency. Acta Paediatr. 1979, 68, 783–785. [Google Scholar] [CrossRef] [PubMed]
  85. Aberer, E.; Stanek, G.; Ertl, M.; Neumann, R. Evidence for spirochetal origin of circumscribed scleroderma (morphea). Acta Derm.-Venereol. 1987, 67, 225–231. [Google Scholar] [CrossRef]
  86. Canales, I.P.; Mas, I.B.; Bauza, F.M.; Armengod, F.S.; Vázquez, F.P.; Cardell, J.A. Determination of antibodies against Borrelia burgdorferi in patients with morphea, lichen sclerosus et atrophicus and erythema chronicum migrans. Rev. Clin. Esp. 1990, 186, 320–323. [Google Scholar]
  87. Raguin, G.; Boisnic, S.; Souteyrand, P.; Baranton, G.; Piette, J.; Godeau, P.; France, C. No evidence for a spirochaetal origin of localized scleroderma. Br. J. Dermatol. 1992, 127, 218–220. [Google Scholar] [CrossRef]
  88. Ranki, A.; Aavik, E.; Peterson, P.; Schauman, K.; Nurmilaakso, P. Successful amplification of DNA specific for Finnish Borrelia burgdorferi isolates in erythema chronicum migrans but not in circumscribed scleroderma lesions. J. Investig. Dermatol. 1994, 102, 339–345. [Google Scholar] [CrossRef]
  89. Alonso-Llamazares, J.; Persing, D.; Anda, P.; Gibson, L.; Rutledge, B.; Iglesias, L. No evidence for Borrelia burgdorferi infection in lesions of morphea and lichen sclerosus et atrophicus in Spain. A prospective study and literature review. Acta Derm.-Venereol. 1997, 77, 299–304. [Google Scholar] [CrossRef]
  90. Weide, B.; Walz, T.; Garbe, C. Is morphoea caused by Borrelia burgdorferi? A review. Br. J. Dermatol. 2000, 142, 636–644. [Google Scholar] [CrossRef]
  91. Dańczak-Pazdrowska, A.; Polańska, A.; Synakiewicz, J.; Gurgul, E.; Molińska-Glura, M.; Ruchała, M.; Żaba, R.; Adamski, Z. Morphea and antithyroid antibodies. Postep. Dermatol. Allergol. 2018, 35, 470–473. [Google Scholar] [CrossRef] [PubMed]
  92. Zulian, F.; Vallongo, C.; Woo, P.; Russo, R.; Ruperto, N.; Harper, J.; Espada, G.; Corona, F.; Mukamel, M.; Vesely, R.; et al. Localized scleroderma in childhood is not just a skin disease. Arthritis Rheum. 2005, 52, 2873–2881. [Google Scholar] [CrossRef] [PubMed]
  93. Lutz, V.; Franc, C.; Bessis, D.; Cosnes, A.; Kluger, N.; Godet, J.; Sauleau, E.; Lipsker, D. High frequency of genital lichen sclerosus in a prospective series of 76 patients with morphea: Toward a better understanding of the spectrum of morphea. Arch. Dermatol. 2012, 148, 24–28. [Google Scholar] [CrossRef] [PubMed]
  94. Vigneron, C.; Bauer, N.; Waisse, W.; Keller, A.; Pop, M.; Clavier, J.-B.; Salze, P.; Noël, G. Morphée du sein radio-induite: Une complication méconnue. Cancer Radiother. 2014, 18, 80. [Google Scholar] [CrossRef]
  95. Spalek, M.; Jonska-Gmyrek, J.; Gałecki, J. Radiation-induced morphea—A literature review. J. Eur. Acad. Dermatol. Venereol. 2015, 29, 197–202. [Google Scholar] [CrossRef]
  96. Alexandrescu, D.T.; Bhagwati, N.S.; Wiernik, P.H. Chemotherapy-induced scleroderma: A pleiomorphic syndrome. Clin. Exp. Dermatol. 2005, 30, 141–145. [Google Scholar] [CrossRef]
  97. Lacombe, M.; Bedane, C.; Delaumenie, S.; Bernard, P. Nivolumab-induced multiple morphea associated with eosinophilic fasciitis. Eur. J. Dermatol. 2021, 31, 844–846. [Google Scholar] [CrossRef]
  98. Trattner, A.; David, M. Morphea and the isomorphic response of koebner. Int. J. Dermatol. 1991, 30, 747. [Google Scholar] [CrossRef]
  99. Toyama, S.; Sato, S.; Asano, Y. Localized scleroderma manifesting with skin lesions associated with mechanical stress. Eur. J. Dermatol. 2019, 29, 439–440. [Google Scholar] [CrossRef]
  100. Mehrtens, S.H.; Fleming, A.; Shall, L. A rare case of a tattoo-induced morphoea reaction. Clin. Exp. Dermatol. 2019, 44, 467–469. [Google Scholar] [CrossRef]
  101. Kreuter, A.; Hyun, J.; Stücker, M.; Sommer, A.; Altmeyer, P.; Gambichler, T. A randomized controlled study of low-dose UVA1, medium-dose UVA1, and narrowband UVB phototherapy in the treatment of localized scleroderma. J. Am. Acad. Dermatol. 2006, 54, 440–447. [Google Scholar] [CrossRef] [PubMed]
  102. Zulian, F.; Culpo, R.; Sperotto, F.; Anton, J.; Avcin, T.; Baildam, E.M.; Boros, C.; Chaitow, J.; Constantin, T.; Kasapcopur, O.; et al. Consensus-based recommendations for the management of juvenile localised scleroderma. Ann. Rheum. Dis. 2019, 78, 1019–1024. [Google Scholar] [CrossRef] [PubMed]
  103. Li, S.C.; Torok, K.S.; Pope, E.; Dedeoglu, F.; Hong, S.; Jacobe, H.T.; Rabinovich, C.E.; Laxer, R.M.; Higgins, G.C.; Ferguson, P.J.; et al. Development of consensus treatment plans for juvenile localized scleroderma: A roadmap toward comparative effectiveness studies in juvenile localized scleroderma. Arthritis Care Res. 2012, 64, 1175–1185. [Google Scholar] [CrossRef]
  104. Constantin, T.; Foeldvari, I.; Pain, C.E.; Pálinkás, A.; Höger, P.; Moll, M.; Nemkova, D.; Weibel, L.; Laczkovszki, M.; Clements, P.; et al. Development of minimum standards of care for juvenile localized scleroderma. Eur. J. Pediatr. 2018, 177, 961–977. [Google Scholar] [CrossRef] [PubMed]
  105. Careta, M.F.; Romiti, R. Localized scleroderma: Clinical spectrum and therapeutic update. An. Bras. Dermatol. 2015, 90, 62–73. [Google Scholar] [CrossRef]
  106. Chen, B.; Wang, X.; Long, X.; Zhang, M.; Huang, J.; Yu, N.; Xu, J. Supportive Use of Adipose-Derived Stem Cells in Cell-Assisted Lipotransfer for Localized Scleroderma. Plast. Reconstr. Surg. 2018, 141, 1395–1407. [Google Scholar] [CrossRef]
  107. Strong, A.L.; Adidharma, W.; Brown, O.H.; Cederna, P.S. Fat Grafting Subjectively Improves Facial Skin Elasticity and Hand Function of Scleroderma Patients. Plast. Reconstr. Surg.-Glob. Open 2021, 9, e3373. [Google Scholar] [CrossRef]
  108. Cardoneanu, A.; Burlui, A.M.; Macovei, L.A.; Bratoiu, I.; Richter, P.; Rezus, E. Targeting Systemic Sclerosis from Pathogenic Mechanisms to Clinical Manifestations: Why IL-6? Biomedicines 2022, 10, 318. [Google Scholar] [CrossRef]
  109. Kreuter, A.; Hyun, J.; Skrygan, M.; Sommer, A.; Bastian, A.; Altmeyer, P.; Gambichler, T. Ultraviolet A1-induced downregulation of human beta-defensins and interleukin-6 and interleukin-8 correlates with clinical improvement in localized scleroderma. Br. J. Dermatol. 2006, 155, 600–607. [Google Scholar] [CrossRef]
  110. Preuss, C.V.; Anjum, F. Tocilizumab. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  111. Khanna, D.; Denton, C.P.; Jahreis, A.; van Laar, J.M.; Frech, T.M.; Anderson, M.E.; Baron, M.; Chung, L.; Fierlbeck, G.; Lakshminarayanan, S.; et al. Safety and efficacy of subcutaneous tocilizumab in adults with systemic sclerosis (faSScinate): A phase 2, randomised, controlled trial. Lancet 2016, 387, 2630–2640. [Google Scholar] [CrossRef]
  112. Lythgoe, H.; Baildam, E.; Beresford, M.W.; Cleary, G.; McCann, L.J.; Pain, C.E. Tocilizumab as a potential therapeutic option for children with severe, refractory juvenile localized scleroderma. Rheumatology 2018, 57, 398–401. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, A.; Nocton, J.; Chiu, Y. A Case of Pansclerotic Morphea Treated with Tocilizumab. JAMA Dermatol. 2019, 155, 388–389. [Google Scholar] [CrossRef] [PubMed]
  114. Martini, G.; Campus, S.; Raffeiner, B.; Boscarol, G.; Meneghel, A.; Zulian, F. Tocilizumab in two children with pansclerotic morphoea: A hopeful therapy for refractory cases? Clin. Exp. Rheumatol. 2017, 35 (Suppl. 106), 211–213. [Google Scholar] [PubMed]
  115. Lonowski, S.; Goldman, N.; Kassamali, B.; Shahriari, N.; LaChance, A.; Vleugels, R.A. Tocilizumab for refractory morphea in adults: A case series. JAAD Case Rep. 2022, 30, 27–29. [Google Scholar] [CrossRef]
  116. Aung, W.W.; Wang, C.; Xibei, J.; Horii, M.; Mizumaki, K.; Kano, M.; Okamura, A.; Kobayashi, T.; Matsushita, T. Immunomodulating role of the JAKs inhibitor tofacitinib in a mouse model of bleomycin-induced scleroderma. J. Dermatol. Sci. 2021, 101, 174–184. [Google Scholar] [CrossRef]
  117. Chakraborty, D.; Šumová, B.; Mallano, T.; Chen, C.-W.; Distler, A.; Bergmann, C.; Ludolph, I.; Horch, R.E.; Gelse, K.; Ramming, A.; et al. Activation of STAT3 integrates common profibrotic pathways to promote fibroblast activation and tissue fibrosis. Nat. Commun. 2017, 8, 1130. [Google Scholar] [CrossRef]
  118. Tang, J.; Zheng, W.; Yang, B. P08 Successful treatment of paediatric morphoea with tofacitinib. Br. J. Dermatol. 2023, 188 (Suppl. S4), ljad113.036. [Google Scholar] [CrossRef]
  119. Damsky, W.; Patel, D.; Garelli, C.J.; Garg, M.; Wang, A.; Dresser, K.; Deng, A.; Harris, J.E.; Richmond, J.; King, B. Jak Inhibition Prevents Bleomycin-Induced Fibrosis in Mice and Is Effective in Patients with Morphea. J. Investig. Dermatol. 2020, 140, 1446–1449.e4. [Google Scholar] [CrossRef]
  120. Available online: https://ec.europa.eu/health/documents/community-register/2017/20170725138407/anx_138407_pl.pdf (accessed on 14 September 2024).
  121. Stausbøl-Grøn, B.; Olesen, A.; Deleuran, B.; Deleuran, M. Abatacept is a Promising treatment for patients with disseminated morphea profunda: Presentation of two cases. Acta Derm.-Venereol. 2011, 91, 686–688. [Google Scholar] [CrossRef]
  122. Fage, S.; Arvesen, K.; Olesen, A. Abatacept Improves Skin-score and Reduces Lesions in Patients with Localized Scleroderma: A Case Series. Acta Derm.-Venereol. 2018, 98, 465–466. [Google Scholar] [CrossRef]
  123. Li, S.C.; Torok, K.S.; Ishaq, S.S.; Buckley, M.; Edelheit, B.; Ede, K.C.; Liu, C.; Rabinovich, C.E. Preliminary evidence on abatacept safety and efficacy in refractory juvenile localized scleroderma. Rheumatology 2021, 60, 3817–3825. [Google Scholar] [CrossRef] [PubMed]
  124. Distler, J.H.W.; Jüngel, A.; Huber, L.C.; Schulze-Horsel, U.; Zwerina, J.; Gay, R.E.; Michel, B.A.; Hauser, T.; Schett, G.; Gay, S.; et al. Imatinib mesylate reduces production of extracellular matrix and prevents development of experimental dermal fibrosis. Arthritis Rheum. 2007, 56, 311–322. [Google Scholar] [CrossRef] [PubMed]
  125. Akhmetshina, A.; Venalis, P.; Dees, C.; Busch, N.; Zwerina, J.; Schett, G.; Distler, O.; Distler, J.H.W. Treatment with imatinib prevents fibrosis in different preclinical models of systemic sclerosis and induces regression of established fibrosis. Arthritis Rheum. 2009, 60, 219–224. [Google Scholar] [CrossRef]
  126. Coelho-Macias, V.; Mendes-Bastos, P.; Assis-Pacheco, F.; Cardoso, J. Imatinib: A novel treatment approach for generalized morphea. Int. J. Dermatol. 2014, 53, 1299–1302. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A,B)—Clinical picture of morphea. (A)—active lesion, characteristic lilac ring, courtesy of Prof. Polańska.
Figure 1. (A,B)—Clinical picture of morphea. (A)—active lesion, characteristic lilac ring, courtesy of Prof. Polańska.
Jcm 13 07134 g001
Figure 2. Pathogenesis of morphea. Factors such as infection and skin trauma (red arrow) can trigger inflammation in predisposed patients. T lymphocytes and eosinophils are involved in this process, appearing in the dermis and also in the vascular area. During the inflammatory phase, the endothelium is damaged, adhesion molecules appear and pro-inflammatory Th1 and Th17 cells are recruited, which activate fibroblasts via cytokines. Th2 cells then recruit T cells that produce profibrotic cytokines such as IL-6, IL-4. This leads to an increase in collagen fibres and subsequent sclerosis. The vessel walls thicken and the vessel lumen narrows. The final phase is atrophy, when the thickness of the epidermis is reduced and melanophages appear. There is a reduction in or atrophy of blood vessels and skin appendages. (The Figure 2 was made using Canva).
Figure 2. Pathogenesis of morphea. Factors such as infection and skin trauma (red arrow) can trigger inflammation in predisposed patients. T lymphocytes and eosinophils are involved in this process, appearing in the dermis and also in the vascular area. During the inflammatory phase, the endothelium is damaged, adhesion molecules appear and pro-inflammatory Th1 and Th17 cells are recruited, which activate fibroblasts via cytokines. Th2 cells then recruit T cells that produce profibrotic cytokines such as IL-6, IL-4. This leads to an increase in collagen fibres and subsequent sclerosis. The vessel walls thicken and the vessel lumen narrows. The final phase is atrophy, when the thickness of the epidermis is reduced and melanophages appear. There is a reduction in or atrophy of blood vessels and skin appendages. (The Figure 2 was made using Canva).
Jcm 13 07134 g002
Table 1. Differential diagnosis of morphea [13,28,29].
Table 1. Differential diagnosis of morphea [13,28,29].
Localized (inflammatory lesions)
  • Lichen sclerosus
  • Granuloma anulare
  • Granulosarcoma
  • Fixed erythema
  • Urticaria pigmentosa
  • Cutaneous radiation syndrome
  • Chronic erythema migrans
  • Mycosis fungoides
Localized (hyperpigmentation lesions)
  • Cafe au lait spots
  • Lichen planus induced by solar radiation
  • Erythema dyschromicum perstans
  • Post-inflammatory hyperpigmentation
Localized (atrophic lesions)
  • Scarring
  • Cellulitis
  • Atrophic dermatitis of the extremities
  • Lichen sclerosus
Localized (sclerosis)
  • Circumscribed myxoedema
  • Morpheaform basal cel carcinoma
  • Necrobiosis lipoidica
Linear
  • Glucocorticosteroids-induced lipoatrophy
  • Site-specific lipoatrophy
  • Progressive lipodystrophy
Deep
  • Lupus panniculitis
  • Inflammation of the subcutis
Generalized
  • Systemic sclerosis
  • GVHD
  • Porphyria cutanea tarda
  • Mixed connective tissue disease
  • Scleredema adultorum of Buschke
  • Scleromyxedema
  • Nephrogenic systemic fibrosis
Table 2. New therapeutic options for morphea.
Table 2. New therapeutic options for morphea.
AuthorsNumber of Patients and Type of MorpheaName of DrugPrior Systemic/Topical TherapiesDose of New Drug/or MedianResponse and Total Time of Therapy/or MedianAdverse Events
Lythgoe (2018) [112]5 P, age btw 6–13 y

Linear morphea
TocilizumabMTX—5, MMF—5, ETP—2, CSA—18 mg/kg for p ≥ 30 kg
12 mg/kg for p < 30 kg

0, 2 and 4 wks, and then at 4-wk intervals
RT—after 6 mth

TT—btw 12–25 mth
None
Lonowski et al. (2022) [115]3 F, age btw 41–69 y
Linear morphea—1, generalized morphea—2
TocilizumabPDN—2, MTX—2, MMF—1, MPDN—1, IVIG—1, RTX—1162 mg/wk sc—2
8 mg/kg iv—1
Median response—3 months

MT—44.6 mth
Hyperlipidemia—1
Zhang et al. (2019) [113] 1 F, 6 y
Pansclerotic morphea
TocilizumabMTX, PDN, MPDN, MMF300 mg every 4 wksmLoSSI—22 to 6
PGA—30 to 17

TT—18 mth
None
Martini et al. (2017) [114]1 F, 1 M
(children)
Pansclerotic morphea
TocilizumabMTX—2, PDN—2, MMF—2, MPDN—2, IMB—18 mg/kg every 4 wksF—after 18 mth,
LoSCAT from 58 to 47
TT—N/A
F—one episode of pneumonia
M—TT—6 mth,
after 30 mth
LoSCAT from 57 to 43
Li et al. (2018) [123]18 P, mean age 13,4 y
(12 F, 6 M)

Linear morphea—12, Deep morphea—1, generalized morphea—1, mixed—5
AbataceptMTX—17, MMF—16, GKS—18, HCQ—2, IMB—2, TA—2, LNLD—1, Biologics (ADA, INF, Tocilizumab)—1, PUVA—1Median—10 mg/kg
iv and sc
After 12 mth, 15 (83%) P

MT—23.2 mth
Mood and/or behavioral issue—2
Fage et al. (2018) [122]13 adults
(11 F, 2 M)

Morphea
AbataceptN/A500 mg < 60 kg or
750 mg > 60 kg
iv on days 1, 15, 30 and thereafter every 4–6 wks
RT btw 3 to 32 mth

Overall, an improvement/reduction in size of lesions or mRSS/mLoSSI score were observed

Total—N/A
Fatigue 1–3 days after infusion, nausea, diarrhea (in some cases with fever)

Withholding treatment—2
1-fatigue, tingling and aching sensation
2—colitis ulcerosa
Stausbøl-Grøn et al. (2011) [121]47 y, F
disseminated morphea
AbataceptPCL—2, PDN—2, CSA—2, TA—1, UVA1—1750 mg iv on days 1, 15 and 30, and thereafter every 4–6 wks (20 drug administration)mRSS—18 to 1
TT-19 mth
1-
Hypertension
38 y, F
Morphea profunda
500 mg iv on day 1, 15, 30, and thereafter every 4 wksmRSS—13 to 6
TT—2.5 mth
treatment had to be stopped after 2.5 mth, patient had a breast cancer
Tang et al. (2023) [118]6 y, F
Linear morphea
TofacitinibMTX—1, TA—1, GLC—1, HCQ—1, UVA1—2, TCS—22 mth on 2.5 mg twice daily, 4 mth on 2.5 mg/d and 2 mth on 2.5 mg/d every other daymLoSSI—6 to 1
LosDi—7 to 2
TT—8 mth
None
13 y, M
Linear morphea
2 mth on 5 mg twice daily, 3 mth on 5 mg/dmLoSSI—7 to 1
LosDi—7 to 2
TT—5 mth
Damsky et al. (2020) [119]54 y, M
Generalized morphea
BaricitinibECP, PDN2 mg dailyResolution of erythema after 2 mth
Improvement in mobility after 6 mth

TT—12 mth
None
Coehlo-Macias et al. (2014) [126]50 y, M Generalized morphea with ulceration on the extremitiesImatinibPTX3 mth on 200 mg/d and 9 mth on 300 mg/dSkin biopsy after 12—dermal thickness from 5 to 4 mm
Improvement of joint mobility; left knee maximum extension went from initial 110° to 130°

TT—12 mth
None
Abbreviations: P—Patient, y—Year old, MTX—Methotrexate, MMF—Mycophenolate-mofetil, ETP—Etanercept, wks—Weeks, wk—Week, RT—Response time, TT—Total time of therapy, mth—Month, Months, btw—Between, CSA—Cyclosporine, PDN—Prednisone, MPDN—Methylprednisolone, IVIG—Intravenous immunoglobulin, sc—Subcutaneous, iv—Intravenous, RTX—Rituximab, F—Female, M—Male, IMB—Imatinib, mLoSSI—The modified Localized Skin Severity Index, PGA—The Physician Global Assessment, LoSCAT—The Localized Scleroderma Assessment Tool, GKS—Glucocorticoids, HCQ—Hydroxychloroquine, TA—Topical agents, LNLD—Lenalidomide, ADA—Adalimumab, INF, Infliximab, PUVA—Psoralen plus ultraviolet A photochemotherapy, mRSS—The modified Rodnan skin score, PCL—Penicillamine, GLC—Glycyrrhizin, TCS—Tacrolimus ointment, LosDi—Localized scleroderma damage index, MT—Median time of therapy, ECP—Extracorporeal photopheresis, PTX—Pentoxifylline, N/A—not available.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Stein, T.; Cieplewicz-Guźla, P.; Iżykowska, K.; Pieniawska, M.; Żaba, R.; Dańczak-Pazdrowska, A.; Polańska, A. What Is New in Morphea—Narrative Review on Molecular Aspects and New Targeted Therapies. J. Clin. Med. 2024, 13, 7134. https://doi.org/10.3390/jcm13237134

AMA Style

Stein T, Cieplewicz-Guźla P, Iżykowska K, Pieniawska M, Żaba R, Dańczak-Pazdrowska A, Polańska A. What Is New in Morphea—Narrative Review on Molecular Aspects and New Targeted Therapies. Journal of Clinical Medicine. 2024; 13(23):7134. https://doi.org/10.3390/jcm13237134

Chicago/Turabian Style

Stein, Tomasz, Paulina Cieplewicz-Guźla, Katarzyna Iżykowska, Monika Pieniawska, Ryszard Żaba, Aleksandra Dańczak-Pazdrowska, and Adriana Polańska. 2024. "What Is New in Morphea—Narrative Review on Molecular Aspects and New Targeted Therapies" Journal of Clinical Medicine 13, no. 23: 7134. https://doi.org/10.3390/jcm13237134

APA Style

Stein, T., Cieplewicz-Guźla, P., Iżykowska, K., Pieniawska, M., Żaba, R., Dańczak-Pazdrowska, A., & Polańska, A. (2024). What Is New in Morphea—Narrative Review on Molecular Aspects and New Targeted Therapies. Journal of Clinical Medicine, 13(23), 7134. https://doi.org/10.3390/jcm13237134

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop