Next Article in Journal
Caffeine and Taurine from Energy Drinks—A Review
Next Article in Special Issue
Evaluation of Teneligliptin and Retagliptin on the Clearance of Melanosome by Melanophagy in B16F1 Cells
Previous Article in Journal
Effectiveness of a Novel Compound HAIR & SCALP COMPLEX on Hair Follicle Regeneration
Previous Article in Special Issue
Dermatological Changes during Menopause and HRT: What to Expect?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in the Pathogenesis and Treatment of Rosacea: A Phenotype-Based Therapeutic Approach

by
Giulia Galluccio
1,†,
Martina D’Onghia
1,†,
Dalma Malvaso
2,3,
Laura Lazzeri
1,
Elisa Cinotti
1,
Giovanni Rubegni
4,
Pietro Rubegni
1,* and
Laura Calabrese
1,2,*
1
Dermatology Unit, Department of Medical, Surgical and Neurological Sciences, University of Siena, 53100 Siena, Italy
2
Dermatologia, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
3
UOC di Dermatologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
4
Ophthalmology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cosmetics 2024, 11(1), 11; https://doi.org/10.3390/cosmetics11010011
Submission received: 17 December 2023 / Revised: 11 January 2024 / Accepted: 15 January 2024 / Published: 17 January 2024
(This article belongs to the Collection Feature Papers in Cosmetics in 2023)

Abstract

:
Rosacea is a common chronic inflammatory skin disorder that mainly affects the central face. It is primarily characterized by recurrent episodes of flushing, persistent erythema, inflammatory papules, telangiectasias, phymatous changes, and ocular symptoms. Its pathogenesis is complex and still not completely understood. It encompasses innate and adaptive immune system dysregulation, neurovascular dysfunction, and genetic and environmental factors. To date, four subtypes of rosacea have been identified, based on the predominant clinical features: erythemato-teleangiectatic, papulopustular, pyhomatous, and ocular rosacea. New insights into this condition have led to several pharmacological treatments, including topical medications, spanning from the conventional azelaic acid, metronidazole, benzoyl peroxide, clindamycin, and erythromycin to new ones including not only brimonidine, oxymetazoline, ivermectine, and minocycline but also systemic drugs such as oral antibiotics, isotretinoin, non-selective β-blockers or α2-adrenergic agonists, and laser- or light-based therapies, together with new therapeutic approaches. The aim of this study was to review the current literature on the pathophysiology of rosacea and to provide an overview of therapeutic approaches that specifically address each clinical subtype.

1. Introduction

Rosacea is a chronic inflammatory skin disorder with a relapsing course that primarily affects the centrofacial and periocular regions. It is characterized by recurrent episodes of flushing, persistent erythema, inflammatory papules, and telangiectasias [1]. Most patients with rosacea also experience ocular involvement, such as blepharoconjunctivitis with eyelid margin inflammation and meibomian gland dysfunction [2]. The disease is oftentimes associated with systemic comorbidities, such as hypertension, dyslipidemia, inflammatory bowel disease, and psychiatric comorbidities, namely anxiety and depression [3,4]. For these reasons, it is not surprising that rosacea has a substantial impact on patients’ quality of life [5] and consequently society and healthcare expenditure [6].
From an epidemiological point of view, rosacea represents a common dermatological disorder with a slight female predominance and a worldwide prevalence of approximately 5% of the general population [7].
Although the real nature of rosacea remains largely elusive, it is known that its pathogenesis encompasses a complex interplay between innate and adaptive immune system dysregulation, neurovascular dysfunction, and genetic and environmental factors [8]. To date, most evidence points towards several triggering factors that may initiate or worsen the disease, including ultraviolet (UV) exposure, local inflammation responses to skin microorganisms (Demodex mites), temperature changes, and stressors [9].
In 2002, the National Rosacea Society Expert Committee (NRSEC) proposed classification of the disease into four categories: erythemato-teleangiectatic, papulopustular, pyhomatous, and ocular rosacea [10]. This classification underwent a revision in 2016 because of new insights into the pathogenesis of rosacea, obtained by establishing a new approach for diagnosis and classification of rosacea based on disease phenotype [11]. Recent findings have suggested that erythemato-telangiectatic and papulopustular rosacea are the most common patterns within the four subtypes, with no differences between the sexes except for the phymatous one, which seems to be more prevalent in men [12].
Despite the burden of rosacea in dermatological clinics, treatment options, which include topical and oral therapies, light devices, skin care, and lifestyle management, often yield poor results [13]. To date, all therapeutic strategies mainly aim to control the clinical symptoms rather than target the causes or prevent the disease.
For all of these reasons, managing rosacea still represents a real challenge for dermatologists, since the choice of the most appropriate treatment strategy not only relies on the clinical characteristics of rosacea lesions but also the patient’s reported symptoms and preferences and the physician’s experience.
The aim of our paper was to review the current literature on the pathophysiology of rosacea and to provide an overview of therapeutic strategies and approaches that specifically address each clinical subtype of rosacea.

2. Pathophysiology

Rosacea is characterized by a multifactorial pathophysiology, resulting from the interaction between environmental influences, genetic factors, immunological, vascular, and neuroinflammatory dysregulation, and the impairment of skin barrier function. A schematic illustration of the main factors playing a role in the pathogenesis of rosacea is shown in Figure 1.

2.1. Environmental Factors

Several environmental factors have been recognized in the initiation or aggravation of rosacea. The association between Demodex mites and the disease is well-known, although the underlying pathogenic mechanisms are still unclear [14]. Demodex follicolorum, a commensal of human skin that mainly colonizes hair follicles, is five to seven times increased in skin biopsies of patients with rosacea [15], particularly in the papulopustular and phymatous subtypes [16,17]. In addition, resident bacteria such as Bacillus oleronius and Staphylococcus epidermidis may worsen the inflammatory reaction by inducing chemotaxis of innate immune cells, such as neutrophils [18]. Dietary habits may play a role in inducing rosacea. In detail, heat, alcohol, capsaicin, and histamine- and cinnamaldehyde-related foods may activate transient receptor potential (TRP) cation channels, resulting in neurogenic vasodilatation, inflammation, and oxidative stress dysfunction [19]. Finally, emotional stress can affect, reveal, or amplify rosacea features by modulating the hypothalamic–pituitary–adrenal axis (HPA) and releasing neuropeptides, neurotrophins, and lymphokines from nerve endings and dermal cells [20].

2.2. Genetic Predisposition

The genetic basis of rosacea is still poorly understood, although family inheritance, twin concordance, and associations with other autoimmune disorders may attest to a genetic predisposition to this skin condition [21]. A null mutation polymorphism in glutathione S-transferase (GST), which plays a key role in cellular defense against electrophilic chemical species and reactive oxygen species (ROS), has been reported in patients with rosacea [22]. Other significant gene polymorphisms include human leukocyte antigen (HLA) class II [23], tachykinin 3 receptor (TACR3) [24], vitamin D receptor (VDR) [25], and vascular endothelial growth factor (VEGF) [26]. Interestingly, Deng et al. [27] presented the integrated results of whole-genome sequencing (WGS) and whole-exome sequencing (WES) in Chinese rosacea families. In their study, the authors identified single rare deleterious variants of LRRC4, SH3PXD2A, and SLC26A8 genes that are linked to neural function, data which support the familial inheritance of neurogenic inflammation in rosacea development [27].

2.3. Immune Dysregulation

Innate immunity is a key player in the initiation and maintenance of rosacea. The dysregulation of toll-like receptor (TLR)2-kallikrein (KLK5)-cathelicidin(LL37) pathways has a crucial role in the pathogenesis of rosacea [28]. TLRs, type I integral transmembrane glycoproteins involved in host cell recognition and responses to microbial pathogens, are among the most characterized innate immune receptors [29]. TLR2 is a member of the TLR protein family, widely expressed in immune, endothelial, and epithelial cells, including keratinocytes and fibroblasts [30]. Following the interaction between pathogen-associated molecular patterns (PAMPs), TLR2 promotes a complex cascade of intracellular signaling [31] that leads to the activation of nuclear factor B (NFκ-B) and the release of proinflammatory cytokines, mainly interleukin (IL)-1, tumor necrosis factor α (TNFα), IL-6, thymic stromal lymphopoietin (TSLP), chemokines, matrix metalloproteinase, and prostaglandins [32]. In addition, TLR2 activation is associated with the release of KLK5, a critical protease involved in the pathogenesis of rosacea [33]. Although the upstream stimulator of TLR2 in rosacea is still unknown, keratinocytes of rosacea patients express higher amounts of this protein than healthy individuals. This fact may partially explain the overreactive response to environmental stimuli [34]. Furthermore, rosacea is associated with increased levels of KLK5, although the ultimate effect of this pathway depends on the cleavage of the active form of the antimicrobial peptide LL-37 [30]. KLK5 is not only involved in the innate immune system within the skin through LL-37 but is also able to directly activate protease-activated receptor 2 (PAR2), triggering the expression of pro-inflammatory cytokines such as IL-8 [35]. The production of LL-37 has been linked to different cell types, including mast cells, which are particularly increased in rosacea lesions [36,37,38]. Interestingly, subcutaneous injections of LL-37 have induced a strong inflammatory response in skin mouse models [33], while telangiectasia, erythema, and inflammation were not featured in mast cell-deficient mice, emphasizing the role of mast cells in the pathologenesis of rosacea [39]. Notably, LL-37 also promotes the release of matrix metalloproteinase (MMP)-9, which in turn activates KLK5, amplifying LL-37 expression, and perpetuating a vicious circle where inflammation is maintained [30]. However, further translational research validating molecular mechanisms in humans is still needed in this field. The complex immunomodulatory role of LL-37 is driven by multiple pathways [40]. Interestingly, LL-37 activates the Janus kinase (JAK)/signal transducer and transcription activator (STAT) signaling pathway, which mediates monocyte chemotaxis and IL-6-, IL-8-, and TNFα expression [41]. Downstream, LL37 exerts its effect through the NFκB pathway which promotes the release of proinflammatory cytokines and chemokines through mTORC1 signaling, which is hyperactivated in rosacea patients. In a positive feedback loop, mTORC1 leads to the activation of LL-37, which in turn activates mTORC1 signaling by binding TLR2 [42,43]. Finally, TLR2 also facilitates the activation of nucleotide binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3), a component of the NLRP3 inflammasome complex, in keratinocytes, which contributes to orchestrating neutrophil chemiotaxis, TNFα-mediated inflammation, prostaglandin e2 synthesis, and IL-1 cleavage and activation [44]. Interestingly, as with other inflammatory skin diseases, evidence is emerging of the role of the IL-1 family of cytokines in rosacea [45].
Both T and B lymphocytes are thought to contribute to the pathogenesis of rosacea, although little is known of the role of adaptive immunity. Histological analysis has revealed predominantly CD4+ T cells around the follicular and perivascular regions in rosacea skin [46]. The T-cell response in all rosacea phenotypes is mainly driven by Th1/Th17-polarized immune cells, as demonstrated by significant upregulation of their cytokines IFN-γ, TNF, IL-17, and IL-22 [47]. Moreover, Th17 promotes vascular endothelial growth factor (VEGF) expression in rosacea, which is also associated with angiogenesis [48]. Recent evidence has pointed to the role of activated B cells, as testified by high levels of B cell markers in skin biopsies of rosacea patients [47]. Additionally, B lymphocytes are responsible for the production of fibrogenic cytokines, such as IL-6 and transforming growth factor beta (TGFb), implicated in the fibrotic changes in phymatous rosacea [8].

2.4. Vascular, Neurovascular, and Neuroinflammatory Dysregulation

Comprehensively, increased skin blood flow, angiogenesis, and vasodilatation are essential processes in rosacea, as demonstrated by typical clinical features such as facial erythema and flushing [13]. The face is one of the few regions of human skin where blood vessels are simultaneously regulated by the sympathetic, parasympathetic, and sensory nerves; thus, vascular dysregulation is the result of a complex interaction. As already mentioned, VEGF and LL-37 are upregulated in rosacea, causing increased vascular permeability, angiogenesis, and vasodilatation [48]. Overall, the pro-angiogenic role of LL-37 is exerted via activation of the formyl peptide type 1 (FPR1) receptor, epidermal growth factor receptor (EGFR), and downstream signaling in epithelial cells [9]. In addition, LL-37 enhances the expression of the adhesion molecules intracellular adhesion molecule-1 (ICAM1), vascular cell adhesion molecule-1 (VCAM-1), and VEGF expression, which actively contribute to the regulation of angiogenic processes [49,50]. Neuroinflammatory and neurovascular dysregulation are also linked to facial erythema, edema, and hyperemia in rosacea patients [51]. The proximity between immune cells in the skin barrier and peripheral nervous system nerves may partially explain the intensive bidirectional crosstalk between those systems through different mediators. Notably, patients with rosacea are susceptible to several stimuli, including UV radiation, cold, heat, or stress, suggesting the relevance of the sensory nervous system in the pathogenesis of rosacea [52]. In detail, TRP ion channels include 27 related molecules that respond to a remarkable variety of chemical and physical factors and are linked to both pro- and anti-inflammatory mechanisms [53]. They are considered important components of the sensory system, mainly located on neuronal and non-neuronal cells, including mast cells, dendritic cells, endothelial cells, and keratinocytes [54]. High numbers of TRP channels seem to be responsible for transducing thermal, chemical, and mechanical rosacea stimuli into clinical manifestations of rosacea. Via the influx of cations, TRP channels lead to the release of vasoactive neuropeptides, such as substance P (SP), the peptide linked to the calcitonin gene (CGRP), the activator polypeptide of l pituitary adenylates cyclase (PACAP), the vasoactive intestinal peptide (VIP), or even bradykinin, which is involved in rosacea associated vasodilation, plasma extravasation, and leukocyte recruitment [55]. Moreover, TRP channels mediate pain sensations and itching [56]. Specifically, the activation of TRP ankyrin type 1 (TRPA1) and TRP vanilloid type 1 (TRPV1) lead to a burning sensation and flushing in rosacea patients [57].

2.5. Other Contributors (Sebaceous Glands, Skin Barrier Dysfunction, and Microbial Dysbiosis)

The sebaceous glands are exocrine glands that produce sebum, a lipid-rich fluid, produced by the holocrine rupture of mature sebocytes and secreted onto the surface of the skin. Sebaceous glands are mainly located on the face, scalp, chest, and back, and their dysfunction has been observed in several dermatological conditions, including acne and rosacea [58,59]. In particular, phymatous rosacea has been associated with thickening of the skin due to sebaceous gland hyperplasia [30]. Interestingly, the role of those glands has emerged from several studies on rosacea patients in which topical isotretinoin resulted in a significant reduction in sebaceous gland volume and sebum production, as well as in improvement of erythema and papulo-pustules [60]. Notably, the sebaceous fatty acid profile in patients with rosacea shows a decrease in long-chain saturated fatty acids that leads to skin dryness and hypersensitivity [61]. In this context, sebocytes could contribute to the general inflammatory status by releasing proinflammatory cytokines and adypokines, including IL-6 [8]. Although the pathophysiological link between chronic inflammation and glandular hyperplasia remains elusive, sebaceous gland dysfunction in rosacea might arise from TLR-mediated inflammatory status, changes in the skin microbiome, alteration in neuronal and endocrine factors, and alteration in skin barrier permeability [62].
Skin integrity is crucial since the skin acts as a barrier to microbes, toxins, and physical stressors, including UV radiation. The stratum corneum and tight junction are two important factors that contribute to maintaining balance in the epidermal barrier [63]. The dryness and hypersensitivity of rosacea skin may be attributed to a reduction in the hydration of the stratum corneous, also due to the decrease in claudin expression, a tight junction membrane protein which forms paracellular barriers and pores, determining tight junction permeability [64]. Indeed, molecular dysfunction in claudin expression has been observed in patients with papulopustular rosacea [62].
Furthermore, an intriguing link between the cutaneous microbiome and rosacea has been described [65]. An imbalance in common skin bacterial species, such as Cutibacterium acnes, Staphylococcus epidermidis, Bacillus oleronius, and Demodex folliculorum, might play a central role in rosacea pathogenesis, through propagation of the inflammation response [66]. However, the microbiological reverberations are not only limited to the skin, but surprisingly, emergent studies have described a potential interplay with the gut microbiome, although the exact mechanisms are still unknown [67,68].
Finally, the association between rosacea and several metabolic conditions, including hypertension, dyslipidemia, obesity, thyroid disorders, and diabetes, has been widely demonstrated in the literature [69,70,71]. Based on current knowledge, rosacea and metabolic-related disorders share increased levels of cathelicidin and inflammatory cytokines, which are responsible for the pathogenesis of both diseases [72].

3. Classification

The diagnosis and classification of rosacea was historically based on the predominant clinical features, which identified four disease subtypes: erythemato-telangiectatic, papulopustular, phymatous, and ocular rosacea [10,73]. Representative clinical pictures for each rosacea subtype are shown in Figure 2. As multiple features tend to present simultaneously and rosacea features can span multiple subtypes, a new phenotype-based approach was then proposed to facilitate clinical practice and improve rosacea management [11]. Indeed, this “phenotype” approach may address rosacea and its treatment in a manner that is more accurate and consistent with the patient’s individual experience. Thus, according to the National Rosacea Society [11], two features are considered as independently diagnostic for rosacea: (i) persistent, centrofacial erythema associated with periodic intensification and (ii) phymatous changes. In addition, minor features include a burning or stinging sensation, erythematous plaques, facial dryness and scaling edema, a peripheral location, and ocular manifestations [11].

4. Treatments

Currently, several treatment options have been approved for rosacea, although considerable differences may be observed within rosacea subtypes [74,75]. Nevertheless, the identification and avoidance of triggering factors (e.g., alcohol, the sun, hot drinks and spicy food) [75], self-care advice, and general skin care measures (e.g., mild facial cleansers and high-SPF broad-spectrum physical sunscreen) [13] remain fundamental for the proper management of all rosacea patients, leading to better treatment results [76]. The key aspects of the phenotype-based therapeutic approaches in rosacea are summarized in Table 1.

4.1. Transient/Persistent Centrofacial Erythema and Telangiectasia

To date, different therapeutic strategies are available for the treatment of flushing and facial erythema, including topical, systemic, or combined approaches; with regard to telangiectasias, vascular lasers and light-based therapies should be preferred [77].
Among the topical treatments, brimonidine tartrate (BT) topical gel is a highly selective α2 agonist approved for moderate-to-severe subtype I rosacea that induces the vasoconstriction of small arteries and veins, reducing vasodilation and edema. Applied once daily, its effects can be reached within 30 min with maximal mitigation in erythema 3–6 h after administration [78]. From two multicentric randomized trials, it emerged that after 1 month of therapy, there was a significant improvement in erythema after the application of brimonidine gel 0.5% in patients with erythemato-telangiectatic rosacea, but it did not significantly affect dilated capillaries [79]. Interestingly, Moore et al. [80] focused on the long-term safety and efficacy of topical BT gel 0.5% applied once a day for 12 months. They found that the most common adverse effects in their patients were flushing (9.1%), followed by worsening erythema (6.5%) and rosacea (3.6%), with the highest efficacy at months 1–3.
Oxymetazoline hydrochloride 1% cream represents another topical option for erythemato-telangiectatic rosacea. It is a potent α-1-adrenergic agonist that has been approved by the U.S. Food and Drug Administration (FDA) for the treatment of persistent erythema in adult patients [81]. Indeed, the results of the REVEAL study, conducted on 440 patients, demonstrate the safety and continued efficacy of oxymetazoline cream 1.0% for moderate-to-severe persistent erythema, although redness may flare after the discontinuation of treatment [82]. Moreover, 8.2% of the entire cohort reported treatment-emergent adverse events (TEAEs), including application-site dermatitis, paresthesia, pain, and pruritus [82]. Moreover, no clinically meaningful changes were observed in skin blanching, inflammatory lesions, or telangiectasia [82].
Several clinical trials on topical calcineurin inhibitors in papulopustular rosacea, such as pimecrolimus and tacrolimus, have shown excellent results in reducing the erythematous inflammatory component [83,84], even in the steroid-induced form of rosacea [85,86]. Conversely, Zhang et al. [87] underline a possible negative effect of pimecrolimus and tacrolimus since these drugs can even induce paradoxical rosacea-like eruptions.
Finally, a few studies have demonstrated that topical metronidazole and topical azelaic acid may reduce erythema in rosacea patients, although their efficacy has only been demonstrated in patients suffering from the papulopustular subtype [88,89,90].
Concerning oral therapies, the role of vasoactive compounds is still controversial. Nonselective β-blockers, especially carvedilol, are thought to decrease erythema and flushing, as well as α2-adrenergic agonist clonidine [91,92]. However, evidence supporting their efficacy is still limited.
Vascular lasers and light-based therapies remain one of the most effective strategies for persistent vascular manifestation, including erythema and telangiectasia. Intense pulsed light (IPL), pulsed dye laser (PDL), and neodymium:yttrium–aluminum–garnet laser (Nd:YAG) are recommended for erythemato-telangiectatic rosacea [93].
IPL has a wavelength between 420 and 1200 nm, which is absorbed by melanin and oxyhemoglobin [94]. Using filters, it emits the required wavelength to target vessels, minimizing energy absorption by melanin, which is not a desired target in rosacea, thus avoiding side effects such as hypopigmentation. This technology works by collating vessels, remodeling collagen, and reorganizing connective tissue [95]. Longer wavelengths are effective for deeper vessels, while shorter ones target more superficial vessels but can interact with melanin and should be avoided in darker skin types [96]. Generally, at least three sessions are required, at intervals of 1–3 weeks, and IPL can be used both in monotherapy and in combination with bipolar radiofrequency [97]. Interestingly, Papageorgiou et al. [98] reported a significant improvement in erythema and telangiectasia after four treatments performed at 3-week intervals, with self-limiting side effects. As regards a PDL, this device emits light in the wavelength of 585–595 nm, with the absorption peak of oxyhemoglobin targeting the superficial capillaries, leading to the obstruction of blood vessels. Used at purpuragenic fluences, it can provide a significant improvement in erythema, symptoms, and quality of life [99], especially in thick, rope-like telangiectasias [100]. Notably, to avoid post-treatment purpura and dyspigmentation of the earlier generation PDLs, a long-pulsed (595 nm) PDL at subpurpuric fluences can be used, with a very favorable safety profile [101]. In addition, fine and superficial telangiectasia seem to respond well to a potassium titanyl phosphate 532 nm laser (KTP laser), although PDL treatments have shown better outcomes [102]. Compared to the microsecond 1064 nm Nd:YAG, the non-purpuragenic 595 nm PDL was more effective for facial erythema in lighter-skinned patients, but microsecond Nd:YAG resulted in being less painful [103]. The synergistic approach to laser therapy, conducted through a sequential delivery of 595 and 1064 nm wavelength radiation with an interpulse delay, has proven to be more effective than standard single-wavelength therapy for facial telangiectasia [104].
In conclusion, patients with prominent telangiectasia will significantly benefit from laser/light-based therapies, but the in case of prominent underlying erythema, which may impair the discrimination of telangiectasia, combination therapies can also be used. Micali et al. [105] proposed a combined approach of sequential treatment for patients showing both marked background erythema and marked telangiectasias. Complete clearing of facial erythema was reached with a session of Nd:YAG laser followed by a first application of brimonidine and a second application 1 month after laser treatment [105].
Moreover, adjuvant topical brimonidine can be used to reduce IPL-induced erythema and associated pain without affecting efficacy [106].
Something that is noteworthy is that another emerging procedural therapy for both erythemato-telangiectatic and papulopustular rosacea is topical photodynamic therapy with 5-aminolevulinic acid (ALA-PDT). It has proved to be safe and effective [107], also in combination with IPL [108]. Lastly, two sessions of fractional microneedling radiofrequency (FMR) treatment at 4-week intervals showed modest improvement in erythema, and it might be used as an alternative or in combination with other treatment methods [109].
An intradermal injection of botulinum toxin (BTX) in large dilution (2 U/0.1 mL, with a total of 20 units of BTX) has recently been used off-label in erythemato-telangiectatic rosacea resistant to previous treatments, with the aim of reducing flushing, erythema, and inflammatory lesions [98]. BTX blocks the release of acetylcholine from the peripheral autonomic nerve endings of cutaneous vessels, thus reducing vascular disease manifestations [99]. Also, it may block the effects of acetylcholine on the erector muscles of the hair and the muscarinic receptors of the sebaceous glands [100]. Finally, BTX inhibits the release of substance P and the calcitonin gene-related peptide, reducing skin inflammation [101]. Interestingly, several studies have reported good outcomes for transient and persistent erythema in patients treated with a different subtype of botulin type A (BTX-A), including onabotulinum toxin A, abobotulinum toxin A, and incobotulinum toxin A [102,103,104].

4.2. Papulopustular Rosacea

Generally, papulopustular rosacea patients show a rapid response to topical medications, including azelaic acid, metronidazole, ivermectin, benzoyl peroxide, clindamycin, and erythromycin [110]. Even though for many years systemic therapies were preferred only for refractory patients, with the introduction of tetracyclines, oral therapy has become more commonly prescribed as a first-line treatment, alone or more often in combination with topical therapies [111].
As regards topical therapies, azelaic acid (AA) is a natural nine-carbon straight-chain saturated dicarboxylic acid widely found in barley, wheat, and rye and produced by Malasezzia furfur [112]. Due to its anti-inflammatory, anti-microbial, antioxidant, anticomedolytic, and antikeratinization activities, AA is indicated for acne, rosacea, melasma, and other dermatological conditions [113]. Topical 15% gel is approved by the Food and Drug Administration for inflammatory papules and pustules in mild-to-moderate rosacea, given its role in reducing ROS production and proinflammatory cytokines (including IL-1, IL-6, and TNFα). Also, it inhibits KLK5 and cathelicidin, and increases serine protease activity [114,115]. A recent meta-analysis demonstrated that erythema intensity and inflammatory lesion counts significantly improved in rosacea patients treated with AA compared with a vehicle after 12 weeks [116]. Moreover, a randomized trial showed that the use of 15% azelaic acid gel twice daily for 15 weeks has higher efficacy compared to 0.75% metronidazole gel, particularly regarding the inflammatory features of rosacea [117]. Another possible formulation of AA is the 20% cream, which seems more effective in papulopustular rosacea [118]. Finally, AA foam 15% is an effective and well-tolerated option that matches the needs of all skin types [119].
Metronidazole (MTZ) is a nitroimidazole antibiotic approved for papulopustular rosacea [120]. It is available in different formulations (as a gel, cream, or lotion). Usually, 0.75–1% cream and gel are the most used treatments. Traditionally applied twice daily, recent evidence has demonstrated that both 0.75% metronidazole cream and 1.0% metronidazole cream used once daily can provide well-tolerated efficacy for moderate-to-severe rosacea [121]. Topical metronidazole gel can also be used in combination with oral therapies, and the prolonged use of gel alone has shown a significantly high disease-free interval and lower recurrence [122]. Compared to AA, MTZ may have a higher tolerable profile, even though 15% azelaic acid gel twice daily for 15 weeks demonstrated significant superiority over 0.75% metronidazole gel in reducing inflammatory lesions and erythema [117].
Ivermectin is a broad-spectrum antiparasitic agent, recently introduced as a topical therapy for papulopustular rosacea, and, in particular, it targets the Demodex mite [123,124]. It also exerts an anti-inflammatory effect, since it upregulates the anti-inflammatory cytokine IL-10 and decreases IL-1β, TNFα, and neutrophilic diapedesis [125]. The application of ivermectin cream 1% once a day has shown clinical superiority and higher patient satisfaction over metronidazole cream 0.75%/twice daily [126].
Other topical therapies include treatments that have been mainly used for acne vulgaris in clinical practice. Despite its therapeutic properties, topical benzoyl peroxide (BPO) in patients with rosacea has traditionally been avoided due to high irritation rates. The FDA recently approved 5.0% microencapsulated benzoyl peroxide (E-BPO) for papulopustular rosacea. This encapsulation forms a barrier between the drug and the skin, with gradual release and the absorption of BPO, reducing tolerability issues and adverse events [127]. A 12-week treatment with minocycline 1.5% foam showed efficacy for moderate-to-severe papulopustular rosacea, maintaining a favorable safety profile without serious treatment-related adverse events [122]. A study conducted on calcineurin inhibitors demonstrated a reduction in facial erythema, although no significant decrease in the number of papulopustular lesions was observed [123]. Regarding topical retinoids, a randomized open trial with 55 rosacea patients demonstrated the efficacy of adapalene gel 0.1% [124]. Additionally, in a randomized double-blind trial, topical tretinoin 0.025% cream appeared to be effective for severe or recalcitrant rosacea compared to low-dose oral isotretinoin [125]. However, retinoids are usually avoided due to their irritant potential. Lastly, clindamycin and erythromycin in cream/gel have also shown good results, but larger studies still need to be carried out [126].
Oral antibiotics and isotretinoin represent the first line agents in severe papulopustular rosacea forms. Tetracyclines and their second-generation semisynthetic derivatives have anti-inflammatory properties and have proven to be effective in the treatment of inflammatory skin conditions [128]. Their primary mechanism of action against bacteria derives from their ability to bind to the bacterial 30S ribosomal subunit, thus inhibiting protein synthesis [128] and reducing inflammation [129]. Also, tetracyclines can exert anti-inflammatory and antioxidative activities by downregulating inflammatory cytokines, leukocyte migration, chemotaxis, and ROS production, as well as antiangiogenic effects via matrix metalloproteinase inhibition. These drugs also showed inhibitory effects on granuloma formation and proteolysis [130,131,132]. The only oral tetracycline approved by the FDA for moderate-to-severe papulopustular rosacea is a subantibiotic dose doxycycline 40 mg modified release capsule once daily [133]. This anti-inflammatory dose of doxycycline is as effective as a standard dose of 100 mg and offers the advantages of increased bioavailability, no bacterial resistance, and candidiasis [134], as well as minimal gastrointestinal side effects [135]. Of note, good and faster effects can be achieved by combining oral modified-release doxycycline (40 mg once/day) and AA 15% or MTZ 1% gel [136]. Oral minocycline 100 mg may represent a good alternative, since it was demonstrated that over a 16-week treatment period, 100 mg of minocycline was non-inferior to 40 mg of doxycycline in terms of efficacy, with comparable safety profiles [137]. Among the emerging tetracycline-derived antibiotics used for acne, lymecycline could also find greater application for the treatment of rosacea and topical steroid-induced rosacea [138,139].
Erythromycin, azithromycin, and clarithromycin have been used effectively and safely for the treatment of papulopustular rosacea, especially in patients not eligible for tetracyclines due to pregnancy, allergy, or intolerance or in cases of a lack of response [140]. Indeed, the management of pregnant women with inflammatory skin diseases is oftentimes challenging. Erythromycin in doses of 250–1000 mg/day is generally used in pregnant women with rosacea [141]; clarithromycin and azithromycin have been shown to have a faster effect with less gastrointestinal distress than erythromycin [142]. Generally, macrolides are considered second-line antibiotic therapies given the high rate of resistance that they have developed in recent years [143]. Furthermore, a double-blind trial (n = 29) showed the superiority of oral metronidazole compared to a placebo after 6 weeks of treatment [141]. Moreover, oral metronidazole (250 mg thrice daily for two weeks) can be added to oral minocycline (50 mg twice daily) to reduce relapses in rosacea patients, with a good safety profile [142].
For severe or antibiotics-recalcitrant papulopustular rosacea, intermittent courses of oral low-dose isotretinoin have also been reported to be effective, due to its anti-inflammatory properties. Oral low-dose isotretinoin (0.2 mg/kg/day) showed superiority compared to a placebo after 4 months of treatment of difficult-to-treat papulopustular rosacea [144]. In another controlled trial, low-dose oral isotretinoin was more effective than a placebo and acted like doxycycline (0.3 mg/kg/day) [145].
A systematic review focused on the widely spread notion that systemic isotretinoin taken within 6 to 12 months of cutaneous surgery could contribute to abnormal scarring or delayed wound healing. In this study, insufficient evidence was found to support delaying cutaneous surgery and most dermatological procedures, except for mechanical dermabrasion and fully ablative laser, which are not recommended in the setting of systemic isotretinoin treatment [146]. Interestingly, combined treatment of recalcitrant papulopustular rosacea involving PDL and fractional microneedling radiofrequency with low-dose isotretinoin demonstrated satisfactory efficacy with reasonable safety profiles in 25 patients with rosacea [147].
In an open clinical trial, the Nd:YAG laser was shown to be safe and effective against papulopustular lesions [145].
Interestingly, novel monoclonal antibodies and oral JAK inhibitors, largely used for the treatment of other inflammatory skin conditions such as psoriasis or atopic dermatitis, have also shown certain degrees of efficacy in papulopustular rosacea [146,147].
In an exploratory, open-label, investigator-initiated clinical trial, secukinumab, a human monoclonal antibody anti-interleukin-17A, led to a significant reduction in both papule count and global severity score (GSS) in patients with moderate-to-severe papulopustular rosacea [148]. Moreover, a recalcitrant case of granulomatous rosacea resolved after subcutaneous injections of adalimumab, a TNFα inhibitor, thus demonstrating the therapeutic potential of this agent for this condition [149].
Finally, a case series of patients with erythemato-telangiectatic and papulopustular rosacea treated with the oral JAK inhibitor tofacitinib was published, with it showing significant regression of rosacea signs and symptoms [150].

4.3. Phymatous Changes

Rhinophyma, a hypertrophy of the nasal soft tissues, presenting with a bulbous ap-pearance, is the most common manifestation of phymatous changes [151]. Similar lesions may occur in different facial locations including the ears (otophyma), forehead (meto-phyma), and chin (gnathophyma).
Physical procedures, such as surgical treatments (e.g., electrosurgery, dermabrasion, cryosurgery, and scalpel excision), and ablative laser surgery with CO2 laser (10,600 nm) and Er:YAG (2940 nm) are often necessary to correct the shape of a deformed nose in phymatous rosacea [152,153,154].
Among the pharmaceutical agents, isotretinoin has been associated with better prognosis of rhinophyma and the shrinkage of the overall volume of phymata by reducing the size of the sebaceous glands. Although it does not appear to be a curative option, it could represent a good strategy after laser treatment to prevent relapse [155,156].

4.4. Ocular Symptoms

Ocular symptoms are mainly represented by chronic blepharoconjunctivitis, episcleritis, chalazion, meibomian gland dysfunction, and corneal complications [2].
Patients with ocular rosacea should be referred to an ophthalmologist if they are experiencing eye discomfort and sticky eye discharge despite frequent topical lubricant use and adequate lid hygiene, especially if they ever experienced symptoms such as reduced vision and ocular pain [75].
While general suggestions such as lifestyle changes, avoidance of triggers, ocular hygiene, and proper ocular moisturization are not sufficient in the management of ocular rosacea, topical or oral treatments are recommended. Topical cyclosporine, topical azithromycin, and topical tacrolimus currently represent the most supported therapies in ocular rosacea [157]. Moreover, a retrospective study reported that seven ocular rosacea patients treated with doxycycline 40 mg, in a slow-release form, experienced a clear improvement after an average of 2.29 months of therapy [158].
Something that is noteworthy is that the long-term use of topical corticosteroids should be avoided due to side-effects such as ocular hypertension, glaucoma, posterior subcapsular cataracts, and tear-film instability [159].
To date, novel therapeutic perspectives include intense pulsed light therapy for the treatment of evaporative dry eye disease with chronic meibomian gland dysfunction [160].

5. Conclusions

In this review, we summarize the main pathophysiological mechanisms and possible therapeutic avenues for rosacea, focusing on a phenotype-based approach. Indeed, a customized and tailored therapeutic strategy should always be pursued in rosacea, mainly based on the clinical phenotype, yet not neglecting the specific needs of each patient.
However, a more precise understanding of the molecular basis of rosacea is still needed and could hopefully lead to the identification of new therapeutic targets addressing the root cause of the disease in the near future.
Interestingly, in addition to commonly used topical and systemic therapies as well as physical approaches, new evidence is slowly emerging on the therapeutic potential of novel agents, such as monoclonal antibodies or oral small molecules (JAK inhibitors) in rosacea. Indeed, given the complex pathogenesis and the role of JAK/STAT in this disease, there is a rationale as to why JAK inhibitors could theoretically prove effective for the treatment of rosacea, as has already happened in psoriasis, atopic dermatitis, and vitiligo [161,162,163]. However, currently available data on the efficacy and safety of these novel therapeutic approaches are still scarce.
For all of these reasons, further clinical and translational research, as well as further clinical studies on novel or existing therapeutic agents for rosacea, should be encouraged.

Author Contributions

Conceptualization, L.C.; methodology, M.D. and G.G.; resources, D.M.; data curation, G.R.; writing—original draft preparation, M.D. and G.G.; writing—review and editing, L.C., M.D. and G.G.; supervision, L.L., E.C. and P.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Written informed consent was obtained from the patients involved in this study to publish this paper.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Van Zuuren, E.J. Rosacea. N. Engl. J. Med. 2017, 377, 1754–1764. [Google Scholar] [CrossRef]
  2. Tavassoli, S.; Wong, N.; Chan, E. Ocular manifestations of rosacea: A clinical review. Clin. Exp. Ophthalmol. 2021, 49, 104–117. [Google Scholar] [CrossRef] [PubMed]
  3. Chang, H.C.; Huang, Y.C.; Lien, Y.J.; Chang, Y.S. Association of rosacea with depression and anxiety: A systematic review and meta-analysis. J. Affect. Disord. 2022, 299, 239–245. [Google Scholar] [CrossRef] [PubMed]
  4. Hung, Y.T.; Le, P.H.; Kuo, C.J.; Tang, Y.C.; Chiou, M.J.; Chiu, C.T.; Kuo, C.F.; Huang, Y.H. The Temporal Relationships and Associations between Cutaneous Manifestations and Inflammatory Bowel Disease: A Nationwide Population-Based Cohort Study. J. Clin. Med. 2021, 10, 1311. [Google Scholar] [CrossRef]
  5. Oussedik, E.; Bourcier, M.; Tan, J. Psychosocial Burden and Other Impacts of Rosacea on Patients’ Quality of Life. Dermatol. Clin. 2018, 36, 103–113. [Google Scholar] [CrossRef] [PubMed]
  6. Romanowicz, M.; Stephenson, J.J.; Del Rosso, J.Q.; Lenhart, G. Healthcare utilization and costs of patients with rosacea in an insured population. J. Drugs Dermatol. 2008, 7, 41–49. [Google Scholar]
  7. Gether, L.; Overgaard, L.K.; Egeberg, A.; Thyssen, J.P. Incidence and prevalence of rosacea: A systematic review and meta-analysis. Br. J. Dermatol. 2018, 179, 282–289. [Google Scholar] [CrossRef]
  8. Chen, C.; Wang, P.; Zhang, L.; Liu, X.; Zhang, H.; Cao, Y.; Wang, X.; Zeng, Q. Exploring the Pathogenesis and Mechanism-Targeted Treatments of Rosacea: Previous Understanding and Updates. Biomedicines 2023, 11, 2153. [Google Scholar] [CrossRef]
  9. Ahn, C.S.; Huang, W.W. Rosacea Pathogenesis. Dermatol. Clin. 2018, 36, 81–86. [Google Scholar] [CrossRef]
  10. Wilkin, J.; Dahl, M.; Detmar, M.; Drake, L.; Feinstein, A.; Odom, R.; Powell, F. Standard classification of rosacea: Report of the National Rosacea Society Expert Committee on the Classification and Staging of Rosacea. J. Am. Acad. Dermatol. 2002, 46, 584–587. [Google Scholar] [CrossRef]
  11. Tan, J.; Almeida, L.M.; Bewley, A.; Cribier, B.; Dlova, N.C.; Gallo, R.; Kautz, G.; Mannis, M.; Oon, H.H.; Rajagopalan, M.; et al. Updating the diagnosis, classification and assessment of rosacea: Recommendations from the global ROSacea COnsensus (ROSCO) panel. Br. J. Dermatol. 2017, 176, 431–438. [Google Scholar] [CrossRef] [PubMed]
  12. Barakji, Y.A.; Ronnstad, A.T.M.; Christensen, M.O.; Zachariae, C.; Wienholtz, N.K.F.; Halling, A.S.; Maul, J.T.; Thomsen, S.F.; Egeberg, A.; Thyssen, J.P. Assessment of Frequency of Rosacea Subtypes in Patients with Rosacea: A Systematic Review and Meta-analysis. JAMA Dermatol. 2022, 158, 617–625. [Google Scholar] [CrossRef] [PubMed]
  13. Sharma, A.; Kroumpouzos, G.; Kassir, M.; Galadari, H.; Goren, A.; Grabbe, S.; Goldust, M. Rosacea management: A comprehensive review. J. Cosmet. Dermatol. 2022, 21, 1895–1904. [Google Scholar] [CrossRef]
  14. Aktas Karabay, E.; Aksu Cerman, A. Demodex folliculorum infestations in common facial dermatoses: Acne vulgaris, rosacea, seborrheic dermatitis. An. Bras. Dermatol. 2020, 95, 187–193. [Google Scholar] [CrossRef] [PubMed]
  15. Forton, F.; Seys, B. Density of Demodex folliculorum in rosacea: A case-control study using standardized skin-surface biopsy. Br. J. Dermatol. 1993, 128, 650–659. [Google Scholar] [CrossRef] [PubMed]
  16. Forton, F.M. Papulopustular rosacea, skin immunity and Demodex: Pityriasis folliculorum as a missing link. J. Eur. Acad. Dermatol. Venereol. 2012, 26, 19–28. [Google Scholar] [CrossRef]
  17. Forton, F.M.N. Rosacea, an infectious disease: Why rosacea with papulopustules should be considered a demodicosis. A narrative review. J. Eur. Acad. Dermatol. Venereol. 2022, 36, 987–1002. [Google Scholar] [CrossRef] [PubMed]
  18. Jarmuda, S.; O’Reilly, N.; Zaba, R.; Jakubowicz, O.; Szkaradkiewicz, A.; Kavanagh, K. Potential role of Demodex mites and bacteria in the induction of rosacea. J. Med. Microbiol. 2012, 61 Pt 11, 1504–1510. [Google Scholar] [CrossRef]
  19. Alia, E.; Feng, H. Rosacea pathogenesis, common triggers, and dietary role: The cause, the trigger, and the positive effects of different foods. Clin. Dermatol. 2022, 40, 122–127. [Google Scholar] [CrossRef]
  20. Reich, A.; Wojcik-Maciejewicz, A.; Slominski, A.T. Stress and the skin. G Ital. Dermatol. Venereol. 2010, 145, 213–219. [Google Scholar]
  21. Awosika, O.; Oussedik, E. Genetic Predisposition to Rosacea. Dermatol. Clin. 2018, 36, 87–92. [Google Scholar] [CrossRef]
  22. Yazici, A.C.; Tamer, L.; Ikizoglu, G.; Kaya, T.I.; Api, H.; Yildirim, H.; Adiguzel, A. GSTM1 and GSTT1 null genotypes as possible heritable factors of rosacea. Photodermatol. Photoimmunol. Photomed. 2006, 22, 208–210. [Google Scholar] [CrossRef]
  23. Chang, A.L.S.; Raber, I.; Xu, J.; Li, R.; Spitale, R.; Chen, J.; Kiefer, A.K.; Tian, C.; Eriksson, N.K.; Hinds, D.A.; et al. Assessment of the genetic basis of rosacea by genome-wide association study. J. Investig. Dermatol. 2015, 135, 1548–1555. [Google Scholar] [CrossRef]
  24. Karpouzis, A.; Avgeridis, P.; Tripsianis, G.; Gatzidou, E.; Kourmouli, N.; Veletza, S. Assessment of Tachykinin Receptor 3′ Gene Polymorphism rs3733631 in Rosacea. Int. Sch. Res. Not. 2015, 2015, 469402. [Google Scholar] [CrossRef] [PubMed]
  25. Fukui, K.; Kariyama, H.; Kashiba, A.; Kato, N.; Kimura, H. Further confirmation of heterogeneity of the rat striatum: Different mosaic patterns of dopamine fibers after administration of methamphetamine or reserpine. Brain Res. 1986, 382, 81–86. [Google Scholar] [CrossRef]
  26. Hayran, Y.; Lay, I.; Mocan, M.C.; Bozduman, T.; Ersoy-Evans, S. Vascular endothelial growth factor gene polymorphisms in patients with rosacea: A case-control study. J. Am. Acad. Dermatol. 2019, 81, 348–354. [Google Scholar] [CrossRef] [PubMed]
  27. Deng, Z.; Chen, M.; Zhao, Z.; Xiao, W.; Liu, T.; Peng, Q.; Wu, Z.; Xu, S.; Shi, W.; Jian, D.; et al. Whole genome sequencing identifies genetic variants associated with neurogenic inflammation in rosacea. Nat. Commun. 2023, 14, 3958. [Google Scholar] [CrossRef]
  28. van Zuuren, E.J.; Arents, B.W.M.; van der Linden, M.M.D.; Vermeulen, S.; Fedorowicz, Z.; Tan, J. Rosacea: New Concepts in Classification and Treatment. Am. J. Clin. Dermatol. 2021, 22, 457–465. [Google Scholar] [CrossRef]
  29. Aluri, J.; Cooper, M.A.; Schuettpelz, L.G. Toll-Like Receptor Signaling in the Establishment and Function of the Immune System. Cells 2021, 10, 1374. [Google Scholar] [CrossRef]
  30. Rodrigues-Braz, D.; Zhao, M.; Yesilirmak, N.; Aractingi, S.; Behar-Cohen, F.; Bourges, J.L. Cutaneous and ocular rosacea: Common and specific physiopathogenic mechanisms and study models. Mol. Vis. 2021, 27, 323–353. [Google Scholar] [PubMed]
  31. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [PubMed]
  32. Oliveira-Nascimento, L.; Massari, P.; Wetzler, L.M. The Role of TLR2 in Infection and Immunity. Front. Immunol. 2012, 3, 79. [Google Scholar] [CrossRef]
  33. Yamasaki, K.; Di Nardo, A.; Bardan, A.; Murakami, M.; Ohtake, T.; Coda, A.; Dorschner, R.A.; Bonnart, C.; Descargues, P.; Hovnanian, A.; et al. Increased serine protease activity and cathelicidin promotes skin inflammation in rosacea. Nat. Med. 2007, 13, 975–980. [Google Scholar] [CrossRef]
  34. Yamasaki, K.; Kanada, K.; Macleod, D.T.; Borkowski, A.W.; Morizane, S.; Nakatsuji, T.; Cogen, A.L.; Gallo, R.L. TLR2 expression is increased in rosacea and stimulates enhanced serine protease production by keratinocytes. J. Investig. Dermatol. 2011, 131, 688–697. [Google Scholar] [CrossRef]
  35. Zhu, Y.; Underwood, J.; Macmillan, D.; Shariff, L.; O’Shaughnessy, R.; Harper, J.I.; Pickard, C.; Friedmann, P.S.; Healy, E.; Di, W.L. Persistent kallikrein 5 activation induces atopic dermatitis-like skin architecture independent of PAR2 activity. J. Allergy Clin. Immunol. 2017, 140, 1310–1322.e5. [Google Scholar] [CrossRef]
  36. Wang, G.; Narayana, J.L.; Mishra, B.; Zhang, Y.; Wang, F.; Wang, C.; Zarena, D.; Lushnikova, T.; Wang, X. Design of Antimicrobial Peptides: Progress Made with Human Cathelicidin LL-37. Adv. Exp. Med. Biol. 2019, 1117, 215–240. [Google Scholar] [CrossRef] [PubMed]
  37. Sanchez-Pena, F.J.; Romero-Tlalolini, M.L.A.; Torres-Aguilar, H.; Cruz-Hernandez, D.S.; Baltierrez-Hoyos, R.; Sanchez-Aparicio, S.R.; Aquino-Dominguez, A.S.; Aguilar-Ruiz, S.R. LL-37 Triggers Antimicrobial Activity in Human Platelets. Int. J. Mol. Sci. 2023, 24, 2816. [Google Scholar] [CrossRef] [PubMed]
  38. Marson, J.W.; Baldwin, H.E. Rosacea: A wholistic review and update from pathogenesis to diagnosis and therapy. Int. J. Dermatol. 2020, 59, e175–e182. [Google Scholar] [CrossRef]
  39. Muto, Y.; Wang, Z.; Vanderberghe, M.; Two, A.; Gallo, R.L.; Di Nardo, A. Mast cells are key mediators of cathelicidin-initiated skin inflammation in rosacea. J. Investig. Dermatol. 2014, 134, 2728–2736. [Google Scholar] [CrossRef]
  40. Sorensen, O.E.; Follin, P.; Johnsen, A.H.; Calafat, J.; Tjabringa, G.S.; Hiemstra, P.S.; Borregaard, N. Human cathelicidin, hCAP-18, is processed to the antimicrobial peptide LL-37 by extracellular cleavage with proteinase 3. Blood 2001, 97, 3951–3959. [Google Scholar] [CrossRef]
  41. Li, T.; Zeng, Q.; Chen, X.; Wang, G.; Zhang, H.; Yu, A.; Wang, H.; Hu, Y. The therapeutic effect of artesunate on rosacea through the inhibition of the JAK/STAT signaling pathway. Mol. Med. Rep. 2018, 17, 8385–8390. [Google Scholar] [CrossRef] [PubMed]
  42. Deng, Z.; Chen, M.; Liu, Y.; Xu, S.; Ouyang, Y.; Shi, W.; Jian, D.; Wang, B.; Liu, F.; Li, J.; et al. A positive feedback loop between mTORC1 and cathelicidin promotes skin inflammation in rosacea. EMBO Mol. Med. 2021, 13, e13560. [Google Scholar] [CrossRef] [PubMed]
  43. Vallabhapurapu, S.; Karin, M. Regulation and function of NF-kappaB transcription factors in the immune system. Annu. Rev. Immunol. 2009, 27, 693–733. [Google Scholar] [CrossRef]
  44. Kelley, N.; Jeltema, D.; Duan, Y.; He, Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int. J. Mol. Sci. 2019, 20, 3328. [Google Scholar] [CrossRef]
  45. Calabrese, L.; Fiocco, Z.; Satoh, T.K.; Peris, K.; French, L.E. Therapeutic potential of targeting interleukin-1 family cytokines in chronic inflammatory skin diseases. Br. J. Dermatol. 2022, 186, 925–941. [Google Scholar] [CrossRef] [PubMed]
  46. Holmes, A.D.; Steinhoff, M. Integrative concepts of rosacea pathophysiology, clinical presentation and new therapeutics. Exp. Dermatol. 2017, 26, 659–667. [Google Scholar] [CrossRef]
  47. Buhl, T.; Sulk, M.; Nowak, P.; Buddenkotte, J.; McDonald, I.; Aubert, J.; Carlavan, I.; Deret, S.; Reiniche, P.; Rivier, M.; et al. Molecular and Morphological Characterization of Inflammatory Infiltrate in Rosacea Reveals Activation of Th1/Th17 Pathways. J. Investig. Dermatol. 2015, 135, 2198–2208. [Google Scholar] [CrossRef]
  48. Lee, H.J.; Hong, Y.J.; Kim, M. Angiogenesis in Chronic Inflammatory Skin Disorders. Int. J. Mol. Sci. 2021, 22, 12035. [Google Scholar] [CrossRef]
  49. Kulkarni, N.N.; Takahashi, T.; Sanford, J.A.; Tong, Y.; Gombart, A.F.; Hinds, B.; Cheng, J.Y.; Gallo, R.L. Innate Immune Dysfunction in Rosacea Promotes Photosensitivity and Vascular Adhesion Molecule Expression. J. Investig. Dermatol. 2020, 140, 645–655.e6. [Google Scholar] [CrossRef]
  50. Melincovici, C.S.; Bosca, A.B.; Susman, S.; Marginean, M.; Mihu, C.; Istrate, M.; Moldovan, I.M.; Roman, A.L.; Mihu, C.M. Vascular endothelial growth factor (VEGF)—Key factor in normal and pathological angiogenesis. Rom. J. Morphol. Embryol. 2018, 59, 455–467. [Google Scholar]
  51. Schwab, V.D.; Sulk, M.; Seeliger, S.; Nowak, P.; Aubert, J.; Mess, C.; Rivier, M.; Carlavan, I.; Rossio, P.; Metze, D.; et al. Neurovascular and neuroimmune aspects in the pathophysiology of rosacea. J. Investig. Dermatol. Symp. Proc. 2011, 15, 53–62. [Google Scholar] [CrossRef] [PubMed]
  52. Guzman-Sanchez, D.A.; Ishiuji, Y.; Patel, T.; Fountain, J.; Chan, Y.H.; Yosipovitch, G. Enhanced skin blood flow and sensitivity to noxious heat stimuli in papulopustular rosacea. J. Am. Acad. Dermatol. 2007, 57, 800–805. [Google Scholar] [CrossRef]
  53. Wang, H.; Siemens, J. TRP ion channels in thermosensation, thermoregulation and metabolism. Temperature 2015, 2, 178–187. [Google Scholar] [CrossRef] [PubMed]
  54. Dietrich, A. Transient Receptor Potential (TRP) Channels in Health and Disease. Cells 2019, 8, 413. [Google Scholar] [CrossRef] [PubMed]
  55. Silverman, H.A.; Chen, A.; Kravatz, N.L.; Chavan, S.S.; Chang, E.H. Involvement of Neural Transient Receptor Potential Channels in Peripheral Inflammation. Front. Immunol. 2020, 11, 590261. [Google Scholar] [CrossRef]
  56. Zhou, X.; Su, Y.; Wu, S.; Wang, H.; Jiang, R.; Jiang, X. The temperature-sensitive receptors TRPV4 and TRPM8 have important roles in the pruritus of rosacea. J. Dermatol. Sci. 2022, 108, 68–76. [Google Scholar] [CrossRef]
  57. Szolcsanyi, J. A pharmacological approach to elucidation of the role of different nerve fibres and receptor endings in mediation of pain. J. Physiol. 1977, 73, 251–259. [Google Scholar]
  58. Shi, V.Y.; Leo, M.; Hassoun, L.; Chahal, D.S.; Maibach, H.I.; Sivamani, R.K. Role of sebaceous glands in inflammatory dermatoses. J. Am. Acad. Dermatol. 2015, 73, 856–863. [Google Scholar] [CrossRef]
  59. Clayton, R.W.; Langan, E.A.; Ansell, D.M.; de Vos, I.; Gobel, K.; Schneider, M.R.; Picardo, M.; Lim, X.; van Steensel, M.A.M.; Paus, R. Neuroendocrinology and neurobiology of sebaceous glands. Biol. Rev. Camb. Philos. Soc. 2020, 95, 592–624. [Google Scholar] [CrossRef]
  60. Khalil, N.Y.; Darwish, I.A.; Al-Qahtani, A.A. Isotretinoin. Profiles Drug Subst. Excip. Relat. Methodol. 2020, 45, 119–157. [Google Scholar] [CrossRef]
  61. Ni Raghallaigh, S.; Bender, K.; Lacey, N.; Brennan, L.; Powell, F.C. The fatty acid profile of the skin surface lipid layer in papulopustular rosacea. Br. J. Dermatol. 2012, 166, 279–287. [Google Scholar] [CrossRef] [PubMed]
  62. Medgyesi, B.; Dajnoki, Z.; Beke, G.; Gaspar, K.; Szabo, I.L.; Janka, E.A.; Poliska, S.; Hendrik, Z.; Mehes, G.; Torocsik, D.; et al. Rosacea Is Characterized by a Profoundly Diminished Skin Barrier. J. Investig. Dermatol. 2020, 140, 1938–1950.e5. [Google Scholar] [CrossRef]
  63. Jensen, J.M.; Proksch, E. The skin’s barrier. G. Ital. Dermatol. Venereol. 2009, 144, 689–700. [Google Scholar] [PubMed]
  64. Gunzel, D.; Yu, A.S. Claudins and the modulation of tight junction permeability. Physiol. Rev. 2013, 93, 525–569. [Google Scholar] [CrossRef]
  65. Daou, H.; Paradiso, M.; Hennessy, K.; Seminario-Vidal, L. Rosacea and the Microbiome: A Systematic Review. Dermatol. Ther. 2021, 11, 1–12. [Google Scholar] [CrossRef]
  66. Condro, G.; Guerini, M.; Castello, M.; Perugini, P. Acne Vulgaris, Atopic Dermatitis and Rosacea: The Role of the Skin Microbiota-A Review. Biomedicines 2022, 10, 2523. [Google Scholar] [CrossRef]
  67. Mahmud, M.R.; Akter, S.; Tamanna, S.K.; Mazumder, L.; Esti, I.Z.; Banerjee, S.; Akter, S.; Hasan, M.R.; Acharjee, M.; Hossain, M.S.; et al. Impact of gut microbiome on skin health: Gut-skin axis observed through the lenses of therapeutics and skin diseases. Gut Microbes 2022, 14, 2096995. [Google Scholar] [CrossRef] [PubMed]
  68. Zhu, W.; Hamblin, M.R.; Wen, X. Role of the skin microbiota and intestinal microbiome in rosacea. Front. Microbiol. 2023, 14, 1108661. [Google Scholar] [CrossRef]
  69. Akin Belli, A.; Ozbas Gok, S.; Akbaba, G.; Etgu, F.; Dogan, G. The relationship between rosacea and insulin resistance and metabolic syndrome. Eur. J. Dermatol. 2016, 26, 260–264. [Google Scholar] [CrossRef]
  70. Gonulal, M.; Teker, K.; Ozturk, A.; Yasar, F.Y. Investigation of thyroid blood tests and thyroid ultrasound findings of patients with rosacea. Dermatol. Ther. 2021, 34, e14632. [Google Scholar] [CrossRef]
  71. Haber, R.; El Gemayel, M. Comorbidities in rosacea: A systematic review and update. J. Am. Acad. Dermatol. 2018, 78, 786–792.e8. [Google Scholar] [CrossRef]
  72. Seremet, S.; Gurel, M.S. Miscellaneous skin disease and the metabolic syndrome. Clin. Dermatol. 2018, 36, 94–100. [Google Scholar] [CrossRef]
  73. Schaller, M.; Almeida, L.M.C.; Bewley, A.; Cribier, B.; Del Rosso, J.; Dlova, N.C.; Gallo, R.L.; Granstein, R.D.; Kautz, G.; Mannis, M.J.; et al. Recommendations for rosacea diagnosis, classification and management: Update from the global ROSacea COnsensus 2019 panel. Br. J. Dermatol. 2020, 182, 1269–1276. [Google Scholar] [CrossRef]
  74. Anzengruber, F.; Czernielewski, J.; Conrad, C.; Feldmeyer, L.; Yawalkar, N.; Hausermann, P.; Cozzio, A.; Mainetti, C.; Goldblum, D.; Lauchli, S.; et al. Swiss S1 guideline for the treatment of rosacea. J. Eur. Acad. Dermatol. Venereol. 2017, 31, 1775–1791. [Google Scholar] [CrossRef] [PubMed]
  75. Hampton, P.J.; Berth-Jones, J.; Duarte Williamson, C.E.; Hay, R.; Leslie, T.A.; Porter, I.; Rauz, S.; Seukeran, D.; Winn, R.T.; Hashme, M.; et al. British Association of Dermatologists guidelines for the management of people with rosacea 2021. Br. J. Dermatol. 2021, 185, 725–735. [Google Scholar] [CrossRef]
  76. Paiva-Santos, A.C.; Goncalves, T.; Peixoto, D.; Pires, P.C.; Velsankar, K.; Jha, N.K.; Chavda, V.P.; Mohammad, I.S.; Cefali, L.C.; Mazzola, P.G.; et al. Rosacea Topical Treatment and Care: From Traditional to New Drug Delivery Systems. Mol. Pharm. 2023, 20, 3804–3828. [Google Scholar] [CrossRef] [PubMed]
  77. Shim, T.N.; Abdullah, A. The effect of pulsed dye laser on the dermatology life quality index in erythematotelangiectatic rosacea patients: An assessment. J. Clin. Aesthet. Dermatol. 2013, 6, 30–32. [Google Scholar] [PubMed]
  78. Anderson, M.S.; Nadkarni, A.; Cardwell, L.A.; Alinia, H.; Feldman, S.R. Spotlight on brimonidine topical gel 0.33% for facial erythema of rosacea: Safety, efficacy, and patient acceptability. Patient Prefer. Adherence 2017, 11, 1143–1150. [Google Scholar] [CrossRef]
  79. Fowler, J.J.; Jackson, M.; Moore, A.; Jarratt, M.; Jones, T.; Meadows, K.; Steinhoff, M.; Rudisill, D.; Leoni, M. Efficacy and safety of once-daily topical brimonidine tartrate gel 0.5% for the treatment of moderate to severe facial erythema of rosacea: Results of two randomized, double-blind, and vehicle-controlled pivotal studies. J. Drugs Dermatol. 2013, 12, 650–656. [Google Scholar]
  80. Moore, A.; Kempers, S.; Murakawa, G.; Weiss, J.; Tauscher, A.; Swinyer, L.; Liu, H.; Leoni, M. Long-term safety and efficacy of once-daily topical brimonidine tartrate gel 0.5% for the treatment of moderate to severe facial erythema of rosacea: Results of a 1-year open-label study. J. Drugs Dermatol. 2014, 13, 56–61. [Google Scholar]
  81. Patel, N.U.; Shukla, S.; Zaki, J.; Feldman, S.R. Oxymetazoline hydrochloride cream for facial erythema associated with rosacea. Expert Rev. Clin. Pharmacol. 2017, 10, 1049–1054. [Google Scholar] [CrossRef] [PubMed]
  82. Draelos, Z.D.; Gold, M.H.; Weiss, R.A.; Baumann, L.; Grekin, S.K.; Robinson, D.M.; Kempers, S.E.; Alvandi, N.; Weng, E.; Berk, D.R.; et al. Efficacy and safety of oxymetazoline cream 1.0% for treatment of persistent facial erythema associated with rosacea: Findings from the 52-week open label REVEAL trial. J. Am. Acad. Dermatol. 2018, 78, 1156–1163. [Google Scholar] [CrossRef] [PubMed]
  83. Koca, R.; Altinyazar, H.C.; Ankarali, H.; Muhtar, S.; Tekin, N.S.; Cinar, S. A comparison of metronidazole 1% cream and pimecrolimus 1% cream in the treatment of patients with papulopustular rosacea: A randomized open-label clinical trial. Clin. Exp. Dermatol. 2010, 35, 251–256. [Google Scholar] [CrossRef]
  84. Kim, M.B.; Kim, G.W.; Park, H.J.; Kim, H.S.; Chin, H.W.; Kim, S.H.; Kim, B.S.; Ko, H.C. Pimecrolimus 1% cream for the treatment of rosacea. J. Dermatol. 2011, 38, 1135–1139. [Google Scholar] [CrossRef] [PubMed]
  85. Lee, D.H.; Li, K.; Suh, D.H. Pimecrolimus 1% cream for the treatment of steroid-induced rosacea: An 8-week split-face clinical trial. Br. J. Dermatol. 2008, 158, 1069–1076. [Google Scholar] [CrossRef]
  86. Meykadeh, N.; Meiss, F.; Marsch, W.C.; Fischer, M. Steroid-aggravated rosacea: Successful therapy with pimecrolimus. Hautarzt 2007, 58, 338–342. [Google Scholar] [CrossRef]
  87. Zhang, H.; Yang, L.; Wang, Y.; Zhang, D.; Tang, K.; Fang, R.; Sun, Q. Topical calcineurin inhibitors as a double-edged sword in rosacea: A systematic review. J. Cosmet. Dermatol. 2022, 21, 1695–1704. [Google Scholar] [CrossRef]
  88. Breneman, D.L.; Stewart, D.; Hevia, O.; Hino, P.D.; Drake, L.A. A double-blind, multicenter clinical trial comparing efficacy of once-daily metronidazole 1 percent cream to vehicle in patients with rosacea. Cutis 1998, 61, 44–47. [Google Scholar]
  89. Kocak, M.; Yagli, S.; Vahapoglu, G.; Eksioglu, M. Permethrin 5% cream versus metronidazole 0.75% gel for the treatment of papulopustular rosacea. A randomized double-blind placebo-controlled study. Dermatology 2002, 205, 265–270. [Google Scholar] [CrossRef]
  90. Bjerke, R.; Fyrand, O.; Graupe, K. Double-blind comparison of azelaic acid 20% cream and its vehicle in treatment of papulo-pustular rosacea. Acta Derm. Venereol. 1999, 79, 456–459. [Google Scholar] [CrossRef]
  91. Hsu, C.C.; Lee, J.Y. Carvedilol for the treatment of refractory facial flushing and persistent erythema of rosacea. Arch. Dermatol. 2011, 147, 1258–1260. [Google Scholar] [CrossRef] [PubMed]
  92. Seo, B.H.; Kim, D.H.; Suh, H.S.; Choi, Y.S. Facial flushing and erythema of rosacea improved by carvedilol. Dermatol. Ther. 2020, 33, e14520. [Google Scholar] [CrossRef]
  93. Hofmann, M.A.; Lehmann, P. Physical modalities for the treatment of rosacea. J. Dtsch. Dermatol. Ges. 2016, 14 (Suppl. S6), 38–43. [Google Scholar] [CrossRef] [PubMed]
  94. Kawana, S.; Ochiai, H.; Tachihara, R. Objective evaluation of the effect of intense pulsed light on rosacea and solar lentigines by spectrophotometric analysis of skin color. Dermatol. Surg. 2007, 33, 449–454. [Google Scholar] [CrossRef]
  95. Sadick, N.S.; Weiss, R. Intense pulsed-light photorejuvenation. Semin. Cutan. Med. Surg. 2002, 21, 280–287. [Google Scholar] [CrossRef] [PubMed]
  96. Mansouri, Y.; Goldenberg, G. Devices and topical agents for rosacea management. Cutis 2014, 94, 21–25. [Google Scholar]
  97. Taub, A.F.; Devita, E.C. Successful treatment of erythematotelangiectatic rosacea with pulsed light and radiofrequency. J. Clin. Aesthet. Dermatol. 2008, 1, 37–40. [Google Scholar]
  98. Papageorgiou, P.; Clayton, W.; Norwood, S.; Chopra, S.; Rustin, M. Treatment of rosacea with intense pulsed light: Significant improvement and long-lasting results. Br. J. Dermatol. 2008, 159, 628–632. [Google Scholar] [CrossRef]
  99. Tan, S.R.; Tope, W.D. Pulsed dye laser treatment of rosacea improves erythema, symptomatology, and quality of life. J. Am. Acad. Dermatol. 2004, 51, 592–599. [Google Scholar] [CrossRef]
  100. Alam, M.; Dover, J.S.; Arndt, K.A. Treatment of facial telangiectasia with variable-pulse high-fluence pulsed-dye laser: Comparison of efficacy with fluences immediately above and below the purpura threshold. Dermatol. Surg. 2003, 29, 681–684; discussion 685. [Google Scholar] [CrossRef]
  101. Bernstein, E.F.; Kligman, A. Rosacea treatment using the new-generation, high-energy, 595 nm, long pulse-duration pulsed-dye laser. Lasers Surg. Med. 2008, 40, 233–239. [Google Scholar] [CrossRef] [PubMed]
  102. West, T.B.; Alster, T.S. Comparison of the long-pulse dye (590-595 nm) and KTP (532 nm) lasers in the treatment of facial and leg telangiectasias. Dermatol. Surg. 1998, 24, 221–226. [Google Scholar] [CrossRef]
  103. Alam, M.; Voravutinon, N.; Warycha, M.; Whiting, D.; Nodzenski, M.; Yoo, S.; West, D.P.; Veledar, E.; Poon, E. Comparative effectiveness of nonpurpuragenic 595-nm pulsed dye laser and microsecond 1064-nm neodymium:yttrium-aluminum-garnet laser for treatment of diffuse facial erythema: A double-blind randomized controlled trial. J. Am. Acad. Dermatol. 2013, 69, 438–443. [Google Scholar] [CrossRef] [PubMed]
  104. Karsai, S.; Roos, S.; Raulin, C. Treatment of facial telangiectasia using a dual-wavelength laser system (595 and 1064 nm): A randomized controlled trial with blinded response evaluation. Dermatol. Surg. 2008, 34, 702–708. [Google Scholar] [CrossRef]
  105. Micali, G.; Dall’Oglio, F.; Verzi, A.E.; Luppino, I.; Bhatt, K.; Lacarrubba, F. Treatment of erythemato-telangiectatic rosacea with brimonidine alone or combined with vascular laser based on preliminary instrumental evaluation of the vascular component. Lasers Med. Sci. 2018, 33, 1397–1400. [Google Scholar] [CrossRef]
  106. Vissing, A.E.; Dierickx, C.; Karmisholt, K.E.; Haedersdal, M. Topical brimonidine reduces IPL-induced erythema without affecting efficacy: A randomized controlled trial in patients with facial telangiectasias. Lasers Surg. Med. 2018, 50, 1002–1009. [Google Scholar] [CrossRef]
  107. Sun, Y.; Chen, L.; Zhang, Y.; Gao, X.; Wu, Y.; Chen, H. Topical photodynamic therapy with 5-aminolevulinic acid in Chinese patients with Rosacea. J. Cosmet. Laser Ther. 2019, 21, 196–200. [Google Scholar] [CrossRef] [PubMed]
  108. Bao, N.; Gu, T.; Zeng, J.; Wu, Y.; Sun, Y.; Gao, X.; Chen, H. Combined therapy of 5-aminolevulinic acid photodynamic therapy and intense pulsed light for rosacea. Lasers Med. Sci. 2022, 38, 17. [Google Scholar] [CrossRef]
  109. Park, S.Y.; Kwon, H.H.; Yoon, J.Y.; Min, S.; Suh, D.H. Clinical and Histologic Effects of Fractional Microneedling Radiofrequency Treatment on Rosacea. Dermatol. Surg. 2016, 42, 1362–1369. [Google Scholar] [CrossRef]
  110. Kang, C.N.; Shah, M.; Tan, J. Rosacea: An Update in Diagnosis, Classification and Management. Skin. Ther. Lett. 2021, 26, 1–8. [Google Scholar]
  111. van Zuuren, E.J.; Fedorowicz, Z.; Tan, J.; van der Linden, M.M.D.; Arents, B.W.M.; Carter, B.; Charland, L. Interventions for rosacea based on the phenotype approach: An updated systematic review including GRADE assessments. Br. J. Dermatol. 2019, 181, 65–79. [Google Scholar] [CrossRef] [PubMed]
  112. Passi, S.; Picardo, M.; Mingrone, G.; Breathnach, A.S.; Nazzaro-Porro, M. Azelaic acid—biochemistry and metabolism. Acta Derm. Venereol. Suppl. 1989, 143, 8–13. [Google Scholar] [CrossRef]
  113. Searle, T.; Ali, F.R.; Al-Niaimi, F. The versatility of azelaic acid in dermatology. J. Dermatolog. Treat. 2022, 33, 722–732. [Google Scholar] [CrossRef] [PubMed]
  114. Gollnick, H.; Layton, A. Azelaic acid 15% gel in the treatment of rosacea. Expert Opin. Pharmacother. 2008, 9, 2699–2706. [Google Scholar] [CrossRef]
  115. Coda, A.B.; Hata, T.; Miller, J.; Audish, D.; Kotol, P.; Two, A.; Shafiq, F.; Yamasaki, K.; Harper, J.C.; Del Rosso, J.Q.; et al. Cathelicidin, kallikrein 5, and serine protease activity is inhibited during treatment of rosacea with azelaic acid 15% gel. J. Am. Acad. Dermatol. 2013, 69, 570–577. [Google Scholar] [CrossRef]
  116. King, S.; Campbell, J.; Rowe, R.; Daly, M.L.; Moncrieff, G.; Maybury, C. A systematic review to evaluate the efficacy of azelaic acid in the management of acne, rosacea, melasma and skin aging. J. Cosmet. Dermatol. 2023, 22, 2650–2662. [Google Scholar] [CrossRef]
  117. Elewski, B.E.; Fleischer, A.B.J.; Pariser, D.M. A comparison of 15% azelaic acid gel and 0.75% metronidazole gel in the topical treatment of papulopustular rosacea: Results of a randomized trial. Arch. Dermatol. 2003, 139, 1444–1450. [Google Scholar] [CrossRef]
  118. Liu, R.H.; Smith, M.K.; Basta, S.A.; Farmer, E.R. Azelaic acid in the treatment of papulopustular rosacea: A systematic review of randomized controlled trials. Arch. Dermatol. 2006, 142, 1047–1052. [Google Scholar] [CrossRef]
  119. Draelos, Z.D.; Elewski, B.; Staedtler, G.; Havlickova, B. Azelaic acid foam 15% in the treatment of papulopustular rosacea: A randomized, double-blind, vehicle-controlled study. Cutis 2013, 92, 306–317. [Google Scholar]
  120. Dall’Oglio, F.; Nasca, M.R.; Gerbino, C.; Micali, G. Advances in pharmacotherapy for rosacea: What is the current state of the art? Expert Opin. Pharmacother. 2022, 23, 1845–1854. [Google Scholar] [CrossRef]
  121. Dahl, M.V.; Jarratt, M.; Kaplan, D.; Tuley, M.R.; Baker, M.D. Once-daily topical metronidazole cream formulations in the treatment of the papules and pustules of rosacea. J. Am. Acad. Dermatol. 2001, 45, 723–730. [Google Scholar] [CrossRef] [PubMed]
  122. Dahl, M.V.; Katz, H.I.; Krueger, G.G.; Millikan, L.E.; Odom, R.B.; Parker, F.; Wolf, J.E.J.; Aly, R.; Bayles, C.; Reusser, B.; et al. Topical metronidazole maintains remissions of rosacea. Arch. Dermatol. 1998, 134, 679–683. [Google Scholar] [CrossRef] [PubMed]
  123. Yucel, A.; Yilmaz, M. Investigation of the prevalance of Demodex folliculorum and Demodex brevis in rosacea patients. Turk. Parazitol. Derg. 2013, 37, 195–198. [Google Scholar] [CrossRef] [PubMed]
  124. Rios-Yuil, J.M.; Mercadillo-Perez, P. Evaluation of Demodex folliculorum as a Risk Factor for the Diagnosis of Rosacea In Skin Biopsies. Mexico’s General Hospital (1975–2010). Indian J. Dermatol. 2013, 58, 157. [Google Scholar] [CrossRef]
  125. Stein, L.; Kircik, L.; Fowler, J.; Tan, J.; Draelos, Z.; Fleischer, A.; Appell, M.; Steinhoff, M.; Lynde, C.; Liu, H.; et al. Efficacy and safety of ivermectin 1% cream in treatment of papulopustular rosacea: Results of two randomized, double-blind, vehicle-controlled pivotal studies. J. Drugs Dermatol. 2014, 13, 316–323. [Google Scholar]
  126. Taieb, A.; Ortonne, J.P.; Ruzicka, T.; Roszkiewicz, J.; Berth-Jones, J.; Peirone, M.H.; Jacovella, J.; Ivermectin Phase, I.I.I.s.g. Superiority of ivermectin 1% cream over metronidazole 0.75% cream in treating inflammatory lesions of rosacea: A randomized, investigator-blinded trial. Br. J. Dermatol. 2015, 172, 1103–1110. [Google Scholar] [CrossRef]
  127. Green, L.J.; Bhatia, N.D.; Toledano, O.; Erlich, M.; Spizuoco, A.; Goodyear, B.C.; York, J.P.; Jakus, J. Silica-based microencapsulation used in topical dermatologic applications. Arch. Dermatol. Res. 2023, 315, 2787–2793. [Google Scholar] [CrossRef]
  128. Garrido-Mesa, N.; Zarzuelo, A.; Galvez, J. Minocycline: Far beyond an antibiotic. Br. J. Pharmacol. 2013, 169, 337–352. [Google Scholar] [CrossRef]
  129. Webster, G.; Del Rosso, J.Q. Anti-inflammatory activity of tetracyclines. Dermatol. Clin. 2007, 25, 133–135. [Google Scholar] [CrossRef]
  130. Parenti, A.; Indorato, B.; Paccosi, S. Minocycline affects human neutrophil respiratory burst and transendothelial migration. Inflamm. Res. 2017, 66, 107–109. [Google Scholar] [CrossRef]
  131. Monk, E.; Shalita, A.; Siegel, D.M. Clinical applications of non-antimicrobial tetracyclines in dermatology. Pharmacol. Res. 2011, 63, 130–145. [Google Scholar] [CrossRef]
  132. Del Rosso, J.Q.; Webster, G.; Weiss, J.S.; Bhatia, N.D.; Gold, L.S.; Kircik, L. Nonantibiotic Properties of Tetracyclines in Rosacea and Their Clinical Implications. J. Clin. Aesthet. Dermatol. 2021, 14, 14–21. [Google Scholar]
  133. Del Rosso, J.Q.; Thiboutot, D.; Gallo, R.; Webster, G.; Tanghetti, E.; Eichenfield, L.F.; Stein-Gold, L.; Berson, D.; Zaenglein, A. Consensus recommendations from the American Acne & Rosacea Society on the management of rosacea, part 3: A status report on systemic therapies. Cutis 2014, 93, 18–28. [Google Scholar] [PubMed]
  134. Del Rosso, J.Q.; Schlessinger, J.; Werschler, P. Comparison of anti-inflammatory dose doxycycline versus doxycycline 100 mg in the treatment of rosacea. J. Drugs Dermatol. 2008, 7, 573–576. [Google Scholar] [PubMed]
  135. Del Rosso, J.Q. Anti-inflamatory dose doxycycline in the treatment of rosacea. J. Drugs Dermatol. 2009, 8, 664–668. [Google Scholar]
  136. Del Rosso, J.Q. Effectiveness and safety of doxycycline 40 mg (30-mg immediate-release and 10-mg delayed-release beads) once daily as add-on therapy to existing topical regimens for the treatment of papulopustular rosacea: Results from a community-based trial. Cutis 2010, 86, 16–25. [Google Scholar] [PubMed]
  137. van der Linden, M.M.D.; van Ratingen, A.R.; van Rappard, D.C.; Nieuwenburg, S.A.; Spuls, P.I. DOMINO, doxycycline 40 mg vs. minocycline 100 mg in the treatment of rosacea: A randomized, single-blinded, noninferiority trial, comparing efficacy and safety. Br. J. Dermatol. 2017, 176, 1465–1474. [Google Scholar] [CrossRef]
  138. James, K.A.; Burkhart, C.N.; Morrell, D.S. Emerging drugs for acne. Expert Opin. Emerg. Drugs 2009, 14, 649–659. [Google Scholar] [CrossRef]
  139. El-Heis, S.; Buckley, D.A. Rosacea-like eruption due to topical pimecrolimus. Dermatol. Online J. 2015, 21. [Google Scholar] [CrossRef]
  140. Gomolin, T.; Cline, A.; Pereira, F. Treatment of rosacea during pregnancy. Dermatol. Online J. 2021, 27, 7. [Google Scholar] [CrossRef]
  141. Lova Navarro, M.; Sanchez-Pedreno Guillen, P.; Victoria Martinez, A.M.; Martinez Menchon, T.; Corbalan Velez, R.; Frias Iniesta, J. Papulopustular Rosacea: Response to Treatment with Oral Azithromycin. Actas Dermosifiliogr. 2018, 109, 529–535. [Google Scholar] [CrossRef] [PubMed]
  142. Torresani, C.; Pavesi, A.; Manara, G.C. Clarithromycin versus doxycycline in the treatment of rosacea. Int. J. Dermatol. 1997, 36, 942–946. [Google Scholar] [CrossRef] [PubMed]
  143. Del Rosso, J.Q.; Kim, G. Optimizing use of oral antibiotics in acne vulgaris. Dermatol. Clin. 2009, 27, 33–42. [Google Scholar] [CrossRef]
  144. Sbidian, E.; Vicaut, E.; Chidiack, H.; Anselin, E.; Cribier, B.; Dreno, B.; Chosidow, O. A Randomized-Controlled Trial of Oral Low-Dose Isotretinoin for Difficult-To-Treat Papulopustular Rosacea. J. Investig. Dermatol. 2016, 136, 1124–1129. [Google Scholar] [CrossRef]
  145. Gollnick, H.; Blume-Peytavi, U.; Szabo, E.L.; Meyer, K.G.; Hauptmann, P.; Popp, G.; Sebastian, M.; Zwingers, T.; Willers, C.; von der Weth, R. Systemic isotretinoin in the treatment of rosacea—Doxycycline- and placebo-controlled, randomized clinical study. J. Dtsch. Dermatol. Ges. 2010, 8, 505–515. [Google Scholar] [CrossRef] [PubMed]
  146. Spring, L.K.; Krakowski, A.C.; Alam, M.; Bhatia, A.; Brauer, J.; Cohen, J.; Del Rosso, J.Q.; Diaz, L.; Dover, J.; Eichenfield, L.F.; et al. Isotretinoin and Timing of Procedural Interventions: A Systematic Review with Consensus Recommendations. JAMA Dermatol. 2017, 153, 802–809. [Google Scholar] [CrossRef] [PubMed]
  147. Kwon, H.H.; Jung, J.Y.; Lee, W.Y.; Bae, Y.; Park, G.H. Combined treatment of recalcitrant papulopustular rosacea involving pulsed dye laser and fractional microneedling radiofrequency with low-dose isotretinoin. J. Cosmet. Dermatol. 2020, 19, 105–111. [Google Scholar] [CrossRef] [PubMed]
  148. Kumar, A.M.; Chiou, A.S.; Shih, Y.H.; Li, S.; Chang, A.L.S. An exploratory, open-label, investigator-initiated study of interleukin-17 blockade in patients with moderate-to-severe papulopustular rosacea. Br. J. Dermatol. 2020, 183, 942–943. [Google Scholar] [CrossRef]
  149. Saal, R.C.; Borda, L.J.; Hoffman, M.L.; Roberts, A.A.; Van Voorhees, A.S. Treatment of granulomatous rosacea with adalimumab. JAAD Case Rep. 2023, 40, 89–91. [Google Scholar] [CrossRef]
  150. Sun, Y.H.; Man, X.Y.; Xuan, X.Y.; Huang, C.Z.; Shen, Y.; Lao, L.M. Tofacitinib for the treatment of erythematotelangiectatic and papulopustular rosacea: A retrospective case series. Dermatol. Ther. 2022, 35, e15848. [Google Scholar] [CrossRef]
  151. Searle, T.; Al-Niaimi, F.; Ali, F.R. Rosacea. Br. J. Hosp. Med. 2021, 82, 1–8. [Google Scholar] [CrossRef] [PubMed]
  152. Gonzalez, L.F.; Herrera, H.; Motta, A. Electrosurgery for the Treatment of Moderate or Severe Rhinophyma. Actas Dermosifiliogr. 2018, 109, e23–e26. [Google Scholar] [CrossRef] [PubMed]
  153. Rohrich, R.J.; Griffin, J.R.; Adams, W.P.J. Rhinophyma: Review and update. Plast. Reconstr. Surg. 2002, 110, 860–869; quiz 870. [Google Scholar] [CrossRef]
  154. Borhani-Khomani, K.; Moller, M.P.; Thomsen, M.V.; Karmisholt, K.; Haedersdal, M.; Bonde, C.T. Treatmentof rhinophyma with laser and surgery. Ugeskr. Laeger 2020, 182, V07190378. [Google Scholar] [PubMed]
  155. Ismail, D.; Asfour, L.; Madan, V. Rhinophyma in women: A case series. Lasers Med. Sci. 2021, 36, 1283–1287. [Google Scholar] [CrossRef]
  156. Baldwin, H.E. Systemic therapy for rosacea. Skin. Ther. Lett. 2007, 12, 9. [Google Scholar]
  157. Malagon-Liceaga, A.; Recillas-Gispert, C.; Ruiz-Quintero, N.C.; Ruelas-Villavicencio, A.L. Treatment of ocular rosacea: A practical review from an interdisciplinary approach. Arch. Soc. Esp. Oftalmol. 2023, 98, 577–585. [Google Scholar] [CrossRef]
  158. Pfeffer, I.; Borelli, C.; Zierhut, M.; Schaller, M. Treatment of ocular rosacea with 40 mg doxycycline in a slow release form. J. Dtsch. Dermatol. Ges. 2011, 9, 904–907. [Google Scholar] [CrossRef]
  159. McGhee, C.N.; Dean, S.; Danesh-Meyer, H. Locally administered ocular corticosteroids: Benefits and risks. Drug Saf. 2002, 25, 33–55. [Google Scholar] [CrossRef]
  160. Vora, G.K.; Gupta, P.K. Intense pulsed light therapy for the treatment of evaporative dry eye disease. Curr. Opin. Ophthalmol. 2015, 26, 314–318. [Google Scholar] [CrossRef]
  161. Calabrese, L.; Malvaso, D.; Chiricozzi, A.; Tambone, S.; D’Urso, D.F.; Guerriero, C.; Peris, K. Baricitinib: Therapeutic potential for moderate to severe atopic dermatitis. Expert Opin. Investig. Drugs 2020, 29, 1089–1098. [Google Scholar] [CrossRef] [PubMed]
  162. Calabrese, L.; Chiricozzi, A.; De Simone, C.; Fossati, B.; D’Amore, A.; Peris, K. Pharmacodynamics of Janus kinase inhibitors for the treatment of atopic dermatitis. Expert Opin. Drug Metab. Toxicol. 2022, 18, 347–355. [Google Scholar] [CrossRef] [PubMed]
  163. D’Urso, D.F.; Chiricozzi, A.; Pirro, F.; Calabrese, L.; Caldarola, G.; Fossati, B.; De Simone, C.; Peris, K. New JAK inhibitors for the treatment of psoriasis and psoriatic arthritis. G Ital. Dermatol. Venereol. 2020, 155, 411–420. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic illustration of the main factors playing a role in the pathogenesis of rosacea.
Figure 1. Schematic illustration of the main factors playing a role in the pathogenesis of rosacea.
Cosmetics 11 00011 g001
Figure 2. Erythemato-telangiectatic rosacea (a) is characterized by flushing, persistent centrofacial erythema, and telangiectasias. Papulopustular rosacea (b) is defined by erythema with a centrofacial distribution, with typically small (<3 mm) erythematous papules and/or pustules, both occurring singly or in clusters and at different stages of development. In more severe forms, inflammatory plaques or oedema may be present. Hypertrophy of the sebaceous glands and connective tissue is the hallmark of phymatous rosacea (c). The predominant form is rhinophyma, which occurs mainly in males and results in skin thickening with a nodular appearance, along with telangiectasias. This subtype may involve the chin (gnathophyma), forehead (metophyma), ears (otophyma), and eyelids (blepharophyma). Ocular rosacea (d), which may overlap with other subtypes, causes telangiectasia of the eyelid margin, conjunctival injection, blepharitis, and occasionally ectropion.
Figure 2. Erythemato-telangiectatic rosacea (a) is characterized by flushing, persistent centrofacial erythema, and telangiectasias. Papulopustular rosacea (b) is defined by erythema with a centrofacial distribution, with typically small (<3 mm) erythematous papules and/or pustules, both occurring singly or in clusters and at different stages of development. In more severe forms, inflammatory plaques or oedema may be present. Hypertrophy of the sebaceous glands and connective tissue is the hallmark of phymatous rosacea (c). The predominant form is rhinophyma, which occurs mainly in males and results in skin thickening with a nodular appearance, along with telangiectasias. This subtype may involve the chin (gnathophyma), forehead (metophyma), ears (otophyma), and eyelids (blepharophyma). Ocular rosacea (d), which may overlap with other subtypes, causes telangiectasia of the eyelid margin, conjunctival injection, blepharitis, and occasionally ectropion.
Cosmetics 11 00011 g002
Table 1. Key aspects of a phenotype-based approach in rosacea.
Table 1. Key aspects of a phenotype-based approach in rosacea.
Rosacea SubtypeFirst Line TherapiesAlternatives
Erythemato-telangectatic
  • Topical brimonidine tartrate 0.5% gel or oxymetazoline hydrochloride 1% cream for persistent facial erythema.
  • IPL, PDL, and Nd:YAG laser for persistent facial erythema and telangectasia.
  • Topical metronidazole, azelaic acid, or calcineurin inhibitors in the case of co-existent inflammatory lesions.
  • Oral non-selective β-blockers carvedilol or α2-adrenergic agonist clonidine to decrease erythema and flushing.
  • Intradermal injection of botulinum toxin A in resistant conditions.
Papulopustular
  • Topic azelaic acid (15% gel or 20% cream and 15% foam), metronidazole (0.75% gel, cream, and lotion and 1% cream and gel), or ivermectin (1% cream).
  • Oral subantibiotic-dose doxycycline 40 mg modified release capsule. Alternatively, tetracycline and minocycline.
  • Intermittent courses of oral low-dose isotretinoin (0.2 mg/kg/day) in the case of antibiotic failure or to avoid long-term therapies.
  • Minocycline 1.5% foam, topical retinoids (adapalene 0.1% gel and tretinoin 0.025% cream) and topical antibacterial (clindamycin, erythromycin. and benzoyl peroxide) for more severe or recalcitrant rosacea.
  • Erythromycin, azithromycin, and clarithromycin in patients not eligible for tetracyclines.
  • Nd:YAG laser for persistent or severe cases.
Phymatous
  • Nasal debulking through surgical approaches or laser ablation.
  • Consider oral isotretinoin to prevent relapse after surgery.
Ocular
  • Proper lid hygiene and ocular lubricants.
  • In the case of persistent symptoms, refer to ophthalmology.
  • Cyclosporine, azithromycin, or tacrolimus as topical agents.
  • Oral subantibiotic-dose doxycycline.
  • IPL for evaporative dry eye disease with meibomian gland dysfunction.
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

Galluccio, G.; D’Onghia, M.; Malvaso, D.; Lazzeri, L.; Cinotti, E.; Rubegni, G.; Rubegni, P.; Calabrese, L. Advances in the Pathogenesis and Treatment of Rosacea: A Phenotype-Based Therapeutic Approach. Cosmetics 2024, 11, 11. https://doi.org/10.3390/cosmetics11010011

AMA Style

Galluccio G, D’Onghia M, Malvaso D, Lazzeri L, Cinotti E, Rubegni G, Rubegni P, Calabrese L. Advances in the Pathogenesis and Treatment of Rosacea: A Phenotype-Based Therapeutic Approach. Cosmetics. 2024; 11(1):11. https://doi.org/10.3390/cosmetics11010011

Chicago/Turabian Style

Galluccio, Giulia, Martina D’Onghia, Dalma Malvaso, Laura Lazzeri, Elisa Cinotti, Giovanni Rubegni, Pietro Rubegni, and Laura Calabrese. 2024. "Advances in the Pathogenesis and Treatment of Rosacea: A Phenotype-Based Therapeutic Approach" Cosmetics 11, no. 1: 11. https://doi.org/10.3390/cosmetics11010011

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