Next Article in Journal
Nicotinamide Mononucleotide Supplementation Alleviates Doxorubicin-Induced Multi-Organ Fibrosis
Previous Article in Journal
AFB1 Toxicity in Human Food and Animal Feed Consumption: A Review of Experimental Treatments and Preventive Measures
Previous Article in Special Issue
Investigation into the Anti-Acne Effects of Castanea sativa Mill Leaf and Its Pure Ellagitannin Castalagin in HaCaT Cells Infected with Cutibacterium acnes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring Acne Treatments: From Pathophysiological Mechanisms to Emerging Therapies

1
Department of Dermatology, International St. Mary’s Hospital, College of Medicine, Catholic Kwandong University, Incheon 22711, Republic of Korea
2
Department of Dermatology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(10), 5302; https://doi.org/10.3390/ijms25105302
Submission received: 22 March 2024 / Revised: 8 May 2024 / Accepted: 10 May 2024 / Published: 13 May 2024

Abstract

:
Acne vulgaris is a common dermatological condition that can present across different ages but predominantly affects adolescents and young adults. Characterized by various lesion types, the pathogenesis of acne is complex, involving genetic, hormonal, microbial, and inflammatory factors. This review comprehensively addresses current and emerging acne management strategies, emphasizing both topical and systemic treatments, procedural therapies, and dietary modifications. Key topical agents include retinoids, benzoyl peroxide, antibiotics, and other specialized compounds. Systemic options like antibiotics, hormonal therapies, and retinoids offer significant therapeutic benefits, particularly for moderate to severe cases. Procedural treatments such as laser devices, photodynamic therapy, chemical peels, and intralesional injections present viable alternatives for reducing acne symptoms and scarring. Emerging therapies focus on novel biologics, bacteriophages, probiotics, and peptides, providing promising future options. This review underscores the importance of personalized approaches to treatment due to the multifaceted nature of acne, highlighting the potential of innovative therapies for improving patient outcomes.

1. Introduction

Acne, a prevalent dermatological condition, affects individuals across all ages, though it predominantly occurs in adolescents and young adults. Commonly, lesions appear on the face, neck, upper back, and chest. Acne manifests in several forms, including neonatal and infantile acne, occupational acne, acne vulgaris, acne conglobata, acne fulminans, acne mechanica, acne excoriee (picker’s acne), chloracne, and acne medicamentosa. Of these, acne vulgaris is the most common, comprising 99% of cases [1,2]. It is characterized by distinct lesion types, categorized as non-inflammatory and inflammatory. Non-inflammatory lesions include both open comedones (blackheads) and closed comedones (whiteheads). Inflammatory lesions include papules, pustules, nodules, and cysts [3].
The pathogenesis of acne is multifactorial, involving sebaceous gland hyperactivity, follicular hyperkeratinization, microbial colonization (particularly by Cutibacterium acnes), and a consequent inflammatory response [4,5,6]. The interplay of these factors, influenced by genetic, hormonal, and environmental variables, underpins the complexity of acne management strategies.
Current treatment modalities for acne are diverse, reflecting the multifaceted nature of the condition. Treatments range from topical agents, such as retinoids and benzoyl peroxide, to systemic therapies, including antibiotics and hormonal agents. The choice of treatment is dictated by the acne type, with milder forms often managed with topical agents and more severe forms requiring systemic treatment. Recent advances have also explored the roles of diet, the gut microbiome, and novel pharmacological targets in managing acne, suggesting a move toward more personalized and effective treatment protocols. This article aims to elucidate the pathophysiological mechanisms of acne and to explore the array of conventional and emerging treatment strategies. This comprehensive review highlights the complexity of acne management and underscores the importance of personalized therapeutic approaches in contemporary dermatological practice.

2. Pathophysiology of Acne

The pathophysiology of acne is complex, making it a multifaceted condition that varies significantly across individuals (Figure 1).

2.1. Genetic and Environmental Factors

2.1.1. Genetic Factors

Genetics plays a crucial role in acne development, with studies indicating a higher prevalence of acne in individuals with a family history of the condition [7,8,9,10,11,12,13,14,15,16,17,18,19,20]. Genes influence several aspects of acne pathogenesis, including sebum production, keratinization processes, and the immune system’s response to C. acnes [21,22,23,24,25,26]. For instance, variations in genes related to the androgen receptor can lead to increased sebum production, a key factor in acne development [23]. Similarly, genetic factors can influence the body’s inflammatory response, contributing to the formation and severity of acne lesions [21,27].

2.1.2. Environmental Factors

Environmental factors are equally influential in the pathogenesis of acne. These include diet, climate, pollution, and lifestyle. Dietary habits, particularly the consumption of high-glycemic-index foods and dairy products, have been linked to acne severity, possibly due to their role in insulin-like growth factor 1 (IGF-1) signaling, which can exacerbate sebum production and inflammation [28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43]. Climate factors, such as humidity and temperature, can affect skin hydration and sebum production, while air pollutants such as NO2 and sulfur dioxide can increase the oxidative stress on the skin, contributing to acne flares [44,45]. Increased humidity cause epidermal keratinocyte swelling, leading to acute hair follicle occlusion, which is a favorable environment for C. acnes [44]. Increased sebum production following an increase in temperature also promotes the growth of lipophilic organisms such as C. acnes [46]. Additionally, stress has been recognized to exacerbate acne through hormonal fluctuations that increase sebum production [47].

2.2. Hormonal Influences

The pathophysiology of acne is profoundly influenced by hormonal factors, particularly those that alter sebaceous gland function. Androgens are pivotal in stimulating sebocyte proliferation, increasing intracellular lipid droplets, and inducing hyperkeratinization of the follicular infrainfundibulum, leading to increased sebum production and acne severity [48]. Hyperseborrhea and dysseborrhea are primary pathological conditions that have been identified, with the former indicating excessive sebum production and the latter changes in sebum composition, both of which contribute to C. acnes proliferation, inflammation, and comedogenesis [49].
Androgens stimulate lipid synthesis and sebocyte differentiation by enhancing the phosphorylation of mammalian target of rapamycin (mTOR), a central player in cellular metabolism, and by activating sterol regulatory-element-binding protein-1 (SREBP-1) via mTORC1, thus promoting lipogenesis [50,51]. Additionally, androgens downregulate the Wnt/β-catenin pathway, upregulating genes like c-MYC and enhancing sebocyte differentiation [52,53,54].
IGF-1 is another crucial hormonal driver, influencing acne by downregulating the nuclear transcription factor forkhead box protein O1 (FoxO1), which, in turn, increases lipogenesis and androgen receptor transduction [5,55,56]. IGF-1 signaling, critical in acne development, regulates the PI3K/Akt cascade and FoxO1 activity, impacting lipogenesis and androgen receptor (AR) transactivation and influencing lipid synthesis and sebaceous secretion [57,58,59,60,61,62,63,64,65].
Neuropeptides, including corticotropin-releasing hormone (CRH), substance P (SP), and the pro-opiomelanocortin (POMC) system, also play a significant role by modulating sebocyte activity and inflammatory responses [48,66,67]. CRH, for instance, is highly expressed in acne-prone skin, supporting a mechanism for stress-induced acne, and it interacts with androgenetic signaling pathways involved in acne pathogenesis [48].
In addition to local hormonal effects on sebaceous glands, systemic hormonal disorders play a crucial role in triggering adult acne. The onset of adult acne often necessitates an investigation into potential underlying endocrine disorders, marked by symptoms of virilization and hormonal imbalances. Clinical signs such as irregular menstrual cycles, clitoral hypertrophy, hirsutism, female-pattern hair loss, and sudden-onset or resistant acne may indicate systemic hyperandrogenism [68]. Conditions like obesity, infertility, metabolic syndrome, and hypothyroidism also merit attention for their association with hormonal disorders. Diagnostic efforts should extend to adrenal hyperplasia, virilizing tumors, and polycystic ovary syndrome (PCOS) in patients with abnormal laboratory findings [69]. Hormonal evaluations, recommended during the early follicular phase of the menstrual cycle, should encompass tests for free and total testosterone, DHEA-S, androstenedione, sex-hormone-binding globulin (SHBG), prolactin, and cortisol [68]. Findings of elevated anti-Müllerian hormone or DHEAS and low SHBG levels may indicate peripheral hyperandrogenism, underscoring the need for a comprehensive hormonal assessment in adult female acne management [68].

2.3. Microbiome

The skin microbiome, including C. acnes, coagulase-negative Staphylococcus, and fungal species, plays a role in maintaining skin health by balancing homeostatic relationships [70]. While the proliferation of C. acnes has been associated with acne, it is the diversity and specific phylotypes of C. acnes that are more directly implicated in acne development [71]. C. acnes is distinguished by unique genomic elements linked to acne pathogenesis [72]. Different strains of C. acnes can induce varying immune responses; acne-associated strains trigger pro-inflammatory cytokines like IFNγ and IL-17, whereas health-associated strains promote the production of anti-inflammatory IL-10 [70].
Microbial dysbiosis, particularly involving C. acnes and Staphylococcus epidermidis, contributes to acne by influencing innate immunity pathways such as PPAR and IFN signaling and TNF, IL, TLR, and MMP activation, leading to immune dysregulation, epithelial dysfunction, and overgrowth of pathogenic microbes [70,73]. C. acnes plays a pivotal role in acne pathogenesis by interacting with innate immunity through the release of extracellular enzymes and reactive oxygen species and the activation of TLRs, alongside influencing sebum production, keratin, filaggrin, and IGF-1 levels. This leads to inflammation and hyperkeratosis, which maintains inflammation through acquired cell-mediated responses via Th1 cells [74].
Increasing evidence also supports an interplay between the skin and gut microbiomes, enriching our understanding of acne pathogenesis beyond the skin’s surface. The intricate ecosystem within the human gut, encompassing a diverse array of bacteria, fungi, viruses, and protozoa, plays a critical role in immune modulation, nutrient synthesis, and defense against pathogens [4,75,76,77]. Dominated primarily by the bacterial phyla Firmicutes and Bacteroidetes, the gut microbiome varies significantly among individuals due to diet, antibiotics, and lifestyle influences [75,78]. This variation is implicated in the development of acne, as alterations in the gut microbiota, known as gut dysbiosis, have been observed in acne patients, characterized by reduced diversity and shifts in the balance of Bacteroidetes to Firmicutes, reflecting dietary patterns associated with a Western diet [73,79,80].
This connection between the gut and skin extends beyond mere coexistence, suggesting direct influences whereby gut microbes and their metabolic products can impact the skin microbiome via the bloodstream [76]. Such microbial interactions and the resultant modulation of cell expansion and metabolism, particularly through the mTOR signaling pathway, underscore a deeper link between gut dysbiosis and acne inflammation [51,76,81]. Furthermore, the role of microbial metabolites in systemic immune regulation is highlighted by the actions of specific gut bacteria, such as Bacteroides fragilis, which produce anti-inflammatory compounds, contrasting with others that might enhance inflammation, underscoring the complexity of the gut–skin axis in acne pathogenesis [76,77]. Emotional stress has been shown to alter the gut microbiome, leading to systemic and skin inflammation, further exacerbating acne [76,77]. Furthermore, gut dysbiosis can influence acne through changes in sebum composition and the promotion of pathogenic C. acnes strains, highlighting the role of the gut microbiome in acne pathogenesis [82].

2.4. Inflammation

The pathogenesis of acne is characterized by complex interactions between the innate and adaptive immune systems, where C. acnes plays a pivotal role. C. acnes triggers the activation of the Toll-like receptors TLR-2 and TLR-4 on sebocytes and keratinocytes, initiating the release of pro-inflammatory cytokines such as IL-6, IL-8, and IL-12 from monocytes, markers that are notably elevated in acne lesions [83,84]. Beyond Toll-like receptor activation, C. acnes also stimulates the Nod-like receptor 3 (NLRP3) inflammasome in monocytic cells, promoting the increased secretion of pro-inflammatory IL-1β [85].
The role of adaptive immunity is also critical in acne. Early acne lesions are characterized by a significant presence of CD4+ T-helper cells. The presence of CD4(+) lymphocytes around follicles in early lesions indicates that inflammation precedes hyperkeratinization [86]. This early inflammatory phase is marked by macrophages and CD3(+) and CD4(+) cells, along with activated vascular adhesion molecules like E-selectin and integrin, emphasizing inflammation’s primacy role in acne development [87]. C. acnes not only drives the proliferation of T-cells but also leads to the accumulation of C. acnes-specific T-cells [88]. These cells are induced to release IFN-γ and IL-17A, fostering a combined Th17 and Th1/Th17 host response that contributes to the inflammatory landscape of acne [85,88].
Moreover, C. acnes’s activation of activator protein 1 (AP1) and nuclear factor (NF)-κB leads to the production of matrix metalloproteinases (MMPs), TNFα, IL-1β, and IL-8 [89]. Elevated IL-17, IL-23, and TNFα levels in acne lesions, coupled with TLR2(+) macrophages and IL-17 cells, reveal a complex network of inflammatory pathways that exacerbate acne severity [85,90]. In the latter stages, the dominance of memory T cells, plasma cells, and B cells in atrophic scars indicates sustained immune responses [91,92]. C. acnes’s enzymatic activity, including lipase, hyaluronidase, and protease activities, potentially contributes to follicular wall breakdown, leading to extensive inflammation, foreign body granulomas, and scarring [93].
Additionally, IGF-1 elevates pro-inflammatory cytokine levels in primary human sebocytes, an effect attenuated by NF-κB inhibitors [94,95]. This suggests IGF-1’s significant role in initiating inflammation within the pilosebaceous unit. Androgens, by raising serum IGF-1 levels, enhance the recruitment of inflammatory cells and cytokines and MMP release [96,97]. MMPs disrupt the follicular membrane, exacerbating inflammation [98]. Furthermore, neutrophils mediate inflammation by increasing hydrogen peroxide generation, underscoring the intricate interplay of immune cells in acne pathogenesis [99].

3. Current Treatment Strategies

Current treatment strategies for acne focus on addressing the various factors contributing to its pathogenesis, including abnormal keratinization, increased sebum production, microbial colonization, and inflammation.

3.1. Topical Treatments

Topical treatments remain a cornerstone of acne management due to their direct action on the skin, minimal systemic side effects, and ease of use. This section reviews the efficacy and mechanisms of action of key topical agents, including retinoids, benzoyl peroxide, antibiotics, and other topical agents (Table 1, Figure 2).

3.1.1. Topical Retinoids

Retinoids exert their effects through their binding to retinoic acid receptors (RARs) in the cell nucleus [100]. There are three RAR subtypes: RAR-α, RAR-β, and RAR-γ; of these, RAR-γ is the most highly expressed in human skin [101]. Retinoids are categorized into four generations based on their molecular structures and specific receptor affinities, with the latest generation progressively reducing toxicity while enhancing efficacy [102,103]. Additionally, topical retinoids are classified into six classes: tretinoin (all-trans retinoic acid), adapalene, tazarotene, trifarotene, alitretinoin, and bexarotene [102]. Among these, the United States of America Food and Drug Administration (FDA) has approved four topical retinoids (tretinoin, adapalene, tazarotene, trifarotene) for acne treatment. Topical retinoids are a cornerstone of acne therapy due to their comedolytic and anti-inflammatory effects. These vitamin A derivatives bind to retinoic acid receptors in skin cells, influencing gene transcription to increase skin cell turnover and reduce oil production [104,105]. Topical therapies offer the advantage of direct application to the affected area, minimizing potential systemic side effects and increasing exposure within the pilosebaceous follicular unit [3].
  • First-Generation Retinoids
Tretinoin became the first topical retinoid to be approved for the treatment of acne by the FDA in 1971 [101]. Tretinoin has a high affinity for all three retinoic acid receptor subtypes, but its binding to RAR-γ is key to its effects [101]. Tretinoin comes in various strengths and formulations (gel, cream, microsphere, lotion) [106]. It functions to normalize the epithelial layer, prevent pilosebaceous unit occlusion, and reduce sebum production [107]. While effective, it can increase sun sensitivity, and potential side effects include redness, dryness, itching, and irritation [108,109].
b.
Second-Generation Retinoids: There are no available second-generation topical formulations of retinoids.
c.
Third-Generation Retinoids
Topical adapalene 0.1% was approved for the treatment of acne in 1996 [101]. Adapalene binds selectively to RAR-β and RAR-γ, but it acts primarily via RAR-γ [101]. A meta-analysis of five clinical studies involving 900 patients with mild to moderate acne compared adapalene 0.1% gel and tretinoin 0.025% gel. After 12 weeks, both showed similar efficacy in reducing lesion counts, but adapalene had better local tolerability [110]. Topical adapalene reduces hyperkeratinization of pilosebaceous follicles and acne-related inflammation, with minimal side effects like redness and irritation [111].
Topical tazarotene gel (0.1%) was approved for treatment of mild to moderate acne in 1997 [101]. Like adapalene, tazarotene binds selectively to RAR-β and RAR-γ. Available in gel, cream, and foam, tazarotene offers greater potency but also potentially greater skin irritation [106,112,113]. A new lotion formulation (0.045%) aims to improve tolerability [106,112,113]. Often prescribed when tretinoin or adapalene are ineffective, tazarotene also has anti-inflammatory properties and targets hyperkeratinization and C. acnes proliferation [114,115]. It may be combined with benzoyl peroxide or antibiotics for enhanced inflammatory acne treatment, though skin irritation and redness are possible side effects [116].
Several studies comparing tazarotene, tretinoin, and adapalene for acne treatment have shown varied results. A trial comparing tazarotene 0.1% gel with tretinoin 0.025% gel found that tazarotene was more effective in reducing non-inflammatory lesion counts but caused more peeling and burning (p < 0.05) [117]. In a 12-week trial, tazarotene 0.1% gel had higher treatment success than adapalene 0.1% gel but also caused more peeling, burning, and erythema [118]. A retrospective analysis of seven studies revealed that tazarotene 0.1% gel had greater significant improvements than adapalene 0.1% gel and tretinoin 0.025% gel [119]. Lastly, a systematic review concluded that tretinoin works faster than tazarotene in reducing inflammatory lesions, while adapalene is better tolerated [104]. Despite these comparisons, factors like concentration and formulation affect tolerability and efficacy, with adapalene often being the most tolerated but least effective and tazarotene being the most effective but least tolerated.
d.
Fourth-Generation Retinoids
Approved by the FDA in 2019 for acne treatment, trifarotene is the most recently approved topical retinoid [101]. Trifarotene is a fourth-generation topical retinoid with selectivity toward the RAR-γ located in the epidermis [103,104,120]. In the 12-week, double-blind, phase III PERFECT 1 and PERFECT 2 studies on 2420 patients with moderate facial and truncal acne, once-daily trifarotene 0.005% cream significantly improved success rates and reduced inflammatory and non-inflammatory lesions compared with the vehicle (p < 0.05) [121]. However, common adverse events, including application-site irritation (7.5% vs. 0.3%) and pruritus (2.4% vs. 0.8%), persisted [101]. Despite its potent, selective binding to the RAR-γ receptor, trifarotene did not fully eliminate the skin irritation often associated with retinoids. Although clinical trials demonstrated that trifarotene 0.005% cream significantly reduces lesions, comparative studies with currently available topical retinoids are yet to be conducted [106]. Theoretically, trifarotene’s RAR-γ selectivity might reduce adverse effects compared with existing topical retinoids; however, further studies are necessary to fully determine its optimal role in clinical practice.
Common side effects of topical retinoids include dryness, irritation, burning, and increased sun sensitivity [109]. To reduce these effects, patients should consider starting with a lower-strength formulation and gradually increasing it, applying the medication every other day or using short-contact therapy (removing the product after a short period) [122,123]. Additionally, the consistent use of moisturizers and sunscreen is essential. It is important to note that acne may initially flare up during the first few weeks of treatment [124]. While topical retinoids are the primary first-line treatment for both non-inflammatory and inflammatory acne, it can take over three months to see the full benefits of the therapy [3].
Topical retinoids are generally not recommended during pregnancy due to the potential risk of fetal malformations [124]. Better-studied options for pregnant patients include azelaic acid or topical clindamycin [124]. Tazarotene, in particular, should be avoided [124].
Research continues to focus on developing new formulations that improve tolerability and potentially increase effectiveness, such as tretinoin 0.05% lotion and tazarotene 0.045% lotion [106,112,113,125].
To maximize outcomes, topical retinoids are often combined with other acne treatments, including benzoyl peroxide and topical antibiotics to address the inflammatory component of acne, and hydrating agents (e.g., Altreno®) to combat the common side effect of dryness [109,126]. As shown in Table 2, tretinoin has been combined with clindamycin for the treatment of acne [102]. The tretinoin 0.1–benzoyl peroxide 3% cream (Twyneo®) received FDA approval as a once-daily application for moderate to severe acne vulgaris but has not yet been approved in the EU [127]. Also, adapalene has been formulated with benzoyl peroxide for treating acne. However, since both retinoids and benzoyl peroxide can cause skin irritation and dryness, combining them can intensify these effects. Gradual titration of such combination products may improve skin tolerance over time [102]. In 2023, FDA approved CabtreoTM (topical clindamycin phosphate 1.2%/adapalene 0.15%/benzoyl peroxide (BPO) 3.1%), the first triple-combination drug for acne [128]. Acne lesion reductions were significantly greater with clindamycin phosphate 1.2%/adapalene 0.15%/BPO 3.1% gel versus its dyads (BPO/adapalene; clindamycin/BPO; and clindamycin/adapalene) and vehicle gel by as early as week 4; however, it has not yet been approved in the EU. Topical 3% minocycline/0.3% adapalene foam is a combination in development for acne [129]. In the 12-week, double-blind, phase III PERFECT 1 and PERFECT 2 studies involving 2420 patients with moderate to severe acne, a combination of 3% minocycline and 0.3% adapalene significantly improved success rates, inflammatory lesion counts, and non-inflammatory lesion counts compared with the vehicle [129].

3.1.2. Topical Benzoyl Peroxide

Topical benzoyl peroxide (BPO) is a widely used first-line antimicrobial treatment for mild to moderate acne [130]. It penetrates the skin and enters the pilosebaceous unit, where it generates free radicals that disrupt the cell walls of C. acnes bacteria [131]. BPO also exhibits mild comedolytic and anti-inflammatory properties and helps prevent the development of bacterial resistance to antibiotics—often offering superior results compared with topical antibiotics alone [132,133,134].
BPO is available as a gel, cream, or wash. Studies indicate that 2.5%, 5%, and 10% formulations are similarly effective, though higher concentrations increase the risk of irritant dermatitis [130]. Typically applied once daily, acne improvement may be seen within 5 days, with more noticeable results after 3 weeks and maximum lesion reduction after 8–12 weeks [130,135].
Common side effects include xerosis (dryness), scaling, erythema (redness), and hypersensitivity reactions [130,135]. BPO can also increase transepidermal water loss, affecting the skin’s barrier function [136]. Since continued use is needed for sustained effects, side effects typically lessen with reduced frequency or lower concentrations [135]. Importantly, patients should be aware that benzoyl peroxide can stain or bleach fabrics.
Combining BPO with other agents like topical retinoids or antibiotics enhances its efficacy in acne treatment [130]. Also, BPO helps reduce bacterial resistance when combined with antibiotics as a result of the bactericidal activity of BPO against antibiotic-resistant C. acnes [137]. For example, Benzamycin combines 3% erythromycin with 5% BPO in a topical gel, offering greater effectiveness and similar tolerability compared with BPO alone [138]. Also, the combination of clindamycin and benzoyl peroxide (BPO) has shown greater efficacy compared with using topical antibiotics alone, with 90% of patients using the combination reporting improvement at 12 weeks compared with 45% of those using clindamycin alone [132,133,134].

3.1.3. Topical Antibiotics

Topical antibiotics are a key component in treating acne, offering both anti-inflammatory and antibacterial effects. They are particularly effective against inflammatory lesions but also reduce biofilm formation and subsequent microcomedones [139]. The American Academy of Dermatology (AAD) recommends topical antibiotics as a first-line treatment for mild acne and in combination with BPO or retinoids to combat antibiotic resistance [135].
Three topical antibiotics have FDA approval for acne treatment in both children and adults:
(a)
Clindamycin: Available in various concentrations and formulations (lotions, foams, gels) [126]. To minimize resistance, clindamycin is frequently combined with BPO (BenzaClin, DUAC®) or tretinoin (Veltin) [126]. While generally well tolerated, dryness and skin irritation are possible side effects.
(b)
Erythromycin: A possible alternative to clindamycin. However, concerns exist about higher rates of C. acnes resistance to topical erythromycin compared with clindamycin [126]. Therefore, it should be combined with other agents like BPO (Benzamycin®). Erythromycin is available as a monotherapy (gels, swabs, solutions). It is generally well tolerated, but can cause skin irritation [126].
(c)
Minocycline: A tetracycline derivative. Topical minocycline 4% foam (Amzeeq™) demonstrates greater effectiveness than its vehicle alone [140,141]. This lipophilic formulation readily moves into the pilosebaceous unit [142]. While its precise mechanism is not fully understood, its potent antibacterial effects are well documented [142]. Studies indicate effectiveness in improving acne within 12 weeks, with continued improvement observed at 52 weeks [142,143]. Minocycline foam is generally well tolerated in trials, with the most frequent side effects being increased creatinine phosphokinase levels and headaches [140,141]. Topical minocycline 4% foam for acne has received FDA approval but has not yet been approved in the EU.
Bacterial resistance to both topical and oral antibiotics can develop within as little as 6 weeks of monotherapy [135,144]. Therefore, topical antibiotic monotherapy is generally discouraged. Combining with BPO minimizes resistance; BPO can be applied concurrently or as a wash prior to antibiotic application [133].

3.1.4. Topical Azelaic Acid

Azelaic acid, a naturally occurring dicarboxylic acid found in barley and wheat, offers multiple benefits in acne treatment [145]. It functions as a comedolytic (preventing clogged pores), antimicrobial (fighting bacteria including C. acnes), anti-inflammatory (reducing acne-related inflammation), anti-keratolytic (regulating skin cell turnover), and antioxidant (protecting against cellular damage) [145,146,147]. Additionally, azelaic acid is effective in treating post-inflammatory hyperpigmentation [146,147].
Azelaic acid is available in both 15% gel (Finacea) and 20% cream (Azelex) formulations and is generally applied twice daily [147]. It is a first-line treatment option for mild to moderate inflammatory and non-inflammatory acne [147]. Azelaic acid is particularly well suited for those with sensitive skin or dyspigmentation concerns [147].
While generally well tolerated, some side effects can occur, including skin irritation, burning, redness, and itching [148]. Close monitoring is recommended in patients with darker complexions, as its lightening effect could be more pronounced [106]. Azelaic acid is considered safe for pregnancy (category B) and for lactating women [146].

3.1.5. Topical Salicylic Acid

Salicylic acid, a beta-hydroxy compound, combats mild acne by acting as a comedolytic, keratolytic, and mild anti-inflammatory, while also possessing fungistatic and bacteriostatic properties [135,149,150].
Salicylic acid is widely available over the counter in concentrations ranging from 0.5% to 2% in both leave-on (gels, creams, lotions) and wash-off (cleansers) products [135]. It is a suitable option for patients who cannot tolerate stronger topical treatments like retinoids or BPO [135]. Salicylic acid can be combined with BPO for enhanced efficacy [146]. Additionally, higher concentrations are used in superficial chemical peels [146].
While generally safe, salicylic acid can sometimes cause skin irritation, dryness, itching, erythema (redness), and peeling [149,151]. In some cases, it might temporarily worsen inflammatory acne lesions [149].
A specific formulation, supramolecular salicylic acid, significantly improves acne and favorably modifies the skin microbiome to resemble that of individuals without acne [152].

3.1.6. Topical Dapsone

Topical dapsone, a sulfone medication, offers antimicrobial, anti-inflammatory, and potentially immunomodulatory actions for treating acne [153,154]. It is available as a 5% or 7.5% gel and reduces both inflammatory and non-inflammatory acne lesions [155,156]. Topical dapsone has received FDA approval but has not yet been approved in the EU. It may be used for inflammatory papulopustular acne when initial therapies fail [146,157].
Topical dapsone is considered a second-line agent. However, for specific patient groups (women with inflammatory acne, those with darker skin tones, or patients with sensitive skin or contraindications to other treatments), it may be an appropriate initial therapy [158,159,160]. Due to its affordability, it could be particularly valuable in developing nations [155,156].
Studies have demonstrated its safety and efficacy, even in patients with glucose-6-phosphate dehydrogenase deficiency or sulfonamide allergies [160,161]. Therefore, unlike oral dapsone, glucose-6-phosphate dehydrogenase testing is not required for topical use [135]. Side effects are generally minimal but may include skin irritation [135].
Avoid combining topical dapsone with BPO, as this can cause temporary orange staining of the skin [135,146]. However, this discoloration can be washed off.

3.1.7. Topical Sulfur and Sodium Sulfacetamide

Sulfur and sodium sulfacetamide are often used in combination to treat acne as their actions complement each other:
(a)
Sulfur: Exhibits mild antibacterial and keratolytic properties, helping loosen dead skin cells to prevent clogged pores [162]. Sulfur interacts with the cysteine within skin cells to form hydrogen sulfide, disrupting disulfide bonds for its keratolytic effect [163]. Newer nanoparticle preparations may offer enhanced effectiveness against Staphylococcus bacteria, a contributor to acne pathogenesis [164,165]. Sulfur on its own can treat mild to moderate acne, but its results are improved when combined with sodium sulfacetamide or BPO [166,167,168,169,170].
(b)
Sodium sulfacetamide: A bacteriostatic agent that disrupts bacterial DNA synthesis by inhibiting para-aminobenzoic acid [126]. Typically formulated as a topical lotion with 10% sodium sulfacetamide and 5% sulfur, it demonstrates significant acne lesion reduction (50–69% after 8 weeks; 78% after 12 weeks) [166,171].
Both sulfur and sodium sulfacetamide have generally well-tolerated side effects, which may include skin dryness, astringency (tightness), and itching [162,166,167]. Sulfur on its own has a noticeable odor, which is why it is often combined with sodium sulfacetamide for scent masking [168].

3.1.8. Topical Clascoterone

Clascoterone (Winlevi®), approved by the FDA, is the first topical antiandrogen available for all sexes [172]. It is a groundbreaking topical acne treatment, the first with a novel mechanism of action since isotretinoin’s introduction in 1982 [173]. It received FDA approval in August 2020 but has not yet been approved in the EU. This topical steroidal antiandrogen offers both anti-inflammatory and antiandrogenic effects, making it a valuable tool for treating acne in both men and women [174,175].
Clascoterone functions by competitively inhibiting dihydrotestosterone (DHT) binding to androgen receptors (ARs) within the skin, particularly in sebaceous glands [173,174,176]. Androgen binding to the AR normally stimulates sebum production. By blocking this action, clascoterone decreases sebum production and reduces the production of pro-inflammatory cytokines [173]. Importantly, upon absorption, clascoterone is rapidly metabolized into an inactive form (cortexolone), minimizing the risk of systemic antiandrogenic effects [176].
Two large-scale, double-blind, vehicle-controlled phase III clinical trials (1440 patients with moderate to severe facial acne) demonstrated the superior efficacy of clascoterone 1% cream compared with the vehicle. Clascoterone significantly reduced both inflammatory and non-inflammatory acne lesions. Treatment success rates at week 12 were significantly higher with clascoterone (18.4–20.3%) compared with a placebo (6.5–9.0%). Also, a pilot study found clascoterone to be comparable to tretinoin 0.05% cream [176]. Importantly, clascoterone was well tolerated, with no systemic adverse effects reported. Local site reactions were primarily mild erythema (11.3–13.1%) and scaling/dryness (8.8–12.2%), occurring at similar rates to the vehicle control [174].
While clascoterone cream is rapidly metabolized, some adrenal dysregulation can still occur. Asymptomatic adrenal suppression was observed in about 7% of patients using high amounts of clascoterone in a phase II trial, which normalized after discontinuation [175]. Though typically used in women, systemic hormone modulation with oral contraceptives or spironolactone can be effective but have systemic side effects [175,176]. These options differ significantly from topical clascoterone.
Further comparative effectiveness studies are needed to fully establish its optimal role. However, it is likely to become a highly beneficial addition to treatment options for a wide range of acne patients. Importantly, this novel mechanism of action allows for combination therapies with other existing treatments.
Table 1. Topical treatments for acne.
Table 1. Topical treatments for acne.
Treatment ExamplesMechanism of ActionIndicationsCommon Side EffectsReferences
Retinoids (e.g., tretinoin, adapalene)Normalizes follicular epithelial desquamation; anti-inflammatoryMild to severe acne, depending on the formulationDryness, photosensitivity, initial acne flare-up[3,103,104,105,106,107,108,109,111,112,113,114,115,116,120,122,123,124,125,126,177,178]
Benzoyl peroxideKills bacteria; peels out the inner lining of the hair follicle, causing skin peelingMild to moderate acneDryness, irritation, potential bleaching of clothes[130,131,132,133,134,135,136,138,146,179,180,181,182,183]
Antibiotics (e.g., clindamycin)Antimicrobial; reduces inflammationMild to moderate inflammatory acneSkin irritation, resistance development[126,135,139,140,141,142,143,144]
Azelaic acidKills bacteria; normalizes keratinization; anti-inflammatoryMild to moderate acnePruritus, burning sensation[106,145,146,147,148]
Salicylic acidHelps break down blackheads and whiteheads; anti-inflammatoryMild acne, comedonal acneSkin irritation, dryness[135,146,149,150,151,152]
Sulfur and sodium sulfacetamideAntibacterial and keratolytic effectsMild to moderate acne, rosaceaDryness, skin irritation[126,162,163,164,165,166,167,168,169,170,171]
Clascoterone (topical antiandrogen)Blocks androgen receptors in the skin; reduces sebum production and inflammationModerate to severe acneLocal irritation, erythema[173,174,175,176]
Table 2. Topical combinations for acne treatment.
Table 2. Topical combinations for acne treatment.
Treatment Formulation, Dose, FrequencyCommon Side EffectsReferences
Tretinoin/ClindamycinGel
0.025%/1.2% daily
Xerosis, irritation, allergic contact dermatitis, erythema[102]
Tretinoin/Benzoyl peroxideCream
0.1%/3% once daily
Xerosis, irritation, allergic contact dermatitis, erythema[127]
Adapalene/Benzoyl peroxideGel
0.1%/2.5% once daily or
0.3%/2.5% once daily
Xerosis, irritation, allergic contact dermatitis, erythema[102]
Benzoyl peroxide/ClindamycinGel
5%/1% once daily or
3.75%/1.2% once daily or
2.5%/1.2% once daily
Xerosis, irritation, allergic contact dermatitis, erythema, bleaching of fabrics[132,133,134]
Benzoyl peroxide/ErythromycinGel
5%/3% once daily
Xerosis, irritation, allergic contact dermatitis, erythema, bleaching of fabrics[138]
Clindamycin/Benzoyl peroxide/Adapalene (IDP-126)Gel
1.2%/3.1%/0.15%
Xerosis, irritation, allergic contact dermatitis, erythema[128]
Novel combined formulation in development:
Minocycline/Adapalene (FCD105)
Foam
3%/0.3%
Xerosis, irritation, allergic contact dermatitis, erythema[129]
Figure 2. Chemical structures of the drugs used in acne therapy.
Figure 2. Chemical structures of the drugs used in acne therapy.
Ijms 25 05302 g002

3.2. Systemic Treatments

Systemic treatments for acne are utilized in moderate to severe cases, or when topical therapies are insufficient or not feasible. These treatments, which include oral antibiotics, hormonal therapies, and isotretinoin, target acne’s deeper pathophysiological mechanisms and offer broad-spectrum action against the factors contributing to acne development (Table 3).

3.2.1. Oral Antibiotics

Oral antibiotics play a key role in managing moderate to severe inflammatory acne. They work by both reducing C. acnes bacteria and through anti-inflammatory actions [144,184]. Their efficacy extends even after discontinuation when used in conjunction with topical retinoids and/or BPO [185,186].
Tetracyclines remain a first-line treatment for acne due to their ability to inhibit bacterial protein synthesis. Among those, lymecycline and doxycycline are recommended as first-line oral antibiotics for the treatment of acne [187]. European guidelines limit the recommendation for systemic antibiotics to lymecycline and doxycycline and restrict the treatment period to three months to prevent the emergence of antibiotic resistance [188]. The US guidelines, Canadian guidelines, UK NICE guidelines, and European guidelines prioritize lymecycline and doxycycline over minocycline due to their comparable effectiveness and the higher incidence of severe adverse events associated with minocycline [188]. However, minocycline and doxycycline are widely used for their convenient once-daily dosing, compatibility with food, and effective penetration of follicles [189,190,191]. Sarecycline is a newer narrow-spectrum tetracycline approved by the FDA in 2018 [172]. However, it has not been approved by the European Medicines Agency (EMA). Sarecycline offers the potential benefits of specifically targeting C. acnes while minimizing disruption to the gut microbiome, potentially reducing the risk of antibiotic resistance [189,192,193,194]. Studies demonstrate sarecycline’s efficacy in treating moderate to severe acne [195].
Photosensitivity is a notable side effect particularly associated with doxycycline, highlighting the necessity for stringent sun protection and comprehensive patient education [196,197]. Moreover, to mitigate the risk of pill esophagitis, it is recommended that tetracyclines be consumed with sufficient water, and patients should refrain from lying down immediately post-ingestion [196,197]. Minocycline, while effective, is associated with a spectrum of potential risks including urticaria, vestibular effects, serum-sickness-like reactions, hyperpigmentation, drug reaction with eosinophilia and systemic symptoms (DRESS), and autoimmune hepatitis [198]. Sarecycline may have a lower incidence of these side effects. Also, doxycycline or lymecycline are often preferred over minocycline due to their comparably effective outcomes and lower incidence of severe side effects [135,199,200,201].
When tetracyclines are unsuitable (e.g., in patients under the age of 8, those with allergies, or other contraindications), other oral antibiotics can be considered. These include macrolides (erythromycin, azithromycin), penicillins (ampicillin, amoxicillin), trimethoprim/sulfamethoxazole, and cephalexin [135,202]. However, it is important to note that the evidence supporting their efficacy in acne treatment may be more limited.
Guidelines emphasize limiting oral antibiotic use to 3–6 months to curb antibiotic resistance, stressing the importance of continued topical treatment to maintain remission after discontinuation [187,199,201,202,203,204]. To minimize resistance, combination therapy with BPO is preferred over antibiotic monotherapy [205]. For adult women, hormonal therapies (spironolactone, combined oral contraceptives) may offer alternatives to antibiotics [195,206].

3.2.2. Hormonal Therapies

Hormonal therapies play a crucial role in acne treatment, especially for women experiencing flares related to their menstrual cycle or those with underlying conditions such as polycystic ovary syndrome (PCOS). These therapies target androgen production and activity, as androgens (like testosterone and DHT) are major contributors to sebum production and acne development [207]. Hormonal therapies for acne include combined oral contraceptives (COCs) and antiandrogens.
COCs are a mainstay in hormonal treatment for acne, utilizing a combination of estrogen and progestin to suppress ovarian androgen production. They also increase levels of sex-hormone-binding globulin, which reduces the circulation of free testosterone, thereby diminishing sebum production [208,209,210]. FDA-approved COCs for acne include Beyaz (drospirenone/ethinyl estradiol/levomefolate), Estrostep Fe (norethindrone acetate and ethinyl estradiol), Ortho Tri-Cyclen (norgestimate and ethinyl estradiol), and Yaz (drospirenone and ethinyl estradiol) [207]. These options not only reduce the visible symptoms of acne but also regulate the menstrual cycle and reduce androgenicity. While improvement can take time, COCs are as effective as oral antibiotics over a longer course and are a first-line treatment for those with PCOS [146,207]. At 3 months, COCs are less effective than oral antibiotics, but by 6 months, they are equally effective in reducing acne [210]. However, their use must be carefully considered against potential risks, especially in patients prone to thromboembolic events [135,208]. Side effects include breast tenderness, mood changes, GI issues, weight gain, headaches, and breakthrough bleeding [208].
Co-cyprindiol (ethinylestradiol/cyproterone acetate) is used as a second-line treatment for severe acne in women, particularly when other treatments have failed [187]. However, it is advised against for long-term use due to safety concerns about the rare but cumulative dose-dependent increased risk of meningioma [187].
Spironolactone, an antiandrogenic diuretic, has proven effective against acne, particularly in female patients, by blocking androgen receptors to reduce sebum production [202,207,211]. Comparable in effectiveness to oral antibiotics, spironolactone is frequently prescribed for long-term management, often alongside oral contraceptives to enhance its antiandrogenic effects and to prevent pregnancy [212,213]. Recent studies suggest that the risk of hyperkalemia in healthy young women taking spironolactone is similar to the baseline risk in this demographic, indicating that routine monitoring of serum potassium may be unnecessary [214]. Common side effects include menstrual irregularities in women not on combined oral contraceptives and potential gynecomastia, though it is not linked to an increased risk of breast or gynecological cancers [213]. Given the risk of feminization of male fetuses, contraception is advised when prescribing spironolactone to women of childbearing potential [215]. Spironolactone can be initiated at doses ranging from 50 to 200 mg/day, adjusted based on clinical response and side effects, and is generally well tolerated [216].
Hormonal therapies are often preferable for long-term acne management compared with antibiotics [210]. Combining them with topical acne treatments can enhance their effectiveness. Careful patient selection is crucial, especially for those with hyperandrogenism or endocrine disorders, who may see the greatest benefit [207]. However, hormonal therapies may not be suitable for everyone due to contraindications like thromboembolic risk factors or specific medical conditions. For these patients, alternative treatments such as antibiotics, retinoids, or isotretinoin may be necessary. Interestingly, some studies suggest that the early initiation of hormonal antiandrogens could potentially decrease the need for prolonged courses of antibiotics in acne management [206].
It is important to be aware that progestin-only contraceptives and implants have the potential to worsen acne [217]. The composition of oral contraceptive pills matters: older generations (first and second) may exacerbate acne, while newer formulations (third and fourth generation) often have beneficial effects [218]. While hormonal treatments are not typically the first choice for acne in women, their use is becoming more widespread.

3.2.3. Oral Retinoids

Oral retinoids are highly effective in treating acne by preventing keratin formation, which can block pores and contribute to acne development [202]. Typically reserved for severe cases that do not respond to topical treatments, well-known oral retinoids used for acne include isotretinoin, acitretin, and tretinoin [219].
Isotretinoin, a first-generation retinoic acid derivative, is indicated for severe recalcitrant nodular acne, but it is often used for resistant moderate to severe cases, acne causing scarring, or acne with a significant psychosocial impact [135]. It works by reducing sebum production, decreasing inflammation, normalizing follicular keratinization (reducing clogged pores), and reducing C. acnes abundance [184,220,221,222]. Isotretinoin has the potential to induce remission, with a cumulative dose of 120–150 mg/kg recommended [223]. Initiating therapy at a low dose may help minimize initial acne flares [204,224]. Some studies suggest continuing treatment for at least 2 months after full resolution to reduce the relapse risk [225,226].
Isotretinoin, known for its teratogenic effects, poses a risk of spontaneous abortion (20%) and embryopathy (18–28%) in exposed pregnancies [227]. Isotretinoin is absolutely contraindicated in pregnancy and lactation, requiring strict contraception during treatment and for one month afterward [68]. Common side effects like skin dryness (72.13%), cheilitis (94.25%), dry eyes (29.49%), and muscle aches (23.05%) are reversible and dose-dependent [228]. Muscle aches can sometimes be associated with elevated creatine phosphokinase (CPK) levels, indicating potential muscle damage [229,230,231,232]. While these elevations are often mild and temporary, monitoring CPK may be advisable for patients experiencing muscle-related symptoms or those engaged in frequent intense physical activity [229,230,231,232]. If significantly elevated CPK levels are detected, a reduction in isotretinoin dosage or a temporary cessation of treatment may be considered until CPK levels normalize [229,230,231,232]. Regular monitoring for liver and lipid abnormalities is recommended [233,234], with guidelines advising against full-blood-count tests [135]. A study among 13,772 acne patients showed frequent mild to moderate reversible lipid and liver enzyme changes, suggesting that baseline and periodic follow-up tests are sufficient [234,235,236]. Less common but significant potential side effects include skeletal effects (hyperostosis, premature epiphyseal closure), inflammatory bowel disease (IBD), and psychiatric effects (depression, emotional instability, anxiety, and, rarely, suicidal thoughts). However, the association with psychiatric effects remains debated, as acne itself carries a risk of depression and suicide [237]. Interestingly, a recent study suggests that isotretinoin use may be associated with a lower suicide rate compared with the general population [238]. Additionally, the association between isotretinoin and IBD remains controversial. Several studies and meta-analyses suggest no increased risk with isotretinoin, and antibiotic use may be a confounding factor in the reported association [239,240,241,242].
In cases where isotretinoin is ineffective or unsuitable for long-term maintenance, acitretin has shown success as a maintenance treatment [243]. Acitretin is a second-generation retinoid primarily indicated for psoriasis and keratinization disorders but offers potential in cases of nodulocystic acne that is resistant to isotretinoin [243]. Acitretin works by inhibiting epidermal growth and differentiation, providing a potential mechanism to control the formation of cysts and reduce inflammation [244]. While systemic retinoids are known for potential side effects like hypertriglyceridemia and hepatotoxicity, careful monitoring and dose adjustments can minimize these risks. Further research is needed to establish optimal dosing and patient selection criteria.
Table 3. Systemic treatments for acne.
Table 3. Systemic treatments for acne.
Treatment ExamplesMechanism of ActionIndicationsCommon Side EffectsReferences
Oral antibiotics (e.g., tetracycline, doxycycline)Antimicrobial, reduce inflammationModerate to severe acneGastrointestinal upset, photosensitivity[135,144,184,185,186,187,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206]
Hormonal therapies (e.g., oral contraceptives, spironolactone)Regulate hormonal imbalance, reduce sebum productionHormonally influenced acne, adult womenMood changes, breast tenderness, thrombosis risk[135,146,207,208,209,210,211,214,215,217,218]
IsotretinoinReduces sebaceous gland size and sebum production, anti-inflammatorySevere scarring acne or acne not responsive to other treatmentsDryness, teratogenicity, elevated liver enzymes[68,135,184,204,220,221,222,223,224,225,226,227,228,233,234,235,236,237,238,239,240,241,242]

3.3. Procedural Therapies

Emerging studies, although limited in scale, suggest that modalities such as laser and light devices, chemical peels, and intralesional steroid injections present viable options for mitigating acne symptoms and reducing scarring [135,245,246] (Table 4). The application of ablative and non-ablative laser technologies for acne scarring introduces options for significant resurfacing or collagen stimulation with less downtime, respectively, signifying that tailored approaches are required to optimize patient outcomes and minimize adverse effects [247].
Photodynamic therapy (PDT) stands out due to its strong evidential backing, which includes a meta-analysis of 13 randomized clinical trials encompassing 701 participants, highlighting its efficacy in reducing inflammatory lesion counts by a mean percentage of 15.97% [246]. PDT uniquely combines light energy with a photosensitizing agent, typically aminolevulinic acid or methyl aminolevulinic acid, to target pilosebaceous units and C. acnes, leveraging the bacterium’s innate production of photosensitizing porphyrins [246,248,249]. PDT has been posited to have a longer-lasting therapeutic effect than IPL [250,251].
LED (light-emitting diode) utilizing red and blue spectra have shown effectiveness in treating inflammatory acne [252,253,254,255]. IPL (intense pulsed light) treats inflammatory acne by killing C. acnes, reducing the size and number of sebaceous glands, and modulating inflammatory markers like TNF-α and TGF-β [250,254,256,257,258]. Pulsed-dye laser (PDL) therapy, alongside its role in inflammatory acne treatment, posits a longer-lasting therapeutic effect compared with IPL [250,251]. The use of the 1450 nm diode laser with a dynamic cooling device has shown moderate success in treating inflammatory acne and reducing excessive sebum production through various delivery methods [259,260,261]. Using stacked-pulse delivery appears to offer slightly better results, though individuals with darker skin (Fitzpatrick types IV–VI) should proceed with caution due to a higher risk of developing hyperpigmentation [259]. Another approach, a dual-regimen treatment that combines the low-fluence targeting of inflammatory lesions with a micropulsed moving mode applied across the entire face, has been found to be more effective than traditional high-fluency stamping. This method is not only less painful but also carries a reduced risk of causing hyperpigmentation [260]. The FDA-cleared AviClearTM (1726-nm laser) presents a targeted approach to treating mild to severe acne by selectively acting on sebaceous glands, with reported manufacturer outcomes suggesting sustained improvements post-treatment [262,263]. Similarly, the FDA has approved the 1726 nm Accure Laser System and TheraClear®, a combination of vacuum and broadband light technology, expanding the arsenal against acne [263].
The FDA’s recent approval of gold nanoparticles for acne treatment offers promising results through the targeted photothermolysis of sebaceous follicles [264,265,266]. Three weekly sessions combining these nanoparticles with diode laser pulses significantly improved inflammatory acne [267], while a similar protocol with a photopneumatic device effectively treated mixed acne [268]. A study also showed that the gold salt auranofin could inhibit the NLRP3 inflammasome in mice, suggesting anti-acne effects [269].
Chemical peels, ranging from superficial to medium depth, including agents such as glycolic acid, salicylic acid, azelaic acid, pyruvic acid, mandelic acid, phytic acid, and Jessner’s solution, have been scrutinized for their effectiveness in treating acne, post-inflammatory hyperpigmentation (PIH), and scarring. Despite the array of options, a robust analysis combining a meta-analysis with multiple split-face studies has yet to establish clear superiority among these treatments [270,271]. Superficial peels, particularly those using glycolic or salicylic acids, are noted for their efficacy in managing comedonal acne. They require a series of treatments for sustained results. A trial comparing these two acids revealed comparable outcomes in lesion reduction over 12 weeks, with the longevity of improvement notably extending up to 8 weeks after discontinuing salicylic acid treatment [271]. However, the application of chemical peels can lead to side effects such as erythema, dyspigmentation, and an increased infection risk. This highlights the necessity for a personalized treatment plan, considering factors like the patient’s skin sensitivity, healing ability, recent treatments, and overall health. Caution is particularly warranted for individuals with darker skin (Fitzpatrick skin types IV and higher) and those on oral retinoids due to a higher dyspigmentation risk [126]. For these patients, medium-depth peels are generally discouraged to avoid adverse pigmentary outcomes [263].
Non-ablative fractionated radiofrequency microneedling (FRFM) and combinations with isotretinoin have been effective in treating active acne and reducing scarring, with the adjunctive use of microneedling facilitating the enhanced delivery of topical treatments (retinoids and salicylic acid) and scar improvement [272,273,274,275]. Using isotretinoin in conjunction with other energy-based devices such as PDL and ablative fractional lasers has also been proven effective in managing active inflammatory acne [276].
Intralesional steroid injections provide an effective remedy for large inflammatory lesions and keloidal scars, albeit with caution due to potential local atrophy [277].
Table 4. Physical and laser therapies for acne.
Table 4. Physical and laser therapies for acne.
Treatment TypeMechanism of ActionIndicationsCommon Side EffectsReferences
Photodynamic therapyActivates photosensitizing agents that kill bacteria and reduce sebum productionSevere acne, acne resistant to other treatmentsRedness, swelling, skin sensitivity to sunlight[246,248,249,250,251]
Blue-light therapyKills C. acnes bacteria using blue light without damaging the skinMild to moderate acneTemporary redness and dryness[252,253,254,255]
Chemical peelCauses the top layer of skin to peel off, helps clear debris from poresMild to severe acne, depending on the peel’s strengthRedness, peeling, potential scarring[126,263,270,271]
Intralesional steroid injectionAnti-inflammatory action and decrease in sebum productionModerate to severe acneLocal atrophy[277]

3.4. Diet Modifications

The impact of diet on acne has been a subject of increased interest and research. Recent studies have begun to shed light on how certain dietary patterns may influence acne development, leading to the incorporation of diet modifications as complementary strategies in acne treatment plans (Table 5).
Previous studies have shown that dairy products and diets high in hyperglycemic carbohydrates boost insulin/IGF-1 signaling and lower insulin-like growth factor binding protein 3 (IGFBP-3) levels [278,279]. IGFBP-3, which binds to the retinoid X receptor-α, promotes cell growth and prevents cell death [280,281]. Meanwhile, IGF-1 reduces the presence of FoxO1 in the nucleus, triggering mTORC1. This activation of mTORC1 leads to an increase in keratinocyte and sebocyte proliferation and lipogenesis, contributing to acne development [282].
Also, adiponectin, known for its anti-inflammatory properties, is inversely associated with the glycemic load and dairy intake, suggesting that a diet with a high glycemic index can lower adiponectin levels, thus worsening acne severity [56]. Studies indicate that patients with acne have lower serum adiponectin levels compared with healthy controls, highlighting the potential of dietary management to modulate inflammation and acne severity [32].
Dietary patterns significantly impact gut diversity and the balance between beneficial and harmful bacteria, with a Western diet shown to decrease microbial diversity and skew the bacterial population in ways that may exacerbate acne. Studies reveal that acne patients exhibit distinct gut microbiota, characterized by decreased Actinobacteria, increased Proteobacteria, and a shift from Firmicutes to Bacteroidetes—an enterotype often associated with a Western diet [80,283]. This diet, rich in dairy, refined carbohydrates, and saturated fats, not only alters the gut microbiome but also triggers metabolic changes conducive to acne development [283]. Conversely, the adoption of a diet low in processed foods and high in vegetables, fruits, and omega-3 fatty acids can mitigate acne by promoting a healthier gut microbiota and reducing inflammation [284,285].
Acne patients often have lower levels of antioxidants like vitamin E, vitamin A, and selenium-dependent enzymes, contributing to acne-related inflammation [286]. Dietary fiber intake and plant-based antioxidants such as catechins in green tea and phytoalexins in red grapes have been shown to improve acne, possibly by reducing sebum production and inhibiting harmful bacteria [4,286]. A high-fiber diet beneficially alters the gut microbiome, improving insulin sensitivity and reducing hyperinsulinemia, thus potentially mitigating acne by decreasing IGF-1 levels, which are linked to increased sebum production and keratinocyte proliferation [4,82,287].
Vitamin D deficiency is more common among acne patients, suggesting a possible link between vitamin D levels and acne [288,289]. While some studies show that vitamin D can influence the expression of inflammatory biomarkers in sebocytes, suggesting a beneficial role in acne management, other research studies have found no significant association between vitamin D levels and acne development [290,291,292]. Additionally, vitamin D supplementation has been observed to positively alter the gut microbiome, potentially impacting skin health [293].
Zinc exhibits antibacterial properties against C. acnes and reduces inflammation [286]. Acne patients often have lower zinc levels, which correlates with the severity and type of acne [294]. Zinc can be used in pregnant women [69,295]. However, excessive iodine intake might exacerbate acne, indicating the need for balanced micronutrient intake for optimal skin health [286].
Biotin is a crucial water-soluble vitamin. Deficiency in biotin can occur as a side effect of isotretinoin treatment for acne, mainly due to its toxic effects on liver function, which reduce biotinidase enzyme activity [296]. This deficiency can lead to mucocutaneous and hair-related adverse effects [296]. A study has shown that supplementation with biotin (10 mg/day) alongside isotretinoin can alleviate these side effects [297]. The study found that biotin not only helps reduce the telogen (resting) hair ratio while increasing the anagen (growing) hair ratio but also assists in maintaining skin hydration [297]. These findings suggest that daily supplementation with 10 mg of biotin could be an effective strategy to mitigate the mucocutaneous adverse effects associated with isotretinoin treatment, making it a valuable addition to dietary modifications for patients undergoing isotretinoin therapy for acne.
Table 5. Dietary supplements for acne.
Table 5. Dietary supplements for acne.
Supplement ExamplesMechanism of ActionIndicationsCommon Side EffectsReferences
Omega-3 fatty acidsAnti-inflammatory, modulates sebum productionVarious acne typesFishy aftertaste, gastrointestinal upset[284,285]
Vitamin DModulates immune system, may reduce skin inflammationVarious acne typesRare, potentially increased calcium levels[288,289,290,291,292,293]
ZincReduces inflammation and bacterial growth and modulates the immune systemVarious acne typesNausea, metallic taste[69,286,294,295]

4. Emerging Therapies

Current research into acne treatment explores various facets, including sebaceous gland activity, inflammation, microbial flora, and even systemic influences like diet, unveiling promising candidates, each targeting different aspects of acne’s multifactorial nature (Table 6).
Hyaluronic acid (HA), a major glycosaminoglycan in the extracellular matrix, plays significant roles in skin cell functions. The HA-binding receptor, CD44, is expressed in human skin sebaceous glands. HA’s ability to downregulate lipid synthesis in a dose-dependent manner and significantly decrease sebum production positions it as a potential acne treatment [298].
Cannabidiol (CBD) is being researched for its potential to interact with the skin’s endocannabinoid system (ECS), which plays a crucial role in maintaining skin health and function [299]. CBD’s potential lies in its anti-inflammatory benefits, making it a candidate for treating inflammatory skin diseases [300]. Specifically, CBD exhibits sebostatic properties, effectively inhibiting the lipogenic actions of compounds like arachidonic acid and a combination of linoleic acid and testosterone. It also suppresses sebocyte proliferation through the activation of transient receptor potential vanilloid-4 (TRPV4) ion channels [301,302,303,304,305]. Given these properties, CBD is a promising candidate for the treatment of acne.
Biologic treatments have been explored in severe acne cases through small-scale studies. Agents targeting TNF-α, such as adalimumab and etanercept, have shown promise in treating aggressive forms of acne, including acne conglobata and fulminans [306,307]. Similarly, treatments inhibiting IL-17 and IL-23, namely secukinumab and risankizumab, respectively, have been effective in managing the stubborn SAPHO syndrome [308,309], underscoring the role of the IL-17/Th17 pathway in acne pathology [85]. However, a recent clinical trial revealed that CJM112, an IL-17A inhibitor with a unique target compared with secukinumab, did not outperform a placebo in reducing inflammatory acne [310]. Additionally, an RCT examining the efficacy of the anti-IL-1β inhibitor gevokizumab (XOMA 052) has been conducted, though results are yet to be published [106].
The gut microbiome has been increasingly implicated in the regulation of systemic inflammation and has an impact on various diseases. A study by Paetzold et al. did not utilize fecal microbiome transplantation but rather transferred healthy skin microbiome components to acne-prone individuals. This approach demonstrated that the recipient’s skin microbiome began to exhibit characteristics similar to that of the donor’s within a week, suggesting a potential for topical microbiome transplantation in dermatological treatments [311]. While this study opens new avenues for treating acne by altering the skin’s microbiome, it primarily focused on microbiome dynamics and engraftment without directly assessing clinical improvements in acne symptoms.
Building upon these insights, the theory suggests that enhancing the natural skin microbiota with topical probiotics could prevent the dominance of harmful C. acnes strains [312]. This approach is thought to work by promoting the production of antimicrobial substances from beneficial bacteria, reducing pro-inflammatory cytokines, and directly curbing the growth of C. acnes [312,313,314,315,316]. Research supports this, with studies showing that lotions containing enterocins from Enterococcus faecalis SL-5 improved inflammatory acne symptoms [317], and Lactobacillus plantarum effectively diminished acne lesion size and redness [318].
Oral probiotics have been found to elevate anti-inflammatory IL-10 levels in individuals with acne [319]. A study highlighted the efficacy of oral probiotics alone, in conjunction with oral minocycline, and as part of combination therapy, showing marked lesion reduction in all scenarios [320]. Furthermore, consuming lactoferrin-enriched fermented milk has been shown to reduce acne lesions [321], and a randomized controlled trial (RCT) demonstrated significant improvements in acne lesions and reduced C. acnes levels with a symbiotic supplement combining multiple probiotic strains and a botanical extract [322]. In a pioneering double-blind pilot study comparing the effects of a Lactobacillus rhamnosus GG supplement against a placebo over three months on 20 adults with acne, significant improvements were observed in the probiotic group [323]. Analysis of skin biopsies from these participants revealed a decrease in IGF-1 levels alongside an increase in FoxO1 gene expression, suggesting a positive regulatory effect on acne-related markers. These results suggest a promising role for probiotics as a beneficial and well-tolerated option for acne management, warranting further research to substantiate these preliminary findings.
Vaccines targeting C. acnes and its virulence factors, like CAMP factor 2, have shown promise in reducing acne-induced inflammation in mice studies [324]. However, currently, there is no clinical evidence for the effectiveness of a C. acnes vaccine in humans.
Bacteriophages, viruses that infect bacteria, offer an innovative approach to targeting acne-causing bacteria. Early research suggests their potential in reducing C. acnes and its associated inflammation, though clinical studies are needed to confirm their therapeutic benefits [325,326].
Designed antimicrobial peptides (dAMPs) represent a novel class of therapeutics, offering direct antimicrobial activity and immune modulation [327,328,329,330]. Their effectiveness against antibiotic-resistant strains of C. acnes in a preclinical model highlights their potential as a future acne treatment modality [331].
Phosphodiesterase (PDE) inhibitors have shown promise in treating acne through their anti-inflammatory properties. By inhibiting PDE, these compounds increase levels of cyclic adenosine monophosphate (cAMP), leading to reduced inflammation. Apremilast, a PDE-4 inhibitor, is one such drug that has demonstrated efficacy in reducing inflammatory lesions in acne, indicating the potential role of PDE inhibitors in acne treatment [332,333].
In light of the recognized role of the Akt/mTOR network in acne, there is an emerging interest in developing novel therapeutic agents targeting the mTORC1- and FoxO1-dependent signaling pathways to mitigate mTORC1/SREBP1-mediated sebum production, which plays a significant role in the pathology of acne [334]. One of the most widely used drug is isotretinoin, a powerful sebum suppressor that operates by increasing the nuclear expression of FoxO1, subsequently inhibiting mTORC1, which leads to the apoptotic death of sebocytes [335]. Beyond traditional drugs, natural dietary polyphenolic ingredients like epigallocatechin-3-gallate (EGCG) and resveratrol are potent mTORC1 inhibitors, effectively reducing sebaceous lipid production and ameliorating clinical acne manifestations [335]. A pilot study demonstrated significant clinical improvement in acne symptoms with the topical application of a resveratrol-containing gel [336].
Table 6. Emerging therapies in acne treatment.
Table 6. Emerging therapies in acne treatment.
Therapy TypeMechanism of ActionResultReferences
Hyaluronic acidActs as a major component of the extracellular matrix, downregulating lipid synthesis in a dose-dependent manner through its interaction with cellular receptors.Significantly decreases sebum production, improving skin hydration and reducing oily skin appearance.[298]
CannabidiolInteracts with the skin’s endocannabinoid system, helping to maintain skin health and function, while exerting anti-inflammatory and sebostatic properties.Reduces inflammation and normalizes sebum production, leading to fewer acne outbreaks.[299,300,301,302,303,304,305]
Biologic treatments (e.g., TNF-α inhibitors like adalimumab; IL-17 and IL-23 inhibitors like secukinumab)Modulates the immune response by targeting and inhibiting specific cytokines involved in inflammation.Reduces inflammation and the severity of acne symptoms, improving the overall skin condition.[85,306,307,308,309,310]
Gut microbiome (microbial transplantation)Transfers a healthy microbiome from a donor to an acne-prone recipient, fostering a microbial environment similar to that of individuals without acne.The recipient’s skin microbiome adopts characteristics beneficial for preventing acne, reducing lesion formation.[311]
Topical probioticsPromotes the production of antimicrobial substances from beneficial bacteria and competes with pathogenic microbes on the skin.Reduces pro-inflammatory cytokine levels and acne severity, promoting a healthier skin barrier.[312,313,314,315,316,317,318]
Oral probioticsEnhances the systemic immune function and modulates local inflammatory responses through increased levels of anti-inflammatory cytokines like IL-10.Demonstrates a decrease in acne lesions and an overall improvement in skin clarity.[319,320,321,322,323]
Vaccines Targets C. acnes and its virulence factors, like CAMP factor 2, to reduce microbial-induced inflammation.Leads to a significant reduction in acne-related inflammation and lesion count.[324]
BacteriophagesSpecifically targets and kills C. acnes bacteria, thereby directly reducing the bacterial load and associated inflammation.Early research suggests a potential reduction in acne severity and improved skin condition.[325,326]
Designed antimicrobial peptides (dAMPs)Provides targeted antimicrobial activity against resistant strains of C. acnes, while also modulating the immune response.Reduces acne outbreaks and severity through effective bacterial control and reduced inflammation.[327,328,329,330,331]
Phosphodiesterase (PDE) inhibitorsIncreases the levels of cyclic adenosine monophosphate (cAMP), which leads to a decreased inflammatory response in the skin.Shows promising reduction in inflammatory acne lesions, contributing to clearer skin.[332,333]

5. Optimal Therapeutic Strategies for Varied Acne Severities

5.1. Acne Severity Grading

The accurate assessment of acne severity through standardized grading and classification systems is pivotal for guiding treatment strategies and evaluating therapeutic outcomes [337]. The Investigator Global Assessment (IGA) is the most widely utilized system in the United States due to its simplicity and proven correlation with patient self-assessments [338,339]. In assessing acne severity, the IGA Scale, sanctioned by the US FDA, serves as a critical tool, delineating five distinct grades. Grade 0 represents clear skin devoid of inflammatory or non-inflammatory lesions. Grade 1, described as “almost clear”, features rare non-inflammatory lesions alongside no more than one small inflammatory lesion. Grade 2, or “mild severity”, encompasses some non-inflammatory lesions with a few inflammatory lesions such as papules and pustules but excludes nodular lesions. Progressing to Grade 3, or “moderate severity”, the skin exhibits more non-inflammatory and some inflammatory lesions, with the inclusion of no more than one small nodular lesion. The highest severity, Grade 4, is categorized as “severe”, with numerous non-inflammatory and inflammatory lesions, including a few nodular lesions. Thus, the progression and escalating severity of acne is encapsulated and clinically assessed using this standardized scale [337,339,340].

5.2. Most Effective Treatment Combinations for Each Type

5.2.1. Mild to Moderate Acne

For the treatment of mild to moderate acne, the combination of benzoyl peroxide and topical retinoids is highly effective, addressing key pathophysiological elements such as abnormal keratinization and microbial colonization while reducing sebum production [3,103,104,105,106,107,108,109,111,112,113,114,115,116,120,122,123,124,125,126,130,131,132,133,134,135,136,138,146,177,178,179,180,181,182,183]. Topical clindamycin in combination with benzoyl peroxide is recommended, leveraging the antimicrobial properties of clindamycin and the resistance-lowering effects of benzoyl peroxide, thereby enhancing therapeutic efficacy [126,135,139,140,141,142,143,144].

5.2.2. Moderate to Severe Acne

In cases of moderate to severe inflammatory acne, a regimen comprising oral antibiotics, such as doxycycline or minocycline, alongside topical retinoids and benzoyl peroxide is advised to target deeper pathophysiological mechanisms, effectively reducing C. acne and mitigating antibiotic resistance [135,144,184,185,186,187,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206]. Hormonal therapies, including oral contraceptives and spironolactone, are particularly effective for women exhibiting acne flares linked to hormonal variations, addressing androgenic contributions to acne pathogenesis [135,146,207,208,209,210,211,214,215,217,218].

5.2.3. Severe, Nodular, or Treatment-Resistant Acne

Oral isotretinoin is recommended for severe, recalcitrant nodular acne or cases resistant to conventional treatments due to its profound impact on sebum production, follicular keratinization, and microbial populations, along with its anti-inflammatory properties [135,184,204,220,221,222,223,224,225,226,227,228,233,234,235,236,237,238,239,240,241,242]. In instances of severe inflammatory acne or acne fulminans where traditional treatments fail, systemic steroids or biologic agents such as TNF-α inhibitors may be utilized, providing significant anti-inflammatory effects and symptomatic relief [85,306,307,308,309,310].

6. Discussion and Conclusions

The complex relationship between the skin microbiome and acne pathophysiology has been highlighted, emphasizing the crucial roles of microbial diversity and specific bacterial species, such as C. acnes and coagulase-negative Staphylococcus, in this dermatological condition. These microorganisms are not merely present; they actively contribute to the development and exacerbation of acne through various mechanisms, including influencing sebum production and eliciting inflammatory responses.
The role of C. acnes, a key bacterium historically linked to acne, has been examined in detail. This bacterium thrives in oily environments provided by sebaceous glands and can trigger inflammation by activating the immune system. Its involvement varies, not just from person to person but also in its strain-specific behavior, which can either provoke or prevent inflammation. Recent studies suggest that specific strains of C. acnes are more commonly associated with healthy skin, while others are linked to chronic acne conditions, indicating the potential for targeted microbial treatments.
Probiotics and bacteriophages represent promising therapeutic avenues. Probiotics can help restore a healthy microbiome balance, potentially reducing acne severity by outcompeting harmful bacteria and reducing inflammation. Bacteriophages, which are viruses that infect bacteria, offer a novel approach to diminishing C. acnes populations without affecting the broader microbiome. This precision could prevent the development of antibiotic resistance, a growing concern with traditional acne treatments.
Environmental factors like diet and pollution were also discussed for their impact on acne. High-glycemic diets and increased dairy intake have been linked to increased sebum production and exacerbated acne symptoms. Pollutants can alter the skin barrier and microbiome, further aggravating or even triggering acne outbreaks. These factors highlight the importance of considering external influences in both the development and management of acne.
The genetic components of acne emphasize that genetic predispositions can influence an individual’s response to external triggers and treatment outcomes. The interplay between genes and the environment, including how they modulate the skin’s immune response and microbial landscape, is critical for understanding individual acne cases and for tailoring treatments accordingly.
In conclusion, this section of the paper advocates a holistic approach to acne treatment, one that incorporates not only topical and systemic therapies but also considers the patient’s lifestyle, environmental exposures, and microbial interactions. It calls for continued research into the gut–skin axis and its implications for acne, suggesting that future therapies could greatly benefit from integrating dietary and lifestyle modifications to support overall skin health and reduce acne severity. The ultimate goal is to move toward more personalized, effective, and sustainable acne management strategies that address the root causes of acne rather than just its symptoms.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Mitchell, B.L.; Saklatvala, J.R.; Dand, N.; Hagenbeek, F.A.; Li, X.; Min, J.L.; Thomas, L.; Bartels, M.; Jan Hottenga, J.; Lupton, M.K.; et al. Genome-wide association meta-analysis identifies 29 new acne susceptibility loci. Nat. Commun. 2022, 13, 702. [Google Scholar] [CrossRef] [PubMed]
  2. Ip, A.; Muller, I.; Geraghty, A.W.A.; McNiven, A.; Little, P.; Santer, M. Young people’s perceptions of acne and acne treatments: Secondary analysis of qualitative interview data. Br. J. Dermatol. 2020, 183, 349–356. [Google Scholar] [CrossRef] [PubMed]
  3. Vasam, M.; Korutla, S.; Bohara, R.A. Acne vulgaris: A review of the pathophysiology, treatment, and recent nanotechnology based advances. Biochem. Biophys. Rep. 2023, 36, 101578. [Google Scholar] [CrossRef] [PubMed]
  4. Clark, A.K.; Haas, K.N.; Sivamani, R.K. Edible plants and their influence on the gut microbiome and acne. Int. J. Mol. Sci. 2017, 18, 1070. [Google Scholar] [CrossRef] [PubMed]
  5. Cong, T.-X.; Hao, D.; Wen, X.; Li, X.-H.; He, G.; Jiang, X. From pathogenesis of acne vulgaris to anti-acne agents. Arch. Dermatol. Res. 2019, 311, 337–349. [Google Scholar] [CrossRef] [PubMed]
  6. Plewig, G.; Kligman, A.M. Acne and Rosacea; Springer Science & Business Media: Berlin, Germany, 2012. [Google Scholar]
  7. Bataille, V.; Snieder, H.; MacGregor, A.; Sasieni, P.; Spector, T. The influence of genetics and environmental factors in the pathogenesis of acne: A twin study of acne in women. J. Investig. Dermatol. 2002, 119, 1317–1322. [Google Scholar] [CrossRef]
  8. Di Landro, A.; Cazzaniga, S.; Parazzini, F.; Ingordo, V.; Cusano, F.; Atzori, L.; Cutrì, F.T.; Musumeci, M.L.; Zinetti, C.; Pezzarossa, E. Family history, body mass index, selected dietary factors, menstrual history, and risk of moderate to severe acne in adolescents and young adults. J. Am. Acad. Dermatol. 2012, 67, 1129–1135. [Google Scholar] [CrossRef] [PubMed]
  9. Evans, D.; Kirk, K.; Nyholt, D.; Novac, C.; Martin, N. Teenage acne is influenced by genetic factors. Br. J. Dermatol. 2005, 152, 579–581. [Google Scholar] [CrossRef] [PubMed]
  10. Friedman, G.D. Twin studies of disease heritability based on medical records: Application to acne vulgaris. Acta Genet. Med. Gemellol. Twin Res. 1984, 33, 487–495. [Google Scholar] [CrossRef]
  11. Ghodsi, S.Z.; Orawa, H.; Zouboulis, C.C. Prevalence, severity, and severity risk factors of acne in high school pupils: A community-based study. J. Investig. Dermatol. 2009, 129, 2136–2141. [Google Scholar] [CrossRef]
  12. Hecht, H. Hereditary Trends in Acne VulgarisPrevention of Acne. Dermatology 1960, 121, 297–307. [Google Scholar] [CrossRef] [PubMed]
  13. Lichtenberger, R.; Simpson, M.A.; Smith, C.; Barker, J.; Navarini, A.A. Genetic architecture of acne vulgaris. J. Eur. Acad. Dermatol. Venereol. 2017, 31, 1978–1990. [Google Scholar] [CrossRef] [PubMed]
  14. Mina-Vargas, A.; Colodro-Conde, L.; Grasby, K.; Zhu, G.; Gordon, S.; Medland, S.E.; Martin, N.G. Heritability and GWAS analyses of acne in Australian adolescent twins. Twin Res. Hum. Genet. 2017, 20, 541–549. [Google Scholar] [CrossRef] [PubMed]
  15. Tan, H.H.; Tan, A.; Barkham, T.; Yan, X.Y.; Zhu, M. Community-based study of acne vulgaris in adolescents in Singapore. Br. J. Dermatol. 2007, 157, 547–551. [Google Scholar] [CrossRef]
  16. Tan, J.K.; Bhate, K. A global perspective on the epidemiology of acne. Br. J. Dermatol. 2015, 172, 3–12. [Google Scholar] [CrossRef] [PubMed]
  17. Wei, B.; Pang, Y.; Zhu, H.; Qu, L.; Xiao, T.; Wei, H.C.; Chen, H.D.; He, C.D. The epidemiology of adolescent acne in North East China. J. Eur. Acad. Dermatol. Venereol. 2010, 24, 953–957. [Google Scholar] [CrossRef] [PubMed]
  18. Xu, S.; Wang, H.; Fan, X.; Sun, L.; Yang, S.; Wang, P.; Xiao, F.; Gao, M.; Cui, Y.; Ren, Y. The familial risk of acne vulgaris in Chinese Hans—A case-control study. J. Eur. Acad. Dermatol. Venereol. 2007, 21, 602–605. [Google Scholar] [CrossRef] [PubMed]
  19. Walton, S.; Wyatt, E.H.; Cunliffe, W.J. Genetic control of sebum excretion and acne—A twin study. Br. J. Dermatol. 1988, 118, 393–396. [Google Scholar] [CrossRef] [PubMed]
  20. Suggs, A.; Loesch, M.; Ezaldein, H.; Christensen, L.; Dawes, D.; Baron, E. An Acne Survey from the World’s Largest Annual Gathering of Twins. J. Drugs Dermatol. 2018, 17, 380–382. [Google Scholar]
  21. He, L.; Wu, W.-J.; Yang, J.-K.; Cheng, H.; Zuo, X.-B.; Lai, W.; Gao, T.-W.; Ma, C.-L.; Luo, N.; Huang, J.-Q.; et al. Two new susceptibility loci 1q24.2 and 11p11.2 confer risk to severe acne. Nat. Commun. 2014, 5, 2870. [Google Scholar] [CrossRef]
  22. Navarini, A.A.; Simpson, M.A.; Weale, M.; Knight, J.; Carlavan, I.; Reiniche, P.; Burden, D.A.; Layton, A.; Bataille, V.; Allen, M.; et al. Genome-wide association study identifies three novel susceptibility loci for severe Acne vulgaris. Nat. Commun. 2014, 5, 4020. [Google Scholar] [CrossRef] [PubMed]
  23. Zhang, M.; Qureshi, A.A.; Hunter, D.J.; Han, J. A genome-wide association study of severe teenage acne in European Americans. Hum. Genet. 2014, 133, 259–264. [Google Scholar] [CrossRef] [PubMed]
  24. Tasli, L.; Turgut, S.; Kaçar, N.; Ayada, C.; Çoban, M.; Akcilar, R.; Ergin, S. Insulin-like growth factor-I gene polymorphism in acne vulgaris. J. Eur. Acad. Dermatol. Venereol. 2013, 27, 254–257. [Google Scholar] [CrossRef]
  25. Yang, J.-K.; Wu, W.-J.; Qi, J.; He, L.; Zhang, Y.-P. TNF-308 G/A polymorphism and risk of acne vulgaris: A meta-analysis. PLoS ONE 2014, 9, e87806. [Google Scholar] [CrossRef] [PubMed]
  26. Zaenglein, A.L. Making the case for early treatment of acne. Clin. Pediatr. 2010, 49, 54–59. [Google Scholar] [CrossRef] [PubMed]
  27. Trivedi, N.R.; Gilliland, K.L.; Zhao, W.; Liu, W.; Thiboutot, D.M. Gene array expression profiling in acne lesions reveals marked upregulation of genes involved in inflammation and matrix remodeling. J. Investig. Dermatol. 2006, 126, 1071–1079. [Google Scholar] [CrossRef]
  28. Smith, R.N.; Mann, N.J.; Braue, A.; Mäkeläinen, H.; Varigos, G.A. The effect of a high-protein, low glycemic–load diet versus a conventional, high glycemic–load diet on biochemical parameters associated with acne vulgaris: A randomized, investigator-masked, controlled trial. J. Am. Acad. Dermatol. 2007, 57, 247–256. [Google Scholar] [CrossRef] [PubMed]
  29. Smith, R.; Mann, N.; Mäkeläinen, H.; Roper, J.; Braue, A.; Varigos, G. A pilot study to determine the short-term effects of a low glycemic load diet on hormonal markers of acne: A nonrandomized, parallel, controlled feeding trial. Mol. Nutr. Food Res. 2008, 52, 718–726. [Google Scholar] [CrossRef] [PubMed]
  30. Burris, J.; Shikany, J.M.; Rietkerk, W.; Woolf, K. A low glycemic index and glycemic load diet decreases insulin-like growth factor-1 among adults with moderate and severe acne: A short-duration, 2-week randomized controlled trial. J. Acad. Nutr. Diet. 2018, 118, 1874–1885. [Google Scholar] [CrossRef]
  31. Smith, R.N.; Braue, A.; Varigos, G.A.; Mann, N.J. The effect of a low glycemic load diet on acne vulgaris and the fatty acid composition of skin surface triglycerides. J. Dermatol. Sci. 2008, 50, 41–52. [Google Scholar] [CrossRef]
  32. Çerman, A.A.; Aktaş, E.; Altunay, İ.K.; Arıcı, J.E.; Tulunay, A.; Ozturk, F.Y. Dietary glycemic factors, insulin resistance, and adiponectin levels in acne vulgaris. J. Am. Acad. Dermatol. 2016, 75, 155–162. [Google Scholar] [CrossRef] [PubMed]
  33. Burris, J.; Rietkerk, W.; Woolf, K. Relationships of self-reported dietary factors and perceived acne severity in a cohort of New York young adults. J. Acad. Nutr. Diet. 2014, 114, 384–392. [Google Scholar] [CrossRef] [PubMed]
  34. Burris, J.; Rietkerk, W.; Shikany, J.M.; Woolf, K. Differences in dietary glycemic load and hormones in New York City adults with no and moderate/severe acne. J. Acad. Nutr. Diet. 2017, 117, 1375–1383. [Google Scholar] [CrossRef] [PubMed]
  35. Penso, L.; Touvier, M.; Deschasaux, M.; Hercberg, S.; Ezzedine, K.; Sbidian, E. Association between adult acne and dietary behaviors: Findings from the NutriNet-Santé Prospective Cohort Study. JAMA Dermatol. 2020, 156, 854–862. [Google Scholar] [CrossRef] [PubMed]
  36. Smith, R.N.; Mann, N.J.; Braue, A.; Mäkeläinen, H.; Varigos, G.A. A low-glycemic-load diet improves symptoms in acne vulgaris patients: A randomized controlled trial. Am. J. Clin. Nutr. 2007, 86, 107–115. [Google Scholar] [CrossRef] [PubMed]
  37. Kaymak, Y.; Adisen, E.; Ilter, N.; Bideci, A.; Gurler, D.; Celik, B. Dietary glycemic index and glucose, insulin, insulin-like growth factor-I, insulin-like growth factor binding protein 3, and leptin levels in patients with acne. J. Am. Acad. Dermatol. 2007, 57, 819–823. [Google Scholar] [CrossRef]
  38. Silverberg, N.B. Whey protein precipitating moderate to severe acne flares in 5 teenaged athletes. Cutis 2012, 90, 70–72. [Google Scholar] [PubMed]
  39. Adebamowo, C.A.; Spiegelman, D.; Berkey, C.S.; Danby, F.W.; Rockett, H.H.; Colditz, G.A.; Willett, W.C.; Holmes, M.D. Milk consumption and acne in adolescent girls. Dermatol. Online J. 2006, 12, 1. [Google Scholar] [CrossRef] [PubMed]
  40. Adebamowo, C.A.; Spiegelman, D.; Berkey, C.S.; Danby, F.W.; Rockett, H.H.; Colditz, G.A.; Willett, W.C.; Holmes, M.D. Milk consumption and acne in teenaged boys. J. Am. Acad. Dermatol. 2008, 58, 787–793. [Google Scholar] [CrossRef]
  41. Okoro, E.O.; Ogunbiyi, A.O.; George, A.O.; Subulade, M.O. Association of diet with acne vulgaris among adolescents in Ibadan, southwest Nigeria. Int. J. Dermatol. 2016, 55, 982–988. [Google Scholar] [CrossRef]
  42. Grossi, E.; Cazzaniga, S.; Crotti, S.; Naldi, L.; Di Landro, A.; Ingordo, V.; Cusano, F.; Atzori, L.; Tripodi Cutrì, F.; Musumeci, M. The constellation of dietary factors in adolescent acne: A semantic connectivity map approach. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 96–100. [Google Scholar] [CrossRef] [PubMed]
  43. Karadağ, A.S.; Balta, I.; Saricaoğlu, H.; Kilic, S.; Kelekçi, K.H.; Yildirim, M.; Arica, D.A.; Öztürk, S.; Karaman, G.; Cerman, A.A. The effect of personal, familial, and environmental characteristics on acne vulgaris: A prospective, multicenter, case controlled study. G. Ital. Dermatol. Venereol. Organo Uff. Soc. Ital. Dermatol. Sifilogr. 2017, 154, 177–185. [Google Scholar] [CrossRef] [PubMed]
  44. Sardana, K.; Sharma, R.C.; Sarkar, R. Seasonal variation in acne vulgaris—Myth or reality. J. Dermatol. 2002, 29, 484–488. [Google Scholar] [CrossRef] [PubMed]
  45. Li, X.; An, S.J.; Liu, X.L.; Ji, A.L.; Cao, Y.; Xiang, Y.; Ma, X.Y.; Hu, Q.; Yuan, Z.Q.; Li, Y.F.; et al. The Association Between Short-Term Air Pollution Exposure and Post-Adolescent Acne: The Evidence from a Time Series Analysis in Xi’an, China. Clin. Cosmet. Investig. Dermatol. 2021, 14, 723–731. [Google Scholar] [CrossRef] [PubMed]
  46. Isler, M.F.; Coates, S.J.; Boos, M.D. Climate change, the cutaneous microbiome and skin disease: Implications for a warming world. Int. J. Dermatol. 2023, 62, 337–345. [Google Scholar] [CrossRef] [PubMed]
  47. Jović, A.; Marinović, B.; Kostović, K.; Čeović, R.; Basta-Juzbašić, A.; Bukvić Mokos, Z. The Impact of Pyschological Stress on Acne. Acta Dermatovenerol. Croat. 2017, 25, 1133–1141. [Google Scholar]
  48. Kurokawa, I.; Danby, F.W.; Ju, Q.; Wang, X.; Xiang, L.F.; Xia, L.; Chen, W.; Nagy, I.; Picardo, M.; Suh, D.H.; et al. New developments in our understanding of acne pathogenesis and treatment. Exp. Dermatol. 2009, 18, 821–832. [Google Scholar] [CrossRef] [PubMed]
  49. Melnik, B.C. Linking diet to acne metabolomics, inflammation, and comedogenesis: An update. Clin. Cosmet. Investig. Dermatol. 2015, 8, 371–388. [Google Scholar] [CrossRef]
  50. Agamia, N.; Abdallah, D.; Sorour, O.; Mourad, B.; Younan, D. Skin expression of mammalian target of rapamycin and forkhead box transcription factor O1, and serum insulin-like growth factor-1 in patients with acne vulgaris and their relationship with diet. Br. J. Dermatol. 2016, 174, 1299–1307. [Google Scholar] [CrossRef]
  51. Monfrecola, G.; Lembo, S.; Caiazzo, G.; De Vita, V.; Di Caprio, R.; Balato, A.; Fabbrocini, G. Mechanistic target of rapamycin (mTOR) expression is increased in acne patients’ skin. Exp. Dermatol. 2016, 25, 153–155. [Google Scholar] [CrossRef]
  52. Cottle, D.L.; Kretzschmar, K.; Schweiger, P.J.; Quist, S.R.; Gollnick, H.P.; Natsuga, K.; Aoyagi, S.; Watt, F.M. c-MYC-induced sebaceous gland differentiation is controlled by an androgen receptor/p53 axis. Cell Rep. 2013, 3, 427–441. [Google Scholar] [CrossRef] [PubMed]
  53. He, T.-C.; Sparks, A.B.; Rago, C.; Hermeking, H.; Zawel, L.; Da Costa, L.T.; Morin, P.J.; Vogelstein, B.; Kinzler, K.W. Identification of c-MYC as a target of the APC pathway. Science 1998, 281, 1509–1512. [Google Scholar] [CrossRef] [PubMed]
  54. Kretzschmar, K.; Cottle, D.L.; Schweiger, P.J.; Watt, F.M. The androgen receptor antagonizes Wnt/β-catenin signaling in epidermal stem cells. J. Investig. Dermatol. 2015, 135, 2753–2763. [Google Scholar] [CrossRef] [PubMed]
  55. Melnik, B.C. FoxO1—The key for the pathogenesis and therapy of acne? JDDG J. Dtsch. Dermatol. Ges. 2010, 8, 105–114. [Google Scholar] [CrossRef] [PubMed]
  56. Hazarika, N. Acne vulgaris: New evidence in pathogenesis and future modalities of treatment. J. Dermatol. Treat. 2021, 32, 277–285. [Google Scholar] [CrossRef] [PubMed]
  57. Ben-Amitai, D.; Laron, Z. Effect of insulin-like growth factor-1 deficiency or administration on the occurrence of acne. J. Eur. Acad. Dermatol. Venereol. 2011, 25, 950–954. [Google Scholar] [CrossRef] [PubMed]
  58. Bohrer, L.R.; Liu, P.; Zhong, J.; Pan, Y.; Angstman, J.; Brand, L.J.; Dehm, S.M.; Huang, H. FOXO1 binds to the TAU5 motif and inhibits constitutively active androgen receptor splice variants. Prostate 2013, 73, 1017–1027. [Google Scholar] [CrossRef] [PubMed]
  59. Chen, C.-C.; Jeon, S.-M.; Bhaskar, P.T.; Nogueira, V.; Sundararajan, D.; Tonic, I.; Park, Y.; Hay, N. FoxOs inhibit mTORC1 and activate Akt by inducing the expression of Sestrin3 and Rictor. Dev. Cell 2010, 18, 592–604. [Google Scholar] [CrossRef] [PubMed]
  60. Fan, W.; Yanase, T.; Morinaga, H.; Okabe, T.; Nomura, M.; Daitoku, H.; Fukamizu, A.; Kato, S.; Takayanagi, R.; Nawata, H. Insulin-like growth factor 1/insulin signaling activates androgen signaling through direct interactions of Foxo1 with androgen receptor. J. Biol. Chem. 2007, 282, 7329–7338. [Google Scholar] [CrossRef]
  61. Guevara-Aguirre, J.; Balasubramanian, P.; Guevara-Aguirre, M.; Wei, M.; Madia, F.; Cheng, C.-W.; Hwang, D.; Martin-Montalvo, A.; Saavedra, J.; Ingles, S. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci. Transl. Med. 2011, 3, 70ra13. [Google Scholar] [CrossRef]
  62. Kamei, Y.; Miura, S.; Suganami, T.; Akaike, F.; Kanai, S.; Sugita, S.; Katsumata, A.; Aburatani, H.; Unterman, T.G.; Ezaki, O. Regulation of SREBP1c gene expression in skeletal muscle: Role of retinoid X receptor/liver X receptor and forkhead-O1 transcription factor. Endocrinology 2008, 149, 2293–2305. [Google Scholar] [CrossRef] [PubMed]
  63. Klinger, B.; Anin, S.; Silbergeld, A.; Eshet, R.; Laron, Z. Development of hyperandrogenism during treatment with insulin-like growth factor-I (IGF-I) in female patients with Laron syndrome. Clin. Endocrinol. 1998, 48, 81–87. [Google Scholar] [CrossRef] [PubMed]
  64. Ma, Q.; Fu, W.; Li, P.; Nicosia, S.V.; Jenster, G.; Zhang, X.; Bai, W. FoxO1 mediates PTEN suppression of androgen receptor N-and C-terminal interactions and coactivator recruitment. Mol. Endocrinol. 2009, 23, 213–225. [Google Scholar] [CrossRef]
  65. Yoon, J.Y.; Kwon, H.H.; Min, S.U.; Thiboutot, D.M.; Suh, D.H. Epigallocatechin-3-gallate improves acne in humans by modulating intracellular molecular targets and inhibiting P. acnes. J. Investig. Dermatol. 2013, 133, 429–440. [Google Scholar] [CrossRef] [PubMed]
  66. Ganceviciene, R.; Graziene, V.; Fimmel, S.; Zouboulis, C. Involvement of the corticotropin-releasing hormone system in the pathogenesis of acne vulgaris. Br. J. Dermatol. 2009, 160, 345–352. [Google Scholar] [CrossRef]
  67. Toyoda, M.; Nakamura, M.; Morohashi, M. Neuropeptides and sebaceous glands. Eur. J. Dermatol. 2002, 12, 422–427. [Google Scholar] [PubMed]
  68. Kutlu, O.; Karadag, A.S.; Wollina, U. Adult acne versus adolescent acne: A narrative review with a focus on epidemiology to treatment. An. Bras. Dermatol. 2023, 98, 75–83. [Google Scholar] [CrossRef]
  69. Dreno, B.; Bagatin, E.; Blume-Peytavi, U.; Rocha, M.; Gollnick, H. Female type of adult acne: Physiological and psychological considerations and management. J. Dtsch. Dermatol. Ges. 2018, 16, 1185–1194. [Google Scholar] [CrossRef]
  70. Ramasamy, S.; Barnard, E.; Dawson, T., Jr.; Li, H. The role of the skin microbiota in acne pathophysiology. Br. J. Dermatol. 2019, 181, 691–699. [Google Scholar] [CrossRef]
  71. Dréno, B.; Pécastaings, S.; Corvec, S.; Veraldi, S.; Khammari, A.; Roques, C. Cutibacterium acnes (Propionibacterium acnes) and acne vulgaris: A brief look at the latest updates. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 5–14. [Google Scholar] [CrossRef]
  72. Scholz, C.F.; Kilian, M. The natural history of cutaneous propionibacteria, and reclassification of selected species within the genus Propionibacterium to the proposed novel genera Acidipropionibacterium gen. nov., Cutibacterium gen. nov. and Pseudopropionibacterium gen. nov. Int. J. Syst. Evol. Microbiol. 2016, 66, 4422–4432. [Google Scholar] [CrossRef]
  73. Dréno, B.; Dagnelie, M.A.; Khammari, A.; Corvec, S. The skin microbiome: A new actor in inflammatory acne. Am. J. Clin. Dermatol. 2020, 21, 18–24. [Google Scholar] [CrossRef]
  74. Beylot, C.; Auffret, N.; Poli, F.; Claudel, J.P.; Leccia, M.T.; Del Giudice, P.; Dreno, B. Propionibacterium acnes: An update on its role in the pathogenesis of acne. J. Eur. Acad. Dermatol. Venereol. 2014, 28, 271–278. [Google Scholar] [CrossRef] [PubMed]
  75. Vaughn, A.R.; Notay, M.; Clark, A.K.; Sivamani, R.K. Skin-gut axis: The relationship between intestinal bacteria and skin health. World J. Dermatol. 2017, 6, 52–58. [Google Scholar] [CrossRef]
  76. Lee, Y.B.; Byun, E.J.; Kim, H.S. Potential role of the microbiome in acne: A comprehensive review. J. Clin. Med. 2019, 8, 987. [Google Scholar] [CrossRef]
  77. Salem, I.; Ramser, A.; Isham, N.; Ghannoum, M.A. The gut microbiome as a major regulator of the gut-skin axis. Front. Microbiol. 2018, 9, 382698. [Google Scholar] [CrossRef]
  78. Kim, S.; Jazwinski, S.M. The gut microbiota and healthy aging: A mini-review. Gerontology 2018, 64, 513–520. [Google Scholar] [CrossRef] [PubMed]
  79. Turnbaugh, P.J.; Bäckhed, F.; Fulton, L.; Gordon, J.I. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 2008, 3, 213–223. [Google Scholar] [CrossRef] [PubMed]
  80. Yan, H.M.; Zhao, H.J.; Guo, D.Y.; Zhu, P.Q.; Zhang, C.L.; Jiang, W. Gut microbiota alterations in moderate to severe acne vulgaris patients. J. Dermatol. 2018, 45, 1166–1171. [Google Scholar] [CrossRef]
  81. Noureldein, M.H.; Eid, A.A. Gut microbiota and mTOR signaling: Insight on a new pathophysiological interaction. Microb. Pathog. 2018, 118, 98–104. [Google Scholar] [CrossRef]
  82. Szántó, M.; Dózsa, A.; Antal, D.; Szabó, K.; Kemény, L.; Bai, P. Targeting the gut-skin axis—Probiotics as new tools for skin disorder management? Exp. Dermatol. 2019, 28, 1210–1218. [Google Scholar] [CrossRef] [PubMed]
  83. Kim, J.; Ochoa, M.-T.; Krutzik, S.R.; Takeuchi, O.; Uematsu, S.; Legaspi, A.J.; Brightbill, H.D.; Holland, D.; Cunliffe, W.J.; Akira, S. Activation of toll-like receptor 2 in acne triggers inflammatory cytokine responses. J. Immunol. 2002, 169, 1535–1541. [Google Scholar] [CrossRef] [PubMed]
  84. Kurokawa, I.; Nakase, K. Recent advances in understanding and managing acne. F1000Research 2020, 9, 792. [Google Scholar] [CrossRef] [PubMed]
  85. Kelhälä, H.-L.; Palatsi, R.; Fyhrquist, N.; Lehtimäki, S.; Väyrynen, J.P.; Kallioinen, M.; Kubin, M.E.; Greco, D.; Tasanen, K.; Alenius, H. IL-17/Th17 pathway is activated in acne lesions. PLoS ONE 2014, 9, e105238. [Google Scholar] [CrossRef] [PubMed]
  86. Layton, A.; Morris, C.; Cunliffe, W.; Ingham, E. Immunohistochemical investigation of evolving inflammation in lesions of acne vulgaris. Exp. Dermatol. 1998, 7, 191–197. [Google Scholar] [CrossRef] [PubMed]
  87. Jeremy, A.H.; Holland, D.B.; Roberts, S.G.; Thomson, K.F.; Cunliffe, W.J. Inflammatory events are involved in acne lesion initiation. J. Investig. Dermatol. 2003, 121, 20–27. [Google Scholar] [CrossRef]
  88. Kistowska, M.; Meier, B.; Proust, T.; Feldmeyer, L.; Cozzio, A.; Kuendig, T.; Contassot, E.; French, L.E. Propionibacterium acnes promotes Th17 and Th17/Th1 responses in acne patients. J. Investig. Dermatol. 2015, 135, 110–118. [Google Scholar] [CrossRef]
  89. Kang, S.; Cho, S.; Chung, J.H.; Hammerberg, C.; Fisher, G.J.; Voorhees, J.J. Inflammation and extracellular matrix degradation mediated by activated transcription factors nuclear factor-κB and activator protein-1 in inflammatory acne lesions in vivo. Am. J. Pathol. 2005, 166, 1691–1699. [Google Scholar] [CrossRef]
  90. Kim, J. Review of the innate immune response in acne vulgaris: Activation of Toll-like receptor 2 in acne triggers inflammatory cytokine responses. Dermatology 2005, 211, 193–198. [Google Scholar] [CrossRef]
  91. Carlavan, I.; Bertino, B.; Rivier, M.; Martel, P.; Bourdes, V.; Motte, M.; Déret, S.; Reiniche, P.; Menigot, C.; Khammari, A. Atrophic scar formation in patients with acne involves long-acting immune responses with plasma cells and alteration of sebaceous glands. Br. J. Dermatol. 2018, 179, 906–917. [Google Scholar] [CrossRef]
  92. Holland, D.; Jeremy, A.; Roberts, S.; Seukeran, D.; Layton, A.; Cunliffe, W. Inflammation in acne scarring: A comparison of the responses in lesions from patients prone and not prone to scar. Br. J. Dermatol. 2004, 150, 72–81. [Google Scholar] [CrossRef] [PubMed]
  93. Kurokawa, I.; Layton, A.M.; Ogawa, R. Updated Treatment for Acne: Targeted Therapy Based on Pathogenesis. Dermatol. Ther. 2021, 11, 1129–1139. [Google Scholar] [CrossRef] [PubMed]
  94. Im, M.; Kim, S.Y.; Sohn, K.C.; Choi, D.K.; Lee, Y.; Seo, Y.J.; Kim, C.D.; Hwang, Y.L.; Zouboulis, C.C.; Lee, J.H. Epigallocatechin-3-gallate suppresses IGF-I-induced lipogenesis and cytokine expression in SZ95 sebocytes. J. Investig. Dermatol. 2012, 132, 2700–2708. [Google Scholar] [CrossRef] [PubMed]
  95. Kim, H.; Moon, S.Y.; Sohn, M.Y.; Lee, W.J. Insulin-like growth factor-1 increases the expression of inflammatory biomarkers and sebum production in cultured sebocytes. Ann. Dermatol. 2017, 29, 20. [Google Scholar] [CrossRef] [PubMed]
  96. Hobbs, C.J.; Plymate, S.R.; Rosen, C.J.; Adler, R.A. Testosterone administration increases insulin-like growth factor-I levels in normal men. J. Clin. Endocrinol. Metab. 1993, 77, 776–779. [Google Scholar] [PubMed]
  97. Mattii, M.; Lovászi, M.; Garzorz, N.; Atenhan, A.; Quaranta, M.; Lauffer, F.; Konstantinow, A.; Küpper, M.; Zouboulis, C.C.; Kemény, L. Sebocytes contribute to skin inflammation by promoting the differentiation of T helper 17 cells. Br. J. Dermatol. 2018, 178, 722–730. [Google Scholar] [CrossRef] [PubMed]
  98. Papakonstantinou, E.; Aletras, A.J.; Glass, E.; Tsogas, P.; Dionyssopoulos, A.; Adjaye, J.; Fimmel, S.; Gouvousis, P.; Herwig, R.; Lehrach, H. Matrix metalloproteinases of epithelial origin in facial sebum of patients with acne and their regulation by isotretinoin. J. Investig. Dermatol. 2005, 125, 673–684. [Google Scholar] [CrossRef]
  99. Akamatsu, H.; Horio, T.; Hattori, K. Increased hydrogen peroxide generation by neutrophils from patients with acne inflammation. Int. J. Dermatol. 2003, 42, 366–369. [Google Scholar] [CrossRef] [PubMed]
  100. Dreno, B.; Kang, S.; Leyden, J.; York, J. Update: Mechanisms of Topical Retinoids in Acne. J. Drugs Dermatol. 2022, 21, 734–740. [Google Scholar] [PubMed]
  101. Baldwin, H.; Webster, G.; Stein Gold, L.; Callender, V.; Cook-Bolden, F.E.; Guenin, E. 50 Years of Topical Retinoids for Acne: Evolution of Treatment. Am. J. Clin. Dermatol. 2021, 22, 315–327. [Google Scholar] [CrossRef]
  102. Motamedi, M.; Chehade, A.; Sanghera, R.; Grewal, P. A Clinician’s Guide to Topical Retinoids. J. Cutan. Med. Surg. 2022, 26, 71–78. [Google Scholar] [CrossRef] [PubMed]
  103. Cosio, T.; Di Prete, M.; Gaziano, R.; Lanna, C.; Orlandi, A.; Di Francesco, P.; Bianchi, L.; Campione, E. Trifarotene: A Current Review and Perspectives in Dermatology. Biomedicines 2021, 9, 237. [Google Scholar] [CrossRef] [PubMed]
  104. Kolli, S.S.; Pecone, D.; Pona, A.; Cline, A.; Feldman, S.R. Topical retinoids in acne vulgaris: A systematic review. Am. J. Clin. Dermatol. 2019, 20, 345–365. [Google Scholar] [CrossRef] [PubMed]
  105. Zasada, M.; Budzisz, E. Retinoids: Active molecules influencing skin structure formation in cosmetic and dermatological treatments. Adv. Dermatol. Allergol./Postępy Dermatol. Alergol. 2019, 36, 392–397. [Google Scholar] [CrossRef] [PubMed]
  106. Drake, L.; Reyes-Hadsall, S.; Barbieri, J.S.; Mostaghimi, A. New Developments in Topical Acne Therapy. Am. J. Clin. Dermatol. 2022, 23, 125–136. [Google Scholar] [CrossRef] [PubMed]
  107. Eichenfield, L.F.; Sugarman, J.L.; Guenin, E.; Harris, S.; Bhatt, V. Novel tretinoin 0.05% lotion for the once-daily treatment of moderate-to-severe acne vulgaris in a preadolescent population. Pediatr. Dermatol. 2019, 36, 193–199. [Google Scholar] [CrossRef] [PubMed]
  108. Lain, E.; Day, D.; Harper, J.; Guenin, E. Tretinoin 0.05% lotion for the once-daily treatment of moderate-to-severe acne vulgaris: Impact of gender and race on efficacy and safety. J. Drugs Dermatol. JDD 2019, 18, 1128–1138. [Google Scholar] [PubMed]
  109. Eichenfield, L.F.; Matiz, C.; Funk, A.; Dill, S.W. Study of the efficacy and tolerability of 0.04% tretinoin microsphere gel for preadolescent acne. Pediatrics 2010, 125, e1316–e1323. [Google Scholar] [CrossRef]
  110. Cunliffe, W.J.; Poncet, M.; Loesche, C.; Verschoore, M. A comparison of the efficacy and tolerability of adapalene 0.1% gel versus tretinoin 0.025% gel in patients with acne vulgaris: A meta-analysis of five randomized trials. Br. J. Dermatol. 1998, 139 (Suppl. S52), 48–56. [Google Scholar] [CrossRef]
  111. Kim, S.Y.; Ochsendorf, F.R. New developments in acne treatment: Role of combination adapalene–benzoylperoxide. Ther. Clin. Risk Manag. 2016, 12, 1497–1506. [Google Scholar] [CrossRef]
  112. Tanghetti, E.A.; Werschler, W.P.; Lain, T.; Guenin, E.; Martin, G.; Pillai, R. Tazarotene 0.045% Lotion for Once-Daily Treatment of Moderate-to-Severe Acne Vulgaris: Results from Two Phase 3 Trials. J. Drugs Dermatol. 2020, 19, 70–77. [Google Scholar] [CrossRef] [PubMed]
  113. Tanghetti, E.A.; Kircik, L.H.; Green, L.J.; Guenin, E.; Harris, S.; Martin, G.; Pillai, R. A Phase 2, Multicenter, Double-Blind, Randomized, Vehicle-Controlled Clinical Study to Compare the Safety and Efficacy of a Novel Tazarotene 0.045% Lotion and Tazarotene 0.1% Cream in the Treatment of Moderate-to-Severe Acne Vulgaris. J. Drugs Dermatol. 2019, 18, 542. [Google Scholar] [PubMed]
  114. Han, G.; Wu, J.J.; Del Rosso, J.Q. Use of topical tazarotene for the treatment of acne vulgaris in pregnancy: A literature review. J. Clin. Aesthetic Dermatol. 2020, 13, E59. [Google Scholar]
  115. Gregoriou, S.; Kritsotaki, E.; Katoulis, A.; Rigopoulos, D. Use of tazarotene foam for the treatment of acne vulgaris. Clin. Cosmet. Investig. Dermatol. 2014, 2014, 165–170. [Google Scholar]
  116. Tamer, F.; Adişen, E. Topical tazarotene use in dermatology: An update. Gazi Med. J. 2020, 31, 86–89. [Google Scholar]
  117. Webster, G.F.; Berson, D.; Stein, L.F.; Fivenson, D.P.; Tanghetti, E.A.; Ling, M. Efficacy and tolerability of once-daily tazarotene 0.1% gel versus once-daily tretinoin 0.025% gel in the treatment of facial acne vulgaris: A randomized trial. Cutis 2001, 67, 4–9. [Google Scholar] [PubMed]
  118. Webster, G.F.; Guenther, L.; Poulin, Y.P.; Solomon, B.A.; Loven, K.; Lee, J. A multicenter, double-blind, randomized comparison study of the efficacy and tolerability of once-daily tazarotene 0.1% gel and adapalene 0.1% gel for the treatment of facial acne vulgaris. Cutis 2002, 69, 4–11. [Google Scholar]
  119. Leyden, J.J.; Shalita, A.; Thiboutot, D.; Washenik, K.; Webster, G. Topical retinoids in inflammatory acne: A retrospective, investigator-blinded, vehicle-controlled, photographic assessment. Clin. Ther. 2005, 27, 216–224. [Google Scholar] [CrossRef] [PubMed]
  120. Thoreau, E.; Arlabosse, J.M.; Bouix-Peter, C.; Chambon, S.; Chantalat, L.; Daver, S.; Dumais, L.; Duvert, G.; Feret, A.; Ouvry, G.; et al. Structure-based design of Trifarotene (CD5789), a potent and selective RARγ agonist for the treatment of acne. Bioorg Med. Chem. Lett. 2018, 28, 1736–1741. [Google Scholar] [CrossRef]
  121. Tan, J.; Thiboutot, D.; Popp, G.; Gooderham, M.; Lynde, C.; Del Rosso, J.; Weiss, J.; Blume-Peytavi, U.; Weglovska, J.; Johnson, S.; et al. Randomized phase 3 evaluation of trifarotene 50 μg/g cream treatment of moderate facial and truncal acne. J. Am. Acad. Dermatol. 2019, 80, 1691–1699. [Google Scholar] [CrossRef]
  122. Tan, J.; Bissonnette, R.; Gratton, D.; Kerrouche, N.; Canosa, J.M. The safety and efficacy of four different fixed combination regimens of adapalene 0.1%/benzoyl peroxide 2.5% gel for the treatment of acne vulgaris: Results from a randomised controlled study. Eur. J. Dermatol. 2018, 28, 502–508. [Google Scholar] [CrossRef] [PubMed]
  123. Bertolani, M.B.; Rodighiero, E.; Gandolfi, M.; Lotti, T.; Pedrazzi, G.; Puviani, M.; Milani, M.; Feliciani, C.; Satolli, F. Efficacy and tolerability of short contact therapy with tretinoin, clindamycin, and glycolic acid gel in acne: A randomized, controlled, assessor-blinded two-center trial: The MASCOTTE study. Dermatol. Ther. 2021, 34, e14724. [Google Scholar] [CrossRef] [PubMed]
  124. Eichenfield, D.Z.; Sprague, J.; Eichenfield, L.F. Management of Acne Vulgaris: A Review. JAMA 2021, 326, 2055–2067. [Google Scholar] [CrossRef] [PubMed]
  125. Tyring, S.K.; Kircik, L.H.; Pariser, D.M.; Guenin, E.; Bhatt, V.; Pillai, R. Novel Tretinoin 0.05% Lotion for the Once-Daily Treatment of Moderate-to-Severe Acne Vulgaris: Assessment of Efficacy and Safety in Patients Aged 9 Years and Older. J. Drugs Dermatol. 2018, 17, 1084–1091. [Google Scholar] [PubMed]
  126. Mohsin, N.; Hernandez, L.E.; Martin, M.R.; Does, A.V.; Nouri, K. Acne treatment review and future perspectives. Dermatol. Ther. 2022, 35, e15719. [Google Scholar] [CrossRef] [PubMed]
  127. Kontzias, C.; Zaino, M.; Feldman, S.R. Tretinoin 0.1% and Benzoyl Peroxide 3% Cream for the Treatment of Facial Acne Vulgaris. Ann. Pharmacother. 2023, 57, 1088–1093. [Google Scholar] [CrossRef] [PubMed]
  128. Stein Gold, L.; Lain, E.; Del Rosso, J.Q.; Gold, M.; Draelos, Z.D.; Eichenfield, L.F.; Sadick, N.; Werschler, W.P.; Gooderham, M.J.; Lupo, M. Clindamycin phosphate 1.2%/adapalene 0.15%/benzoyl peroxide 3.1% gel for moderate-to-severe acne: Efficacy and safety results from two randomized phase 3 trials. J. Am. Acad. Dermatol. 2023, 89, 927–935. [Google Scholar] [CrossRef] [PubMed]
  129. Del Rosso, J.Q.; Gold, L.S.; Draelos, Z.; Raoof, T.J.; Hooper, D.; Stuart, I. A prospective, multicenter, randomized, double-blind, vehicle-controlled phase 2 study to evaluate the safety and efficacy of a combination of 3% minocycline and 0.3% adapalene topical foam formulation for the treatment of moderate-to-severe acne. SKIN J. Cutan. Med. 2020, 4, s90. [Google Scholar] [CrossRef]
  130. Sagransky, M.; Yentzer, B.A.; Feldman, S.R. Benzoyl peroxide: A review of its current use in the treatment of acne vulgaris. Expert. Opin. Pharmacother. 2009, 10, 2555–2562. [Google Scholar] [CrossRef]
  131. Hegemann, L.; Toso, S.; Kitay, K.; Webster, C. Anti-inflammatory actions of benzoyl peroxide: Effects on the generation of reactive oxygen species by leucocytes and the activity of protein kinase C and calmodulin. Br. J. Dermatol. 1994, 130, 569–575. [Google Scholar] [CrossRef]
  132. Mohd Nor, N.H.; Aziz, Z. A systematic review of benzoyl peroxide for acne vulgaris. J. Dermatol. Treat. 2013, 24, 377–386. [Google Scholar] [CrossRef] [PubMed]
  133. Leyden, J.J.; Wortzman, M.; Baldwin, E.K. Antibiotic-resistant Propionibacterium acnes suppressed by a benzoyl peroxide cleanser 6%. Cutis 2008, 82, 417–421. [Google Scholar]
  134. Kumar, S.; Devi, B.; Goud, V. A comparative study of acne vulgaris with special reference to therapeutic options. IP Indian J. Clin. Exp. Dermatol. 2019, 5, 306–311. [Google Scholar] [CrossRef]
  135. Zaenglein, A.L.; Pathy, A.L.; Schlosser, B.J.; Alikhan, A.; Baldwin, H.E.; Berson, D.S.; Bowe, W.P.; Graber, E.M.; Harper, J.C.; Kang, S.; et al. Guidelines of care for the management of acne vulgaris. J. Am. Acad. Dermatol. 2016, 74, 945–973.e33. [Google Scholar] [CrossRef]
  136. Thiboutot, D.; Del Rosso, J.Q. Acne Vulgaris and the Epidermal Barrier: Is Acne Vulgaris Associated with Inherent Epidermal Abnormalities that Cause Impairment of Barrier Functions? Do Any Topical Acne Therapies Alter the Structural and/or Functional Integrity of the Epidermal Barrier? J. Clin. Aesthet. Dermatol. 2013, 6, 18–24. [Google Scholar]
  137. Boonchaya, P.; Rojhirunsakool, S.; Kamanamool, N.; Khunkhet, S.; Yooyongsatit, S.; Udompataikul, M.; Taweechotipatr, M. Minimum contact time of 1.25%, 2.5%, 5%, and 10% benzoyl peroxide for a bactericidal effect against Cutibacterium acnes. Clin. Cosmet. Investig. Dermatol. 2022, 2022, 403–409. [Google Scholar] [CrossRef]
  138. Leyden, J.J.; Hickman, J.G.; Jarratt, M.T.; Stewart, D.M.; Levy, S.F. The efficacy and safety of a combination benzoyl peroxide/clindamycin topical gel compared with benzoyl peroxide alone and a benzoyl peroxide/erythromycin combination product. J. Cutan. Med. Surg. 2001, 5, 37–42. [Google Scholar] [CrossRef]
  139. Mosler, E.L.; Leitner, C.; Gouda, M.A.; Carter, B.; Layton, A.M.; KhalafAllah, M.T. Topical antibiotics for acne. Cochrane Database Syst. Rev. 2018, CD012263. [Google Scholar] [CrossRef]
  140. Raoof, T.J.; Hooper, D.; Moore, A.; Zaiac, M.; Sullivan, T.; Kircik, L.; Lain, E.; Jankicevic, J.; Stuart, I. Efficacy and safety of a novel topical minocycline foam for the treatment of moderate to severe acne vulgaris: A phase 3 study. J. Am. Acad. Dermatol. 2020, 82, 832–837. [Google Scholar] [CrossRef]
  141. Gold, L.S.; Dhawan, S.; Weiss, J.; Draelos, Z.D.; Ellman, H.; Stuart, I.A. A novel topical minocycline foam for the treatment of moderate-to-severe acne vulgaris: Results of 2 randomized, double-blind, phase 3 studies. J. Am. Acad. Dermatol. 2019, 80, 168–177. [Google Scholar] [CrossRef]
  142. Paik, J. Topical Minocycline Foam 4%: A Review in Acne Vulgaris. Am. J. Clin. Dermatol. 2020, 21, 449–456. [Google Scholar] [CrossRef] [PubMed]
  143. Stein Gold, L.; Dhawan, S.; Weiss, J.; Draelos, Z.D.; Ellman, H.; Stuart, I. Open-label Extension Study Evaluating Long-term Safety and Efficacy of FMX101 4% Minocycline Foam for Moderate-to-Severe Acne Vulgaris. J. Clin. Aesthet. Dermatol. 2019, 12, 16–23. [Google Scholar]
  144. Patel, M.; Bowe, W.P.; Heughebaert, C.; Shalita, A.R. The development of antimicrobial resistance due to the antibiotic treatment of acne vulgaris: A review. J. Drugs Dermatol. JDD 2010, 9, 655–664. [Google Scholar] [PubMed]
  145. Tomić, I.; Miočić, S.; Pepić, I.; Šimić, D.; Filipović-Grčić, J. Efficacy and safety of azelaic acid nanocrystal-loaded in situ hydrogel in the treatment of acne vulgaris. Pharmaceutics 2021, 13, 567. [Google Scholar] [CrossRef] [PubMed]
  146. Zaenglein, A.L. Acne Vulgaris. N. Engl. J. Med. 2018, 379, 1343–1352. [Google Scholar] [CrossRef] [PubMed]
  147. Webster, G. Combination azelaic acid therapy for acne vulgaris. J. Am. Acad. Dermatol. 2000, 43, S47–S50. [Google Scholar] [CrossRef]
  148. Hashim, P.W.; Chen, T.; Harper, J.C.; Kircik, L.H. The Efficacy and Safety of Azelaic Acid 15% Foam in the Treatment of Facial Acne Vulgaris. J. Drugs Dermatol. JDD 2018, 17, 641–645. [Google Scholar] [PubMed]
  149. Chauhan, P.N.; Sharma, A.; Rasheed, H.; Mathur, H.; Sharma, P. Treatment Opportunities and Technological Progress Prospective for Acne Vulgaris. Curr. Drug Deliv. 2023, 20, 1037–1048. [Google Scholar] [CrossRef] [PubMed]
  150. Draelos, Z.; Lewis, J.; McHugh, L.; Pellegrino, A.; Popescu, L. Novel retinoid ester in combination with salicylic acid for the treatment of acne. J. Cosmet. Dermatol. 2016, 15, 36–42. [Google Scholar] [CrossRef]
  151. Calvisi, L. Efficacy of a combined chemical peel and topical salicylic acid-based gel combination in the treatment of active acne. J. Cosmet. Dermatol. 2021, 20, 2–6. [Google Scholar] [CrossRef]
  152. Bilal, H.; Xiao, Y.; Khan, M.N.; Chen, J.; Wang, Q.; Zeng, Y.; Lin, X. Stabilization of acne vulgaris-associated microbial dysbiosis with 2% supramolecular salicylic acid. Pharmaceuticals 2023, 16, 87. [Google Scholar] [CrossRef] [PubMed]
  153. Stotland, M.; Shalita, A.R.; Kissling, R.F. Dapsone 5% gel: A review of its efficacy and safety in the treatment of acne vulgaris. Am. J. Clin. Dermatol. 2009, 10, 221–227. [Google Scholar] [CrossRef] [PubMed]
  154. Beheshti-Mall, L.; Shafaroodi, H.; Jafariazar, Z.; Afshar, M. A novel hydrogel-thickened microemulsion of dapsone for acne treatment: Development, characterization, physicochemical stability and ex vivo permeation studies. Marmara Pharm. J. 2018, 22, 267–276. [Google Scholar] [CrossRef]
  155. Al Sabaa, H.; Mady, F.M.; Hussein, A.K.; Abdel-Wahab, H.M.; Ragaie, M.H. Dapsone in topical niosomes for treatment of acne vulgaris. Afr. J. Pharm. Pharmacol. 2018, 12, 221–230. [Google Scholar]
  156. Mohamed, G.N.; Gharib, K.M.; Samir, M.A. Emerging role of dapsone in the management of acne vulgaris. Egypt. J. Hosp. Med. 2022, 87, 1204–1207. [Google Scholar] [CrossRef]
  157. Lucky, A.W.; Maloney, J.M.; Roberts, J.; Taylor, S.; Jones, T.; Ling, M.; Garrett, S. Dapsone gel 5% for the treatment of acne vulgaris: Safety and efficacy of long-term (1 year) treatment. J. Drugs Dermatol. 2007, 6, 981–987. [Google Scholar] [PubMed]
  158. Lynde, C.W.; Andriessen, A. Cohort study on the treatment with dapsone 5% gel of mild to moderate inflammatory acne of the face in women. Skinmed 2014, 12, 15–21. [Google Scholar] [PubMed]
  159. Alexis, A.F.; Burgess, C.; Callender, V.D.; Herzog, J.L.; Roberts, W.E.; Schweiger, E.S.; Stockton, T.C.; Gallagher, C.J. The Efficacy and Safety of Topical Dapsone Gel, 5% for the Treatment of Acne Vulgaris in Adult Females With Skin of Color. J. Drugs Dermatol. 2016, 15, 197–204. [Google Scholar] [PubMed]
  160. Piette, W.W.; Taylor, S.; Pariser, D.; Jarratt, M.; Sheth, P.; Wilson, D. Hematologic safety of dapsone gel, 5%, for topical treatment of acne vulgaris. Arch. Dermatol. 2008, 144, 1564–1570. [Google Scholar] [CrossRef]
  161. Draelos, Z.D.; Carter, E.; Maloney, J.M.; Elewski, B.; Poulin, Y.; Lynde, C.; Garrett, S.; United States/Canada Dapsone Gel Study Group. Two randomized studies demonstrate the efficacy and safety of dapsone gel, 5% for the treatment of acne vulgaris. J. Am. Acad. Dermatol. 2007, 56, 439.e1–439.e10. [Google Scholar] [CrossRef]
  162. Akhavan, A.; Bershad, S. Topical acne drugs: Review of clinical properties, systemic exposure, and safety. Am. J. Clin. Dermatol. 2003, 4, 473–492. [Google Scholar] [CrossRef]
  163. Pace, W.E. A Benzoyl Peroxide-Sulfur Cream for acne vulgaris. Can. Med. Assoc. J. 1965, 93, 252–254. [Google Scholar]
  164. Hashem, N.M.; Hosny, A.; Abdelrahman, A.A.; Zakeer, S. Antimicrobial activities encountered by sulfur nanoparticles combating Staphylococcal species harboring sccmecA recovered from acne vulgaris. AIMS Microbiol. 2021, 7, 481–498. [Google Scholar] [CrossRef]
  165. Del Rosso, J.Q.; Leyden, J.J.; Thiboutot, D.; Webster, G.F. Antibiotic use in acne vulgaris and rosacea: Clinical considerations and resistance issues of significance to dermatologists. Cutis 2008, 82, 5–12. [Google Scholar]
  166. Breneman, D.L.; Ariano, M.C. Successful treatment of acne vulgaris in women with a new topical sodium sulfacetamide/sulfur lotion. Int. J. Dermatol. 1993, 32, 365–367. [Google Scholar] [CrossRef] [PubMed]
  167. Wilkinson, R.D.; Adam, J.E.; Murray, J.J.; Craig, G.E. Benzoyl peroxide and sulfur: Foundation for acne management. Can. Med. Assoc. J. 1966, 95, 28–29. [Google Scholar]
  168. Gupta, A.K.; Nicol, K. The use of sulfur in dermatology. J. Drugs Dermatol. 2004, 3, 427–431. [Google Scholar] [PubMed]
  169. Danto, J.L.; Maddin, W.S.; Stewart, W.D.; Nelson, A.J. A controlled trial of benzoyl peroxide and precipitated sulfur cream in acne vulgaris. Appl. Ther. 1966, 8, 624–625. [Google Scholar] [PubMed]
  170. Del Rosso, J.Q. The use of sodium sulfacetamide 10%-sulfur 5% emollient foam in the treatment of acne vulgaris. J. Clin. Aesthet. Dermatol. 2009, 2, 26–29. [Google Scholar]
  171. Draelos, Z.D. The multifunctionality of 10% sodium sulfacetamide, 5% sulfur emollient foam in the treatment of inflammatory facial dermatoses. J. Drugs Dermatol. 2010, 9, 234–236. [Google Scholar]
  172. Auffret, N.; Claudel, J.P.; Leccia, M.T.; Ballanger, F.; Dreno, B. Novel and emerging treatment options for acne vulgaris. Eur. J. Dermatol. 2022, 32, 451–458. [Google Scholar] [PubMed]
  173. Rosette, C.; Agan, F.J.; Mazzetti, A.; Moro, L.; Gerloni, M. Cortexolone 17α-propionate (Clascoterone) Is a Novel Androgen Receptor Antagonist that Inhibits Production of Lipids and Inflammatory Cytokines from Sebocytes In Vitro. J. Drugs Dermatol. 2019, 18, 412–418. [Google Scholar] [PubMed]
  174. Hebert, A.; Thiboutot, D.; Stein Gold, L.; Cartwright, M.; Gerloni, M.; Fragasso, E.; Mazzetti, A. Efficacy and Safety of Topical Clascoterone Cream, 1%, for Treatment in Patients With Facial Acne: Two Phase 3 Randomized Clinical Trials. JAMA Dermatol. 2020, 156, 621–630. [Google Scholar] [CrossRef] [PubMed]
  175. Barbieri, J.S. A New Class of Topical Acne Treatment Addressing the Hormonal Pathogenesis of Acne. JAMA Dermatol. 2020, 156, 619–620. [Google Scholar] [CrossRef] [PubMed]
  176. Trifu, V.; Tiplica, G.S.; Naumescu, E.; Zalupca, L.; Moro, L.; Celasco, G. Cortexolone 17α-propionate 1% cream, a new potent antiandrogen for topical treatment of acne vulgaris. A pilot randomized, double-blind comparative study vs. placebo and tretinoin 0·05% cream. Br. J. Dermatol. 2011, 165, 177–183. [Google Scholar] [CrossRef] [PubMed]
  177. Zaenglein, A.L.; Thiboutot, D.M. Expert committee recommendations for acne management. Pediatrics 2006, 118, 1188–1199. [Google Scholar] [CrossRef] [PubMed]
  178. Chowdhury, M.; Mamun, S.; Uddin, M.; Khan, R.M.; Hoque, M.; Shikder, M. Oral azithromycin pulse therapy and daily topical adapalene in the treatment of acne vulgaris: An open randomized noncomparative study. Med. Today 2017, 10, 52–56. [Google Scholar] [CrossRef]
  179. Krakowski, A.C.; Stendardo, S.; Eichenfield, L.F. Practical considerations in acne treatment and the clinical impact of topical combination therapy. Pediatr. Dermatol. 2008, 25, 1–14. [Google Scholar] [CrossRef] [PubMed]
  180. Iijima, S.; Tsunoda, T. Twenty cases of allergic contact dermatitis due to benzoyl peroxide in acne patients in Japan. J. Cutan. Immunol. Allergy 2019, 2, 108–112. [Google Scholar] [CrossRef]
  181. Garg, A.K.; Maddiboyina, B.; Alqarni, M.H.S.; Alam, A.; Aldawsari, H.M.; Rawat, P.; Singh, S.; Kesharwani, P. Solubility enhancement, formulation development and antifungal activity of luliconazole niosomal gel-based system. J. Biomater. Sci. Polym. Ed. 2021, 32, 1009–1023. [Google Scholar] [CrossRef]
  182. Cebrián, R.; Arévalo, S.; Rubiño, S.; Arias-Santiago, S.; Rojo, M.D.; Montalbán-López, M.; Martínez-Bueno, M.; Valdivia, E.; Maqueda, M. Control of Propionibacterium acnes by natural antimicrobial substances: Role of the bacteriocin AS-48 and lysozyme. Sci. Rep. 2018, 8, 11766. [Google Scholar] [CrossRef] [PubMed]
  183. Zheng, Y.; Yin, S.; Xia, Y.; Chen, J.; Ye, C.; Zeng, Q.; Lai, W. Efficacy and safety of 2% supramolecular salicylic acid compared with 5% benzoyl peroxide/0.1% adapalene in the acne treatment: A randomized, split-face, open-label, single-center study. Cutan. Ocul. Toxicol. 2019, 38, 48–54. [Google Scholar] [CrossRef] [PubMed]
  184. Tzellos, T.; Zampeli, V.; Makrantonaki, E.; Zouboulis, C.C. Treating acne with antibiotic-resistant bacterial colonization. Expert. Opin. Pharmacother. 2011, 12, 1233–1247. [Google Scholar] [CrossRef]
  185. Poulin, Y.; Sanchez, N.; Bucko, A.; Fowler, J.; Jarratt, M.; Kempers, S.; Kerrouche, N.; Dhuin, J.C.; Kunynetz, R. A 6-month maintenance therapy with adapalene-benzoyl peroxide gel prevents relapse and continuously improves efficacy among patients with severe acne vulgaris: Results of a randomized controlled trial. Br. J. Dermatol. 2011, 164, 1376–1382. [Google Scholar] [CrossRef] [PubMed]
  186. Thiboutot, D.M.; Shalita, A.R.; Yamauchi, P.S.; Dawson, C.; Kerrouche, N.; Arsonnaud, S.; Kang, S. Adapalene gel, 0.1%, as maintenance therapy for acne vulgaris: A randomized, controlled, investigator-blind follow-up of a recent combination study. Arch. Dermatol. 2006, 142, 597–602. [Google Scholar] [CrossRef] [PubMed]
  187. Santer, M.; Burden-Teh, E.; Ravenscroft, J. Managing acne vulgaris: An update. Drug Ther. Bull. 2023, 62, 6–10. [Google Scholar] [CrossRef]
  188. Dessinioti, C.; Katsambas, A. Antibiotics and Antimicrobial Resistance in Acne: Epidemiological Trends and Clinical Practice Considerations. Yale J. Biol. Med. 2022, 95, 429–443. [Google Scholar] [PubMed]
  189. Zhanel, G.; Critchley, I.; Lin, L.-Y.; Alvandi, N. Microbiological profile of sarecycline, a novel targeted spectrum tetracycline for the treatment of acne vulgaris. Antimicrob. Agents Chemother. 2019, 63, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  190. Leyden, J.J.; Del Rosso, J.Q. Oral antibiotic therapy for acne vulgaris: Pharmacokinetic and pharmacodynamic perspectives. J. Clin. Aesthetic Dermatol. 2011, 4, 40. [Google Scholar]
  191. Leyden, J.J.; Kaidbey, K.; Gans, E. The antimicrobial effects in vivo of minocycline, doxycycline and tetracycline in humans. J. Dermatol. Treat. 1996, 7, 223–225. [Google Scholar] [CrossRef]
  192. Deeks, E.D. Sarecycline: First Global Approval. Drugs 2019, 79, 325–329. [Google Scholar] [CrossRef]
  193. Marson, J.W.; Baldwin, H.E. An Overview of Acne Therapy, Part 1: Topical therapy, Oral Antibiotics, Laser and Light Therapy, and Dietary Interventions. Dermatol. Clin. 2019, 37, 183–193. [Google Scholar] [CrossRef] [PubMed]
  194. Moore, A.Y.; Charles, J.E.M.; Moore, S. Sarecycline: A narrow spectrum tetracycline for the treatment of moderate-to-severe acne vulgaris. Future Microbiol. 2019, 14, 1235–1242. [Google Scholar] [CrossRef] [PubMed]
  195. Moore, A.; Green, L.J.; Bruce, S.; Sadick, N.; Tschen, E.; Werschler, P.; Cook-Bolden, F.E.; Dhawan, S.S.; Forsha, D.; Gold, M.H.; et al. Once-Daily Oral Sarecycline 1.5 mg/kg/day Is Effective for Moderate to Severe Acne Vulgaris: Results from Two Identically Designed, Phase 3, Randomized, Double-Blind Clinical Trials. J. Drugs Dermatol. 2018, 17, 987–996. [Google Scholar] [CrossRef]
  196. Kadayifci, A.; Gulsen, M.; Koruk, M.; Savas, M. Doxycycline-induced pill esophagitis. Dis. Esophagus 2004, 17, 168–171. [Google Scholar] [CrossRef] [PubMed]
  197. Bjellerup, M.; Ljunggren, B. Double blind cross-over studies on phototoxicity to three tetracycline derivatives in human volunteers. Photo-dermatology 1987, 4, 281–287. [Google Scholar]
  198. Pariser, D.M.; Green, L.J.; Lain, E.L.; Schmitz, C.; Chinigo, A.S.; McNamee, B.; Berk, D.R. Safety and tolerability of sarecycline for the treatment of acne vulgaris: Results from a phase III, multicenter, open-label study and a phase I phototoxicity study. J. Clin. Aesthetic Dermatol. 2019, 12, E53. [Google Scholar] [CrossRef]
  199. Nast, A.; Dréno, B.; Bettoli, V.; Bukvic Mokos, Z.; Degitz, K.; Dressler, C.; Finlay, A.Y.; Haedersdal, M.; Lambert, J.; Layton, A. European evidence-based (S3) guideline for the treatment of acne–update 2016–short version. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 1261–1268. [Google Scholar] [CrossRef]
  200. Asai, Y.; Baibergenova, A.; Dutil, M.; Humphrey, S.; Hull, P.; Lynde, C.; Poulin, Y.; Shear, N.H.; Tan, J.; Toole, J. Management of acne: Canadian clinical practice guideline. Cmaj 2016, 188, 118–126. [Google Scholar] [CrossRef]
  201. Xu, J.; Mavranezouli, I.; Kuznetsov, L.; Murphy, M.S.; Healy, E. Management of acne vulgaris: Summary of NICE guidance. BMJ 2021, 374, n1800. [Google Scholar] [CrossRef]
  202. Fox, L.; Csongradi, C.; Aucamp, M.; Du Plessis, J.; Gerber, M. Treatment modalities for acne. Molecules 2016, 21, 1063. [Google Scholar] [CrossRef] [PubMed]
  203. Corcoran, L.; Muller, I.; Layton, A.M.; Rucinski, G.; Venkatess, V.; Sufraz, A.; Dove, S.; Lown, M.; Stuart, B.; Francis, N.; et al. Systematic review of clinical practice guidelines for acne vulgaris published between January 2017 and July 2021. Skin Health Dis. 2023, 3, e240. [Google Scholar] [CrossRef] [PubMed]
  204. Thiboutot, D.M.; Dréno, B.; Abanmi, A.; Alexis, A.F.; Araviiskaia, E.; Cabal, M.I.B.; Bettoli, V.; Casintahan, F.; Chow, S.; da Costa, A. Practical management of acne for clinicians: An international consensus from the Global Alliance to Improve Outcomes in Acne. J. Am. Acad. Dermatol. 2018, 78, S1–S23.e21. [Google Scholar] [CrossRef] [PubMed]
  205. Walsh, T.R.; Efthimiou, J.; Dréno, B. Systematic review of antibiotic resistance in acne: An increasing topical and oral threat. Lancet Infect. Dis. 2016, 16, e23–e33. [Google Scholar] [CrossRef] [PubMed]
  206. Park, J.H.; Bienenfeld, A.; Orlow, S.J.; Nagler, A.R. The use of hormonal antiandrogen therapy in female patients with acne: A 10-year retrospective study. Am. J. Clin. Dermatol. 2018, 19, 449–455. [Google Scholar] [CrossRef] [PubMed]
  207. Elsaie, M.L. Hormonal treatment of acne vulgaris: An update. Clin. Cosmet. Investig. Dermatol. 2016, 9, 241–248. [Google Scholar] [CrossRef]
  208. Eichenfield, L.F.; Krakowski, A.C.; Piggott, C.; Del Rosso, J.; Baldwin, H.; Friedlander, S.F.; Levy, M.; Lucky, A.; Mancini, A.J.; Orlow, S.J. Evidence-based recommendations for the diagnosis and treatment of pediatric acne. Pediatrics 2013, 131, S163–S186. [Google Scholar] [CrossRef] [PubMed]
  209. Zimmerman, Y.; Eijkemans, M.; Coelingh Bennink, H.; Blankenstein, M.; Fauser, B. The effect of combined oral contraception on testosterone levels in healthy women: A systematic review and meta-analysis. Hum. Reprod. Update 2014, 20, 76–105. [Google Scholar] [CrossRef] [PubMed]
  210. Koo, E.B.; Petersen, T.D.; Kimball, A.B. Meta-analysis comparing efficacy of antibiotics versus oral contraceptives in acne vulgaris. J. Am. Acad. Dermatol. 2014, 71, 450–459. [Google Scholar] [CrossRef]
  211. Gabbard, R.D.; Hoopes, R.R.; Kemp, M.G. Spironolactone and XPB: An old drug with a new molecular target. Biomolecules 2020, 10, 756. [Google Scholar] [CrossRef]
  212. Kim, G.K.; Del Rosso, J.Q. Oral Spironolactone in Post-teenage Female Patients with Acne Vulgaris: Practical Considerations for the Clinician Based on Current Data and Clinical Experience. J. Clin. Aesthet. Dermatol. 2012, 5, 37–50. [Google Scholar] [PubMed]
  213. Charny, J.W.; Choi, J.K.; James, W.D. Spironolactone for the treatment of acne in women, a retrospective study of 110 patients. Int. J. Women’s Dermatol. 2017, 3, 111–115. [Google Scholar] [CrossRef] [PubMed]
  214. Plovanich, M.; Weng, Q.Y.; Mostaghimi, A. Low Usefulness of Potassium Monitoring Among Healthy Young Women Taking Spironolactone for Acne. JAMA Dermatol. 2015, 151, 941–944. [Google Scholar] [CrossRef] [PubMed]
  215. Liszewski, W.; Boull, C. Lack of evidence for feminization of males exposed to spironolactone in utero: A systematic review. J. Am. Acad. Dermatol. 2019, 80, 1147–1148. [Google Scholar] [CrossRef] [PubMed]
  216. Layton, A.M.; Eady, E.A.; Whitehouse, H.; Del Rosso, J.Q.; Fedorowicz, Z.; van Zuuren, E.J. Oral Spironolactone for Acne Vulgaris in Adult Females: A Hybrid Systematic Review. Am. J. Clin. Dermatol. 2017, 18, 169–191. [Google Scholar] [CrossRef] [PubMed]
  217. Ebede, T.L.; Arch, E.L.; Berson, D. Hormonal treatment of acne in women. J. Clin. Aesthetic Dermatol. 2009, 2, 16. [Google Scholar]
  218. Dagnelie, M.A.; Poinas, A.; Dréno, B. What is new in adult acne for the last 2 years: Focus on acne pathophysiology and treatments. Int. J. Dermatol. 2022, 61, 1205–1212. [Google Scholar] [CrossRef] [PubMed]
  219. Scott-Emuakpor, R.; Vuthaluru, K.; Nagre, A.; Jawed, I.; Patel, P.A.; Sidhu, H.K. Role of Oral Retinoids in Treatment of Acne Vulgaris With a Bioinformatics-Based Perspective of Personalized Medicine. Cureus 2023, 15, e38019. [Google Scholar] [CrossRef] [PubMed]
  220. Wang, S.; Jiang, R.; Meng, T.; Zhang, F.; Li, J.; Jin, Y.; Lee, J.; Zhu, M.; Jiang, J. Stem cell membrane-coated isotretinoin for acne treatment. J. Nanobiotechnol. 2020, 18, 106. [Google Scholar] [CrossRef]
  221. Landis, M.N. Optimizing isotretinoin treatment of acne: Update on current recommendations for monitoring, dosing, safety, adverse effects, compliance, and outcomes. Am. J. Clin. Dermatol. 2020, 21, 411–419. [Google Scholar] [CrossRef]
  222. Bagatin, E.; Costa, C.S. The use of isotretinoin for acne–an update on optimal dosing, surveillance, and adverse effects. Expert. Rev. Clin. Pharmacol. 2020, 13, 885–897. [Google Scholar] [CrossRef]
  223. Tan, J.; Knezevic, S.; Boyal, S.; Waterman, B.; Janik, T. Evaluation of evidence for acne remission with oral isotretinoin cumulative dosing of 120–150 mg/kg. J. Cutan. Med. Surg. 2016, 20, 13–20. [Google Scholar] [CrossRef] [PubMed]
  224. Thiboutot, D.; Dréno, B.; Sanders, V.; Rueda, M.J.; Gollnick, H. Changes in the management of acne: 2009–2019. J. Am. Acad. Dermatol. 2020, 82, 1268–1269. [Google Scholar] [CrossRef] [PubMed]
  225. Rademaker, M. Making sense of the effects of the cumulative dose of isotretinoin in acne vulgaris. Int. J. Dermatol. 2016, 55, 518–523. [Google Scholar] [CrossRef] [PubMed]
  226. Fallah, H.; Rademaker, M. Isotretinoin in the management of acne vulgaris: Practical prescribing. Int. J. Dermatol. 2021, 60, 451–460. [Google Scholar] [CrossRef] [PubMed]
  227. Sladden, M.J.; Harman, K.E. What is the chance of a normal pregnancy in a woman whose fetus has been exposed to isotretinoin? Arch. Dermatol. 2007, 143, 1187–1188. [Google Scholar] [CrossRef] [PubMed]
  228. Hodgkiss-Harlow, C.J.; Eichenfield, L.F.; Dohil, M.A. Effective monitoring of isotretinoin safety in a pediatric dermatology population: A novel “patient symptom survey” approach. J. Am. Acad. Dermatol. 2011, 65, 517–524. [Google Scholar] [CrossRef]
  229. Kaymak, Y. Creatine phosphokinase values during isotretinoin treatment for acne. Int. J. Dermatol. 2008, 47, 398–401. [Google Scholar] [CrossRef] [PubMed]
  230. Manfredini, M.; Bettoli, V.; Forconi, R.; Pacetti, L.; Farnetani, F.; Corazza, M.; Pellacani, G. Creatine Phosphokinase Values during Low Starting Dose Isotretinoin Therapy. Skin Appendage Disord. 2020, 6, 142–146. [Google Scholar] [CrossRef]
  231. Busetti, B.M.; Azulay, D.R.; Aguinaga, F.; Cordova, E.O. Evaluation of CPK levels during acne treatment with oral isotretinoin. An. Bras. Dermatol. 2021, 96, 626–627. [Google Scholar] [CrossRef]
  232. Rivillas, J.A.; Santos Andrade, V.A.; Hormaza-Jaramillo, A.A. Myositis Induced by Isotretinoin: A Case Report and Literature Review. Am. J. Case Rep. 2020, 21, e917801. [Google Scholar] [CrossRef] [PubMed]
  233. Hansen, T.J.; Lucking, S.; Miller, J.J.; Kirby, J.S.; Thiboutot, D.M.; Zaenglein, A.L. Standardized laboratory monitoring with use of isotretinoin in acne. J. Am. Acad. Dermatol. 2016, 75, 323–328. [Google Scholar] [CrossRef] [PubMed]
  234. Zane, L.T.; Leyden, W.A.; Marqueling, A.L.; Manos, M.M. A population-based analysis of laboratory abnormalities during isotretinoin therapy for acne vulgaris. Arch. Dermatol. 2006, 142, 1016–1022. [Google Scholar] [CrossRef] [PubMed]
  235. Opel, D.; Kramer, O.; Chevalier, M.; Bigby, M.; Albrecht, J. Not every patient needs a triglyceride check, but all can get pancreatitis: A systematic review and clinical characterization of isotretinoin-associated pancreatitis. Br. J. Dermatol. 2017, 177, 960–966. [Google Scholar] [CrossRef] [PubMed]
  236. Barbieri, J.S.; Shin, D.B.; Wang, S.; Margolis, D.J.; Takeshita, J. The clinical utility of laboratory monitoring during isotretinoin therapy for acne and changes to monitoring practices over time. J. Am. Acad. Dermatol. 2020, 82, 72–79. [Google Scholar] [CrossRef] [PubMed]
  237. Singer, S.; Tkachenko, E.; Sharma, P.; Barbieri, J.S.; Mostaghimi, A. Psychiatric Adverse Events in Patients Taking Isotretinoin as Reported in a Food and Drug Administration Database From 1997 to 2017. JAMA Dermatol. 2019, 155, 1162–1166. [Google Scholar] [CrossRef] [PubMed]
  238. Droitcourt, C.; Nowak, E.; Rault, C.; Happe, A.; Le Nautout, B.; Kerbrat, S.; Balusson, F.; Poizeau, F.; Travers, D.; Sapori, J.M.; et al. Risk of suicide attempt associated with isotretinoin: A nationwide cohort and nested case-time-control study. Int. J. Epidemiol. 2019, 48, 1623–1635. [Google Scholar] [CrossRef] [PubMed]
  239. Etminan, M.; Bird, S.T.; Delaney, J.A.; Bressler, B.; Brophy, J.M. Isotretinoin and risk for inflammatory bowel disease: A nested case-control study and meta-analysis of published and unpublished data. JAMA Dermatol. 2013, 149, 216–220. [Google Scholar] [CrossRef] [PubMed]
  240. Lee, S.Y.; Jamal, M.M.; Nguyen, E.T.; Bechtold, M.L.; Nguyen, D.L. Does exposure to isotretinoin increase the risk for the development of inflammatory bowel disease? A meta-analysis. Eur. J. Gastroenterol. Hepatol. 2016, 28, 210–216. [Google Scholar] [CrossRef]
  241. Margolis, D.J.; Fanelli, M.; Hoffstad, O.; Lewis, J.D. Potential association between the oral tetracycline class of antimicrobials used to treat acne and inflammatory bowel disease. Am. J. Gastroenterol. 2010, 105, 2610–2616. [Google Scholar] [CrossRef]
  242. Barbieri, J.S. Isotretinoin and risk of inflammatory bowel disease: More data to support lack of meaningful risk. J. Am. Acad. Dermatol. 2021, 84, 228–229. [Google Scholar] [CrossRef] [PubMed]
  243. Scheman, A.J. Nodulocystic acne and hidradenitis suppurativa treated with acitretin: A case report. Cutis 2002, 69, 287–288. [Google Scholar] [PubMed]
  244. Pilkington, T.; Brogden, R.N. Acitretin. A review of its pharmacology and therapeutic use. Drugs 1992, 43, 597–627. [Google Scholar] [CrossRef] [PubMed]
  245. Handler, M.Z.; Bloom, B.S.; Goldberg, D.J. Energy-based devices in treatment of acne vulgaris. Dermatol. Surg. 2016, 42, 573–585. [Google Scholar] [CrossRef] [PubMed]
  246. Tang, X.; Li, C.; Ge, S.; Chen, Z.; Lu, L. Efficacy of photodynamic therapy for the treatment of inflammatory acne vulgaris: A systematic review and meta-analysis. J. Cosmet. Dermatol. 2020, 19, 10–21. [Google Scholar] [CrossRef]
  247. Sadick, N.S.; Cardona, A. Laser treatment for facial acne scars: A review. J. Cosmet. Laser Ther. 2018, 20, 424–435. [Google Scholar] [CrossRef]
  248. Posadzki, P.; Car, J. Light therapies for acne. JAMA Dermatol. 2018, 154, 597–598. [Google Scholar] [CrossRef] [PubMed]
  249. Mavranezouli, I.; Daly, C.H.; Welton, N.J.; Deshpande, S.; Berg, L.; Bromham, N.; Arnold, S.; Phillippo, D.M.; Wilcock, J.; Xu, J. A systematic review and network meta-analysis of topical pharmacological, oral pharmacological, physical and combined treatments for acne vulgaris. Br. J. Dermatol. 2022, 187, 639–649. [Google Scholar] [CrossRef]
  250. Choi, Y.; Suh, H.; Yoon, M.; Min, S.; Lee, D.; Suh, D. Intense pulsed light vs. pulsed-dye laser in the treatment of facial acne: A randomized split-face trial. J. Eur. Acad. Dermatol. Venereol. 2010, 24, 773–780. [Google Scholar] [CrossRef]
  251. Seaton, E.; Charakida, A.; Mouser, P.; Grace, I.; Clement, R.; Chu, A. Pulsed-dye laser treatment for inflammatory acne vulgaris: Randomised controlled trial. Lancet 2003, 362, 1347–1352. [Google Scholar] [CrossRef]
  252. Papageorgiou, P.; Katsambas, A.; Chu, A. Phototherapy with blue (415 nm) and red (660 nm) light in the treatment of acne vulgaris. Br. J. Dermatol. 2000, 142, 973–978. [Google Scholar] [CrossRef]
  253. Lim, W.; Lee, S.; Kim, I.; Chung, M.; Kim, M.; Lim, H.; Park, J.; Kim, O.; Choi, H. The anti-inflammatory mechanism of 635 nm light-emitting-diode irradiation compared with existing COX inhibitors. Lasers Surg. Med. Off. J. Am. Soc. Laser Med. Surg. 2007, 39, 614–621. [Google Scholar] [CrossRef] [PubMed]
  254. Sami, N.A.; Attia, A.T.; Badawi, A.M. Phototherapy in the treatment of acne vulgaris. J. Drugs Dermatol. JDD 2008, 7, 627–632. [Google Scholar] [PubMed]
  255. Li, J.; Li, J.; Zhang, L.; Liu, X.; Cao, Y.; Wang, P.; Wang, X. Comparison of red light and blue light therapies for mild-to-moderate acne vulgaris: A randomized controlled clinical study. Photodermatol. Photoimmunol. Photomed. 2022, 38, 459–464. [Google Scholar] [CrossRef] [PubMed]
  256. Chang, S.E.; Ahn, S.J.; Rhee, D.Y.; Choi, J.H.; Moon, K.C.; Suh, H.S.; Soyun-Cho, M. Treatment of facial acne papules and pustules in Korean patients using an intense pulsed light device equipped with a 530-to 750-nm filter. Dermatol. Surg. 2007, 33, 676–679. [Google Scholar]
  257. Barakat, M.T.; Moftah, N.H.; El Khayyat, M.A.; Abdelhakim, Z.A. Significant reduction of inflammation and sebaceous glands size in acne vulgaris lesions after intense pulsed light treatment. Dermatol. Ther. 2017, 30, e12418. [Google Scholar] [CrossRef] [PubMed]
  258. Taylor, M.; Porter, R.; Gonzalez, M. Intense pulsed light may improve inflammatory acne through TNF-α down-regulation. J. Cosmet. Laser Ther. 2014, 16, 96–103. [Google Scholar] [CrossRef] [PubMed]
  259. Uebelhoer, N.S.; Bogle, M.A.; Dover, J.S.; Arndt, K.A.; Rohrer, T.E. Comparison of stacked pulses versus double-pass treatments of facial acne with a 1450-nm laser. Dermatol. Surg. 2007, 33, 552–559. [Google Scholar] [PubMed]
  260. Kwon, H.H.; Choi, S.C.; Jung, J.Y.; Bae, Y.I.; Park, G.H. Comparison of novel dual mode vs conventional single pass of a 1450-nm diode laser in the treatment of acne vulgaris for Korean patients: A 20-week prospective, randomized, split-face study. J. Cosmet. Dermatol. 2018, 17, 1063–1068. [Google Scholar] [CrossRef]
  261. De Vries, F.; Meulendijks, A.; Driessen, R.; van Dooren, A.; Tjin, E.; van de Kerkhof, P. The efficacy and safety of non-pharmacological therapies for the treatment of acne vulgaris: A systematic review and best-evidence synthesis. J. Eur. Acad. Dermatol. Venereol. 2018, 32, 1195–1203. [Google Scholar] [CrossRef]
  262. Scopelliti, M.G.; Kothare, A.; Karavitis, M. A novel 1726-nm laser system for safe and effective treatment of acne vulgaris. Lasers Med. Sci. 2022, 37, 3639–3647. [Google Scholar] [CrossRef] [PubMed]
  263. Cruz, S.; Vecerek, N.; Elbuluk, N. Targeting Inflammation in Acne: Current Treatments and Future Prospects. Am. J. Clin. Dermatol. 2023, 24, 681–694. [Google Scholar] [CrossRef] [PubMed]
  264. Paithankar, D.Y.; Sakamoto, F.H.; Farinelli, W.A.; Kositratna, G.; Blomgren, R.D.; Meyer, T.J.; Faupel, L.J.; Kauvar, A.N.; Lloyd, J.R.; Cheung, W.L. Acne treatment based on selective photothermolysis of sebaceous follicles with topically delivered light-absorbing gold microparticles. J. Investig. Dermatol. 2015, 135, 1727–1734. [Google Scholar] [CrossRef] [PubMed]
  265. Park, K.Y.; Han, H.S.; Hong, J.Y.; Seo, S.J.; Lee, S.J. Gold nanoshell-mediated photothermal therapy for acne vulgaris. Dermatol. Ther. 2020, 33, e13189. [Google Scholar] [CrossRef] [PubMed]
  266. Artounian, K.; Bundogji, N.; Hoss, E.; Boen, M. Applications of Gold and Silver Nanoparticles in the Treatment of Acne Vulgaris: A Systematic Review. J. Drugs Dermatol. JDD 2021, 20, 666–670. [Google Scholar] [PubMed]
  267. Fuchs, C.S.K.; Bay, C.; Adatto, M.; Lomholt, H.; Haedersdal, M. Acne treatment with light absorbing gold microparticles and optical pulses: An open-label European multi-centered study in moderate to moderately severe acne vulgaris patients. Lasers Surg. Med. 2019, 51, 686–693. [Google Scholar] [CrossRef] [PubMed]
  268. Suh, D.H.; Park, T.J.; Jeong, J.Y.; Kim, K.E.; Park, J.H.; Lee, S.J.; Kim, H.J.; Ryu, H.J. Photothermal therapy using gold nanoparticles for acne in Asian patients: A preliminary study. Dermatol. Ther. 2021, 34, e14918. [Google Scholar] [CrossRef] [PubMed]
  269. Yang, G.; Lee, S.J.; Kang, H.C.; Cho, Y.-Y.; Lee, H.S.; Zouboulis, C.C.; Han, S.-H.; Ma, K.-H.; Jang, J.-K.; Lee, J.Y. Repurposing auranofin, an anti-rheumatic gold compound, to treat acne vulgaris by targeting the NLRP3 inflammasome. Biomol. Ther. 2020, 28, 437. [Google Scholar] [CrossRef]
  270. Kempiak, S.J.; Uebelhoer, N. Superficial chemical peels and microdermabrasion for acne vulgaris. Semin. Cutan. Med. Surg. 2008, 27, 212–220. [Google Scholar] [CrossRef]
  271. Kessler, E.; Flanagan, K.; Chia, C.; Rogers, C.; Glaser, D.A. Comparison of alpha- and beta-hydroxy acid chemical peels in the treatment of mild to moderately severe facial acne vulgaris. Dermatol. Surg. 2008, 34, 45–50; discussion 51. [Google Scholar] [CrossRef]
  272. Lee, S.J.; Goo, J.W.; Shin, J.; Chung, W.S.; Kang, J.M.; Kim, Y.K.; Cho, S.B. Use of fractionated microneedle radiofrequency for the treatment of inflammatory acne vulgaris in 18 Korean patients. Dermatol. Surg. 2012, 38, 400–405. [Google Scholar] [CrossRef] [PubMed]
  273. Kim, S.T.; Lee, K.H.; Sim, H.J.; Suh, K.S.; Jang, M.S. Treatment of acne vulgaris with fractional radiofrequency microneedling. J. Dermatol. 2014, 41, 586–591. [Google Scholar] [CrossRef] [PubMed]
  274. Kim, J.; Lee, Y.I.; Kim, J.; Jung, J.Y.; Lee, W.J.; Lee, J.H. Safety of combined fractional microneedle radiofrequency and CO2 as an early intervention for inflammatory acne and scarring treated with concomitant isotretinoin. Dermatol. Surg. 2020, 46, e71–e77. [Google Scholar] [CrossRef] [PubMed]
  275. Yang, D.; Chen, M.; Sun, Y.; Jin, Y.; Lu, C.; Pan, X.; Quan, G.; Wu, C. Microneedle-mediated transdermal drug delivery for treating diverse skin diseases. Acta Biomater. 2021, 121, 119–133. [Google Scholar] [CrossRef] [PubMed]
  276. Kim, J.; Jongudomsombat, T.; Lee, Y.I.; Kim, J.; Oh, S.H.; Hong, J.W.; Lee, J.H. Combined use of energy-based interventions with low-dose isotretinoin for the treatment of inflammatory acne: An retrospective cohort analysis. J. Cosmet. Dermatol. 2022, 21, 4383–4391. [Google Scholar] [CrossRef] [PubMed]
  277. Levine, R.M.; Rasmussen, J.E. Intralesional corticosteroids in the treatment of nodulocystic acne. Arch. Dermatol. 1983, 119, 480–481. [Google Scholar] [CrossRef] [PubMed]
  278. Melnik, B.C.; John, S.M.; Schmitz, G. Over-stimulation of insulin/IGF-1 signaling by western diet may promote diseases of civilization: Lessons learnt from laron syndrome. Nutr. Metab. 2011, 8, 41. [Google Scholar] [CrossRef] [PubMed]
  279. Hoppe, C.; Mølgaard, C.; Dalum, C.; Vaag, A.; Michaelsen, K.F. Differential effects of casein versus whey on fasting plasma levels of insulin, IGF-1 and IGF-1/IGFBP-3: Results from a randomized 7-day supplementation study in prepubertal boys. Eur. J. Clin. Nutr. 2009, 63, 1076–1083. [Google Scholar] [CrossRef] [PubMed]
  280. Berra, B.; Rizzo, A.M. Glycemic index, glycemic load: New evidence for a link with acne. J. Am. Coll. Nutr. 2009, 28, 450S–454S. [Google Scholar] [CrossRef]
  281. Hoyt, G.; Hickey, M.S.; Cordain, L. Dissociation of the glycaemic and insulinaemic responses to whole and skimmed milk. Br. J. Nutr. 2005, 93, 175–177. [Google Scholar] [CrossRef]
  282. Melnik, B. Dietary intervention in acne: Attenuation of increased mTORC1 signaling promoted by Western diet. Derm.-Endocrinol. 2012, 4, 20–32. [Google Scholar] [CrossRef]
  283. Deng, Y.; Wang, H.; Zhou, J.; Mou, Y.; Wang, G.; Xiong, X. Patients with acne vulgaris have a distinct gut microbiota in comparison with healthy controls. Acta Derm. Venereol. 2018, 98, 783–790. [Google Scholar] [CrossRef] [PubMed]
  284. Jung, J.Y.; Kwon, H.H.; Hong, J.S.; Yoon, J.Y.; Park, M.S.; Jang, M.Y.; Suh, D.H. Effect of dietary supplementation with omega-3 fatty acid and gamma-linolenic acid on acne vulgaris: A randomised, double-blind, controlled trial. Acta Derm. Venereol. 2014, 94, 521–525. [Google Scholar] [CrossRef] [PubMed]
  285. Siddiqui, R.; Makhlouf, Z.; Khan, N.A. The increasing importance of the gut microbiome in acne vulgaris. Folia Microbiol. 2022, 67, 825–835. [Google Scholar] [CrossRef] [PubMed]
  286. Kucharska, A.; Szmurło, A.; Sińska, B. Significance of diet in treated and untreated acne vulgaris. Adv. Dermatol. Allergol. Postępy Dermatol. Alergol. 2016, 33, 81–86. [Google Scholar] [CrossRef] [PubMed]
  287. Kumar, J.; Rani, K.; Datt, C. Molecular link between dietary fibre, gut microbiota and health. Mol. Biol. Rep. 2020, 47, 6229–6237. [Google Scholar] [CrossRef] [PubMed]
  288. Yildizgören, M.T.; Togral, A.K. Preliminary evidence for vitamin D deficiency in nodulocystic acne. Derm.-Endocrinol. 2014, 6, e983687. [Google Scholar] [CrossRef] [PubMed]
  289. Alhetheli, G.; Elneam, A.I.A.; Alsenaid, A.; Al-Dhubaibi, M. Vitamin D levels in patients with and without acne and its relation to acne severity: A case-control study. Clin. Cosmet. Investig. Dermatol. 2020, 2020, 759–765. [Google Scholar] [CrossRef]
  290. Lee, W.J.; Choi, Y.H.; Sohn, M.Y.; Lee, S.-J. Expression of inflammatory biomarkers from cultured sebocytes was influenced by treatment with vitamin D. Indian J. Dermatol. 2013, 58, 327. [Google Scholar] [CrossRef]
  291. Al-Taiar, A.; AlKhabbaz, M.; Rahman, A.; Al-Sabah, R.; Shaban, L.; Akhtar, S. Plasma 25-hydroxy vitamin D is not associated with acne vulgaris. Nutrients 2018, 10, 1525. [Google Scholar] [CrossRef]
  292. Stewart, T.J.; Bazergy, C. Hormonal and dietary factors in acne vulgaris versus controls. Derm.-Endocrinol. 2018, 10, e1442160. [Google Scholar] [CrossRef] [PubMed]
  293. Bashir, M.; Prietl, B.; Tauschmann, M.; Mautner, S.I.; Kump, P.K.; Treiber, G.; Wurm, P.; Gorkiewicz, G.; Högenauer, C.; Pieber, T.R. Effects of high doses of vitamin D3 on mucosa-associated gut microbiome vary between regions of the human gastrointestinal tract. Eur. J. Nutr. 2016, 55, 1479–1489. [Google Scholar] [CrossRef] [PubMed]
  294. Rostami Mogaddam, M.; Safavi Ardabili, N.; Maleki, N.; Soflaee, M. Correlation between the severity and type of acne lesions with serum zinc levels in patients with acne vulgaris. BioMed Res. Int. 2014, 2014, 474108. [Google Scholar] [CrossRef] [PubMed]
  295. Cervantes, J.; Eber, A.E.; Perper, M.; Nascimento, V.M.; Nouri, K.; Keri, J.E. The role of zinc in the treatment of acne: A review of the literature. Dermatol. Ther. 2018, 31, e12576. [Google Scholar] [CrossRef] [PubMed]
  296. Reyes-Hadsall, S.; Ju, T.; Keri, J.E. Use of Oral Supplements and Topical Adjuvants for Isotretinoin-Associated Side Effects: A Narrative Review. Skin Appendage Disord. 2023, 10, 1–9. [Google Scholar] [CrossRef] [PubMed]
  297. Aksac, S.E.; Bilgili, S.G.; Yavuz, G.O.; Yavuz, I.H.; Aksac, M.; Karadag, A.S. Evaluation of biophysical skin parameters and hair changes in patients with acne vulgaris treated with isotretinoin, and the effect of biotin use on these parameters. Int. J. Dermatol. 2021, 60, 980–985. [Google Scholar] [CrossRef] [PubMed]
  298. Ewurum, A.; Alur, A.A.; Glenn, M.; Schnepf, A.; Borchman, D. Hyaluronic acid-lipid binding. BMC Chem. 2021, 15, 36. [Google Scholar] [CrossRef] [PubMed]
  299. Lowe, H.; Toyang, N.; Steele, B.; Bryant, J.; Ngwa, W. The endocannabinoid system: A potential target for the treatment of various diseases. Int. J. Mol. Sci. 2021, 22, 9472. [Google Scholar] [CrossRef]
  300. Tóth, K.F.; Ádám, D.; Bíró, T.; Oláh, A. Cannabinoid Signaling in the Skin: Therapeutic Potential of the “C (ut) annabinoid” System. Molecules 2019, 24, 918. [Google Scholar] [CrossRef]
  301. Oláh, A.; Tóth, B.I.; Borbíró, I.; Sugawara, K.; Szöllõsi, A.G.; Czifra, G.; Pál, B.; Ambrus, L.; Kloepper, J.; Camera, E. Cannabidiol exerts sebostatic and antiinflammatory effects on human sebocytes. J. Clin. Investig. 2014, 124, 3713–3724. [Google Scholar] [CrossRef]
  302. Mounessa, J.S.; Siegel, J.A.; Dunnick, C.A.; Dellavalle, R.P. The role of cannabinoids in dermatology. J. Am. Acad. Dermatol. 2017, 77, 188–190. [Google Scholar] [CrossRef] [PubMed]
  303. Marks, D.H.; Friedman, A. The therapeutic potential of cannabinoids in dermatology. Skin Ther. Lett. 2018, 23, 1–5. [Google Scholar]
  304. Eagleston, L.R.; Kalani, N.K.; Patel, R.R.; Flaten, H.K.; Dunnick, C.A.; Dellavalle, R.P. Cannabinoids in dermatology: A scoping review. Dermatol. Online J. 2018, 24, 1. [Google Scholar] [CrossRef]
  305. Sheriff, T.; Lin, M.J.; Dubin, D.; Khorasani, H. The potential role of cannabinoids in dermatology. J. Dermatol. Treat. 2020, 31, 839–845. [Google Scholar] [CrossRef] [PubMed]
  306. Sand, F.L.; Thomsen, S.F. Adalimumab for the treatment of refractory acne conglobata. JAMA Dermatol. 2013, 149, 1306–1307. [Google Scholar] [CrossRef] [PubMed]
  307. Yiu, Z.; Madan, V.; Griffiths, C. Acne conglobata and adalimumab: Use of tumour necrosis factor-α antagonists in treatment-resistant acne conglobata, and review of the literature. Clin. Exp. Dermatol. 2015, 40, 383–386. [Google Scholar] [CrossRef] [PubMed]
  308. Flora, A.; Holland, R.; Smith, A.; Frew, J.W. Rapid and sustained remission of synovitis, acne, pustulosis, hyperostosis, and osteitis (SAPHO) syndrome with IL-23p19 antagonist (risankizumab). JAAD Case Rep. 2021, 14, 33–36. [Google Scholar] [CrossRef] [PubMed]
  309. Wang, G.; Zhuo, N.; Li, J. Off-label Use of Secukinumab: A Potential Therapeutic Option for SAPHO Syndrome. J. Rheumatol. 2022, 49, 656. [Google Scholar] [CrossRef]
  310. Thiboutot, D.M.; Craft, N.; Rissmann, R.; Gatlik, E.; Souquières, M.; Jones, J.; Loesche, C. Anti-IL-17A blockade did not significantly reduce inflammatory lesions in a placebo-controlled pilot study in adult patients with moderate to severe acne. J. Dermatol. Treat. 2023, 34, 2138691. [Google Scholar] [CrossRef]
  311. Paetzold, B.; Willis, J.R.; Pereira de Lima, J.; Knödlseder, N.; Brüggemann, H.; Quist, S.R.; Gabaldón, T.; Güell, M. Skin microbiome modulation induced by probiotic solutions. Microbiome 2019, 7, 95. [Google Scholar] [CrossRef]
  312. Goodarzi, A.; Mozafarpoor, S.; Bodaghabadi, M.; Mohamadi, M. The potential of probiotics for treating acne vulgaris: A review of literature on acne and microbiota. Dermatol. Ther. 2020, 33, e13279. [Google Scholar] [CrossRef] [PubMed]
  313. Gueniche, A.; Benyacoub, J.; Philippe, D.; Bastien, P.; Kusy, N.; Breton, L.; Blum, S.; Castiel-Higounenc, I. Lactobacillus paracasei CNCM I-2116 (ST11) inhibits substance P-induced skin inflammation and accelerates skin barrier function recovery in vitro. Eur. J. Dermatol. 2010, 20, 731–737. [Google Scholar] [PubMed]
  314. Cosseau, C.; Devine, D.A.; Dullaghan, E.; Gardy, J.L.; Chikatamarla, A.; Gellatly, S.; Yu, L.L.; Pistolic, J.; Falsafi, R.; Tagg, J.; et al. The commensal Streptococcus salivarius K12 downregulates the innate immune responses of human epithelial cells and promotes host-microbe homeostasis. Infect. Immun. 2008, 76, 4163–4175. [Google Scholar] [CrossRef] [PubMed]
  315. Bowe, W.P.; Filip, J.C.; DiRienzo, J.M.; Volgina, A.; Margolis, D.J. Inhibition of propionibacterium acnes by bacteriocin-like inhibitory substances (BLIS) produced by Streptococcus salivarius. J. Drugs Dermatol. 2006, 5, 868–870. [Google Scholar] [PubMed]
  316. Oh, S.; Kim, S.H.; Ko, Y.; Sim, J.H.; Kim, K.S.; Lee, S.H.; Park, S.; Kim, Y.J. Effect of bacteriocin produced by Lactococcus sp. HY 449 on skin-inflammatory bacteria. Food Chem. Toxicol. 2006, 44, 552–559. [Google Scholar] [CrossRef] [PubMed]
  317. Kang, B.S.; Seo, J.G.; Lee, G.S.; Kim, J.H.; Kim, S.Y.; Han, Y.W.; Kang, H.; Kim, H.O.; Rhee, J.H.; Chung, M.J.; et al. Antimicrobial activity of enterocins from Enterococcus faecalis SL-5 against Propionibacterium acnes, the causative agent in acne vulgaris, and its therapeutic effect. J. Microbiol. 2009, 47, 101–109. [Google Scholar] [CrossRef]
  318. Muizzuddin, N.; Maher, W.; Sullivan, M.; Schnittger, S.; Mammone, T. Physiological effect of a probiotic on skin. J. Cosmet. Sci. 2012, 63, 385–395. [Google Scholar]
  319. Rahmayani, T.; Putra, I.B.; Jusuf, N.K. The Effect of Oral Probiotic on the Interleukin-10 Serum Levels of Acne Vulgaris. Open Access Maced. J. Med. Sci. 2019, 7, 3249–3252. [Google Scholar] [CrossRef] [PubMed]
  320. Jung, G.W.; Tse, J.E.; Guiha, I.; Rao, J. Prospective, randomized, open-label trial comparing the safety, efficacy, and tolerability of an acne treatment regimen with and without a probiotic supplement and minocycline in subjects with mild to moderate acne. J. Cutan. Med. Surg. 2013, 17, 114–122. [Google Scholar] [CrossRef]
  321. Kim, J.; Ko, Y.; Park, Y.K.; Kim, N.I.; Ha, W.K.; Cho, Y. Dietary effect of lactoferrin-enriched fermented milk on skin surface lipid and clinical improvement of acne vulgaris. Nutrition 2010, 26, 902–909. [Google Scholar] [CrossRef]
  322. Rinaldi, F.; Marotta, L.; Mascolo, A.; Amoruso, A.; Pane, M.; Giuliani, G.; Pinto, D. Facial Acne: A Randomized, Double-Blind, Placebo-Controlled Study on the Clinical Efficacy of a Symbiotic Dietary Supplement. Dermatol. Ther. 2022, 12, 577–589. [Google Scholar] [CrossRef] [PubMed]
  323. Fabbrocini, G.; Bertona, M.; Picazo, Ó.; Pareja-Galeano, H.; Monfrecola, G.; Emanuele, E. Supplementation with Lactobacillus rhamnosus SP1 normalises skin expression of genes implicated in insulin signalling and improves adult acne. Benef. Microbes 2016, 7, 625–630. [Google Scholar] [CrossRef] [PubMed]
  324. Liu, P.F.; Nakatsuji, T.; Zhu, W.; Gallo, R.L.; Huang, C.M. Passive immunoprotection targeting a secreted CAMP factor of Propionibacterium acnes as a novel immunotherapeutic for acne vulgaris. Vaccine 2011, 29, 3230–3238. [Google Scholar] [CrossRef]
  325. Lam, H.Y.P.; Lai, M.J.; Chen, T.Y.; Wu, W.J.; Peng, S.Y.; Chang, K.C. Therapeutic Effect of a Newly Isolated Lytic Bacteriophage against Multi-Drug-Resistant Cutibacterium acnes Infection in Mice. Int. J. Mol. Sci. 2021, 22, 7031. [Google Scholar] [CrossRef] [PubMed]
  326. Van Steensel, M.A.M.; Goh, B.C. Cutibacterium acnes: Much ado about maybe nothing much. Exp. Dermatol. 2021, 30, 1471–1476. [Google Scholar] [CrossRef] [PubMed]
  327. Clemens, L.E.; Jaynes, J.; Lim, E.; Kolar, S.S.; Reins, R.Y.; Baidouri, H.; Hanlon, S.; McDermott, A.M.; Woodburn, K.W. Designed Host Defense Peptides for the Treatment of Bacterial Keratitis. Investig. Ophthalmol. Vis. Sci. 2017, 58, 6273–6281. [Google Scholar] [CrossRef] [PubMed]
  328. Woodburn, K.W.; Jaynes, J.M.; Clemens, L.E. Evaluation of the Antimicrobial Peptide, RP557, for the Broad-Spectrum Treatment of Wound Pathogens and Biofilm. Front. Microbiol. 2019, 10, 1688. [Google Scholar] [CrossRef] [PubMed]
  329. Woodburn, K.W.; Clemens, L.E.; Jaynes, J.; Joubert, L.M.; Botha, A.; Nazik, H.; Stevens, D.A. Designed Antimicrobial Peptides for Recurrent Vulvovaginal Candidiasis Treatment. Antimicrob. Agents Chemother. 2019, 63, 10–1128. [Google Scholar] [CrossRef]
  330. Gordon, Y.J.; Romanowski, E.G.; McDermott, A.M. A review of antimicrobial peptides and their therapeutic potential as anti-infective drugs. Curr. Eye Res. 2005, 30, 505–515. [Google Scholar] [CrossRef]
  331. Woodburn, K.W.; Jaynes, J.; Clemens, L.E. Designed Antimicrobial Peptides for Topical Treatment of Antibiotic Resistant Acne Vulgaris. Antibiotics 2020, 9, 23. [Google Scholar] [CrossRef]
  332. Adamo, S.; Nilsson, J.; Krebs, A.; Steiner, U.; Cozzio, A.; French, L.E.; Kolios, A.G.A. Successful treatment of SAPHO syndrome with apremilast. Br. J. Dermatol. 2018, 179, 959–962. [Google Scholar] [CrossRef] [PubMed]
  333. Sánchez-Velázquez, A.; Falkenhain-López, D.; Arroyo-Andrés, J.; Montero-Menárguez, J.; García-Donoso, C.; Postigo-Llorente, C. Apremilast: A novel adjuvant treatment for refractory isotretinoin-induced acne fulminans. Dermatol. Ther. 2022, 35, e15637. [Google Scholar] [CrossRef]
  334. Melnik, B.C.; Schmitz, G. Are therapeutic effects of antiacne agents mediated by activation of F ox O 1 and inhibition of m TORC 1? Exp. Dermatol. 2013, 22, 502–504. [Google Scholar] [CrossRef] [PubMed]
  335. Roy, T.; Boateng, S.T.; Uddin, M.B.; Banang-Mbeumi, S.; Yadav, R.K.; Bock, C.R.; Folahan, J.T.; Siwe-Noundou, X.; Walker, A.L.; King, J.A.; et al. The PI3K-Akt-mTOR and Associated Signaling Pathways as Molecular Drivers of Immune-Mediated Inflammatory Skin Diseases: Update on Therapeutic Strategy Using Natural and Synthetic Compounds. Cells 2023, 12, 1671. [Google Scholar] [CrossRef] [PubMed]
  336. Fabbrocini, G.; Staibano, S.; De Rosa, G.; Battimiello, V.; Fardella, N.; Ilardi, G.; La Rotonda, M.I.; Longobardi, A.; Mazzella, M.; Siano, M. Resveratrol-containing gel for the treatment of acne vulgaris: A single-blind, vehicle-controlled, pilot study. Am. J. Clin. Dermatol. 2011, 12, 133–141. [Google Scholar] [CrossRef] [PubMed]
  337. Reynolds, R.V.; Yeung, H.; Cheng, C.E.; Cook-Bolden, F.; Desai, S.R.; Druby, K.M.; Freeman, E.E.; Keri, J.E.; Stein Gold, L.F.; Tan, J.K.L.; et al. Guidelines of care for the management of acne vulgaris. J. Am. Acad. Dermatol. 2024, 90, 1006.e1–1006.e30. [Google Scholar] [CrossRef] [PubMed]
  338. Cho, S.I.; Yang, J.H.; Suh, D.H. Analysis of trends and status of physician-based evaluation methods in acne vulgaris from 2000 to 2019. J. Dermatol. 2021, 48, 42–48. [Google Scholar] [CrossRef]
  339. Bae, I.H.; Kwak, J.H.; Na, C.H.; Kim, M.S.; Shin, B.S.; Choi, H. A Comprehensive Review of the Acne Grading Scale in 2023. Ann. Dermatol. 2024, 36, 65–73. [Google Scholar] [CrossRef]
  340. US_FDA Guidance for Industry Acne Vulgaris: Developing Drugs for Treatment. Available online: https://downloads.regulations.gov/FDA-1975-N-0012-0317/attachment_250.pdf (accessed on 6 May 2024).
Figure 1. Acne pathogenesis and therapeutic targets.
Figure 1. Acne pathogenesis and therapeutic targets.
Ijms 25 05302 g001
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

Kim, H.J.; Kim, Y.H. Exploring Acne Treatments: From Pathophysiological Mechanisms to Emerging Therapies. Int. J. Mol. Sci. 2024, 25, 5302. https://doi.org/10.3390/ijms25105302

AMA Style

Kim HJ, Kim YH. Exploring Acne Treatments: From Pathophysiological Mechanisms to Emerging Therapies. International Journal of Molecular Sciences. 2024; 25(10):5302. https://doi.org/10.3390/ijms25105302

Chicago/Turabian Style

Kim, Hyun Jee, and Yeong Ho Kim. 2024. "Exploring Acne Treatments: From Pathophysiological Mechanisms to Emerging Therapies" International Journal of Molecular Sciences 25, no. 10: 5302. https://doi.org/10.3390/ijms25105302

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