Open access peer-reviewed chapter

Skin Pigmentation and Cosmetic Considerations for Even Skin Tone

Written By

Anita Damodaran and Nirmala Nair

Submitted: 12 October 2022 Reviewed: 21 October 2022 Published: 16 November 2022

DOI: 10.5772/intechopen.108693

From the Edited Volume

Pigmentation Disorders - Etiology and Recent Advances in Treatments

Edited by Shahin Aghaei

Chapter metrics overview

190 Chapter Downloads

View Full Metrics

Abstract

The pigment polymer, melanin is the major determinant of visible pigmentation of skin, hair, and eyes. Its synthesis within organelles called melanosomes in melanocytes and transfer to and distribution within keratinocytes in the epidermis regulates skin pigmentation. Sunlight and its ultraviolet radiation component have a well-established role in skin tanning, through increasing epidermal melanin. Additionally, linked to the pigmentary system are disorders of pigmentation, resulting in problems ranging from hypopigmentation to hyperpigmentation. This chapter provides an overview of the prominent hyperpigmentary manifestations such as post-inflammatory hyperpigmentation (e.g., that associated with acne), solar lentigo, melasma, and peri-orbital hyperpigmentation and recent advances in cosmetic interventions borne out of strong scientific understanding and consumer clinical studies.

Keywords

  • hyperpigmentation
  • niacinamide
  • resorcinol
  • cosmetic
  • even skin tone

1. Introduction

Visual appearance is the reflection of one’s inner self and is hence associated with self-esteem. To achieve skin devoid of imperfections has been an age-old quest, as it plays an important role in social acceptability. One such important aspect of appearance is skin color and its associated disorders. Skin color is determined by the amount and type of melanin (i.e., eumelanin and pheomelanin) synthesized in melanocytes and distributed within epidermal layers. It is also widely agreed that the type of melanin and its distribution in the epidermis are the most important factors in the protection of human skin from the detrimental effects of ultraviolet radiation (UVR). Constitutive skin color is what a person is born with and is genetically determined, while environmental factors such as UVR and pollution and physiological changes such as inflammation, hormonal changes, age, etc., influence facultative skin color.

Many constitutive pigmentation genes have been identified through spontaneous mutations causing a visible change in hair or skin color phenotype in mice or humans. All these genes are associated with either regulation of the pigmentation process in melanocytes or its development, survival, differentiation, and/or responses to stimuli [1]. In recent studies, using a genome-wide association approach complemented with targeted resequencing, scientists have identified previously unreported non-canonical skin pigmentation pathways in African populations, suggesting that the architecture of skin pigmentation can vary across humans subjected to different local evolutionary pressures [2, 3, 4]. Thus, novel variants in genes not previously linked to pigmentation are being discovered, and mounting evidence appears to suggest that there could be many more variants yet to be identified.

On the other hand, the molecular regulation of facultative pigmentation also called hyperpigmentation, has not been very well characterized. Limited knowledge exists on the etiology and the key genes/proteins involved in the induction, characteristics, and maintenance of the pathology of these conditions. Facial hyper-pigmentary conditions like melasma, post-inflammatory hyperpigmentation (PIH), periorbital darkening, and solar lentigines (SL, age spots) are common cosmetic concerns across populations, especially in individuals of skin of color. Some of these conditions are described in the next section with the latest understanding of their underlying biology.

Advertisement

2. Hyper pigmentary conditions and biology

The common facial melanotic conditions prevalent in humans are shown in Figure 1.

Figure 1.

Various facial hyper melanosis prevalent in humans. (A) Periorbital hyperpigmentation (POH); (B) solar/senile lentigines (SL); (C) post inflammatory hyperpigmentation (PIH); (D) melasma.

2.1 Solar lentigines

Solar lentigines (SL) is a common hyperpigmentary disease occurring in approximately 90% of old Caucasians and 70% of old Asians [5]. It is reported to develop more frequently in men than women [6, 7]. It presents a well-demarcated circumscribed yellow or brown macule with a diameter of 1–3 cm, predominantly on sun-exposed areas such as the face and the dorsal aspects of the hands and forearms [8]. However, unlike UV tanning, which may disappear with time in the absence of further sun exposure, SL is not known to resolve on its own, probably due to irreversible damage resulting from repeated sun exposure. Besides sun exposure, environmental factors such as constituents of ambient air pollution; may also contribute to SL development [9].

Histopathological assessment of SL has been carried out by several independent investigators. Prominent features include hyperpigmentation of the basal cell layer with a characteristic club-shaped elongation of rete ridges, numerous melanocytes, and increased melanin production without cellular atypia. A significant increase in both the epidermal area and the number of melanocytes, compared to that with a similar degree of photodamage, but frequently lacking the rete ridge hyperplasia classically associated with lentigines from other anatomic sites have been reported for facial SL [10, 11]. Classification of human SL progressive stages, based on the degree of melanin deposition and the depth and complexity of the rete ridges has been proposed with early stages of SL presenting with lower melanin levels as well as shorter and simpler rete ridges, whereas later stages show an accumulation of melanin and intricated rete ridges protruding into the thinning dermis and epidermis [12, 13, 14]. More recent studies have also pointed to the role of underlying inflammation in SL pathogenesis [15].

In recent years, insights generated through several qualitative and quantitative studies have significantly advanced our understanding of the underlying mechanisms in SL pathogenesis. Gene profiling studies reported upregulation of genes related to inflammation, fatty-acid metabolism, and melanogenesis, and downregulation of cornified envelope-related genes, indicating that solar lentigo is likely induced by mutagenic effects of repeated ultraviolet light exposures, thereby increasing melanin production, with a concomitant reduction in proliferation and differentiation of keratinocytes [14, 16]. The observed reduction in keratinocyte proliferation has been corroborated in a subsequent study, wherein the authors propose that early events may involve pigment-related genes, but with increasing time, increased levels of keratins 5 and 10 may put pressure on the basement membrane with basal keratinocytes loaded with melanin, pushing down toward the dermis to form rete ridges instead of moving upwards toward the stratum corneum as in normal skin [17]. Analysis of 160 skin biopsy samples from the lesional skin of SL showed a gradual increase in the expression of specific microRNAs from photo protected to peri-lesional skin to SL [18]. The profile reveals a significant change in microRNAs that regulate genes involved in lipid and fatty acid metabolism as well as inflammation.

Several independent studies have implicated cell types besides melanocytes in SL pathogenesis. In a study to decipher morphological and immunohistochemical changes of keratinocytes in facial SL, it was shown that individual keratinocytes were larger in size and showed increased p16 staining, pointing to a role for senescent changes underlying the pathogenesis [19]. More recently, in a study on 190 SL subjects, Notch1-dependent keratinocyte malfunction was suggested as the cause of the development of SL [20]. Besides the epidermal compartment, several independent studies have also reported changes in epithelial-mesenchymal crosstalk in SL pathogenesis. Prominent among these are endothelin-1 (ET-1), hepatocyte growth factor (HGF), keratinocyte growth factor (KGF), and stem cell factor (SCF) [21, 22]. An interesting difference between UVB melanosis and SL is that in the former, ET-1 and SCF are stimulated by interleukin-1 alpha (IL-1α), while in SL, they are stimulated by tumor necrosis factor-alpha (TNFα) [23]. Observed degradation of heparan sulfate chains owing to increased heparinase at the dermal-epidermal junction (DEJ), may exacerbate the transfer of growth factors and cytokines between the epidermis and dermis, contributing to hyperpigmentation in SL [24]. Studies have also pointed to increased blood flow and vasculature in SL using immunohistochemistry [25], 3D microvascular analysis [26], and optical coherence tomography angiography [27]. Most recently, a dysregulation of Nrf2 signaling has been shown to be associated with SL pathogenesis [28]. Taken together, these pieces of evidence point to a complex network of cells and secreted factors that are likely involved in the development and progression of SL.

2.2 Postinflammatory hyperpigmentation

Postinflammatory hyperpigmentation (PIH) is one of the most common causes of altered normal skin color [29]. It is an acquired hyper-melanosis and a common sequela of various inflammatory episodes. It primarily afflicts patients with darker skin types (Fitzpatrick types III–VI), although all skin types can develop PIH [30]. As compared to 25% in white patients, the prevalence of PIH in Hispanic and black patients is 48 and 65%, respectively [31]. Additionally, it has been reported to occur with equal incidence in males and females of all ages [32]. Although PIH is easily diagnosed from the patient’s history and the presence of inflammation, several dermatoses lead to PIH without noticeable inflammation. Hence, while visual assessment can aid in PIH evaluation, the use of non-invasive methods can further aid diagnosis [33].

PIH presents itself in two forms—epidermal (light to dark brown) which usually disappears spontaneously and dermal (blue-gray coloration) which has a more prolonged course and may either take years to resolve or may be permanent [33, 34]. Unfortunately, a vicious cycle may emerge leading to new areas of hyperpigmentation if the underlying inflammatory disorder is not resolved [34]. In many cases, the extent of damage to the skin may result in scars and keloids.

Causative factors can be divided into endogenous and exogenous factors. The former includes PIH from inherited diseases such as incontinentia pigmenti, cutaneous diseases such as acne, lichen planus, erythema dyschromia persistans, and facial melanoses, as well as systemic diseases such as morphea, porphyria, and biliary cirrhosis [29]. The latter on the other hand include mechanical trauma, extremes of temperature, ionizing and non-ionizing radiation, phototoxic reactions, laser resurfacing [35, 36], and cases of contact dermatitis [37]. Noteworthy is the PIH commonly accompanying acne, often considered more bothersome than acne itself [38] and that accompanying axillary darkening because of shaving, plucking, and/or antiperspirant use [39].

Different histological patterns of PIH emerge, depending on which skin layer is involved. For the epidermal type, there appears to be an increase in the number (hyperplasia), size (hypertrophy), and activity of melanocytes, leading to increased melanin content, with little dermal changes [40]. On the other hand, in the dermal type, melanin enters the dermis, contributing to dermal pigmentation, with an accompanying perivascular lymphocytic infiltrate [40, 41].

Likely pathogenic mechanisms underlying PIH include inflammatory mediators such as arachidonic acid metabolites, nitric oxide, etc., and crosstalk between melanocytes and neighboring keratinocytes and fibroblasts, resulting in the exchange of several melanogenic factors [42].

2.3 Periorbital hyperpigmentation

Periorbital hyperpigmentation (POH) is a common dermatological condition that presents as a dark periorbital area beneath or around the eyes. While it afflicts about 78% of the global population, the majority of the affected individuals are of Asian and African origin [43]. It is reported to occur in both sexes with a greater frequency in females with early onset at age 16–25 [44, 45]. It can also frequently be seen in multiple members of the same family [46].

While the etiology is multifactorial, prominent causative factors include familial, UV, inadequate sleep, post-inflammatory hyperpigmentation following atopic dermatitis, allergic contact dermatitis, lichen planus pigmentosus, erythema dyschromicum perstans [47] tear-trough depression, and periorbital edema [43, 48, 49]. Basis the clinical pattern of pigmentation and vasculature, POH can be classified into (a) constitutional, which involves the presence of a curved band of dark brown to black pigmentation on the skin of the lower eyelids, (b) postinflammatory pigmentation, marked by the presence of irregular patches of dark brownish or gray pigmentation on the skin of the lower, upper, or both the eyelids with lichenifed eczema in surrounding areas, (c) vascular, involving bluish discoloration of the lower eyelids and visible greenish blue veins that become more prominent on stretching of the overlying skin, (d) the shadow effect, involving the presence of deep tear trough over the medial aspect of the inferior orbital rim that disappears with direct lighting, and (e) mixed highlighted by presence of periorbital blue, purple, or pink hue with puffiness associated with palpebral bags, blepharoptosis, and loss of fat with bony prominence [43, 50, 51].

The pathogenesis of POH may be due to one of many factors, notably, dermal melanocytosis [52], an extension of pigmentary demarcation lines of the face [45], postinflammatory hyperpigmentation, superficial location of the vasculature [53], tear-trough depression [52], and thinning of skin [54]. Ultraviolet-induced damage stimulates melanogenesis through multiple pathways [55] or melasma, if it appears on the eyelids, leading to dark eyelids and a tanned lower eyelid region [56].

Histological examination reveals that the most consistent features of POH are hyper melanization of the basal layer and lower malpighian layer with increased melanin granules together with dermal melanin incontinence, dermal melanophages, and perivascular lymphocytic infiltrate [53, 57, 58]. Additionally, the dilation of dermal blood vessels may contribute to the severity of POH [58]. In a study on Asian subjects, the group reported dermal melanocytosis along with melanophages in patients with POH as detected by immune-histochemical staining with S-100 antigen [59]. The almost universal presence of dermal melanin is likely to influence poor treatment outcomes of POH.

Recently, in a study on Caucasian, Asian, and African subjects, it was confirmed through various instrumental measurements that the three features associated with the occurrence of infraorbital dark circles, were hyperpigmentation, a tendency for more dilated, thicker, or increased number of capillaries and thinner skin in the under-eye area in Caucasian subjects. These trends were also observed in the African and far east Asian subjects [60]. However, detailed molecular mechanisms involved in POH continue to elude us.

2.4 Melasma

Melasma is defined as a chronic, acquired disorder of hyperpigmentation, presenting as symmetrical, light to dark brown and ashen gray-brown macules and patches on sun-exposed areas of the face and neck [61]. It is also sometimes referred to in medical literature as the “mask of pregnancy or chloasma,” the latter originating from the Greek word chloazein, meaning to be green [62]. It is primarily a disease of adult women, with reported cases in men being only 10% [63], presenting mostly in the facial area of darker-complexioned individuals (skin types IV–VI) of Hispanic, East Asian, and Southeast Asian origin exposed to intense ultraviolet radiation [62] and shorter wavelengths of visible light [64]. In a multicenter survey from nine countries, the mean onset of age was 34 years [65]. It negatively impacts the quality of life; often, poor therapeutic responses pose a huge challenge to dermatologists and escalate the cost of treatment for affected individuals [66]. Predisposing factors include pregnancy, hormonal therapies (including oral contraceptives), phototoxic and anti-epileptic medications, intense sun exposure [67], and genetic predisposition [68, 69]. Recent studies suggest that thyroid hormones may play a key role in melasma [70].

There are two broad ways to classify melasma: (a) based on the distribution of lesions, three clinical patterns of melasma have been recognized—Centro facial, malar, and mandibular; (b) based on the distribution of melanin in the epidermis and dermis using a Woods lamp into epidermal, dermal, mixed, and indeterminate types [62].

As with other hyperpigmentary conditions, the pathogenesis is multifactorial and poorly understood. Some of the prominently reported mechanisms underlying melasma pathogenesis include melanocyte activation, melanin and melanosome retention in the epidermis and dermis, basement membrane damage, solar elastosis, increased mast cell count and increased vascularization [71]. Overall, the epidemiological and pathophysiological data seem to suggest that melasma is a photoaging disorder [64].

Transcriptional profiling revealed that a subset of Wnt signaling modulators, including Wnt inhibitory factor-1 (WIF-1), secreted frizzled-related protein 2 (sFRP2), and Wnt5a, were upregulated in lesional melasma skin [72]. The downregulation of WIF-1 which may occur in keratinocytes and fibroblasts can influence melasma pathogenesis through the up-regulation of canonical and non-canonical Wnt signaling [73]. Given that UV is implicated in melasma pathogenesis, the upregulation of several of the UV-induced cytokines in melasma is not surprising.

Likewise, melanocytes in melasma can also be activated by hormones. Notably, lesional skin shows increased expression of progesterone receptors [74, 75] and estrogen receptor beta expression in the dermis [74]. Significant advances have been made in elucidating the influence of estrogens on melanogenesis [76].

The number of blood vessels, vessel size, and vessel density are greater in lesional melasma skin than in perilesional skin [77, 78, 79]. Factors that could influence vascularization and pigmentation in melasma are upregulated in lesional areas of melasma. Prominent among these are vascular endothelial growth factor (VEGF), stem cell factor (SCF), and inducible nitric oxide synthase (iNOs) [77, 80, 81]. Endothelin-1 (ET-1) released from endothelial cells stimulates pigmentation through endothelin receptor B activation at the surface of melanocytes [82]. Concerning solar elastosis, the current consensus is that 83–93% of melasma patients show a variable degree of solar elastosis with abnormal elastotic material [83, 84]. An increased number of mast cells, particularly around elastotic areas of the dermis in lesional melasma [83] is believed to exacerbate solar elastosis, besides promoting basement membrane disruption and extracellular matrix (ECM) degradation through the release of tryptase, vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2, and transforming growth factor-beta (TGF-β) [85].

Basement membrane damage has been reported in melasma patients via periodic acid-Schiff-diastase (D-PAS) staining and anti-collagen type IV immunohistochemistry, respectively [84]. This predisposes melanocytes to hang into the dermal compartment—the characteristic pendulous melanocytes observed in histology leading to dermal incontinence [86]. Increased expression of matrix metalloproteinases such as MMP2 and 9 in response to chronic UV exposure is likely to contribute to basement membrane damage [87]. The outcomes of these events result in melanin or melanophages in the dermis [84, 88]. Recently, melanin content, location and distribution in melasma, and elastosis severity were confirmed using multiphoton microscopy [89]. Dermal incontinence is a challenge across hyperpigmentation and one, that is likely to significantly contribute to the recalcitrant nature of these conditions.

While we continue to investigate the pathology and biology of various hyperpigmentary conditions, the other key question that needs to be addressed is the factors that make an individual susceptible to a particular hyperpigmentary disorder, besides family history. An in-depth analysis of the epidemiology and population genetics data may help in identifying key population or individual biology, which will in turn support the designing of better preventive options.

Advertisement

3. Role of UV and visible light in skin pigmentation

From the discussion on hyperpigmentary disorders prevalent in humans in the previous section, the role of UV is quite evident and seems to be central to all conditions, so it is worthwhile to discuss the effects of UV and solar spectrum on the skin in the following section.

Ultraviolet radiation on the earth’s surface, both UVB and UVA radiation, constitutes about 5% of total sunlight received. In skin phototypes I–II, exposure to UVB radiation, leads to erythema and sunburns [90]. Skin pigmentation or tanning induced by UV radiation in skin phototypes III–IV, occurs in three different phases-immediate pigment darkening (IPD) and persistent pigment darkening (PPD), both thought to be the result of oxidation or redistribution of melanin while delayed tanning (DT), a characteristic of UVB is because of new melanin synthesis [91, 92]. There is limited data on the skin types V–VI and their response behavior to UV. While erythema, IPD, and DT are the noticeable effects of UV-induced damage to the skin in a short period, repeated exposure may result in one of the chronic effects of sun exposure such as photo-aging which results in the development of deep wrinkles and spots on the exposed skin [93].

UVB (290–320 nm) being more energetic directly damages DNA by causing cyclobutane pyrimidine dimerization and the formation of 6–4 photoproducts. UVA rays (320–400 nm) on the other hand generate reactive oxygen species (ROS) that, in turn, cause indirect DNA damage and activation of several cellular pathways [94]. At the molecular level, UV has been shown to cause DNA damage with subsequent accumulation of p53 protein which regulates the expression of the transcription factor hepatocyte nuclear factor-1α in melanocytes, which in turn regulates MITF and tyrosinase levels [95] and the transcription of the pro-opiomelanocortin (POMC) gene in keratinocytes which eventually regulates MC1R induced melanogenesis in melanocytes [96]. GWAS, linkage, and association-based studies on European and non-European populations suggest the role of distinct genes like glutamate metabotropic receptor 6 (GRM6), activating transcription factor 1 (ATF1), WNT1, and pre melanosome protein 17 (SILV/Pmel17) in observed differential tanning response [1, 97] pointing to the fact that the genetic makeup of the population could drive the nature and the molecular events leading to a tan.

In the past decade, the focus has shifted to the other wavelengths of light namely visible light (400–700 nm) and infrared radiation (IR; 700–1400 nm) which form a major part of the solar spectrum, 45 and 54%, respectively. These longer wavelengths penetrate deeper into the skin and can induce ROS and potentially cause erythema in light skin and pigmentary changes in individuals with darker skin types [92, 98, 99, 100, 101]. Furthermore, susceptibility to pigmentation by visible and IR radiation was shown to reside in darker skin types more than Caucasian skin [92, 102, 103]. Visible light and IR activate metalloproteinases and decrease collagen production by inducing oxidative stress [104, 105]. Damodaran et al. [106] demonstrated that among variable parts of the visible light spectrum, blue wavelength contributes maximally to pigmentation in human subjects (Figure 2A and B). As the blue region is adjacent to UV radiation in terms of wavelength and frequency, blue light is expected to induce photobiological effects like UV radiation, including photoaging and skin pigmentation [101, 107]. In the field of ophthalmology, researchers have identified that “excessive blue light exposure” may cause photokeratitis and retinal injury [108]. Interestingly, the prevalence of photoreceptors like opsins, which recognize blue and green wavelengths of visible light in the eye, have been demonstrated in human skin [109, 110] including in melanocytes, which could be modulated by light exposure.

Figure 2.

Effect of visible light on skin pigmentation—clinical evidence. (A) Effect of different wavelengths of light on skin pigmentation; (B) effect of visible light on color (L*, b*, and a*), hydration, TEWL, antioxidants (catalase, total antioxidants [TOAX], superoxide dismutase [SOD], and free fatty acids [faa]), and inflammation markers (IL-1a) as measured from tape strips.

Multiple studies also suggest that sun exposure (UVA, B, and visible light) could have a role in the induction or exacerbation of hyperpigmentary conditions like freckles, solar lentigines, PIH, melasma, and dark circles [45, 111, 112, 113]. In a randomized study with melasma subjects, Castanedo-Cazares et al. [114] showed that UV sunscreens with visible light protection along with 4%HQ treatment significantly improve MASI scores as against just UV protection with HQ treatment [114, 115].

Therefore, it can be assumed that protection is mandatory not only against UV but against the whole spectrum of light as an adjuvant to regular therapy for hyperpigmentation [99, 116]. In addition to UV, the role of blue light /high-energy visible light from non-solar sources like digital devices also needs to be investigated for its contribution to the exacerbation of hyperpigmentation.

Advertisement

4. Cosmetic agents for hyperpigmentation

The consumer desire to achieve uniform skin tone or complexion has been actively supported and promoted by the cosmetic industry for several decades. The commonly used agents for the treatment of hyperpigmentation are hydroquinone, arbutin, kojic acid, and their derivatives, various derivatives of vitamin C, tretinoin, and natural extracts [117] besides chemical peels, lasers, and light-based therapies. This section will focus on topical solutions recommended for hyperpigmentation.

Hydroquinone (HQ) has been the gold standard in the treatment of hyperpigmentation for over five decades although due to its adverse effects (including toxicity and mutagenicity), it is now banned from use in cosmetics or for over-the-counter (OTC) sales in many countries. Currently, HQ is banned in OTC and available only under prescriptions [118, 119], Considering its deleterious effects, consumer interest is shifting to safer and naturally derived actives.

Over time, HQ has been combined with other drugs, with the intent of improving its efficacy. It was Albert Kligman who first developed and demonstrated the efficacy of a formula containing 0.1% tretinoin, 5% hydroquinone, and 0.1% dexamethasone in 1975. A modified Kligman’s formula containing 4% HQ , 0.05% tretinoin, and 0.01% fluocinolone acetonide is currently in use, with the backing of successful clinical on melasma patients [120, 121, 122]. This mix is popularly called Tri-Luma or the triple combination, can be used across all Fitzpatrick phototypes, and is reported to be better tolerated.

Hydroquinone’s action is mostly suggested to be via its ability to inhibit melanin formation by blocking tyrosinase enzyme activity [123, 124]. Deri et al. [125], by visualization studies of HQ in the active site of tyrosinase protein crystals, together with molecular modeling, binding constant analysis, and kinetic experiments, have confirmed that HQ can act both as a tyrosinase enzyme substrate and as an inhibitor. As a substrate, instead of forming melanin, it gets metabolized to quinones and free radicals which cause lipid peroxidation, damaging the melanocyte membrane and leading to melanocyte cytotoxicity and depigmentation [126, 127]. However, in a recent study designed to compare the effects of arbutin, HQ , and kojic acid with numerous resorcinol vis-à-vis inhibition of recombinant human tyrosinase [128, 129], HQ was found to be ineffective against tyrosinase enzyme as compared to resorcinol (Figure 3). Thus, although considered to be a tyrosinase inhibitor for a long, it appears that the cytotoxic potential of HQ may largely contribute to its depigmentation effect.

Figure 3.

Inhibition of recombinant human tyrosinase enzyme activity by different resorcinols and known tyrosinase inhibitors. Dose-dependent inhibition of tyrosinase enzyme activity was performed using synthetic recombinant human tyrosinase enzyme protein and DOPA as the substrate.

Other tyrosinase inhibitors like arbutin, a derivative of hydroquinone, and kojic acid, have lesser adverse effects, unlike hydroquinone. Glycolic acid, on the other hand, works by causing the desquamation of keratinocytes or epidermolysis when used at higher concentrations [130].

A range of vitamins and their derivatives with enhanced stability or improved dermal penetration are also in use in various cosmetic and pharmaceutical products for skin tone benefits [131, 132].

Vitamin C (VC), also known as ascorbic acid, is a water-soluble vitamin, essential for several processes in human skin, such as dermal collagen synthesis, cell turnover, ROS scavenging, and many more [133]. Emerging evidence indicates that VC and its derivatives can exert therapeutic effects on recalcitrant melasma and facial hyperpigmentation, especially with improved dermal delivery [134, 135]. Vitamin C and its derivatives, magnesium ascorbyl phosphate, and 3-O-ethyl-L-ascorbic acid, exert their depigmenting effect by acidification of the melanocyte cytoplasm, which in turn suppresses the catalytic activity of tyrosinase. Cytoplasmic pH is critical for the catalytic activity of tyrosinase and for the assembly of Pmel17 protein fibrils in the melanosome to form the scaffold upon which melanin is deposited, and any change would therefore influence the melanogenesis process in melanocytes [136, 137]. Vitamin C seems to induce the expression of sodium-dependent VC transporter-2 that further facilitates the transmembrane transport of VC which in turn leads to acidification of the cytoplasm [138]. In addition, 3-O-ethyl-L-ascorbic acid was also shown to modulate the function through Nrf2-mediated α-MSH inhibition in UVA-irradiated keratinocytes and by autophagy induction and inhibition of α-MSH-stimulated melanogenesis in melanocytes [139].

The amide form of vitamin B3, niacinamide has been shown to inhibit melanosome transfer from the melanocytes to the epidermal keratinocytes [140, 141, 142]. Various other functions of niacinamide, including improved immunity, barrier function, etc., make it a popular ingredient in multiple cosmetic preparations [132, 143].

Oral and topical tranexamic acid is a recent addition to dermatologists’ treatment options to tackle recalcitrant conditions such as melasma and PIH, in combination with other skin tone agents, though more successful studies are needed to back its topical use [144, 145]. In addition, superficial chemical peeling alone or in combination with other topical procedures is also key in the treatment of hyper-pigmentary conditions [146]. In such treatment, avoidance of sun exposure and the use of sunscreens is a must for a successful outcome. Then there are laser and light-based therapies like intense pulsed light, low fluence Q-switched lasers, and non-ablative fractionated lasers that are currently prevalent for use for melasma. Though effective these are also associated with an increased risk of recurrence and PIH [146].

In the next section, we will focus on currently popular cosmetic ingredients such as niacinamide, resorcinol, and retinoids and their potential for the future.

4.1 Niacinamide

Niacinamide has been reported to possess numerous properties, for instance, anti-inflammatory, antimicrobial, immunity booster, bioenergetics modulator, and antioxidant, which makes it suitable for various dermatological uses [132, 143, 147]. Niacinamide can effectively treat various skin conditions viz. aging, hyperpigmentation, acne, rosacea, psoriasis, pruritus, dermatitis, etc. [132, 143]. It increases the biosynthesis of ceramides, as well as other stratum corneum lipids and thus enhances epidermal permeability barrier function [148]. Studies have shown that niacinamide modulated NFkB-mediated inflammatory response, which could support its anti-aging activity [149]. In addition, niacinamide protected against collagen loss in the dermis by preventing its glycation and activation of fibroblast function [150].

Niacinamide is also a well-established and commonly used skin-brightening agent and has been reported to have a significant effect on various hyperpigmentary conditions [142, 151, 152]. Navarrete-Solis et al. [152] showed significant improvement in MASI scores in melasma subjects treated with 4% niacinamide same as for 4% hydroquinone, with less melanin and inflammatory infiltration on histological evaluation of biopsies. However, the lightening effect of hydroquinone was evident as early as the first month of treatment, albeit with some adverse events, whereas with niacinamide, it was noted only in the second month of treatment The effect of niacinamide on melasma was attributed to its anti-inflammatory, melanosome transfer inhibition, and antiaging effects on elastosis [152].

Another study was carried out by a group from Iran [153], on the efficacy of topical 4% niacinamide and 1% clindamycin gels in a randomized, double-blind clinical trial on acne patients with moderate inflammatory acne vulgaris. The mean grade of acne and mean count of papules in patients were comparable in both treatments at the end of the study. In a double-blind study carried out on Indian subjects with acne marks, the application of 1.25% of niacinamide significantly reduced acne marks within 6 weeks as compared to baseline, assessed visually using a skin color scale by an expert dermatologist (Figure 4A and B) [154].

Figure 4.

Clinical benefits of niacinamide for acne marks. (A) The average extent of lightening of blemishes after product usage for week 2/4/6/8. Statistically significant reduction in the intensity of blemish color at week 2/4/6/8. The negative y-axis values indicate a brightening effect. (B) The facial images of subjects at weeks 0 and 8, after the application of a niacinamide-containing formulation. These photographs are true evidence from the study and are representative of some of the best responders from the study.

In an axillary depigmentation trial in phototype III–V, the treatment of 4% niacinamide and desonide 0.05%, significantly reduced the hyperpigmentation of the axillae in 9 weeks versus the placebo group. However, the efficacy of the topical steroid was significantly better than niacinamide treatment and the effect was apparent in desonide-treated subjects by 6 weeks of application. There were also reduced mononuclear and phagocytic cell infiltrates, as well as melanin expression in niacinamide-treated subjects, compared with baseline as determined from the biopsies. However, desonide, in addition, improved the basement membrane as seen in histopathology [155].

Studies have supported the use of anti-inflammatory and depigmenting agents for treatments of PIH, however very few have objectively evaluated PIH development. Damodaran et al. [154] used the sodium dodecyl sulfate (SDS) occluded patch test human trial model of irritation [156] to evaluate the effects of inflammation on pigmentation and the efficacy of niacinamide on inflammation and PIH. The study showed that the pigment or L* value increased with an increasing irritation score (Figure 5A). Pre-application of niacinamide reduced inflammation resulting from SDS and the subsequent development of pigmentation (Figure 5B and C).

Figure 5.

SDS-induced human occluded patch test. (A) Irritation score induced by SDS follows the trends as with pigmentation (L* increase); (B) inhibition of SDS-induced inflammation by pre-application of niacinamide in a dose-dependent manner; (C) niacinamide-containing formulation reduced PIH induced by SDS.

A possible molecular mechanism in the efficacy of niacinamide on PIH could be attributed to the modulation of a protein called SERPINB3 in the skin [157]. Serpin Family B Member 3 (SERPINB3) is an endogenous protease inhibitor known for its role in maintaining epidermal barrier homeostasis and is implicated in inflammatory skin conditions including acne [158, 159]. Nair et al. [157] demonstrated that the dermal fibroblasts secrete SERPINB3 in response to inflammatory cytokine interleukin-1 alpha, that in turn induced melanin production in melanocytes (Figure 6A). Niacinamide could blunt the SERPINB3 secretion by fibroblasts (Figure 6B) suggesting the novel role of SERPINB3 in its anti-inflammatory and anti-PIH activity for the potential amelioration of acne and acne marks.

Figure 6.

Effect of SERPINEB3 on pigmentation and its modulation by niacinamide. (A) 24 h of IL-1α treatment increased SERPINB3 protein secretion (25% vs. control) from dermal fibroblasts. (B) Pre-treatment of cells with 5 mM niacinamide significantly reduced (29% vs. IL-1α) the SERPINB3 secretion of cells. The values are mean ± SE of cells from three independent donors. *p < 0.05 vs. control, #p < 0.05 vs. IL-1α.

4.2 Resorcinol

The synthesis and distribution of melanin contribute to skin and hair color in mammals. In human melanocytes, melanin is synthesized within melanosomes by the enzyme tyrosinase, which catalyzes the rate-limiting reaction ie. the conversion of tyrosine to DOPA quinone [160, 161, 162].

Tyrosinase, also known as polyphenol oxidase (PPO), is a copper-containing mixed-function oxidase, widely distributed in micro-organisms, animals, and plants. These oxidases catalyze two distinct reactions of melanin synthesis: hydroxylation of a tyrosine a monophenol (monophenolase activity) and the oxidation of DOPA-an o-diphenol to the corresponding o-quinone (diphenolase activity). The quinones formed being highly reactive, polymerize spontaneously to form high molecular weight compounds or pigments—melanin [163]. Melanin is then secreted by melanocyte cells, which are distributed in keratinocytes in the basal layer of the epidermis in the skin. However, abnormal accumulation of melanin has been associated with hyper-pigmentary conditions, including melasma, freckles, and senile lentigines as covered in the earlier section.

Identification of potent and specific tyrosinase inhibitors has been ongoing for several decades due to its wider implications for the food, pharmaceutical, and cosmetic industries [164, 165]. However, most of the initial discoveries were made using mushroom tyrosinase as a model system [166, 167]. It is only in recent times that more potent tyrosinase inhibitors have been identified using enzymes sourced from human melanocytes or recombinant human tyrosinase protein with superior efficacy on melanotic conditions like solar lentigines and melasma, [129, 168]. The commonly used compounds in the class of tyrosinase inhibitors include hydroquinone, arbutin, kojic acid, thiols, and resorcinol and among them, resorcinol derivatives are the most potent in enzyme inhibition [128, 129]. It has been shown that resorcinol is a mono-oxygenase substrate and gets oxidized to hydroxy intermediate; 3-hydroxy-ortho-quinone, which results in irreversible elimination of Cu (0) of tyrosinase enzyme leading to inactivation of the enzyme [169, 170]. Tyrosinase inhibition activity, though, is mainly attributed to its resorcinol moiety, the efficacy can be further modulated by the various substitution at the 4-position of resorcinol [129, 168, 170]. The levels of tyrosinase inhibition by various 4-resorcinols vary with respect to their alkyl chain length (Figure 3).

Phenylethyl resorcinol (PER, 4-(1-phenylethyl)1,3-benzenediol) is a potent inhibitor of tyrosinase [171, 172, 173]. PER has been in use as a skin lightener in cosmetics for more than a decade, but the clinical demonstration of its efficacy on hyperpigmentation is limited by its chemical instability to UV light [174, 175].

The clinical efficacy of 4-butyl resorcinol (BR) on the other hand, has been very well established in multiple human trials [173, 176, 177, 178, 179]. A randomized, single-blind clinical study was conducted on female subjects with 0.3% 4-butyl resorcinol, 0.3% 4-hexyl resorcinol, and 0.5% 4-phenylethy resorcinol for 12 weeks. 4-butyl resorcinol significantly reduced the appearance of age spots on the forearm after 8 weeks, while others did in 12 weeks. Thereafter, 1% 4-butyl resorcinol was clinically evaluated on age spots on the forearm again; enhancement in skin tone was noticeable by the end of the 16 weeks and was judged to be significant after 4 weeks [173]. The 0.1% 4-butyl resorcinol cream showed rapid efficacy with excellent tolerability in patients with melasma [177, 180]. Liposome-encapsulated 4-butyl resorcinol had a significantly improved effect on the melanin index after 8 weeks of application, without any occurrence of adverse events [178]. In a study on Indian subjects to assess the efficacy, safety, and tolerability of 0.3% 4-butyl resorcinol in melasma patients, a significant decrease in MASI score was observed with no evidence of adverse effect [179].

Another alkyl resorcinol in the skin tone benefit market is 4-hexyl resorcinol (4-HR), an effective tyrosinase inhibitor that was used earlier in the food and medical industries [181, 182, 183]. 4-HR has been demonstrated through in-vitro and human trials to improve hyperpigmentation through its tyrosinase inhibition activity and its anti-inflammatory potential [184, 185]. 4-HR could regulate collagen and elastin production by inhibiting NF-kB activity in fibroblasts and improving photodamaged skin and clinical signs of aging [186]. Chaudhuri [187] reported multiple benefits of 4-HR, including evidence for its anti-microbial, antioxidant, and anti-glycation effects, along with its effect on extracellular matrix proteins. In a face study by Won et al. [188], 4-HR successfully improved overall skin tone when used in an emulsion over 12 weeks.

The most recent entrant in the skin tone market is the imidazolyl derivative of resorcinol, thiamidol, identified through a high throughput screening of a large compound library on human tyrosinase protein. With an IC50 of 1 μM for tyrosinase enzyme inhibition, it is the most potent inhibitor known so far [129, 168]. The group has also demonstrated the efficacy of thiamidol on various hyperpigmentary conditions like melasma [189, 190, 191], solar lentigines [192], PIH [193], and UV-induced skin damage [194] and demonstrated its efficacy to be superior to HQ [190].

The availability of recombinant human tyrosinase protein [128, 129] combined with new platforms that allow for structure-active site prediction based on amino acid sequence [168] will hopefully promote further investigations on tyrosinase chemistry, leading to the identification of inhibitors with greater potency and reduced safety concerns.

In general, as apparent with hydroquinone, the efficacy of a molecule gets compromised when safety becomes paramount. The usage of high doses of ingredients is also a concern in terms of formulation compatibility, sensory, and cost. In such cases, efficacy could be boosted by combining it with other potent agents with a different mode of action and examples do exist where combination therapies have been shown to work for the treatment of recalcitrant hyperpigmentary conditions such as melasma, with a clear example being Kligman’s triple combination.

Shariff et al. [128] successfully adopted a combinatorial approach to demonstrate the superior clinical efficacy of a cosmetic formula containing 4-hexyl resorcinol and niacinamide. Based on the in vitro evaluation, appropriate ingredients, and doses were selected to develop a formulation which was then evaluated for spot lightening and anti-aging on Chinese subjects. In the double-blind split-face study comparing 3% niacinamide with 0.4% 4-HR+ 3% niacinamide over 12 weeks of application, significant improvement was achieved for spot lightening and fine lines and wrinkles in crow’s feet and perioral areas by the combination of 4HR+ niacinamide over niacinamide alone. This highlighted that combining molecules with different modes of action on pigmentation, resorcinol as a tyrosinase inhibitor and niacinamide as a melanosome transfer and anti-inflammatory molecule, may lead to better and safer skin tone formulas with efficacy on par or better than prescription drugs like hydroquinone.

Another ingredient with a different and established mode of action is retinoid and will be covered in the next section.

4.3 Retinoids

Topical retinoids are recommended by the American academy of dermatology as the first line of treatment for acne and hyperpigmentation, based on strong levels of evidence from well-controlled clinical trials [195, 196]. For issues related to irritation from retinoids, the use of moisturizers is recommended. Dermatologists use tretinoin along with other topical agents and procedures such as superficial chemical peels, to treat hyperpigmentation and improve outcomes [197].

Retinoids have been shown to bind and activate retinoid X nuclear receptors and mediate their cellular function which includes inhibition of keratinocyte proliferation and normalization of follicular differentiation which restores normal desquamation and helps to unclog pores [198]. Retinoids are also anti-inflammatory and suppress toll-like receptors, cytokine, and nitric oxide production [199, 200]. It also corrects pigmentation by inhibiting melanosome transfer and the rate of epidermal turnover [196].

Though tretinoin is recommended for the treatment of dark circles [201], melasma [202], and acne [203], some new retinoid derivatives such as adapalene, tazarotene, and trifarotene have also come into the market for dermatological usage [204, 205].

The effects of retinoids on hyperpigmentation have been evaluated in various studies, either for efficacy or safety assessment, with favorable results [195]. Tretinoin was the first retinoid specifically evaluated on hyperpigmentation in patients with melanin-rich skin. Tretinoin reduced hyperpigmented lesions mostly due to PIH in phototype IV–VI skin subjects, with improvement evident as early as week 4 [206]. However, irritation continued to be an issue and was managed by emollients and moisturizers.

The new derivative, tazarotene has FDA approval at 0.1% levels for use as an adjunct for the treatment of fine lines and wrinkles, mottled hyperpigmentation, and lentigines on the face [207]. In an acne-induced PIH or marks study with skin types III-VI, statistically significant reductions were achieved with tazarotene versus vehicle on hyperpigmented lesions, [208]. However, adverse events were still observed with tazarotene as in the case of tretinoin. Trifarotene 0.005% cream is a fourth-generation retinoid approved for the treatment of facial and truncal acne [209], but its clinical effects on PIH in patients with skin of color have not yet been reported.

Combination therapies are the mainstay of retinoid therapies which also help counteract associated irritation issues, essentially either with other topical drugs like HQ , clindamycin or light/laser therapies [196, 197].

Retinyl esters, such as retinyl propionate, are known to be less irritating and less efficacious than retinol or tretinoin and this was hypothesized to be the result of rapid degradation of retinoic acid generated endogenously from esters, by P450 (CYP) protein. Adamus et al. [210] demonstrated the use of climbazole a weak pan-inhibitor P450 (CYP) to prevent loss due to the degradation of retinoic acid thus boosting the efficacy of retinyl propionate in vitro. This was followed by a human trial which confirmed the hypothesis and demonstrated that retinyl propionate alone, and in combination with climbazole, was significantly less irritating than a similar serum containing retinol. The retinyl propionate with climbazole serum treatment also showed equal, or better efficacy than retinol alone serum on fine and coarse lines and wrinkles, as well as pigmentation. Additionally, the combined retinol/climbazole was shown to modulate histological biomarkers of skin aging viz. epidermal thickness, procollagen-1 protein, Ki-67, and retinoid activity (CRABP2, KRT4) [211].

Thus, milder but efficacious retinoids like retinyl propionate with climbazole could be another ingredient with a different mode of activity for the exploration of potential combinations in the future.

Advertisement

5. Conclusion

Melanin in the skin provides better photoprotection to melanated skin. However, abnormal accumulation of melanin as seen in hyperpigmentary conditions, viz. melasma, freckles, senile lentigines, etc. is undesirable. Hyperpigmentary conditions are by and large refractory and recurrent in nature, particularly to traditional cosmetic actives. Multiple risk factors, together with the complex and less understood biology of these conditions, largely contribute to poor treatment outcomes. The most effective interventions for their management are mostly drug-based preparations and physical methods such as lasers that are not very well tolerated, particularly in skin of color.

A comprehensive understanding of hyperpigmentation biology combined with the development of potent mixes that target multiple pathogenic mechanisms is likely the way to provide prescription strength efficacy for its control, without associated safety concerns and side effects. Also, as our understanding of the impact of sunlight and its various components (besides UV) on skin pigmentation and health improves, it is imperative to educate consumers on the diligent use of more comprehensive sun protection measures, regardless of skin phototype.

References

  1. 1. Pavan WJ, Strum RA. The genetics of human skin and hair pigmentation. Annual Review of Genomics and Human Genetics. 2019;20:41-72. DOI: 10.1146/annurev-genom-083118-015230
  2. 2. Naik PP, Farrukh SN. Influence of ethnicities and skin color variations in different populations: A review. Skin Pharmacology and Physiology. 2022;35:65-76. DOI: 10.1159/000518826
  3. 3. Deng L, Xu S. Adaptation of human skin color in various populations. Hereditas. 2018;155:1-12. DOI: 10.1186/s41065-017-0036-2
  4. 4. Martin AR, Lin M, Granka JM, Myrick JW, Liu X, Sockell A, et al. An unexpectedly complex architecture for skin pigmentation in Africans. Cell. 2017;171:1340-1353. DOI: 10.1016/j.cell.2017.11.015
  5. 5. Nip J, Potterf B, Rocha S, Vora S, Bosko C. The new face of pigmentation and aging. In: Farage MA, Miller KW, Maibach HI, editors. Textbook of Aging Skin. Springer Publishers; 2010. pp. 1039-1054. DOI: 10.1007/978-3-662-47398-6_53
  6. 6. Bastiaens MT, Westendorp RG, Vermeer BJ, Bavinck JN. Ephelides are more related to pigmentary constitutional host factors than solar lentigines. Pigment Cell Research. 1999;12:316-322. DOI: 10.1111/j.1600-0749
  7. 7. Bastiaens MT, Hoefnagel J, Westendorp R, Vermeer BJ, Bavinck JN. Solar lentigines are strongly related to sun exposure in contrast to ephelides. Pigment Cell Research. 2004;17:225-229. DOI: 10.1111/j.1600-0749.2004.00131.x
  8. 8. Rahman SB, Bhawan J. Lentigo. International Journal of Dermatology. 1996;35:229-239. DOI: 10.1111/j.1365-4362.1996.tb02994.x
  9. 9. Nakamura M, Morita A, Seite S, Stemmann TH, Beck SG, Krutmann J. Environment-induced lentigines: Formation of solar lentigines beyond ultraviolet radiation. Experimental Dermatology. 2015;24:407-411. DOI: 10.1111/exd.12690
  10. 10. Andersen WK, Labadie RR, Bhawan J. Histopathology of solar lentigines of the face: A quantitative study. Journal of the American Academy of Dermatology. 1997;36:444-447. DOI: 10.1016/s0190-9622(97)80224-1
  11. 11. Yonei N, Kaminaka C, Kimura A, Furukawa F, Yamamoto Y. Two patterns of solar lentigines: A histopathological analysis of 40 Japanese women. The Journal of Dermatology. 2012;39:829-832. DOI: 10.1111/j.1346-8138.2012.01574.x
  12. 12. Lin CB, Hu Y, Rossetti D, Chen N, David C, Slominski A, et al. Immuno-histochemical evaluation of solar lentigines: The association of KGF/KGFR and other factors with lesion development. Journal of Dermatological Science. 2010;59:91-97. DOI: 10.1016/j.jdermsci.2010.06.006
  13. 13. Cario-Andre M, Lepreux S, Pain C, Nizard C, Noblesse E, Taieb A. Perilesional vs. lesional skin changes in senile lentigo. Journal of Cutaneous Pathology. 2004;31:441-447. DOI: 10.1111/j.0303-6987.2004.00210.x
  14. 14. Aoki H, Moro O, Tagami H, Kishimoto J. Gene expression profiling analysis of solar lentigo in relation to immunohistochemical characteristics. The British Journal of Dermatology. 2007;156:1214-1223. DOI: 10.1111/j.1365-2133.2007.07830.x
  15. 15. Jung JM, Yang H, Lee WJ, Won CH, Lee MW, Choi JH, et al. Inflammatory features and rete ridge patterns of facial solar lentigo may guide laser treatment. Dermatologic Therapy. 2020;33:e13464. DOI: 10.1111/dth.13464
  16. 16. Hafner C, Stoehr R, van Oers JM, Zwarthoff EC, Hofstaedter F, Landthaler M, et al. FGFR3 and PIK3CA mutations are involved in the molecular pathogenesis of solar lentigo. The British Journal of Dermatology. 2009;160:546-551. DOI: 10.1111/j.1365-2133.2008.08963.x
  17. 17. Choi W, Yin L, Smuda C, Batzer J, Hearing VJ, Kolbe L. Molecular and histological characterization of age spots. Dermatologica. 2017;26:242-248. DOI: 10.1111/exd.13203
  18. 18. Ganesh C, Damodaran A, Green MR, Rocha S, Pauloski N, Vora S. Multiple genes and diverse hierarchical pathways affect human pigmentation. In: Smith J, Haworth M, editors. Skin Pigmentation. Nova Science Publishers; 2013. pp. 89-119
  19. 19. Shin J, Park JY, Kim SJ, Kang HY. Characteristics of keratinocytes in facial solar lentigo with flattened rete ridges: Comparison with melasma. Clinical and Experimental Dermatology. 2015;40:489-494. DOI: 10.1111/ced.12621
  20. 20. Barysch MJ, Braun RP, Kolm I, Ahlgrimm-Siesz V, Hofmann-Wellenhof R, Duval C, et al. Keratinocytic malfunction as a trigger for the development of solar lentigines. Dermatopathology (Basel). 2019;6:1-11. DOI: 10.1159/000495404
  21. 21. Kovacs D, Cardinali G, Aspite N, Cota C, Luzi F, Bellei B, et al. Role of fibroblast-derived growth factors in regulating hyperpigmentation of solar lentigo. The British Journal of Dermatology. 2010;163:1020-1027. DOI: 10.1111/j.1365-2133.2010.09946.x
  22. 22. Goorochurn R, Viennet C, Tissot M, Locatelli F, Granger C, Varin-Blank N, et al. Differential morphological and functional features of fibroblasts explanted from solar lentigo. The British Journal of Dermatology. 2017;1177:109-e111. DOI: 10.1111/bjd.15386
  23. 23. Imokawa G. Melanocyte activation mechanisms and rational therapeutic treatments of solar lentigos. International Journal of Molecular Sciences. 2019;20:3666. DOI: 10.3390/ijms20153666
  24. 24. Iriyama S, Ono T, Aoki H, Amano S. Hyperpigmentation in human solar lentigo is promoted by heparanase-induced loss of heparan sulfate chains at the dermal-epidermal junction. Journal of Dermatological Science. 2011;64:223-228. DOI: 10.1016/j.jdermsci.2011.09.007
  25. 25. Hasegawa K, Fujiwara R, Sato K, Park JY, Kim SJ, Kim M, et al. Increased blood flow and vasculature in solar lentigo. The Journal of Dermatology. 2016;43:1209-1213. DOI: 10.1111/1346-8138.13458
  26. 26. Shibata T, Kajiya K, Sato K, Yoon J, Kang HY. 3D microvascular analysis reveals irregularly branching blood vessels in the hyperpigmented skin of solar lentigo. Pigment Cell & Melanoma Research. 2018;31:725-727. DOI: 10.1111/pcmr.12731
  27. 27. Hara Y, Yamashita T, Ninomiya M, Kubo Y, Katagiri C, Saeki S, et al. Vascular morphology in facial solar lentigo assessed by optical coherence tomography angiography. Journal of Dermatological Science. 2021;102:193-195. DOI: 10.1016/j.jdermsci.2021.04.001
  28. 28. Kerns ML, Chien AL, Kang S. A role for NRF2-signaling in the treatment and prevention of solar lentigines. Plastic and Reconstructive Surgery. 2021;148:27S-31S. DOI: 10.1097/PRS.0000000000008783
  29. 29. Epstein JH. Postinflammatory hyperpigmentation. Clinics in Dermatology. 1989;7:55-65. DOI: 10.1016/0738-081x(89)90057-6
  30. 30. Taylor S, Grimes P, Lim J, Im S, Lui H. Postinflammatory hyperpigmentation. Journal of Cutaneous Medicine and Surgery. 2009;13:183-191. DOI: 10.2310/7750.2009.08077
  31. 31. Perkins AC, Cheng CE, Hillebrand GG, Miyamoto K, Kimball AB. Comparison of the epidemiology of acne vulgaris among Caucasian, Asian, Continental Indian and African American women. Journal of the European Academy of Dermatology and Venereology. 2011;25:1054-1060. DOI: 10.1111/j.1468-3083.2010.03919.x
  32. 32. Ruiz-Maldonado R, Orozco-Covarrubias ML. Postinflammatory hypopigmentation and hyperpigmentation. Seminars in Cutaneous Medicine and Surgery. 1997;16:36-43. DOI: 10.1016/s1085-5629(97)80034-x
  33. 33. Silpa-Archa N, Kohli I, Chaowattanapanit S, Lim HW, Hamzavi I. Postinflammatory hyperpigmentation: A comprehensive overview: Epidemiology, pathogenesis, clinical presentation, and non-invasive assessment technique. Journal of the American Academy of Dermatology. 2017;77:591-605. DOI: 10.1016/j.jaad.2017.01.035
  34. 34. Lacz NL, Vafaie J, Kihiczak NI, Schwartz RA. Postinflammatory hyperpigmentation: A common but troubling condition. International Journal of Dermatology. 2004;3:362-365. DOI: 10.1111/j.1365-4632.2004.02267.x
  35. 35. Alster TS, Nanni CA, Williams CM. Comparison of four carbon dioxide resurfacing lasers: A clinical and histopathologic evaluation. Dermatologic Surgery. 1999;25:153-159. DOI: 10.1046/j.1524-4725.1999.08126.x
  36. 36. Bernstein LJ, Kauvar ANB, Grossman MC, Geronemus RG. The short- and long-term side effects of carbon dioxide laser resurfacing. Dermatologic Surgery. 1997;23:519-525. DOI: 10.1111/j.1524-4725.1997.tb00677.x
  37. 37. Harari Z, Sommer I, Knobel B. Multifocal contact dermatitis to nitroderm TTS 5 with extensive postinflammatory hypermelanosis. Dermatologica. 1987;174:249-252. DOI: 10.1159/000249190
  38. 38. Abad-Casintahan F, Chow SK, Goh CL, Kubba R, Hayashi N, Noppakun N, et al. Frequency and characteristics of acne-related post-inflammatory hyperpigmentation. Asian Acne Board. Journal of Dermatology. 2016;3:826-828. DOI: 10.1111/1346-8138.13263
  39. 39. Evans RL, Marriott RE, Harker M. Axillary skin: Biology and care. International Journal of Cosmetic Science. 2012;34:389-395. DOI: 10.1111/j.1468-2494.2012.00729.x
  40. 40. Park JY, Park JH, Kim SJ, Kwon JE, Kang HY, Lee ES, et al. Two histopathological patterns of postinflammatory hyperpigmentation: Epidermal and dermal. Journal of Cutaneous Pathology. 2017;44:118-124. DOI: 10.1111/cup.12849
  41. 41. Masu S, Seiji M. Pigmentary incontinence in fixed drug eruptions. Histologic and electron microscopic findings. Journal of the American Academy of Dermatology. 1983;8:525-532. DOI: 10.1016/s0190-9622(83)70060-5
  42. 42. Maghfour J, Olayinka J, Hamzavi IH, Mohammad TF. A focused review on the pathophysiology of post-inflammatory hyperpigmentation. Pigment Cell & Melanoma Research. 2022;35:320-327. DOI: 10.1111/pcmr.13038
  43. 43. Roh MR, Chung KY. Infraobital dark circles: Definition, causes, and treatment options. Dermatologic Surgery. 2009;35:1163-1171. DOI: 10.1111/j.1524-4725.2009.01213.x
  44. 44. Grimes PE. Aesthetics and Cosmetic Surgery for Darker Skin Types. 1st ed. Lippincott Williams & Wilkins (LWW); 2007
  45. 45. Sheth PB, Shah HA, Dave JN. Periorbital hyperpigmentation: A study of its prevalence, common causative factors and its association with personal habits and other disorders. Indian Journal of Dermatology. 2014;59:151-157. DOI: 10.4103/0019-5154.127675
  46. 46. Freitag FM, Cestari TF. What causes dark circles under the eyes? Journal of Cosmetic Dermatology. 2007;6:211-215. DOI: 10.1111/j.1473-2165.2007.00324.x
  47. 47. Sardana K, Rajpal M, Garg V, Mishra D. Periorbital hyperpigmentation mimicking fixed drug eruption: A rare presentation of erythema dyschromicum perstans in a paediatric patient. Journal of the European Academy of Dermatology and Venereology. 2006;20:1381-1383. DOI: 10.1111/j.1468-3083.2006.01735.x
  48. 48. Lim HGY, Fischer AH, Sung S, Kang S, Al C. Periocular dark circles: Correlates of severity. Annals of Dermatology. 2021;33:393-401. DOI: 10.5021/ad.2021.33.5.393
  49. 49. Goldberg RA, McCann JD, Fiaschetti D, Simon GJ. What causes eyelid bags? Analysis of 114 consecutive patients. Plastic and Reconstructive Surgery. 2005;115:1395-1402. DOI: 10.1097/01.prs.0000157016.49072.61
  50. 50. Sawant O, Khan T. Management of periorbital hyperpigmentation: An overview of nature-based agents and alternative approaches. Dermatologic Therapy. 2020;33:e13717. DOI: 10.1111/dth.13717
  51. 51. Ranu H, Thng S, Goh BK, Burger A, Goh CL. Periorbital hyperpigmentation in Asians: An epidemiologic study and a proposed classification. Dermatologic Surgery. 2011;37:1297-1303. DOI: 10.1111/j.1524-4725.2011.02065.x
  52. 52. Sarkar R, Ranjan R, Garg S, Garg VK, Sonthalia S, Bansal S. Periorbital hyperpigmentation: A comprehensive review. The Journal of Clinical and Aesthetic Dermatology. 2016;9:49-55
  53. 53. Sarkar R, Das A. Periorbital hyperpigmentation: What lies beneath? Indian Dermatology Online Journal. 2018;9:229-230. DOI: 10.4103/idoj.IDOJ_303_17
  54. 54. Daroach M, Kumaran MS. Periorbital hyperpigmentation − An overview of the enigmatous condition. Pigment International. 2018;5:1-3
  55. 55. Nouveau S, Agrawal D, Kohli M, Bernerd F, Misra N. Skin hyperpigmentation in Indian population: Insights and best practice. Indian Journal of Dermatology. 2016;61:487-495. DOI: 10.4103/0019-5154.190103
  56. 56. Grimes PE, Ijaz S, Nashawati R, Kwak D. New oral and topical approaches for the treatment of melasma. International Journal of Women's Dermatology. 2019;5:30-36. DOI: 10.1016/j.ijwd.2018.09.004
  57. 57. Watanabe S, Nakai K, Ohnishi T. Condition known as “dark rings under the eyes” in the Japanese population is a kind of dermal melanocytosis which can be successfully treated by Q-switched ruby laser. Dermatologic Surgery. 2006;32:785-789. DOI: 10.1111/j.1524-4725.2006.32161.x
  58. 58. Graziosi AC, Quaresma MR, Michalany NS, Ferreira LM. Cutaneous idiopathic hyperchromia of the orbital region (CIHOR): A histopathological study. Aesthetic Plastic Surgery. 2013;37:434-438. DOI: 10.1007/s00266-012-0048-2
  59. 59. Kurita M, Kato H, Yoshimura K. A therapeutic strategy based on histological assessment of hyperpigmented skin lesions in Asians. Journal of Plastic, Reconstructive & Aesthetic Surgery. 2009;62:955-963. DOI: 10.1016/j.bjps.2007.10.079
  60. 60. Mac-Mary S, Solinis IZ, Predine O, Sainthillier JM, Sladen C, Bell M, et al. Identification of three key factors contributing to the aetiology of dark circles by clinical and instrumental assessments of the infraorbital region. Clinical, Cosmetic and Investigational Dermatology. 2019;18:919-929. DOI: 10.2147/CCID.S217956
  61. 61. Perez BA, Perez MA, Camacho F. Management of facial hyperpigmentation. American Journal of Clinical Dermatology. 2000;1:261-268. DOI: 10.2165/00128071-200001050-00001
  62. 62. Sheth VM, Pandya AG. Melasma: A comprehensive update Part 1. Journal of the American Academy of Dermatology. 2011;65:689-697. DOI: 10.1016/j.jaad.2010.12.046
  63. 63. Grimes PE. Melasma: Etiologic and therapeutic considerations. Archives of Dermatology. 1995;131:1453-1457. DOI: 10.1001/archderm.131.12.1453
  64. 64. Passeron T, Picardo M. Melasma, a photoaging disorder. Pigment Cell & Melanoma Research. 2018;31(4):461-465. DOI: 10.1111/pcmr.12684
  65. 65. Ortonne JP, Arellano I, Berneburg M, Cestari T, Chan H, Grimes P, et al. A global survey of the role of ultraviolet radiation and hormonal influences in the development of melasma. Journal of the European Academy of Dermatology and Venereology. 2009;23:1254-1262. DOI: 10.1111/j.1468-3083.2009.03295.x
  66. 66. Handel AC, Miot LD, Miot HA. Melasma: A clinical and epidemiological review. Anais Brasileiros de Dermatologia. 2014;89:771-782. DOI: 10.1590/abd1806-4841.20143063
  67. 67. Khanna N, Rasool S. Facial melanoses: Indian perspective. Indian Journal of Dermatology, Venereology and Leprology. 2011;77:552. DOI: 10.4103/0378-6323.84046
  68. 68. Lee AY. Recent progress in melasma pathogenesis. Pigment Cell & Melanoma Research. 2015;28:648-660. DOI: 10.1111/pcmr.12404
  69. 69. Suryaningsih BE, Sadewa AH, Wirohadidjojo YW, Soebono H. Association between heterozygote Val92Met MC1R gene polymorphisms with incidence of melasma: A study of Javanese women population in Yogyakarta. Clinical, Cosmetic and Investigational Dermatology. 2019;12:489-495. DOI: 10.2147/CCID.S206115
  70. 70. Kheradmand M, Afshari M, Damiani G, Abediankenari S, Moosazadeh M. Melasma and thyroid disorders: A systematic review and meta-analysis. International Journal of Dermatology. 2019;58:1231-1238. DOI: 10.1111/ijd.14497
  71. 71. Artzi O, Horovitz T, Bar-Ilan E, Shehadeh W, Koren A, Zusmanovitch L, et al. The pathogenesis of melasma and implications for treatment. Journal of Cosmetic Dermatology. 2021;20:3432-3445. DOI: 10.1111/jocd.14382
  72. 72. Kang HY, Suzuki I, Lee DJ, Ha J, Reiniche P, Aubert J, et al. Transcriptional profiling shows altered expression of WNT pathway- and lipid metabolism-related genes as well as melanogenesis-related genes in melasma. The Journal of Investigative Dermatology. 2011;131:1692-1700. DOI: 10.1038/jid.2011.109
  73. 73. Kim JY, Lee TR, Lee AY. Reduced WIF-1 expression stimulates skin hyperpigmentation in patients with melasma. The Journal of Investigative Dermatology. 2013;133:191-200. DOI: 10.1038/jid.2012.270
  74. 74. Jang YH, Lee JY, Kang HY, Lee ES, Kim YC. Oestrogen and progesterone receptor expression in melasma: An immunohistochemical analysis. Journal of the European Academy of Dermatology and Venereology. 2010;24:1312-1316. DOI: 10.1111/j.1468-3083.2010.03638.x
  75. 75. Tamega Ade A, Miot HA, Moc ONP, Silva MG, Marques ME, Miot LD. Gene and protein expression of oestrogen-b and progesterone receptors in facial melasma and adjacent healthy skin in women. International Journal of Cosmetic Science. 2015;37:222-228. DOI: 10.1111/ics.12186
  76. 76. Kim NH, Cheong KA, Lee TR, Lee AY. PDZK1 upregulation in estrogen-related hyperpigmentation in melasma. The Journal of Investigative Dermatology. 2012;132:2622-2631. DOI: 10.1038/jid.2012.175
  77. 77. Kim EH, Kim YC, Lee ES, Kang HY. The vascular characteristics of melasma. Journal of Dermatological Science. 2007;46:111-116. DOI: 10.1016/j.jdermsci.2007.01.009
  78. 78. Kang HY, Bahadoran P, Suzuki I, Zugaj D, Khemis A, Passeron T, et al. In vivo reflectance confocal microscopy detects pigmentary changes in melasma at a cellular level resolution. Experimental Dermatology. 2010;19:e228-e233. DOI: 10.1111/j.1600-0625.2009.01057.x
  79. 79. Passeron T. Long-lasting effect of vascular targeted therapy of melasma. Journal of the American Academy of Dermatology. 2013;69:e141-e142. DOI: 10.1016/j.jaad.2013.02.022
  80. 80. Jo HY, Kim CK, Suh IB, Ryu SW, Ha KS, Kwon YG, et al. Co-localization of inducible nitric oxide synthase and phosphorylated Akt in the lesional skins of patients with melasma. The Journal of Dermatology. 2009;36:10-16. DOI: 10.1111/j.1346-8138.2008.00579.x
  81. 81. Kang HY, Hwang JS, Lee JY, Ahn JH, Kim JY, Lee ES, et al. The dermal stem cell factor and c-kit are overexpressed in melasma. The British Journal of Dermatology. 2006;154:1094-1099. DOI: 10.1111/j.1365-2133.2006.07179.x
  82. 82. Regazzetti C, De Donatis GM, Ghorbel HH, Cardot-Leccia N, Ambrosetti D, Bahadoran P, et al. Endothelial cells promote pigmentation through Endothelin Receptor B activation. The Journal of Investigative Dermatology. 2015;135:3096. DOI: 10.1038/jid.2015.332
  83. 83. Hernandez-Barrera R, Torres-Alvarez B, Castanedo-Cazares JP, Oros-Ovalle C, Moncada B. Solar elastosis and presence of mast cells as key features in the pathogenesis of melasma. Clinical and Experimental Dermatology. 2008;33:305-308. DOI: 10.1111/j.1365-2230.2008.02724. x
  84. 84. Torres-Álvarez B, Mesa-Garza IG, Castanedo-Cázares JP, Fuentes-Ahumada C, Oros-Ovalle C, Navarrete-Solis J, et al. Histochemical and immunohistochemical study in melasma: Evidence of damage in the basal membrane. The American Journal of Dermatopathology. 2011;33:291-295. DOI: 10.1097/DAD.0b013e3181ef2d45
  85. 85. Iddamalgoda A, Le QT, Ito K, Tanaka K, Kojima H, Kido H. Mast cell tryptase and photoaging: Possible involvement in the degradation of extra cellular matrix and basement membrane proteins. Archives of Dermatological Research. 2008;300(Suppl. 1):S69-S76. DOI: 10.1007/s00403-007-0806-1
  86. 86. Lee DJ, Park KC, Ortonne JP, Kang HY. Pendulous melanocytes: A characteristic feature of melasma and how it may occur. The British Journal of Dermatology. 2012;166:684-686. DOI: 10.1111/j.1365-2133.2011.10648.x
  87. 87. Inomata S, Matsunaga Y, Amano S, Takada K, Kobayashi K, Tsunenaga M, et al. Possible involvement of gelatinases in basement membrane damage and wrinkle formation in chronically ultraviolet B-exposed hairless mouse. The Journal of Investigative Dermatology. 2003;120:128-134. DOI: 10.1046/j.1523-1747.2003.12021.x
  88. 88. Kang WH, Yoon KH, Lee ES, Kim J, Lee KB, Yim H, et al. Melasma: Histopathological characteristics in 56 Korean patients. The British Journal of Dermatology. 2002;146:228-237. DOI: 10.1046/j.0007-0963.2001.04556.x
  89. 89. Lentsch G, Balu M, Williams J, Lee S, Harris R, König K, et al. In vivo multiphoton microscopy of melasma. Pigment Cell & Melanoma Research. 2019;32:403-411. DOI: 10.1111/pcmr.12756
  90. 90. Christensen L, Suggs A, Baron E. Ultraviolet photobiology in dermatology. Advances in Experimental Medicine and Biology. 2017;996:89-104. DOI: 10.1007/978-3-319-56017-5_8
  91. 91. Kohli I, Sakamaki T, Tian WD, Moyal D, Hamzavi IH, Kollias N. The dynamics of pigment reactions of human skin to ultraviolet A radiation. Photodermatology, Photoimmunology & Photomedicine. 2019;35:387-392. DOI: 10.1111/phpp.12497
  92. 92. Mahmoud BH, Ruvolo E, Hexsel CL, Liu Y, Owen MR, Kollias N, et al. Impact of long-wavelength UVA and visible light on melanocompetent skin. The Journal of Investigative Dermatology. 2010;130:2092-2097. DOI: 10.1038/jid.2010.95
  93. 93. Amaro-Ortiz A, Yan B, D’Orazio JA. Ultraviolet radiation, aging, and the skin: Prevention of damage by topical cAMP manipulation. Molecules. 2014;19:6202-6219. DOI: 10.3390/molecules19056202
  94. 94. Cadet J, Douki T. Formation of UV-induced DNA damage contributing to skin cancer development. Photochemical & Photobiological Sciences. 2018;17:1816-1841. DOI: 10.1039/c7pp00395a
  95. 95. Box NF, Terzian T. The role of p53 in pigmentation, tanning and melanoma. Pigment Cell & Melanoma Research. 2008;21:525-533. DOI: 10.1111/j.1755-148X.2008.00495.x
  96. 96. Cui R, Widlund HR, Feige E, Lin JY, Wilensky DL, Igras VE, et al. Central role of p53 in the suntan response and pathologic hyperpigmentation. Cell. 2007;128:853-864. DOI: 10.1016/j.cell.2006.12.045
  97. 97. Paik SH, Kim H-J, Son H-Y, Lee S, Im S-W, Ju YS, et al. Gene mapping study for constitutive skin color in an isolated Mongolian population. Experimental & Molecular Medicine. 2012;44:241-249. DOI: 10.3858/emm.2012.44.3.020
  98. 98. Austin E, Geisler AN, Nguyen J, Kohli I, Hamzavi I, Lim HW, et al. Part I: Properties and cutaneous effects of visible light. Journal of the American Academy of Dermatology. 2022;84:1219-1231. DOI: 10.1016/j.jaad.2021.02.048
  99. 99. Lim HW, Kohli I, Ruvolo E, Kolbe L, Hamzavi IH. Impact of visible light on skin health: The role of antioxidants and free radical quenchers in skin protection. Journal of the American Academy of Dermatology. 2022;86(3S):S27-S37. DOI: 10.1016/j.jaad.2021.12.024
  100. 100. Ramasubramaniam R, Roy A, Sharma B, Nagalakshmi S. Are there mechanistic differences between ultraviolet and visible radiation induced skin pigmentation? Photochemical & Photobiological Sciences. 2011;10:1887-1893. DOI: 10.1039/c1pp05202k
  101. 101. Gosh Dastidar S, Rajkumar S, Rajgopal R, and Damodaran A. Impact of high energy visible light on skin appearance. Journal of the American Academy of Dermatology. 2018;79(3): AB159. DOI: https://doi.org/10.1016/j.jaad.2018.05.653. Annual meeting of American Academy of Dermatology (AAD), 16-20 February 2018, San Diego, California, USA
  102. 102. Kim S, Rainer BM, Qi J, Brown I, Ogurtsova A, Leung S, et al. Clinical and molecular change induced by repeated low-dose visible light exposure in both light-skinned and dark-skinned individuals. Photodermatology, Photoimmunology & Photomedicine. 2022:1-9. DOI: 10.1111/phpp.12819
  103. 103. Moreiras H, O’Connor C, Bell M, Tobin DJ. Visible light and human skin pigmentation: The importance of skin phototype. Experimental Dermatology. 2021;30:1324-1331. DOI: 10.1111/exd.14400
  104. 104. Liebel F, Kaur S, Ruvolo E, Kollias N, Southall MD. Irradiation of skin with visible light induces reactive oxygen species and matrix-degrading enzymes. The Journal of Investigative Dermatology. 2012;132:1901-1907. DOI: 10.1038/jid.2011.476
  105. 105. Rajkumar S, Roy A, Surendra N, Gupta AK, Joshi M, and Damodaran A. Visible light effects on skin function and pigmentation. In: 8th annual meeting of ASPCR, 12-13 November 2016, Kaohsiung, Taiwan
  106. 106. Damodaran A, Rajkumar S, Shruthi PB, Patil N, Kumar S, and Kale V. Novel role of visible and blue light in modulation of inflammation and molecular clock genes in skin cells, in vitro. In: 24th World Congress of Dermatology; 10-15 June 2019, Milan, Italy
  107. 107. Campiche R, Curpen SJ, Lutchmanen-Kolanthan V, Gougeon S, Cherel M, Laurent G, et al. Pigmentation effects of blue light irradiation on skin and how to protect against them. International Journal of Cosmetic Science. 2020;42:399-406. DOI: 10.1111/ics.12367
  108. 108. Zhao Z, Zhou Y, Tan G, Li J. Research progress about the effect and prevention of blue light on eyes. International Journal of Ophthalmology. 2018;11:1999-2003. DOI: 10.18240/ijo.2018.12.20
  109. 109. Suh S, Choi EH, Mesinkovska NA. The expression of opsins in the human skin and its implications for photobiomodulation: A systematic review. Photodermatology, Photoimmunology & Photomedicine. 2020;36:329-338. DOI: 10.1111/phpp.12578
  110. 110. Ozdeslika RN, Olinskib LE, Trieuc MM, Oprianc DD, Oanceaa E. Human nonvisual opsin 3 regulates pigmentation of epidermal melanocytes through functional interaction with melanocortin 1 receptor. Proceedings of the National Academy of Sciences of the United States of America. 2019;116:11508-11517. DOI: 10.1073/pnas.1902825116
  111. 111. Cohen L, Brodsky MA, Zubair R, Kohli I, Hamzavi I, Sadeghpour M. Cutaneous interaction with visible light: What do we know. Journal of the American Academy of Dermatology. 2020. DOI: 10.1016/j.jaad.2020.03.115. PMID: 32289393 [Epub ahead of print]
  112. 112. Fatima S, Braunberger T, Mohammad TF, Kohli I, Hamzavi IH. The role of sunscreen in melasma and postinflammatory hyperpigmentation. Indian Journal of Dermatology. 2020;65:5-10. DOI: 10.4103/ijd.IJD_295_18
  113. 113. Praetorius C, Strum RA, Steingrimsson E. Sun-induced freckling: Ephelides and solar lentigines. Pigment Cell & Melanoma Research. 2014;27:339-350. DOI: 10.1111/pcmr.12232
  114. 114. Castanedo-Cazares JP, Hernandez-Blanco D, Carlos-Ortega B, Fuentes-Ahumada C, Torres-Álvarez B. Near-visible light and UV photoprotection in the treatment of melasma: A double-blind randomized trial. Photodermatology, Photoimmunology & Photomedicine. 2014;30:35-42. DOI: 10.1111/phpp.12086
  115. 115. Boukari F, Jourdan E, Fontas E, Montaudié H, Castela E, Lacour JP, et al. Prevention of melasma relapses with sunscreen combining protection against UV and short wavelengths of visible light: A prospective randomized comparative trial. Journal of the American Academy of Dermatology. 2015;72:189-190. DOI: 10.1016/j.jaad.2014.08.023
  116. 116. Geisler AN, Austin E, Nguyen J, Hamzavi I, Jagdeo J, Lim HW. Visible light part II. Photoprotection against visible and ultraviolet light. Journal of the American Academy of Dermatology. 2021;84:1233-1244. DOI: 10.1016/j.jaad.2020.11.074
  117. 117. Halder RM, Richards GM. Management of dyschromias in ethnic skin. Dermatologic Therapy. 2004;17:151-157. DOI: 10.1111/j.1396-0296.2004.04015.x
  118. 118. Levitt J. The safety of hydroquinone: A dermatologist’s response to the 2006 Federal Register. Journal of the American Academy of Dermatology. 2007;57:854-872. DOI: 10.1016/j.jaad.2007.02.020
  119. 119. Schwartz C, Jan A, and Zito PM. Hydroquinone. In: StatPearls. Treasure Island, FL: StatPearls Publishing; 2022 https://www.ncbi.nlm.nih.gov/books/NBK539693/
  120. 120. Gupta AK, Gover MD, Nouri K, Taylor S. The treatment of melasma: A review of clinical trials. Journal of the American Academy of Dermatology. 2006;55:1048-1065. DOI: 10.1016/j.jaad.2006.02.009
  121. 121. Torok HM, Jones T, Rich P, Smith S, Tschen E. Hydroquinone 4%, tretinoin 0.05%, fluocinolone acetonide 0.01%: A safe and efficacious 12-month treatment for melasma. Cutis. 2005;75:57-62
  122. 122. Chan R, Park KC, Lee MH, Lee E-S, Chang SE, Leow YH, et al. A randomized controlled trial of the efficacy and safety of a fixed triple combination (fluocinolone acetonide 0.01%, hydroquinone 4%, tretinoin 0.05%) compared with hydroquinone 4% cream in Asian patients with moderate to severe melasma. The British Journal of Dermatology. 2008;159:697-703. DOI: 10.1111/j.1365-2133.2008.08717.x
  123. 123. Chiari ME, Vera DMA, Palacios SM, Carpinella MC. Tyrosinase inhibitory activity of a 6-isoprenoid-substituted flavanone isolated from Dalea elegans. Bioorganic & Medicinal Chemistry. 2011;19:3474-3482. DOI: 10.1016/j.bmc.2011.04.025
  124. 124. Chawla S, deLong MA, Visscher MO, Wickett RR, Manga P, Boissy RE. Mechanism of tyrosinase inhibition by deoxyarbutin and its second-generation derivatives. The British Journal of Dermatology. 2008;159:1267-1274. DOI: 10.1111/j.1365-2133.2008.08864.x
  125. 125. Deri B, Kanteev M, Goldfeder M, Lecina D, Guallar V, Adir N, et al. The unravelling of the complex pattern of tyrosinase inhibition. Scientific Reports. 2016;6:34993. DOI: 10.1038/srep34993
  126. 126. Smith CJ, O’Hare KB, Allen JC. Selective cytotoxicity of hydroquinone for melanocyte-derived cells is mediated by tyrosinase activity but independent of melanin content. Pigment Cell Research. 1988;1:386-389. DOI: 10.1111/j.1600-0749.1988.tb00140.x
  127. 127. Arck PC, Overall R, Spatz K, Liezman C, Handjiski B, Klapp BF, et al. Towards a “free radical theory of graying”: Melanocyte apoptosis in the aging human hair follicle is an indicator of oxidative stress induced tissue damage. The FASEB Journal. 2006;20:1567-1569. DOI: 10.1096/fj.05-4039fje
  128. 128. Shariff R, Du Y, Dutta M, Kumar VS, Thimmaiah S, Doraiswamy C, et al. Superior even skin tone and anti-ageing benefit of a combination of 4-hexylresorcinol and niacinamide. International Journal of Cosmetic Science. 2022;44:1-15. DOI: 10.1111/ics.12759
  129. 129. Mann T, Gerwat W, Batzer J, Eggers K, Scherner C, Wenck H, et al. Inhibition of human tyrosinase requires molecular motifs distinctively different from mushroom tyrosinase. Journal of Investigative Dermatology. 2018;138:1601-1608. DOI: 10.1016/j.jid.2018.01.019
  130. 130. Nautiyl A, Wairkar S. Management of hyperpigmentation: Current treatments and emerging therapies. Pigment Cell & Melanoma Research. 2021;34:1000-1014. DOI: 10.1111/pcmr.12986
  131. 131. Sanadi RM, Deshmukh RS. The effect of Vitamin C on melanin pigmentation – A systematic review. Journal of Oral and Maxillofacial Pathology. 2020;24:374-382. DOI: 10.4103/jomfp.JOMFP_207_20
  132. 132. Boo YC. Mechanistic basis and clinical evidence for the applications of nicotinamide (niacinamide) to control skin aging and pigmentation. Antioxidants (Basel). 2021;10:1315-1339. DOI: 10.3390/antiox10081315
  133. 133. Pullar JM, Carr AC, Vissers MCM. The roles of Vitamin C in skin health. Nutrients. 2017;9:866-893. DOI: 10.3390/nu9080866
  134. 134. Huh C-H, Seo K-II, Park J-Y, Lim J-G, Eun H-C, Park K-C. A randomized, double-blind, placebo-controlled trial of vitamin C iontophoresis in melasma. Dermatologica. 2003;206:316-320. DOI: 10.1159/000069943
  135. 135. Lee GSK. Intravenous vitamin C in the treatment of post-laser hyperpigmentation for melasma: A short report. Journal of Cosmetic and Laser Therapy. 2008;10:234-236. DOI: 10.1080/14764170802187193
  136. 136. Fuller BB, Spaulding DT, Smith DR. Regulation of the catalytic activity of pre-existing tyrosinase in black and Caucasian human melanocyte cell cultures. Experimental Cell Research. 2001;262:197-208. DOI: 10.1006/excr.2000.5092
  137. 137. McGlinchey RP, Jiang Z, Lee JC. Molecular origin of pH-dependent fibril formation of a functional amyloid. ChemBioChem. 2014;15:1569-1572. DOI: 10.1002/cbic.201402074
  138. 138. Miao F, Su M, Jiang S, Luo L, Shi Y, Lei T. Intramelanocytic acidification plays a role in the antimelanogenic and antioxidative properties of vitamin C and its derivatives. Oxidative Medicine and Cellular Longevity. 2019;2019:2084805, 14 pages. DOI: 10.1155/2019/2084805
  139. 139. Chen S, Hseu Y, Gowrisankar YV, Chung Y, Zhang Y, Way T, et al. The anti-melanogenic effects of 3-O-ethyl ascorbic acid via Nrf2-mediated α-MSH inhibition in UVA-irradiated keratinocytes and autophagy induction in melanocytes. Free Radical Biology & Medicine. 2021;173:151-169. DOI: 10.1016/j.freeradbiomed.2021.07.030
  140. 140. Lin H, Shieh B, Lu M, Chen J, Chang L, Chao C. A method for quantifying melanosome transfer efficacy from melanocytes to keratinocytes in vitro. Pigment Cell & Melanoma Research. 2008;21:559-564. DOI: 10.1111/j.1755-148X.2008.00487.x
  141. 141. Ma H, Zhao G, Zi S, Li D, Liu W, Yang Q. Efficacy of quantifying melanosome transfer with flow cytometry in a human melanocyte–HaCaT keratinocyte co-culture system in vitro. Experimental Dermatology. 2010;19:e282-e285. DOI: 10.1111/j.1600-0625.2009.00956.x
  142. 142. Greatens A, Hakozaki T, Koshoffer A, Epstein H, Schwemberger S, Babcock G, et al. Effective inhibition of melanosome transfer to keratinocytes by lectins and niacinamide is reversible. Experimental Dermatology. 2005;14:498-508. DOI: 10.1111/j.0906-6705.2005.00309.x
  143. 143. Madaan P, Sikka P, Malik DS. Cosmeceutical aptitudes of niacinamide: A review. Recent Advances in Anti-Infective Drug Discovery. 2021;16:196-208. DOI: 10.2174/2772434416666211129105629
  144. 144. Desai S, Ayres E, Bak H, et al. Effect of a tranexamic acid, kojic acid, and 509 niacinamide containing serum on facial dyschromia: A clinical evaluation. Journal of Drugs in Dermatology. 2019;18:454-459
  145. 145. Ebrahimi B, Naeini FF. Topical tranexamic acid as a promising treatment for melasma. Journal of Research in Medical Sciences: The Official Journal of Isfahan University of Medical Sciences. 2014;19:753-757
  146. 146. Ko D, Wang RF, Ozog D, Lim HW, and Mohammad TF. Disorders of hyperpigmentation. Part II. Review of management and treatment options for hyperpigmentation. Journal of the American Academy of Dermatology. 2022;S0190-9622(22):00252-00253. DOI: https://doi.org/10.1016/j.jaad.2021.12.065
  147. 147. Mathapathi MS, Mallemalla P, Vora S, Iyer V, Tiwari JK, Chakrabortty A, et al. Niacinamide leave-on formulation provides long-lasting protection against bacteria in vivo. Experimental Dermatology. 2017;26:827-829. DOI: 10.1111/exd.13285
  148. 148. Tanno O, Ota Y, Kitamura N, Katsube T, Inoue S. Nicotinamide increases biosynthesis of ceramides as well as other stratum corneum lipids to improve the epidermal permeability barrier. The British Journal of Dermatology. 2000;143:524-531. DOI: 10.1111/j.1365-2133.2000.03705.x
  149. 149. Ahn KS, Moon KY, Lee J, Kim YS. Downregulation of NFkappaB activation in human keratinocytes by melanogenic inhibitors. Journal of Dermatological Science. 2003;31:193-201. DOI: 10.1016/s0923-1811(03)00039-2
  150. 150. Bissett DL, Miyamoto K, Sun P, Li J, Berge CA. Topical niacinamide reduces yellowing, wrinkling, red blotchiness, and hyperpigmented spots in aging facial skin. International Journal of Cosmetic Science. 2004;26:231-238. DOI: 10.1111/j.1467-2494.2004.00228.x
  151. 151. Hakozaki T, Minwalla L, Zhuang J, Chhoa M, Matsubara A, Miyamoto K, et al. The effect of niacinamide on reducing cutaneous pigmentation and suppression of melanosome transfer. The British Journal of Dermatology. 2002;147:20-31. DOI: 10.1046/j.1365-2133.2002.04834.x
  152. 152. Navarrete-Solís J, Castanedo-Cázares JP, Torres-Álvarez B, Oros-Ovalle C, Fuentes-Ahumada C, González FJ, et al. Double-blind, randomized clinical trial of niacinamide 4% versus hydroquinone 4% in the treatment of melasma. Dermatology Research and Practice. 2011;2011:379173. DOI: 10.1155/2011/379173
  153. 153. Khodaeiani E, Fouladi RF, Amirnia M, Saeidi M, Karimi ER. Topical 4% nicotinamide vs. 1% clindamycin in moderate inflammatory acne vulgaris. International Journal of Dermatology. 2013;52:999-1004. DOI: 10.1111/ijd.12002
  154. 154. Damodaran A, Nair N, Shariff R, Damle A, Chandramowli G, Joshi P, Krishnamuthy H, and Vora S. Multiple modes of action of niacinamide in regulating skin pigmentation. In: Pigment Cell & Melanoma Research. 2014;27:849-1001. DOI: 10.1111/pcmr.12292. XXII International Pigment Cell Conference (IPCC); 4-7 September 2014; Singapore
  155. 155. Castanedo-Cazares JP, Lárraga-Piñones G, Ehnis-Pérez A, Fuentes-Ahumada C, Oros-Ovalle C, Smoller BR, et al. Topical niacinamide 4% and desonide 0.05% for treatment of axillary hyperpigmentation: A randomized, double-blind, placebo-controlled study. Clinical, Cosmetic and Investigational Dermatology. 2013;6:29-36. DOI: 10.2147/CCID.S39246
  156. 156. Engel K, Reuter J, Seiler C, Mönting JS, Jakob T, Schempp CM. Anti-inflammatory effect of pimecrolimus in the sodium lauryl sulphate test. Journal of the European Academy of Dermatology and Venereology. 2008;22(4):447-450. DOI: 10.1111/j.1468-3083.2007.02477.x
  157. 157. Nair N, Dutta M, Karu J, Lim F-L, Damodaran A, and Logt P. Regulation of postinflammatory hyperpigmentation by niacinamide through potential modulation of the protease inhibitor, SERPINB3. In: Journal of the American Academy of Dermatology. 2022;85(Supplement):AB99. DOI: https://doi.org/10.1016/j.jaad.2021.06.417. Annual meeting of American Academy of Dermatology (AAD), 19-23 March 2021, San Francisco, USA
  158. 158. Cork MJ, Danby SG, Vasilopoulos Y, Hadgraft J, Lane ME, Moustafa M, et al. Epidermal barrier dysfunction in AD. The Journal of Investigative Dermatology. 2009;129:1892-1908. DOI: 10.1038/jid.2009.133
  159. 159. Kelhälä HL, Palatsi R, Fyhrquist N, Lehtimäki S, Väyrynen JP, KallioinenM KME, et al. IL-17/Th17 pathway is activated in acne lesions. PLoS One. 2014;9:e105238. DOI: 10.1371/journal.pone.0105238
  160. 160. Hearing VJ, Jimenez M. Mammalian tyrosinase-the critical regulatory control point in melanocyte pigmentation. The International Journal of Biochemistry. 1987;19:1141-1147. DOI: 10.1016/0020-711x(87)90095-4
  161. 161. Sanchez-Ferrer A, Rodrıguez-Lopez JN, Garcıa-Canovas F, García-Carmona F. Tyrosinase: A comprehensive review of its mechanism. Biochimica et Biophysica Acta. 1995;1247:1-11. DOI: 10.1016/0167-4838(94)00204-t
  162. 162. Cooksey CJ, Garratt PJ, Land EJ, Pavel S, Ramsden CA, Riley PA, et al. Evidence of the indirect formation of the catecholic intermediate substrate responsible for the autoactivation kinetics of tyrosinase. The Journal of Biological Chemistry. 1997;272:26226-26235. DOI: 10.1074/jbc.272.42.26226
  163. 163. Ramsden CA, Riley PA. Tyrosinase: The four oxidation states of the active site and their relevance to enzymatic activation, oxidation and inactivation. Bioorganic & Medicinal Chemistry. 2014;22:2388-2395. DOI: 10.1016/j.bmc.2014.02.048
  164. 164. Zolghadri S, Bahrami A, Hasan Khan MT, Munoz-Munoz J, Garcia-Molina F, Garcia-Canovas F, et al. A comprehensive review on tyrosinase inhibitors. Journal of Enzyme Inhibition and Medicinal Chemistry. 2019;34(1):279-309. DOI: 10.1080/14756366.2018.1545767
  165. 165. Pillaiyar T, Manickam M, Namasivayam V. Skin whitening agents: Medicinal chemistry perspective of tyrosinase inhibitors. Journal of Enzyme Inhibition and Medicinal Chemistry. 2017;32:403-425. DOI: 10.1080/14756366.2016.1256882
  166. 166. Espin JC, Varon R, Fenoll LG, Gilabert MA, García-Ruíz PA, Tudela J, et al. Kinetic characterization of the substrate specificity and mechanism of mushroom tyrosinase. European Journal of Biochemistry. 2000;267:1270-1279. DOI: 10.1046/j.1432-1327.2000.01013.x
  167. 167. Muñoz-Muñoz JL, Acosta-Motos JR, Garcia-Molina F, Varon R, Garcia-Ruíz PA, Tudela J, et al. Tyrosinase inactivation in its action on dopa. Biochimica et Biophysica Acta. 2010;1804:1467-1475. DOI: 10.1016/j.bbapap.2010.02.015
  168. 168. Mann T, Scherner C, Rohm KH, Kolbe L. Structure-activity relationships of thiazolyl resorcinols, potent and selective inhibitors of human tyrosinase. International Journal of Molecular Sciences. 2018;19:690-704. DOI: 10.3390/ijms19030690
  169. 169. Stratford MRL, Ramsden CA, Riley PA. Mechanistic studies of the inactivation of tyrosinase by resorcinol. Bioorganic & Medicinal Chemistry. 2013;21:1166-1173. DOI: 10.1016/j.bmc.2012.12.031
  170. 170. Shimizu K, Kondo R, Sakai K. Inhibition of tyrosinase by flavonoids, stilbenes and related 4-substituted resorcinols: Structure activity investigations. Planta Medica. 2000;66:11-15. DOI: 10.1055/s-2000-11113
  171. 171. Schmaus G, Vielhaber G, Jacobs K, Franke H. 4-(1-Phenylethyl) 1,3-benzenediol: A new highly potent lightening agent. Journal of Cosmetic Science. 2006;57:197-198
  172. 172. Vielhaber G, Schmaus G, Jacobs K, Franke H, Lange S, Herrmann M, et al. 4-(1-Phenylethyl)1,3-benzenediol: A new, highly efficient lightening agent. International Journal of Cosmetic Science. 2007;29:63-65. DOI: 10.1111/j.1467-2494.2007.00355_6.x
  173. 173. Kolbe L, Mann T, Gerwat W, Batzer J, Ahlheit S, Scherner C, et al. 4-n-butylresorcinol, a highly effective tyrosinase inhibitor for the topical treatment of hyperpigmentation. Journal of the European Academy of Dermatology and Venereology. 2013;27:19-23. DOI: 10.1111/jdv.12051
  174. 174. Limsuwan T, Boonme P, Khongkow P, Amnuaikit T. Ethosomes of phenylethyl resorcinol as vesicular delivery system for skin lightening applications. Biomedical Research. 2017;2017:8310979. DOI: 10.1155/2017/8310979
  175. 175. Fan H, Li Y, Huang Y, Liu G, and Xia Q. Preparation and evaluation of phenylethyl resorcinol liposome. Integrated Ferroelectrics 2014;151: 89-98. DOI: https://doi.org/10.1080/10584587.2014.899873
  176. 176. Kim DS, Kim SY, Park SH, Choi YG, Kwon SB, Kim MK, et al. Inhibitory effects of 4-n-butylresorcinol on tyrosinase activity and melanin synthesis. Biological & Pharmaceutical Bulletin. 2005;28:2216-2219. DOI: 10.1248/bpb.28.2216
  177. 177. Huh SY, Shin JW, Na J, Huh C, Youn S, Park K. The efficacy and safety of 4-n-butylresorcinol 0.1% cream for the treatment of melasma: A randomized controlled split-face trial. Annals of Dermatology. 2010;22(1):21-25. DOI: 10.5021/ad.2010.22.1.21
  178. 178. Huh SY, Shin JW, Na J, Huh C, Youn S, Park K. Efficacy and safety of liposome-encapsulated 4-n-butylresorcinol 0.1% cream for the treatment of melasma: A randomized controlled split-face trial. The Journal of Dermatology. 2010;37:311-315. DOI: 10.1111/j.1346-8138.2010.00787.x
  179. 179. Mohan MT, Gowda A, Jaiswal AK, Sharath Kumar BC, Shilpashree P, Gangaboraiah B, et al. Assessment of efficacy, safety, and tolerability of 4-n-butylresorcinol 0.3% cream: An Indian multicentric study on melasma. Clinical, Cosmetic and Investigational Dermatology. 2016;9:21-27. DOI: 10.2147/CCID.S89451
  180. 180. Khemis A, Kaiafa A, Queille-Roussel C, Duteil L, Ortonne JP. Evaluation of efficacy and safety of rucinol serum in patients with melasma: A randomized controlled trial. The British Journal of Dermatology. 2007;156:997-1004. DOI: 10.1111/j.1365-2133.2007.07814.x
  181. 181. Chang TS. An updated review of tyrosinase inhibitors. International Journal of Molecular Sciences. 2009;10:2440-2475. DOI: 10.3390/ijms10062440
  182. 182. Makino ET, Mehta RC, Banga A, Jain P, Sigler ML, Sonti S. Evaluation of a hydroquinone-free skin brightening product using in-vitro inhibition of melanogenesis and clinical reduction of ultraviolet-induced hyperpigmentation. Journal of Drugs in Dermatology. 2013;12:S16-S20
  183. 183. Frankos VH, Schmitt DF, Haws LC, McEvily AJ, Iyengar R, Miller SA, et al. Generally recognized as safe (GRAS) evaluation of 4-hexylresorcinol for use as a processing aid for prevention of melanosis in shrimp. Regulatory Toxicology and Pharmacology. 1991;14:202-212. DOI: 10.1016/0273-2300(91)90007-i
  184. 184. Kim SG, Lee SW, Park YW, Jeong JH, Choi JY. 4-Hexylresorcinol inhibits NF-κB phosphorylation and has a synergistic effect with cisplatin in KB cells. Oncology Reports. 2011;26:1527-1532. DOI: 10.3892/or.2011.1436
  185. 185. Fidalgo J, Deglesne PA, Arroya R, Ranneva E, Deprez P. 4-Hexylresorcinol a new molecule for cosmetic application. Journal of Biomolecular Research and Therapeutics. 2018;8:170-172. DOI: 10.35248/2167-7956.19.8.170
  186. 186. Kaur S, Kizoulis M, Fantasia J, Oddos T, Bigot N, Galera P, et al. 4-Hexyl-1,3-phenylenediol, a nuclear factor-κB inhibitor, improve photodamaged skin and clinical signs of ageing in a double-blinded, randomized controlled trial. The British Journal of Dermatology. 2015;173:218-226. DOI: 10.1111/bjd.13747
  187. 187. Chaudhuri R. Hexylresorcinol providing skin benefits by modulating multiple molecular targets. In: Sivamani RK, Jagdeo J, Elsner P, Maibach HI, editors. Cosmeceuticals and Active Cosmetics. 3rd ed. Boca Raton, FL: CRC Press; 2015. pp. 71-82
  188. 188. Won Y, Loy C, Randhawa M, Southall MD. Clinical efficacy and safety of 4-hexyl-1,3-phenylenediol for improving skin hyperpigmentation. Archives of Dermatological Research. 2014;306:455-465. DOI: 10.1007/s00403-014-1439-9
  189. 189. Arrowitz C, Schoelermann AM, Mann T, Jiang LI, Weber T, Kolbe L. Effective tyrosinase inhibition by thiamidol results in significant improvement of mild to moderate melasma. The Journal of Investigative Dermatology. 2019;139:1691-1698.e6. DOI: 10.1016/j.jid.2019.02.013
  190. 190. Lima PB, Dias JAF, Cassiano DP, Esposito ACC, Miot LDB, Bagatin E, et al. Efficacy and safety of topical isobutylamido thiazolyl resorcinol (thiamidol) vs. 4% hydroquinone cream for facial melasma: An evaluator-blinded, randomized controlled trial. Journal of the European Academy of Dermatology and Venereology. 2021;35:1881-1887. DOI: 10.1111/jdv.17344
  191. 191. Disphanurat W, Srisantithum B. Efficacy and safety of 0.15% isobutylamido thiazolyl resorcinol combined with hyaluronic acid vs 0.15% isobutylamido thiazolyl resorcinol or hyaluronic acid alone in melasma treatment: A randomized evaluator-blind trial. Journal of Cosmetic Dermatology. 2021:1-10. DOI: 10.1111/jocd.14031
  192. 192. Philipp-Dormston WG, Echagüe AV, Pérez Damonte SH, Riedel J, Filbry A, Warnke K, et al. Thiamidol containing treatment regimens in facial hyperpigmentation: An international multi-centre approach consisting of a double-blind, controlled, split-face study and of an open-label, real-world study. International Journal of Cosmetic Science. 2020;42:377-387. DOI: 10.1111/ics.12626
  193. 193. Roggenkamp D, Dlova N, Mann T, Batzer J, Riedel J, Kausch M, et al. Effective reduction of post-inflammatory hyperpigmentation with the tyrosinase inhibitor isobutylamido-thiazolyl-resorcinol (Thiamidol). International Journal of Cosmetic Science. 2021;43:292-301. DOI: 10.1111/ics.12694
  194. 194. Vachiramon V, Kositkuljorn C, Leerunyakul K, Chanprapaph K. Isobutylamido thiazolyl resorcinol for prevention of UVB induced hyperpigmentation. Journal of Cosmetic Dermatology. 2021;20:987-992. DOI: 10.1111/jocd.13615
  195. 195. Kang HY, Valerio L, Bahadoran P, Ortonne JP. The role of topical retinoids in the treatment of pigmentary disorders: An evidence-based review. American Journal of Clinical Dermatology. 2009;10:251-260. DOI: 10.2165/00128071-200910040-00005
  196. 196. Ortonne JP. Retinoid therapy of pigmentary disorders. Dermatologic Therapy. 2006;19:280-288. DOI: 10.1111/j.1529-8019.2006.00085.x
  197. 197. Rivas S, Pandya AG. Treatment of melasma with topical agents, peels and lasers: An evidence-based review. American Journal of Clinical Dermatology. 2013;14:359-376. DOI: 10.1007/s40257-013-0038-4
  198. 198. Kurlandsky SB, Xiao JH, Duell EA, Voorhees JJ, Fisher GJ. Biological activity of all-trans retinol requires metabolic conversion to all-trans retinoic acid and is mediated through activation of nuclear retinoid receptors in human keratinocytes. The Journal of Biological Chemistry. 1994;269:32821-32827
  199. 199. Riahi R, Bush A, Cohen PR. Topical retinoids: Therapeutic mechanisms in the treatment of photodamaged skin. American Journal of Clinical Dermatology. 2016;17:265-276. DOI: 10.1007/s40257-016-0185-5
  200. 200. Davis EC, Callender VD. Postinflammatory hyperpigmentation: A review of the epidemiology, clinical features, and treatment options in skin of colour. The Journal of Clinical and Aesthetic Dermatology. 2010;3:20-31
  201. 201. Momosawa A, Kurita M, Ozaki M, Miyamoto S, Kobayashi Y, Ban I, et al. Combined therapy using Q-switched ruby laser and bleaching treatment with tretinoin and hydroquinone for periorbital skin hyperpigmentation in Asians. Plastic and Reconstructive Surgery. 2008;121:282-288. DOI: 10.1097/01.prs.0000293869.00522.ec
  202. 202. Kimbrough-Green CK, Griffiths CE, Finkel LJ, Hamilton TA, Bulengo-Ransby SM, Ellis CN, and Voorhees JJ. Topical retinoic acid (tretinoin) for melasma in Black patients: A vehicle controlled clinical trial. Archives of Dermatology 1994;130:727-733. DOI: https://doi.org/10.1001/ARCHDERM.1994
  203. 203. Mohsin N, Hernandez LE, Martin MR, Does AV, Nouri K. Acne treatment review and future perspectives. Dermatologic Therapy. 2022;35:e15719. DOI: 10.1111/dth.15719
  204. 204. Woolery-Lloyd HC, Keri J, Doig S. Retinoids and azelaic acid to treat acne and hyperpigmentation in skin of color. Journal of Drugs in Dermatology. 2013;12:434-437
  205. 205. Callender VD, Barbosa V, Burgess CM, Heath C, McMichael AJ, Ogunleye T, et al. Approach to treatment of medical and cosmetic facial concerns in skin of color patients. Cutis. 2017;100:375-380
  206. 206. Bulengo-Ransby SM, Griffiths C, Kimborough-Green CK, Finkel LJ, Hamilton TA, Ellis CN, et al. Topical tretinoin (retinoic acid) therapy for hyperpigmented lesions caused by inflammation of the skin in black patients. The New England Journal of Medicine. 1993;328:1438-1443. DOI: 10.1056/NEJM199305203282002
  207. 207. Phillips TJ, Gottlieb AB, Leyden JJ, Lowe NJ, Lew-Kaya DA, Sefton J, et al. Efficacy of 0.1% tazarotene cream for the treatment of photodamage: A 12-month, multicenter, randomized trial. Archives of Dermatology. 2002;138:1486-1493. DOI: 10.1001/archderm.138.11.1486
  208. 208. Grimes P, Callender V. Tazarotene cream for postinflammatory hyperpigmentation and acne vulgaris in darker skin: A double blind, randomized, vehicle-controlled study. Cutis. 2006;77:45-50
  209. 209. Tan J, Thiboutot D, Popp G, Gooderham M, Lynde C, Del Rosso J, et al. Randomized phase 3 evaluation of trifarotene 50 μg/g cream treatment of moderate facial and truncal acne. Journal of the American Academy of Dermatology. 2019;80:1691-1699. DOI: 10.1016/j.jaad.2019.02.044
  210. 210. Adamus J, Feng L, Hawkins S, Kalleberg K, Lee JM. Climbazole boosts activity of retinoids in skin. International Journal of Cosmetic Science. 2017;39:411-418. DOI: 10.1111/ics.12390
  211. 211. Hawkins S, Adamus J, Chiang C-Y, Covell E, O’Leary J, Lee J-M. Retinyl propionate and climbazole combination demonstrates clinical improvement to the appearance of hyperpigmentation and deep wrinkling with minimal irritation. International Journal of Cosmetic Science. 2017;39:589-599. DOI: 10.1111/ics.12412

Written By

Anita Damodaran and Nirmala Nair

Submitted: 12 October 2022 Reviewed: 21 October 2022 Published: 16 November 2022