You are on page 1of 10

PIGMENT CELL RES 16: 101–110.

2003 Copyright  Blackwell Munksgaard 2003


Printed in UK—all rights reserved ISSN 0893-5785

Review: Innovative Technology


Chemical and Instrumental Approaches to Treat Hyperpigmentation

STEFANIA BRIGANTI, EMANUELA CAMERA and MAURO PICARDO

San Gallicano Dermatological Institute, Via Elio Chianesi, Rome, Italy


*Address reprint requests to Mauro Picardo, San Gallicano Dermatological Institute, Via Elio Chianesi 52, 00144 Rome, Italy.
E-mail: picardo@ifo.it
Received 23 December 2002; in final form 16 January 2003

Many modalities of treatment for acquired skin hyperpigmen- necessary. Moreover, the evidence that bleaching compounds
tation are available including chemical agents or physical are fairly ineffective on dermal accumulation of melanin has
therapies, but none are completely satisfactory. Depigmenting prompted investigations on the effectiveness of physical ther-
compounds should act selectively on hyperactivated melano- apies, such as lasers. This review which describes the different
cytes, without short- or long-term side-effects, and induce a approaches to obtain depigmentation, suggests a classification
permanent removal of undesired pigment. Since 1961 hydro- of whitening molecules on the basis of the mechanism by which
quinone, a tyrosinase inhibitor, has been introduced and its they interfere with melanogenesis, and confirms the necessity to
therapeutic efficacy demonstrated, and other whitening agents apply standardized protocols to evaluate depigmenting treat-
specifically acting on tyrosinase by different mechanisms have ments.
been proposed. Compounds with depigmenting activity are now
numerous and the classification of molecules, based on their Key words: Melanin, Melanocyte, Melanogenesis, Hyperpig-
mechanism of action, has become difficult. Systematic studies mentation, Depigmenting agents, Classification, Tyrosinase,
to assess both the efficacy and the safety of such molecules are TRP-1, ROS, Peroxidase, Melanosome transfer, Laser

INTRODUCTION
Increased production and accumulation of melanins char- accumulation of melanin, physical therapies, such as
acterize a large number of skin diseases, which include lasers, have been proposed and are currently under
acquired hyperpigmentation, such as melasma, postinflam- investigation.
matory melanoderma, solar lentigo, etc. (1, 2). Epidermal This review reports different approaches to achieve depig-
and dermal hyperpigmentation can be dependent on either mentation, classifies the active molecules on the basis of their
increased numbers of melanocytes or activity of melano- mechanisms of interference with melanogenesis and focuses
genic enzymes (3). Ultraviolet light, chronic inflammation, on innovative drugs.
and rubbing of the skin as well as abnormal a-melanocyte
stimulating hormone (a-MSH) release, are triggering
DEPIGMENTING AGENTS
factors for these disorders (4, 5). As a result of their
prevalent localization in photo-exposed areas, acquired The ideal depigmenting compound should have a potent,
hyperpigmentation have psychosocial and cosmetic rele- rapid and selective bleaching effect on hyperactivated mel-
vance, and many efforts have been devoted to screening anocytes, carry no short- or long-term side-effects and lead to
recognized and putative depigmenting agents. Moreover, a permanent removal of undesired pigment, acting at one or
as bleaching compounds are fairly ineffective on dermal more steps of the pigmentation process.

Abbreviations – a-MSH, a-melanocyte stimulating hormone; TRP-1, tyrosinase related protein-1; TRP-2, tyrosinase related protein-2; MITF,
microphthalmia transcription factor; ERK, extracellular signal-regulated kinase; AKT, serine-threonine kinase; PKB, protein kinase-B; PKC, protein
kinase C; ATRA, tretinoin, all-trans retinoic acid; l-DOPA, l-3,4-dihydroxyphenylalanine; HQ, hydroquinone, dihydroxybenzene; ROS, reactive
oxygen species; 4-SCAP, 4-S-cystaminylphenol; DHI, 5,6-dihydroxyindole; DHICA, 5,6-dihydroxyindole-2-carboxylic acid; NF-kB, nuclear factor
kappa B; MG, methyl ester of gentisic acid, 2,5-dihidroxybenzoic acid; MBEH, hydroquinone monobenzyl ether; EA, ellagic acid; AZA, azelaic acid;
AsA , ascorbic acid; VC-PMG , magnesium-l-ascorbyl-2-phosphate; a-Toc, a-tocopherol; a-Toc-F, a-tocopherol ferulate; PAR-2, protease-activated
receptor 2; STI, soybean trypsin inhibitor; BBI, Bowman-Birk protease inhibitor; ET-1, endothelin)1; ECE, endothelin-1 converting enzyme.

Pigment Cell Res. 16, 2003 101


Fig. 1. Schematic illustration of the possible
approaches to interfere with melanogenesis
pathway. Tyr, tyrosinase; M, melanosomes;
ROS, reactive oxygen species.

Depigmentation can be achieved by regulating (i) the melanocytes (10) whereas it enhances the pigmentation of low
transcription and activity of tyrosinase, tyrosinase related melanized (S91 murine) melanoma cells, and decreases that of
protein-1 (TRP-1), tyrosinase related protein-2 (TRP-2), highly pigmented normal human melanocytes after UV
and/or peroxidase; (ii) the uptake and distribution of
melanosomes in recipient keratinocytes and (iii) melanin
Table 1. Classification of depigmenting agents. Compounds have been
and melanosome degradation and turnover of ÔpigmentedÕ divided in categories on the basis of the reported mechanisms of
keratinocytes (Fig. 1, Table 1). interference with melanin synthesis and deposition

Before melanin synthesis


REGULATION OF MELANOGENIC ENZYMES Tyrosinase transcription
C2-ceramide Tretinoin
As a result of the key role played by tyrosinase in melanin Tyrosinase glycosylation
PaSSO3Ca
biosynthesis, most whitening agents act specifically to reduce
the function of this enzyme by means of several mechanisms: During melanin synthesis
(i) interference with its transcription and/or glycosylation, Tyrosinase inhibition
Hydroquinone Kojic acid
(ii) inhibition by different modalities, (iii) reduction of 4-hydroxy-anisole Methyl Gentisate
by-products and (iv) post-transcriptional control. 4-S-CAP & derivatives Ellagic Acid
Arbutin Resveratrol
Aloesin Oxyresveratrol
Transcriptional Control of Tyrosinase Expression Azelaic acid
Peroxidase inhibition
Transcription of genes encoding tyrosinase and TRP-1 is Methimazole Phenols/catechols
under transcriptional control of the microphthalmia Product reduction and ROS scavengers
Ascorbic acid a-Toc
transcription factor (MITF) (6–8). Substances able to inhibit Ascorbic Acid Palmitate D, L-a TF
MITF expression and activity, as well as the extracellular VC-PMG Hydrocumarins
signal-regulated kinase (ERK) and serine-threonine kinase Thioctic acid
(AKT)/protein kinase B (PKB) pathways, could represent After melanin synthesis
depigmenting agents. Tyrosinase degradation
Linoleic acid a-Linolenic acid
Among these, tretinoin (all-trans retinoic acid; ATRA), Inhibition of melanosome transfer
acting on retinoid-activated transcription factors, interferes Serine protease inhibitors Niacinamide
with melanocyte development and melanogenesis. ATRA has Lecthins and Neoglycoproteins RW-50353
bimodal effects on melanocytes: it stimulates the differenti- Soybean/milk extracts
Skin turnover acceleration
ation of melanocyte precursors, inducing transcription of Lactic acid Retinoic acid
tyrosinase by protein kinase C (PKC) activation and MITF Glycolic acid Linoleic acid
expression, and removes differentiated melanocytes by pro- Liquiritin
moting apoptosis via caspase-3 pathway and bcl-2 down-
PaSSO3Ca, Calcium D-pantetheine-S-sulphonate; 4-SCAP, 4-S-cyst-
modulation (9). ATRA does not inhibit melanogenesis in skin aminylphenol; VC-PMG, magnesium-L-ascorbyl-2-phosphate; a-Toc,
equivalents or monolayer cultures of murine and human a-tocopherol; a-Toc-F, a-Tocopherol ferulate.

102 Pigment Cell Res. 16, 2003


exposure (11). Topical ATRA stimulates tyrosinase activity active affecting the intracellular transport or deposition
only in individuals with low skin type and (12) improves the within melanosomes. Glucosamine or tunicamycin, specific
irregular hyperpigmentation associated with photo-aging or inhibitors of lipid carrier-dependent glycosylation of protein,
inflammation (13, 14). Agents able to decrease MITF expres- have been found to induce, in melanoma cells, a marked loss
sion include C2-ceramide, which inhibits cell proliferation and of pigmentation associated with ultrastructural alterations
melanogenesis in a mouse melanocyte cell line, probably of melanogenic compartments (19). Calcium d-pantetheine-
through an initial inhibition and a late activation of AKT/ S-sulphonate (PaSSO3Ca), probably through the alteration
PKB and ERK pathways (15, 16). of tyrosinase and TRP-1 glycosylation, causes an inhibition
of melanogenic enzymes, without modifying their expression,
and produces a reversible loss of pigmentation in normal
Glycosylation and Maturation of Melanogenic Enzymes
human melanocytes and in melanoma cell lines (20).
The major post-translational modification of the melano-
genic enzymes consists of glycosylation at asparagine resi-
Control of Tyrosinase Activity
dues. The inhibition of N-glycosylation by a variety of agents
results in the reduction of melanosome maturation, and of The classification of tyrosinase inhibitors (Table 2) is difficult
melanosomal enzyme activities (17, 18). The modification of because of the capability of several compounds to work in
the glycosylation pattern makes the protein inactive or less different ways, interacting with both catalytic and regulatory

Table 2. Chemical structures of modulators of melanogenic enzyme activity. Most of the compounds show chemical analogy with l-tyrosinase the
natural substrate of tyrosinase. In the table adjunctive effects, which could contribute to the depigmenting action have been reported

a
Kinetics study on mushroom tyrosinase, bKinetics study on melanocyte or melanoma cell cultures.

Pigment Cell Res. 16, 2003 103


sites of the enzyme or being metabolized to a product that, in tyrosinase oxidation rate and cytotoxic effects (42). 4-S-
turn, can act as either a non-competitive or a competitive cystaminylphenol (4-SCAP) and its N-acetylated derivatives
inhibitor. Tyrosine hydroxylation is the rate limiting step for have both depigmenting and anti-melanoma effects. Topical
melanogenesis, but requires l-3,4-dihydroxyphenylalanine treatment in Yucatan pigs induces a stable but reversible
(l-DOPA), the substrate of tyrosinase’s second activity, as depigmentation with slight irritant effects when compared
a cofactor. Thus, tyrosinase activity cannot be easily exam- with HQ (43, 44). The activity seems to be related to: (i) a
ined through kinetic analyses. Moreover controversial results marked decrease of the functioning melanocytes and melan-
were obtained when purified mushroom tyrosinase or cell ized melanosome number, (ii) a reduced transfer of melano-
lysates of melanoma cells were employed. Most of the somes to keratinocytes, and (iii) a selective degeneration of
inhibitors are phenol/catechol derivatives, structurally sim- melanocytes and deposition of melanin–like material in the
ilar to tyrosine or DOPA, which act as alternative substrates Golgi apparatus, where tyrosinase is located (45). Catechols
of tyrosinase without producing pigment (21, 22). (o-diphenols) bearing electron donor groups in position para
were found to be more effective in inhibiting tyrosinase and
inducing melanocytotoxic effects than mono-phenolic deriv-
Alternative Phenolic Substrates
atives (21, 46). However, when a derivative with methyl
The most popular depigmenting agent is hydroquinone groups in position ortho and meta, such as 2,5,6-trimethyl-p-
(dihydroxybenzene; HQ) introduced for clinical use since hydroxy-methoxybenzene, is used as substrate of mushroom
1961. Several trials have demonstrated its therapeutic efficacy tyrosinase, the oxidation rate is low and the initial acceler-
alone or in association with other compounds (23–28). HQ ation (lag period) is not observed (47). Arbutin, a naturally
seems to exert its effects mainly in melanocytes with active occurring HQ b-d-gluconopyranoside, which is depigmented
tyrosinase activity, such as epidermal hyperactivated mel- at non-cytotoxic concentrations (48), acts similar to HQ
anocytes (29) and has been considered as a reference (IC50 ¼ 17 mM) (49). In both normal human melanocytes
standard in evaluating depigmenting agents (30, 31). The and melanoma cells, arbutin induces a decrease of tyrosinase
oxidation products are quinones and reactive oxygen species activity without affecting messenger RNA (mRNA) expres-
(ROS), which lead to an oxidative damage of membrane sion, inhibits the 5,6-dihydroxyindole-2-carboxylic acid
lipids and proteins, including tyrosinase, and depletion of (DHICA) polymerase activity (pmel17/silver protein) and
glutathione contributes to the lightening action (32). HQ may exerts an inhibitory effect on melanosome maturation (50).
interfere with pigmentation even through: (i) the covalent An interference with the uptake of tyrosine into melanocytes
binding to histidine or interaction with coppers at the active has also been postulated (51). However, despite encouraging
site of tyrosinase (33), (ii) the inhibition of DNA and RNA initial results (52) other authors have not confirmed the
synthesis and (iii) the alteration of melanosome formation activity on intact melanocytes and in clinical trial (30). Kojic
and melanization extent (34). However, because of the acid (5-hydroxy-2-hydroxymethyl-4H-pyran-4-one) is an
hazard of long-term treatments (35), the use of hydroquinone antibiotic produced by many species of Asperigillus and
in cosmetics has been banned by the European Committee Penicillum. The depigmenting action is attributed to the
(24th Dir. 2000/6/EC) and formulations are available only by chelating ability (53), even if an interference with different
prescription of physicians and dermatologists. HQ mono- steps of melanin synthesis (54) and inhibition of nuclear
benzyl ether (MBEH) is metabolized to reactive free radicals factor-kappa B (NF-jB) activation in keratinocytes, con-
inside the cells, causing permanent depigmentation even at trasting with the hyperpigmentation associated with inflam-
sites distant from those of application. The clinical use is matory response, have been described (55). Clinical trials
limited to obtain a generalized depigmentation in patients have reported a skin lightening effect of kojic acid (56). The
with diffuse vitiligo (36). Several other phenolic compounds methyl ester of gentisic acid (2,5-dihidroxybenzoic acid;
have been screened as depigmenting agents and studies have MG), a natural product of Gentiana’s root, inhibits melanin
been performed on the relationship between chemical struc- synthesis in murine melanocytes at concentrations unable to
ture and tyrosinase inhibitory activity. The presence of induce cytotoxicity (57). MG can act as pro-drug, inhibiting
hydroxylic group and of an electron donator group in tyrosinase following the intracellular release of HQ, even if
position para has been suggested as a fundamental require- kinetic studies have demonstrated a direct effect on the
ment for an effective action as alternative substrate of enzyme, possibly through the binding of the copper ions
tyrosinase. In fact, in para-hydroxylated monophenols, (30). Clinical trials have demonstrated the efficacy of the
methylation of one hydroxyl favours nucleophilic attack at compound (58). Aloesin, a natural hydroxychromone deriv-
the active site enhancing the catalytic efficiency (37). The fact ative isolated from Aloe Vera, inhibits mushroom (59) and
that 4-hydroxyanisole (para-hydroxy-methoxybenzene), melanoma tyrosinase at non-cytotoxic concentration (60, 61)
oxidized by tyrosinase (Km ¼ 62–80 lM), exhibits strong probably acting as a competitive inhibitor on DOPA
melanocytotoxicity and clinical effects have been demonstra- oxidation and as an non-competitive on tyrosine hydroxy-
ted in the treatment of solar lentiges and related hyperpig- lase activity. In vivo, aloesin and arbutin co-treatment
mented lesions, in combination with tretinoin (38–40). inhibits UV-induced melanogenesis in a synergistic manner
4-Tertiary butylphenol has been reported to induce in human (62).
melanocytes a competitive inhibition of both tyrosine Most hydroxystilbene compounds like resveratrol, natur-
hydroxylase and DOPA oxidase properties of tyrosinase ally contained in herbal medicines, have potent inhibitory
without cytotoxicity (41). The presence of thio-ether side effects on mushroom tyrosinase. Resveratrol, one of the
chains in para position on the phenolic ring enhances ingredients of red wine, possesses several biological properties,

104 Pigment Cell Res. 16, 2003


such as free radical scavenging, anti-inflammatory and exerts inhibitory action towards both mushroom tyrosinase
anticancer activities (63, 64). A dose-dependent inhibition and peroxidase (81) and, in brown guinea-pigs, induces mild
of l-tyrosine oxidation by tyrosinase, without affecting the to moderate inhibition of melanization, with morphologic
protein expression and synthesis, has been described in B-16 changes of melanocytes.
murine melanoma. Resveratrol is probably metabolized by
tyrosinase leading to the formation of an o-hydroxy deriv-
Redox Agents and ROS Scavengers
ative, which is a competitive inhibitor of the enzyme (65).
Oxyresveratrol, extracted from Morus alba, is a stronger Compounds with redox properties can have depigmenting
inhibitor (IC50 ¼ 1 lM) on mushroom DOPA oxidase effects by interacting with o-quinones, thus avoiding the
activity than resveratrol (IC50 ¼ 54.6 lM) (66), suggesting oxidative polymerization of melanin intermediates, or with
that the number and the position of hydroxyl substituents the copper at the active site. Moreover, redox agents, by
have an important role in determining the activity. Ellagic scavenging ROS generated in the skin following UV expo-
acid (EA), a polyphenol widely distributed in plants, is sure, can inhibit possible second messengers which is able to
capable of preventing pigmentation caused by sunburn (67). stimulate epidermal melanogenesis either directly or indi-
EA inhibits tyrosinase non-competitively in a dose-depend- rectly (82).
ent manner, through its capacity to chelate copper, even if Ascorbic acid (AsA) interferes with the different steps of
other mechanisms, such as a scavenger effect have been melanization, by interacting with copper ions at the
suggested. Interestingly, in brownish guinea-pigs, EA tyrosinase active site (83) and reducing dopaquinone and
induced a reversible inhibition of melanin synthesis only in by blocking DHICA oxidation (84). However, AsA is
UV-activated melanocytes. highly instable, being quickly oxidized and decomposed in
aqueous solution and, because of its prevalent hydrophilic
nature, has a low degree of penetration into the skin.
Azelaic Acid
Different esters, such as magnesium-l-ascorbyl-2-phosphate
Compounds able to bind either amino or carboxyl groups (VG-PMG), stable in water and in alkaline medium and
may block the access of tyrosine to the active site, behaving absorbed percutaneously, have been used. These com-
as competitive inhibitors. Among these, azelaic acid (AZA), pounds are able to induce a lightening effect in normal
a naturally occurring 9-carbon dicarboxylic acid compound and in hyperactive melanocytes (85). a-Tocopherol (a-Toc)
isolated from cultures of Pityrosporum Ovale, is included. derivatives inhibit tyrosinase in vitro (86) and melanogenesis
AZA inhibits tyrosinase activity in vitro (Ki ¼ 2.73 · in epidermal melanocytes (87). The antioxidant properties
10)3 M) (68) and may also interfere with DNA synthesis of a-Toc, which interferes with lipid peroxidation of
and mitochondria activity in hyperactive and abnormal melanocyte membranes and increases the intracellular
melanocytes (69). AZA has been used to treat melasma and glutathione content, could explain its depigmenting effect
postinflammatory hyperpigmentation (23) and to arrest the (88, 89). Co-treatment with AsA, which regenerates a-Toc
progression of lentigo maligna to melanoma (70). Topical from its radical form, results in more effective and
azelaic acid (15–20%) has been reported to be efficacious in longlasting antioxidant response. Topical application of
improving melasma (23, 71), even if not all the authors are a-Toc and AsA, in vivo, decreases the tanning response
in agreement with these therapeutic effects. inhibiting the UV-induced melanogenesis and proliferation
of melanocytes (90). An alternative compound is a-toco-
pherol ferulate (a-Toc-F), a derivative of a-Toc linked by
Peroxidase Inhibitors
an ester bond to ferulic acid, an antioxidant, which provides
The involvement of peroxidase in the polymerization of stabilization to a-Toc, similar to AsA. The a-Toc-F inhibits
melanogenic intermediates has been suggested by the high melanin synthesis in normal human melanocytes without
efficiency of peroxidase in the oxidation of 5,6-dihydroxyin- interfering with the tyrosinase pathway (87).
dole (DHI) with the generation of hydrogen peroxide (H2O2) 6-Hydroxy-3,4-dihydrocumarins, novel antioxidants with
as a by-product (72, 73). Intracellular H2O2, generated after a-Toc like chemical structures (91) have been recently
UV irradiation or in response to cytokines, such as tumor reported to have an anti-melanogenic activity in cultured
necrosis factor-a (TNF-a) or transforming growth factor-b normal human melanocytes at non-cytotoxic concentration
(TGF-b) (74–76), can induce a transient reduction of without interfering with tyrosinase activity. The acceleration
tyrosinase and other melanogenic protein activities, through of the biosynthesis of glutathione within melanocytes,
the down-regulation of the MITF transcription factor (77). leading to the inhibition of tyrosinase transfer to premelan-
The inhibition of peroxidase, however, results in depigmen- osomes, could be the mechanism of action (92). Thioctic acid
tation, reducing the polymerization rate of eumelanin in (a-lipoic acid), a disulfide derivative of octanoic acid, exhibits
different experimental models (78). Moreover, several mel- several biological effects, which include the quenching of
anocytotoxic phenol/catechol derivatives, are good sub- ROS, metal chelation, interaction and regeneration of other
strates of either tyrosinase or peroxidase (21), and the antioxidants, redox regulation of protein thiol groups, and
inhibition of peroxidase could play a role in their mechanism effects on gene expression and apoptosis (93). Thioctic acid
of action as well as of depigmenting agents, such as has been reported to prevent UV-induced photo-oxidative
glucocorticoids or ascorbic acid, that do not exhibit any damage, mainly through the down-modulation of NF-jB
effects against the melanogenic enzymes (79, 80). Methimaz- activation and to inhibit tyrosinase activity probably by
ole, an antithyroid agent belonging to the thionamide group, chelating the copper ions (94).

Pigment Cell Res. 16, 2003 105


can be useful in the treatment of melasma (106). Liquiritin, a
Post-Transcriptional Control of Tyrosinase
flavonoid glycoside of liquorice, has been found to induce a
Substances which are able to regulate melanin synthesis significant improvement of hyperpigmentation in patients
without affecting the expression of melanogenic proteins are with bilateral and symmetrical idiopathic epidermal melasma
likely to exert a post-transcriptional control of melanogenic (107). The mechanisms proposed involve melanin dispersion
enzymes. Unsaturated fatty acids, such as oleic acid (C18:1), by means of the pyran ring of its flavonoidal nucleus, and
linoleic acid (C18:2), or a-linolenic acid (C18:3), suppress acceleration of epidermal renewal.
pigmentation, in vitro, correlated with the degree of unsat-
uration, whereas saturated fatty acids, such as palmitic acid,
Inhibitors of Inflammation-Induced Melanogenic Response
increase the rate of melanogenesis (95). Linoleic acid in vivo
showed the greatest lightening effect in UVB induced Some mediators produced by keratinocytes after exposure to
pigmentation, without toxic effects on melanocytes (95). pro-inflammatory stimuli or UV exposure, such as interleu-
The evidence that no changes in TRP-1 or TRP-2 proteins kin-1a (IL-1a) and endothelin 1 (ET-1), are able to promote
have been found, suggests that tyrosinase is selectively melanogenesis. Therefore, anti-inflammatory compounds
targeted by fatty acids, which seem to act on the degradation could be useful for the prevention or treatment of postin-
of the enzyme during the physiologic proteasome-dependent flammation hyperpigmentation. ET-1 shows an unique
mechanism (96), leading to the alteration of tyrosinase behavior in exerting stimulatory effects both on DNA
protein content in hyperactive melanocytes. Linoleic acid synthesis and melanization by human melanocytes (108).
accelerates the process whereas palmitic acid works in an Activation of epidermal ETs is determined by the enzymatic
antagonistic manner mimicking protease inhibitors (97). cleavage of inactive prepolypeptides by an endopeptidase
termed ET converting enzyme (ECE), regulated by the pro-
inflammatory cytokine IL-1a (109). Topical application of
Inhibition Melanosome Transfer
M. chamomilla extract inhibits UVB-induced pigmentation,
Several studies have focused on the identification of by avoiding ET-1-induced DNA synthesis, but not the IL-a-
regulatory factors of the melanosome movement in dend- induced ET-1 production and tyrosinase activation (108).
rites and of the interaction between keratinocytes and Among anti-inflammatory drugs, topical corticosteroids
melanocytes during the transfer process (98, 99). The have been included in several clinical trials for the treatment
activation of protease-activated receptor 2 (PAR-2), a seven of melasma even to decrease the irritation caused by
trans-membrane G-protein coupled receptor, which is depigmenting agents (110–112). The possible mechanism is
expressed in keratinocytes and not in melanocytes, was the suppression of cytokines through the inhibition of NF-kB
found to activate keratinocyte phagocytosis, enhancing the activation, which may explain their short-lived effect with a
pigment transfer (100). Inhibition of PAR-2 cleavage by transient loss of color. Glabridin, the main component of
serine protease inhibitors, such as RWJ-50353, completely hydrophobic fraction of liquorice extracts decreases tyrosin-
avoids the UVB-induced pigmentation of epidermis analogs ase activity in B16 melanoma cells, without affecting DNA
(101, 102). In dark skinned Yucatan microswine, topical synthesis, and, in guinea-pigs, inhibits UVB-induced skin
treatment with serine protease inhibitors, such as the pigmentation, as well as erythema (113). The capability to
soybean trypsin inhibitor (STI) or Bowman–Birk protease inhibit cyclooxygenase activity and superoxide anion pro-
inhibitor (BBI) in a liposomal delivery system, exerts a duction suggests that the anti-inflammatory effect involves
significant skin lightening effect without toxic effects on an interference with the arachidonic acid cascade and that
melanocytes. Soybean milk extracts reduced the melanin protection against oxidative stress has a key role in modu-
deposition within the swine epidermis and prevented UVB lating melanogenesis. Variations in the chemical structure
induced pigmentation in vivo, similar to STI, without the modulate the effect, because melanogenesis inhibition is lost
addition of any delivery vehicle (103). Even niacinamide, by replacing both hydroxyl groups and reduced by substitu-
the biologically active form of vitamin B3, inhibits melan- tion at the 4¢ position.
ogenesis, both in vitro and in vivo, probably through the
suppression of melanosome transfer. Purified tyrosinase, in
Laser Treatment of Pigmented Lesions
fact, is not inhibited and pigmentation is reduced in co-
culture system or in reconstructed epidermis and not in Therapeutic options, other than chemical agents, have been
melanocyte monocultures (104). proposed for hyperpigmentation, such as cryotherapy with
liquid nitrogen, laser surgery, chemical peeling and superfi-
cial dermoabrasion (116, 117). Since the first demonstration
Melanin Dispersion and Acceleration of Skin Turnover
(118), treatments of pigmented lesions with argon laser, CO2
The capacity of several compounds to disperse melanin laser, Nd:YAG laser or Q-switched ruby laser have been
pigment and/or accelerate epidermal turnover can result in reported (114). The theory of a selective photothermolysis
skin lightening. Chemical substances used as exfoliantes, suggests that laser therapy would allow discriminating
such a-hydroxy acids, free fatty acids and retinoic acid, destruction of pigment without injuring the surrounding
stimulates cell renewal facilitating the removal of melanized tissue (119). Selective melanin photothermolysis can be
keratinocyte, leading to melanin pigment loss (10, 95). obtained with any laser light having a wavelength in the
Topical application has been shown to reduce the visibility absorption spectrum of melanin and sufficient energy levels
of age spots, without reducing their size or number (105), and to target melanosomes (120). Laser induces extreme heating

106 Pigment Cell Res. 16, 2003


of melanosomes with subsequent thermal expansion, local ruby laser, both epidermal repigmentation and an increased
vaporization and generation of acoustic waves that damage number of dermal macrophages have been observed (121).
the nucleus and eventually destroy the pigment-laden cells. The poor response may be referred to the hyperactivation of
The released melanin is then removed through trans-epider- melanocytes and to the laser-mediated inflammatory
mal elimination or phagocytosis by dermal macrophages. To response, that leads to rapid reoccurrence or even darkening
be effective and specific, wavelengths that avoid absorption of the treated areas. To improve the efficacy, the develop-
by other skin chromophores and penetrate to the desired ment of a combination laser treatment has been proposed. A
depth have to be used. Different laser wavelengths induce pilot study has described the effectiveness of a combination
similar melanosome alterations, but the threshold doses and laser therapy (pulsed CO2 laser followed by Q-switched
penetration are substantially different. Shorter wavelengths alexandrite laser) in treatment of dermal-type melasma (122).
(<600 nm) damage pigmented cells with lower energy Given the epidermal trauma and worsening in pigmentation
fluences, while longer wavelengths (>600 nm) penetrate induced by laser impact, it is not surprising that laser
deeper into the skin but need more energy to induce treatment of postinflammatory hyperpigmentation has been
melanosome disruption. Thus, short wavelengths damage discouraging (123).
only superficial pigmented lesions, leaving deeper structures
intact, while longer wavelengths can target pigmented lesions
CONCLUSIONS
in the dermis. Equally important is the pulse duration, which
should be shorter than the thermal relaxation time of The knowledge of melanocyte biology and processes under-
melanin. Clinically, the immediate effect of sub-microsecond lying melanin synthesis has made remarkable progresses over
pulses in pigmented skin is tissue-whitening, which corres- the last years opening new paths in the pharmacologic
ponds to melanosome disruption and is an end-point during approach to the treatment of hyperpigmentation. At the
treatment. There are four main short-pulsed pigment-select- same time, the topic has become more complex and
ive lasers in clinical use nowadays: the pigmented lesion the classification of the molecules more difficult. Moreover,
dye laser (PLDL) (510 nm, 300 ns), the Q-switched ruby the pathogenetic mechanisms underlying acquired hyperpig-
laser (694 nm, 24–40 ns), the Q-switched alexandrite laser mentation have not been completely clarified, and the
(755 nm, 50–100 ns), and the Q-switched Nd:YAG laser therapeutic approaches are focused on the outcome of the
(1064 nm, 5–10 ns), which can be frequency-doubled to emit process. Numerous are the candidate depigmenting agents
a green light at 532 nm of the same pulse duration. and, deeper studies and clinical trials are needed to assess
Epidermal melasma responds to laser treatment similar to their safety. HQ is one of the most potent whitening agents
the use of depigmenting agents or chemical peels, but the first discovered, but since its introduction some adverse
dermal and mixed types are resistant to laser therapy. effects have been recognized. Recent molecules, even if very
However, several months after treatment with a Q-switched promising, need extended follow-up and could disclose

Fig. 2. Possible multi-step protocol for the


evaluation of putative depigmenting com-
pounds.

Pigment Cell Res. 16, 2003 107


undesired effects, not yet identified, or secondary effects, 11. Romero C, Aberdam E, Larnier C, Ortonne JP. Retinoic acid as
which may accompany the main function. The diverse sites of modulator of UVB-induced melanocyte differentiation. J Cell Sci
1994;107:1095–1103.
action lead to a difficult comparison of their efficacy. To 12. Talwar HS, Griffiths EM, Fisher GJ, Russman A, Krach K,
render the scenario more complex, different models and Benrazavi S, Voorhees JJ. Differential regulation of tyrosinase
methods have been used to assay the depigmenting activity. activity in skin of white and black individuals in vivo by topical
retinoic acid. J Invest Dermatol 1993;100:800–805.
From a methodological point of view, according to Virador 13. Rafal KS, Griffiths EM, Ditre CM, Finkel LJ, Hamilton TA, Ellis
et al. (124) and Lei et al. (125) a multi-step process could be CN, Voorhees JJ. Topical tretinoin (retinoic acid) treatment for liver
covered (Fig. 2). Initial evaluation of depigmenting proper- spot associted with photodamage. N Engl J Med 1992;326:386–374.
14. Bulengo-Ransby SM, Griffiths EM, Kimbrough-Green CK, Finkel
ties should be performed on purified tyrosinase and other LJ, Hamilton TA, Ellis CN, Voorhees JJ. Topical tretinoin (reinoic
melanogenic enzymes including peroxidase, thereafter acid) therapy for hyperpigmented lesions caused by inflammation of
employing melanocyte cultures, cytotoxicity and effects on the skin in black patients. N Engl J Med 1993;328:1438–1443.
melanin synthesis, should be assayed. For the biological 15. Kim DS, Kim SY, Moon SJ, Chung JH, Kim KH, Cho KH, Park
KC. Ceramide inhibits cell proliferation trough Akt/PKB inactiva-
evaluation, co-culture systems and reconstructed skin seem tion and decreases melanin synthesis in Mel-Ab Cells. Pigment Cell
to be reliable methods to screen the capability of new agents Res 2001;14:110–115.
to interfere with the pigmentary process, in particular 16. Kim DS, Kim SY, Chung JH, Kim KH, Eun HC, Park KC.
Delayed ERK activation by ceramide reduces melanin synthesis in
following triggering stimuli such as UV irradiation, or human melanocytes. Cell Signal 2002;14:779–785.
exposure to a-MSH or proinflammatory mediators. Finally, 17. Negriou G, Branza-Nichita N, Petrescu AJ, Dwek RA, Petrescu
the in vivo activity of hypopigmenting formulation should be SM. Protein specific N-glycosylation of tyrosinase and tyrosinase-
assessed by using non-invasive techniques, such as remittance related protein-1 in B16 mouse melanoma cells. Biochem J
1999;344:659–665.
spectrophotometry and UV light photography to obtain 18. Imokawa G, Mishima Y. Functional analysis of tyrosinase isozymes
comparable values (126). od cultured malignant melanoma cells during the recovery period
Based on the review of the literature and on our clinical following interrupted melanogenesis induced by glycosilation inhib-
itors. J Invest Dermatol 1984;83:196–201.
experience, we can conclude that the identification of the ideal 19. Mishima Y, Imokawa G. Selective aberration and pigment loss in
depigmenting agent is difficult and, for a clinical use, a melanosomes of malignant melanoma cells in vitro by glycosilation
formulation which include compounds acting at different steps inhibitors: premelanosomes as glycoprotein. J Invest Dermatol
1983;81:106–114.
of pigmentation should be advantageous. In mixtures, the 20. Franchi J. Depigmenting effects of calcium d-pantetheine-S-sulfo-
possible synergistic effect of different compounds could allow nate on human melanocytes. Pigment Cell Res 2000;13:165–171.
application of lower concentrations of each agent, possibly 21. Passi S, Nazzaro-Porro M. Molecular basis of substrate and
reducing adverse effects and increasing the clinical results. inhibitory specificity of tyrosinase: phenolic compounds. Br J
Dermatol 1981;104:659–665.
22. Menter JM, Etemadi AA, Chapman W, Hollins TD, Willis I. In vivo
depigmentation by hydroxybenzene derivatives. Melanoma Res
REFERENCES 1993;6:443–449.
1. Urabe K, Nakayama J, Hori Y. Mixed epidermal and dermal 23. Perez-Bernal A, Munoz-Perez MA, Camacho F. Management of
hypermelanoses. In: Norlund JJ, Boissy RE, Hearing VJ, King RA, facial hyperpigmentation. Am J Clin Dermatol 2000;1:261–268.
Ortonne JP. The Pigmentary System: Physiology and Pathophysi- 24. Fisher AA. The safety of bleaching creams containing hyroquinone.
ology. New York: Oxford University Press; 1998. pp. 909–911. Cutis 1998;61:303–304.
2. Cullen MK. Genetic epidermal syndromes: disorders characterized 25. Sanchez JL, Vazquez M. A hydroquinone solution in the treatment
by lentigines. In: Norlund JJ, Boissy RE, Hearing VJ, King RA, of melasma. Int J Dermatol 1982;21:55–58.
Ortonne JP. The Pigmentary System: Physiology and Pathophysi- 26. Kauh YC, Zachian TF. Melasma. Adv Exp Med Biol 1999;455:191–
ology. New York: Oxford University Press; 1998. pp. 760–766. 499.
3. Ortonne JP, Nordlund JJ. Mechanisms that cause abnormal skin 27. Kang WH, Chun SC, Lee S. Intermittent therapy for melasma in
color. In: Norlund JJ, Boissy RE, Hearing VJ, King RA, Ortonne Asian patients with combined topical agents (retinoic, hydroquinone
JP. The Pigmentary System: Physiology and Pathophysiology. New and hydrocortisone): clinical and histological studies. J Dermatol
York: Oxford University Press; 1998. pp. 489–502. 1998;25:587–589.
4. Im S, Kim J, On WY, Kang WH. Increased expression of 28. Guevara IL, Pandya AG. Melasma treated with hydroquinone,
a-melanocyte-stimulating-hormone in the lesional skin of melasma. tretinoin and a fluorinated steroid. Int J Dermatol 2001;40:212–215.
Br J Dermatol 2002;146:165–167. 29. Smith CJ, O’Hare KB, Alle JC. Selective cytotoxicity of hydroqui-
5. Kang WH, Yoon KH, Lee ES, Kim J, Lee KB, Yim H, Sohn S, Im none for melanocyte-derived cells is mediated by tyrosinase activity
S. Melasma: histopathological characteristics in 56 Korean patients. but independent of melanin content. Pigment Cell Res 1988;1:386–
Br J Dermatol 2002;146:228–237. 389.
6. Yasumoto K, Yokoyama K, Takahashi K et al. Functional analysis 30. Curto EV, Kwong C, Hermersdorfer H, Glatt H, Santis C, Virador
of microphtalmia-associated transcription factor in pigment cell- V, Hearing VJ Jr, Dooley TP. Inhibitors of mammalian melanocyte
specific transcription of the human tyrosinase damily genes. J Biol tyrosinase: in vitro comparisons of alkyl esters of gentisic acid with
Chem 1997;272:503–509. other putative inhibitors. Biochem Pharmacol 1999;57:663–672.
7. Tachibana M. MITF: a stream flowing for pigment cells. Pigment 31. Palumbo A, d’Ischia M, Misuraca G, Prota G. Mechanism of
Cell Res 2000;13:230–240. inhibition of melanogenesis by hydroquinone. Biochim Biophys
8. Fang D, Tsuji Y, Setaluri V. Selective donw-regulation of tyros- Acta 1991;23:85–90.
inase family gene TYRP1 by inhibition of the activity of 32. Bolognia JL, Sodi SA, Obser MP, Pawelek JM. Enhancement of the
melanocyte transcription factor, MITF. Nucl Acids Res 2002;30: depigmenting effect of hydroquinone by cystamine and buthionine
3096–3106. sulfoxime. Br J Dermatol 1995;133:349–357.
9. Watabe H, Soma Y, Ito M, Kawa Y, Mizoguchi M. All-trans- 33. Prota G. Melanins and melanogenesis. New York: Academic Press:
retinoic acid induces differentatiation and apoptosis of murine 1992. pp. 1–290.
melanocyte precursors with induction of the microphtalmia-associ- 34. Penney KB, Smith CJ, Allen JC. Depigmenting action of hydro-
ated transcription factor. J Inv Dermatol 2002;118:35–42. quinone depends on disruption of fundamental cell processes. J
10. Yoshimura K, Tsukamoto K, Okazaki M, Virador VM, Lei TC, Invest Dermatol 1984;82:308–310.
Suzuki Y, Uchida G, Kitano Y, Harii K. Effects of all-trans retinoic 35. De Caprio AP. The toxicology of hydroquinone-relevance to
acid on melanogenesis in pigmented skin equivalents and monolayer occupational and environmental exposure. Crit Rev Toxicol
culture of melanocytes. J Dermatol Sci 2001;27:68s–75s. 1999;29:283–330.

108 Pigment Cell Res. 16, 2003


36. Njoo MD, Westerhof W, Bos JD, Bossuyt PM. The development of 59. Piao LZ, Patk HR, Park YK, Lee SK, Park JH, Park MK.
guideline for the treatment of vitiligo. Arch Dermatol Mushroom tyrosinase inhibition activity of some chromones. Chem
1999;135:1514–1521. Pherm Bull 2002;50:309–311.
37. Fenoll LG, Rodriguez-Lopez JN, Varon R, Garcia-Ruiz PA, 60. Jones K, Hughes J, Hong M, Jia Q, Orndorff S. Modulation of
Garcia-Canovas F, Tudela J. Action mechanism of tyrosinase on melanogenesis by aloesin: a competitive inhibitor of tyrosinase.
meta-and para-hydroxylated monophenols. Biol Chem 2000;38:313– Pigment Cell Res 2002;15:335–340.
320. 61. Jin YH, Lee SJ, Chung MH et al. Aloesin and arbutin inhibit
38. Fleicher AB, Schwartzel EH, Colby SI, Altman DJ. The combina- tyrosinase activity in a synergistic manner via a different action
tion of 2% 4-hydroxyanisole (Mequinol) and 0.01% tretinoin is mechanism. Arch Pharm Res 1999;22:232–236.
effective in improving the appearance of solar lentigines and related 62. Choi S, Park YI, Lee SK, Kim JE, Chung MH. Aloesin inhibits
hyperpigmented lesions in two phase double-blind multicenter hyperpigmentation induced by UV radiation. Exp Dermatol
clinical studies. J Am Acad Dermatol 2000;42:459–467. 2002;27:513–515.
39. Espin JC, Varon R, Tudela J, Garcia-Canovas F. Kinetic study of 63. Jang M, Cai L, Udeani GO, Slowing KV, Thomas CF, Beecher CW,
the oxidation of 4-hydroxyanisole catalized by tyrosinase. Biochem Fong HH, Farnsworth NR, Kinghorn AD, Mehta RG, Moon RC,
Mol Biol Int 1997;41:1256–1276. Pezzuto JM. Cancer chemopreventive activity of resveratrol, a
40. Rodriguez-Vicente J, Vicente-Ortega V, Canteras-Jordana M, natural product derived from grapes. Science 1997;275:218–220.
Calderon-Rubiales F. Relationship between 4-hydroxyanisole and 64. Subbaramaiah K, Chung WJ, Michaluart P, Telang N, Tanabe T,
dopa oxidase activity for three melanoma cell lines. Melanoma Res Inoue H, Jang M, Pezzuto JM, Dannenberg AJ. Resveratrol inhibits
1997;7:373–381. cycloxygenase-2 transcription and activity in phorbol ester-treated
41. Yang F, Boissy RE. Effects of 4-tertiary butylphenol on the human mammary epithelial cells. J Biol Chem 1998;273:21875–
tyrosinase activity in human melanocytes. Pigment Cell Res 21882.
1999;12:237–245. 65. Bernard P, Berthon JY. Resveratrol: an original mechanism on
42. Riley PA, Cooksey CJ, Johnson CI, Land EJ, Latter AM, Ramsden tyrosinase inhibition. Int J Cosmetic Sci 2000;22:219–226.
CA. Melanogenesis-targeted anti-melanoma pro-drug development: 66. Kim YM, Yun J, Lee CK, Lee H, Min KR, Kim Y. Oxyresveratrol
effect of side-chain variations on the cytotoxicity of and hydroxystilbene compounds, inhibitory effect on tyrosinase
tyrosinase-generated ortho-quinones in a model screening system. and mechanism of action. J Biol Chem 2002;277:16340–16344.
Eur J Cancer 1997;33:135–143. 67. Shimogaki H, Tanaka Y, Tamai H, Masuda M. In vitro and in vivo
43. Thomas DP, Kishi H, Cao H, Ota M, Yamashita T, Singh S, evaluation of ellagic acid on melanogenesis inhibition. Int J
Jimbow K. Selective incorporation and specific cytocidal wffwcts as Cosmetic Sci 2000;22:291–303.
the cellular basis for the antimelanoma action of sulphur containing 68. Lemic-Stoicevic L, Nias AH, Breathnach AS. Effect of azelaic acid
tyrosine analogs. J Invest Dermatol 1999;113:928–934. on melanoma cell in culture. Exp Dermatol 1995;4:79–81.
44. Gili A, Thomas PD, Ota M, Jimbow K. Comparison of in vitro 69. Nazzaro-Porro M. Azelaic acid. J Am Acad Dermatol
cytotoxicity of N-acetyl and N-propionyl derivatives of phenolic 1987;17:1033–1041.
thioether amines in melanoma and neuroblastoma cells and the 70. Nazzaro-Porro M, Passi S, Zina G, Breathnach AS. Then year’s
relationship to tyrosinase and tyrosine hydroxylase enzyme activity. experience of treating lentigo maligna with topical azelaic acid. Acta
Melanoma Res 2000;10:9–15. Derm Venereol (Stockh). 1989;143 (Suppl.):49–57.
45. Jimbow M, Marusyk H, Jimbow K. The in vivo melanocytoxicity 71. Sarkar R, Bhalla M, Kanwar AJ. A comparative study of 20%
and depigmenting potency of N-2,4-acetoxyphenyl thioethyl aceta- azelaic acid cream monotherapy versus a sequential therapy in the
mide in the skin and hair. Br J Dermatol 1995;133:526–536. treatment of melasma in dark-skinned patients. Dermatology
46. Thorneby-Andersson K, Olov S, Hansson C. Tyrosinase-mediated 2002;205:249–254.
formation of a reactive quinone from the depigmenting agents, 72. Nappi AJ, Vass E. Hydrogen peroxide generation associated with
4-tert-butylphenol and 4-tert-butylcatechol. Pigment Cell Res the oxidation of the eumelanin precursors 5,6-dihydroxyindole and
2000;13:33–38. 5,6-dihydroxy-2-carboxylic acid. Melanoma Res 1996;6:341–349.
47. Naish-Byfield S, Riley PA. Tyrosinase kinetics. failure of acceler- 73. d’Ischia M, Napolitano A, Prota G. Peroxidase as an alternative to
ation in oxidation of ring-blocked monohydric phenol substrate. tyrosinase in the oxidative polymerization of 5,6-dihydroxyindoles
Pigment Cell Res 1998;11:94–97. to melanin(s). Biochim Biophys Acta 1991;1073:423–430.
48. Sugai T. Clinical effects of arbutin in patients with chloasma (in 74. Meier B, Radeke HH, Selle S, Younes M, Sies H, Resch K,
Japanese). Hifu (Skin Res) 1992;34:522–529. Habermehl GG. Human fibroblasts release reactive oxygen species
49. Maeda K, Fukuda M. Arbutin: mechanism of its depigmenting in response to interleukin-1 or tumour necrosis factor-a. Biochem J
action in human melanocyte culture. J Pharmacol Exp Ter 1989;263:539–545.
1996;276:765–769. 75. Thannickal VJ, Fanburg BL. Activation of an H2O2-generating
50. Chakraborty AK, Funasaka Y, Komoto M, Ichihashi M. Effect of NADH oxidase in human lung fibroblasyts by transforming growth
arbutin on melanogenic protiens in human melanocytes. Pigment factor-b1. J Biol Chem 1995;270:30334–30338.
Cell Res 1998;11:206–212. 76. Lo YYC, Wong JMS, Cruz TF. Reactive oxygen species mediate
51. Bolognia JL, Sodi SA, Pawelek JM. Two separate pathways in the cytokine activation of c-jun NH2-terminal kinases. J Biol Chem
regulation of pigmentation. J Invest Dermatol 1991;96:632. 1996;271:15703–15707.
52. Cameli N, Marmo W, Gaeta A, Calderini G, Picardo M. Evaulation 77. Jimenez-Cervantes C, Martinez-Esparza M, Perez C, Daum N,
of clinically efficacy of a misture of depigmenting agents. Pigment Solano F. Garcia-Borron JC. Inhibition of melanogenesis in
Cell Res 2001;14:406. response to oxidative stress: transient downregulation of melanocyte
53. Battaini G, Monzani E, Casella L, Santagostini L, Pagliarin R. differentiation markers and possible involvement of microphtalmia
Inhibition of the catecholase activity of biomimetic dinuclear copper transcription factor. J Cell Sci 2001;14:2335–2344.
complexes by kojic acid. J Biol Inorg Chem 2000;5:262–268. 78. Kasraee B. Peroxidase-mediated mechanisms are involved in the
54. Kahn V. Effect of kojic acid on the oxidation of dl-DOPA, melanocytotoxic and melanogenesis-inhibiting effects of chemical
norepinephrine, and dopamine by mushroom tyrosinase. Pigment agents. Dermatology 2002;205:329–339.
Cell Res 1995;8:234–240. 79. Bandyopadhyay U, Biswas K, Bandyopadhyay D, Ganguly CK,
55. Moon KY, Ahn KS, Lee J, Kim YS. Kojic acid, a potential Banerjeen RK. Dexamethasone makes the gastric mucosa suscept-
inhibitor of NF-kappa B activation intransfectant human HaCaT ible to ulceration by inhibiting prostaglandin synthetase and
and SCC13 cells. Arch Pram Res 2001;24:307–311. peroxidase-two important gastroprotective enzymes. Mol Cell
56. Lim JT. Treatment of melasma using kojig acid in a gel Biochem 1999;202:31–36.
containing hydroquinone and glycolic acid. Dermatol Surg 80. Rogozhin VV, Vercoturov VV. Effect of antioxidants (digoxin,
1999;25:282–284. quercetin, and ascorbic acid) on catalytic properties of horseradish
57. Dooley TP, Gadwood RC, Kilgore K, Thomasco LM. Development peroxidase. Biochemistry 1998;63:657–661.
of an in vitro primary screen for skin depigmentation and 81. Kasraee K. Depigmentation of brown guinea pig skin by topical
antimelanoma agents. Skin Pharmacol 1994;7:188–200. application of methimazole. J Invest Dermatol 2002;118:205–207.
58. Bocchietto E, Pecis L, D’Abrosca F, Losio N, Picardo M. In vitro 82. Karg E, Odh G, Wittbjer A, Rosengren E, Rorsman H. Hydrogen
testing of innovative depigmenting and low toxicity topic compo- peroxide as inducer of elevated tyrosinase level in melanoma cells. J
sitions. Pigment Cell Res 2001;14:405. Invest Dermatol 1993;100:209s–213s.

Pigment Cell Res. 16, 2003 109


83. Gukasyan GS. Study of the kinetics of oxidation of monophenols 103. Paine C, Sharlow E, Liebel F, Eisinger M, Shapiro S, Seiberg M. An
by tyrosinase. The effect of reducers. Biochemistry (Mosc) alternative approach to depigmentation by soybean extracts via
2002;67:277–280. inhibition of the PAR-2 pathway. J Invest Dermato 2001;116:587–
84. Ros JR, Rodriguez-Lopes JN, Garcia-Canovas F. Effect of 595.
l-ascorbic acid on the monophelase activity of tyrosinase. Biochem 104. Hakozaki T, Minwalla L, Zhuang J, Chhoa M, Matsubara A,
J 1993;295:309–312. Miayamoto K, Greatens A, Hillebrand GG, Bissett DL, Boissy RE.
85. Kameyama K, Sakai C, Kondoh S, Yonemoto K, Nishiyama S, The effect of niacinamide on reducing cutaneous pigmentation and
Tagawa M, Murata T, Ohnuma T, Quigley J, Dorsky A, Bucks D, suppression of melanosome transfer. Br J Dermatol 2002;147:20–31.
Blanock K. Inhibitory effect magnesium l-ascorbyl-2-phosphate 105. Smith W. The effects of topical L (+) lactic acid and ascorbic acid
(VC-PMG) on melanogenesis in vitro and in vivo. J Am Acad on skin withening. Int J Cosmetic Sci 1999;21:33–40.
Dermatol 1996;34:29–33. 106. Jahaveri SM, Handa S, Kaur I, Kumar B. Safety and efficacy of
86. Shimizu K, Kondo R, Sakai K, Takeda N, Nagahata T, Oniki T. glycolic acid facial peel in Indian women with melasma. Int J
Novel vitamin E derivative with 4-substituited resorcinol moiety has Dermatol 2001;40:354–357.
both antioxidant and tyrosinase inhibitory properties. Lipids 107. Amer M, Metwalli M. Topical liquiritin improves melasma. Int J
2001;36:1321–1326. Dermatol 2000;39:299–301.
87. Ichihashi M, Funasaka Y, Ohashi A, Chakraborty A, Ahmed NU, 108. Imokawa G, Kobayashi T, Miyagishi M, Higashi K, Yada Y. The
Ueda M, Osaka T. The inhibitory effect of DL-alpha-tocopheryl role of endothelin-1 in epidermal hyperpigmentation and signalling
ferulate in lecithin on melanogenesis. Anticancer Res 1999;19:3769– mechanisms of mitogenesis and melanogenesis. Pigment Cell Res
3774. 1997;10:218–228.
88. Ochiai Y, Okano Y, Funasaka Y, Ichihashi M. Effects of a-Toc 109. Hachiya A, Kobayashi T, Takema Y, Imokawa G. Biochemical
on In Vitro Aged Melanocytes. The 52nd Annual Meeting of the characterization of endothelin-converting enzyme-1a in cultured
Central division of Japanese Dermatological Association 2001, skin-derived cells and its postulated role in the stimulation of
Tokyo. p. 119. melanogenesis in human epidermis. J Biol Chem 2002;277:5395–
89. Marmol VD, Solano F, Sels A, Huez G, Libert A, Lejeune F, 5403.
Ghanem G. Glutathione depletion increases tyrosinase activity in 110. Kligman AM, Willis I. A new formula for depigmenting human
human melanoma cells. J Invest Dermatol 1993;101:871–874. skin. Arch Dermatol 1975;111:40–48.
90. Quevedo Jr WC, Holstein TJ, Dyckman J, Mcdonald CJ, Isaacson 111. Westerhof W, Njoo MD. In: Katsambas AD, Lotti TM. European
EL. Inhibition of tunning response and immunosuppression by Handbook of Dermatological Treatments. Berlin: Springer; 1995.
topical application of Vitamin C and vitamin E to the skin of pp. 217–223.
hairless (hr/hr) mice. Pigment Cell Res 2000;13:89–98. 112. Katsambas A, Antoniou C, Melasma. Classification and treatment.
91. Nishiyama T, Ohnishi J, Hashiguchi Y. Fused heterocyclic antioxi- J Eur Acad Dermatol 1995;4:217–223.
dants. antioxidative activities of hydrocumarins in a homogeneous 113. Yokota T, Nishio H, Kubota Y, Mizoguchi M. The inhibitory effect
solution. Biosci Biotechnol Biochem 2001;65:1127–1133. of glabridin from liquorice extracts on melanogenesis and inflam-
92. Yamamura T, Onishi J, Nishiyama T. Antimelanogenic activity of mation. Pigment Cell Res 1998;11:355–361.
hydrocumarins in cultured normal human melanocytes by stimula- 114. Stratigos AJ, Dover JS, Arndt KD. Laser treatment of pigmented
ting intracellular glutathione synthesis. Arch Dermatol Res lesions-2000. Arch Dermatol 2000;136:915–921.
2002;294:349–354. 115. Anderson RR. Laser in dermatology-a critical update. J Dermatol
93. Packer L, Witt H, Tritscheler H. Lipoic acid as a biological 2000;27:700–705.
antioxidant. Free Rad Biol Med 1995;19:227–250. 116. Li YT, Yang KC. Comparison of the frequency-doubled Q-switched
94. Saliou C, Kitazawa M, McLaughlin L, Yang JP, Lodge JK, Tetsuka Nd:YAG laser and 35% trichloroacetic acid for the treatment of
T, Iwasaki K, Cillard J, Okamoto T, Packer L. Antioxidants face lentigines. Dermatol Surg 1999;25:202–204.
modulate acute solar ultraviolet radiation-induced NF-Kappa-B 117. Kunachak S, Leelaudomlipi P, Wongwaisayawan S. Dermabrasion:
activation in a human keratinocyte cell line. Free Rad Biol Med a curative treatment for melasma. Aesthetic Plast Surg 2001;25:114–
1999;26:174–183. 117.
95. Ando H, Ryu A, Hashimoto A, Oka M, Ichashi M. Linoleic and 118. Goldman L, Rockwell RJ, Meyer R, Otten R, Wilson RG,
a-linoleic acid lightens ultraviolet-induced hyperpigmentation of the Kitzmiller KW. Laser treatment of tatoos: a preliminary survey of
skin. Arch Dermatol Res 1998;290:375–381. three years’ clinical experience. JAMA 1967;201:841–844.
96. Halaban E, Cheng E, Zhang Y, Moellmann D, Hanlon D, Michalak 119. Anderson RR, Parish JA. Selective photothermolysis. precise
M, Setaluri V, Hebert DN. Aberrant retention of tyrosinase in the microsurgery by selective absorption of pulsed radiation. Science
endoplasmic reticulum mediates accelerated degradation of the 1983;220:524–527.
enzyme and contributes to the dedifferentiated phenotype of amela- 120. Brazzini B, Hautmann G, Ghersetich I, Hercogova J, Lotti T. Laer
notic melanoma cells. Proc Natl Acad Sci USA 1997;94:6210–6215. tissue interaction in epidermal pigmented lesions. J Eur Acad
97. Ando H, Funasaka Y, Oka M, Ohashi A, Furumura M, Matsunaga Dermatol Venereol 2001;15:388–391.
J, Matsunaga N, Hearing VJ, Ichihashi M. Possible involvement of 121. Taylor CR, Anderson RR. Ineffective treatment of refractory
proteolytic degradation of tyrosinase in the regulatory effect of fatty melasma and post-inflammatory hyperpigmentation by Q-switched
acids on melanogenesis. J Lipid Res 1999;40:1312–1316. ruby laser. J Dermatol Surg Oncol 1994;20:592–597.
98. Seiberg M. Keratinocyte-melanocyte interaction during melano- 122. Nouri K, Bowes L, Charter T, Romagnosa R, Spencer J. Combi-
some transfer. Pigment Cell Res 2001;14:236–242. nation treatment of melasma with pulsed CO2 laser followed by
99. Minwalla L, Zhao Y, Cornelius J, Babcock GF, Wickett RR, Le Q-switched alexandrite laser: a pilot study. Dermatol Surg
Poole IC, Boissy RE. Inhibition of melanosome transfer from 1999;25:494–497.
melanocytes to keratinocytes by lectins and neoglycoproteins in an 123. Taylor CR, Anderson RR. Ineffective treatment of refractory
in vitro model system. Pigment Cell Res 2001;14:185–194. melasma and post-inflammatory hyperpigmentation by swithched
100. Sharlow ER, Paine C, Babiarz L, Eisinger M, Shapiro SS, Seiberg ruby laser. J Dermatol Surg Oncol 1994;20:592–597.
M. The protease activated receptor2 upregulates keratinocyte 124. Virador VM, Kobayashi N, Matsunaga J, Hearing VJ. A standard-
phagocytosis. J Cell Sci 2000;113:3093–3101. ized protocol for assessing regulators of pigmentation. Anal
101. Seiberg M, Paine C, Sharlow E, Costanzo M, Andrade-Gordon P, Biochem 1999;270:207–219.
Eisinger M, Shapiro S. The proteasome-activated receptor 2 125. Lei TC, Virador VM, Vieira WD, Hearing VJ. A melanocyte-
regulates pigmentation via keratinocyte–melanocyte interaction. keratinocyte coculture model to assess regulators of pigmentation
Exp Cell Res 2000;254:25–32. in vitro. Anal Biochem 2002;305:260–268.
102. Seiberg M, Paine C, Sharlow E, Costanzo M, Andrade-Gordon P, 126. Hermanns JF, Petit L, Pierard-Franchimont C, Paquet P, Pierard
Eisinger M, Shapiro SS. Inhibition of melanosome transfer results in GE. Assessment of topical hypopigmenting agents on solar lenti-
skin lightening. J Invest Dermatol 2000;115:162–167. gines of Asian wome. Dermatology 2002;204:281–286.

110 Pigment Cell Res. 16, 2003

You might also like