Principles and Practice of Photoprotection
Principles and Practice of Photoprotection
Principles and Practice of Photoprotection
Lim
Editors
Principles and
Practice of
Photoprotection
Principles and Practice of Photoprotection
Steven Q. Wang Henry W. Lim
Editors
v
Acknowledgments
This book is dedicated to our families: Judy and Kevin and Mamie. We thank them
for their patience and sacrifice throughout the course of this project.
vii
Contents
Part I
Part II
ix
x Contents
Part III
Part IV
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 477
Part I
Chapter 1
Clinical and Biological Relevance of Visible
and Infrared Radiation
Key Points
Biologically, visible radiation has been shown to induce erythema, pig-
mentation, free radical production, and DNA damage, while infrared radia-
tion has been shown to induce erythema, thermal pain, photoaging,
cytotoxicity, DNA damage, and oxidative stress.
Visible light has been shown to be an action spectrum in solar urticaria,
chronic actinic dermatitis, and porphyrias; it is used for the treatment of
hyperbilirubinemia. Infrared radiation can cause erythema ab igne and
squamous cell carcinoma.
Lasers with wavelengths in the visible and infrared spectrum can be used
to treat vascular and pigmented lesions, keloids, etc. IPL, LLLT, and PDT
are other light sources with wavelengths in the visible and infrared spec-
trum that are also used to treat numerous dermatologic conditions.
New imaging techniques that use visible and infrared radiation have been
recently developed. The data is promising and could greatly impact the
field of dermatology in the future.
Active research is ongoing on effective photoprotective measures against
visible light and infrared radiation.
1.1 Introduction
wavelengths penetrate deeper than shorter wavelengths. Therefore, blue light, which
is at the shorter end of the wavelength spectrum of visible light, can be used clini-
cally for lesions contained in the epidermis, while red light, which has a longer
wavelength, is useful for thick lesions or to target deeper structures [2, 3].
A variety of molecules can act as chromophores, some examples being amino
acids, lipids, porphyrins, photosensitizing drugs, DNA, hemoglobin, bilirubin,
melanin, and water. When a chromophore absorbs a photon, the chromophore
transitions to an excited state, transiently. The chromophore releases energy, in the
form of heat or light, when it returns to the ground state. The chromophore can then
transfer this energy to another molecule or undergo chemical changes. Multiple
photons are necessary to produce sufficient energy to cause cellular changes, which
then leads to a clinical effect [2, 4]. The amount of absorption depends on the
chromophores in the skin and the wavelength of light used. The energy absorbed is
also known as the energy density, or fluence, and is measured in joules per square
centimeter [5].
Visible light is the portion of the electromagnetic radiation responsible for general
illumination and is visible to the human eye. The wavelength of the visible radia-
tion spectrum is from 400 to 700 nanometers (nm). Each color of light represents
a different wavelength, with blue being at the shorter end of the spectrum and red
at the longer end (Fig. 1.1). See Table 1.1 for more details on specific
wavelengths.
Visible Infrared
400nm 700nm 1mm
Stratum Corneum
Epidermis 0.1 mm
Dermis
3-4 mm
Subcutis
Fig. 1.1 The wavelengths and their corresponding depth of penetration in the skin of each band
within the visible and infrared spectrum
6 K. Lawrence et al.
1.2.1.1 Erythema
1.2.1.2 Pigmentation
Kollias and Baqer used a polychromatic light source with wavelength from 390
to 1700 nm, which consists of the visible spectrum and part of the spectrum of infra-
red radiation. They were able to induce pigmentary changes; however, they did not
notice any erythema or thermal changes, even after 3 h of irradiation with a total
dose of 270 W cm2. IPD was present, and pigmentation that lasted for 10 weeks
was observed when doses greater than 720 J cm2 were used [15]. Rosen et al.
showed that visible radiation up to 470 nm can induce an IPD response; this study
was performed by using a xenon-mercury arc lamp with grating holographic mono-
chromator to select for wavelengths of 334, 365, 405, 435, or 549 nm and spectro-
photometric analysis of skin reflectance [16]. Pathak et al. identified the peak IPD
response to be between 380 and 500 nm using a fixed exposure of 45 J cm2 [17].
Ramasubramaniam et al. used midday sunlight in Bangalore, India, with filters to
determine the cutaneous effects of visible light (greater than 420 nm) and UV light
(less than 400 nm) on pigmentation on Fitzpatrick skin types IV and V. They found
there is not a significant difference in the IPD produced by UV and visible light. They
identified similarly shaped action spectra for IPD and PPD when comparing UV and
visible light. However, UV radiation is much more efficient in producing IPD, and the
PPD response by visible light is much less intense. Since UV and visible light pro-
duced similar action spectra, though, they believe it is likely the same melanin precur-
sor that UV and visible light are interacting with in order to induce these effects [18].
Mahmoud et al., using a light source that emits 98.3 % visible light, also found
that visible radiation induced immediate pigmentation on volunteers with Fitzpatrick
skin types IVVI, with the lowest effective dose being 40 J cm2 [9]. The pigment
was darker as the dose was increased. They noted that the pigment was most intense
in type V skin type volunteers. The pigmentation was still present at 2 weeks, the
end point of their study, even at the lower doses. However, they found that no
pigmentation was induced in skin type II individuals, using the same light source
and doses. The pigmentation induced in this study was more intense and lasted
longer than the pigmentation described by Ramasubramaniam et al. (ref). However,
the light source in Mahmoud et al. was artificial, while natural sunlight was used in
the study done by Ramasubramaniam et al., and the dose used was four times higher
in the study by Mahmoud et al., which could account for the differences [9, 18].
Confocal microscopy used by Mahmoud et al. showed that visible radiation induced
redistribution of melanin from the basal layer to the upper epidermis. Diffuse
reflectance spectroscopy also showed increased melanin content directly related to
the visible radiation dose [9].
Of note, Duteil et al. showed recently that not all wavelengths of visible light
have the same effect on pigmentation. Healthy volunteers of skin types III and IV
were irradiated with wavelengths from both ends of the visible spectrum and the
results compared. Blue-violet light (415 nm) induced pronounced and longlasting
pigmentation (up to 3 months) in both skin types, while red light (630 nm) did not
induce pigmentation [19].
Porges et al. used a solar stimulator to expose individuals with Fitzpatrick skin
types II, III, and IV to light from 385 to 690 nm and observed IPD and DT as well
as erythema. The IPD and erythema faded over 24 h. The DT remained unchanged
8 K. Lawrence et al.
for 10 days. The threshold for PPD (greater than 80 J cm2) was slightly higher than
that for IPD (between 40 and 80 J cm2), while the threshold for DT was higher than
the threshold for IPD. Porges et al. were able to induce pigmentation in lighter skin
types, while Mahmoud et al. were not. These differences could be due to the small
amount of wavelengths outside the visible spectrum UV from 385 to 400 nm in the
study done by Porges et al. or from the limited amount of infrared radiation in the
light source in the study done by Mahmoud et al. [9, 13].
Visible light-induced pigmentation, especially in darker skin types, may be
clinically relevant by potentially playing a role in pigmentation disorders. Melasma
and post-inflammatory hyperpigmentation are much more prominent in darker
skinned individuals. This is consistent with the clinical observation that sunscreens,
which protect against UV but not visible radiation, do not fully protect the
progression of these conditions [6].
It has been well described that UVB is the predominant spectrum causing direct
DNA damage, and indirect DNA damage through ROS is predominantly induced by
UVA. Recently, the effects of visible light on DNA damage were studied. Edstrom
1 Clinical and Biological Relevance of Visible and Infrared Radiation 9
et al. irradiated normal skin with 126 J cm2 visible light which corresponds to about
a half hour outside on a Sweden summer day. An Osram xenon arc lamp with two
filters was used to block out all but the visible spectrum. This was done three times
weekly for 4 weeks while taking intermittent punch biopsies. They found that
visible light increased p53-positive cells as well as proliferation in the epidermis,
although to a lesser extent than UVA1 (340400 nm). p53 normally downregulates
bcl-2, but interestingly they found a slight increase in bcl-2 in the epidermis, which
could potentially mean the p53 gene was mutated [26].
Kielbassa et al. used a xenon arc lamp with grid monochromator and/or cutoff filters
(to make monochromatic radiation) to study the spectrum in which dimers and oxida-
tive DNA modification occur in hamster cells. From UVA1 range into the visible light
spectrum, oxidative DNA damage was observed, with a peak between 400 and 450 nm
[27]. Hoffmann-Dorr et al. analyzed the effect of visible light on direct and indirect
DNA damage on melanoma cells and human skin fibroblasts. Visible light induces
ROS, which indirectly damages DNA. They concluded that the oxidative damage from
400 to 500 nm accounted for 10 % of the total indirect damage that occurs with sunlight
exposure [28]. Liebel et al. showed that visible light radiation induced production of
ROS, proinflammatory cytokines, and MMP-1 expression. However, neither thymine
dimers are produced from visible light radiation nor TNF-alpha expression induced
[25]. Now that visible light is being used more clinically, in lasers and photodynamic
therapy (PDT), the long-term effects on DNA are becoming clinically relevant.
1.2.2.3 Porphyrias
1.2.2.4 Hyperbilirubinemia
Acne lesions have been reported to decrease after exposure to blue, red, violet, or
UV light. Some individuals report an improvement in their acne after sun exposure.
The exact mechanism of action has not been completely elucidated; however, it is
believed that the light works through anti-inflammatory and antibacterial
mechanisms. Furthermore, it is known that porphyrins are produced by
Propionibacterium acnes; therefore, exposure to Soret band results in the destruction
of the bacteria. In fact, this is the rational for the use of photodynamic therapy in the
treatment of acne vulgaris [5, 4648].
Erythema
IR can cause erythema, typically lasting less than 1 h, and is believed to be due to
vasodilation secondary to a thermal effect. By 24 h, no erythema or pigmentation is
observed [6]. The erythema observed has been used to determine standardized ways
to measure IR doses. The minimal response dose and minimal heating dose have
been described [50, 51].
Thermal Pain
Photoaging
Photoaging is a term used to describe the characteristic changes that occur to the
skin after chronic exposure to sunlight, originally believed to be solely due to chronic
UV radiation. Some common symptoms of photoaging include wrinkles, telangiec-
tasias, solar lentigines, laxity, and a change of the texture to leathery. IR was first
found to contribute to photoaging when it was shown in albino guinea pigs that UV
plus IR exposure induced more photoaging than just UV radiation alone [52].
There are multiple mechanisms by which IR, mostly IR-A (7601400 nm), is
suggested to induce photoaging. Increased expression of MMP-1 is one of these
mechanisms, which leads to increased degradation of collagen [53]. It has also been
proposed that IR disturbs the electron flow in the mitochondria, which results in
insufficient energy production in dermal fibroblasts. Different signaling pathways
are then triggered, and alterations in functional and structural aspects of the skin
occur [54]. Additionally, IR has been shown to cause decreased antioxidant enzyme
activity, to stimulate angiogenesis, and to increase the number of mast cells, all of
which have been found associated with photoaging [55, 56].
12 K. Lawrence et al.
IR has not been found to induce DNA damage alone [6]. IR appears to have a pro-
tective effect on UV-induced cytotoxicity and DNA damage. Menezes et al. found a
longlasting partial protection from UVA- and UVB-induced cytotoxic damage after
prior radiation with IR light [57]. Jantschitsch et al. irradiated in vivo mouse skin
with IR-A prior to UVB radiation and found decreased UVB-induced apoptosis and
DNA damage compared to irradiation with UVB alone. Decreased UVB-induced
DNA damage was seen in in vitro human skin fibroblasts after IR radiation [58].
Markers of Damage
Due to acute and chronic adverse effects described above that can occur from IR
exposure, indicators are needed in order to better understand the tissue threshold for
damage. The expression of matrix metalloproteinase (MMP)-1 has been proposed a
useful marker of early IR damage at the cellular level. MMP-1 expression increases
in response to over-warming of tissue, UV overexposure, or mechanical stress.
Other markers that have been explored include heat shock proteins, ROS, and
apoptosis-related proteins. However, results of these investigations are contradictory
in many cases, so specific conclusions cannot be elucidated at this time [49].
Oxidative Stress
IR has been shown to induce oxidative stress both by increasing formation of free radi-
cals and decreasing the antioxidant content in human skin. Zastrow et al. found that
the amount of excess free radical formation was not only dependent on the dose of
radiation but also on the skin temperature increase due to IR radiation (7601600 nm).
Using an in vitro human fibroblast model, Jung et al. showed that IR radiation at 37 C
did not induce excess free radical production, while at 39 C or higher, production of
excess free radicals was observed. Now that the detrimental effects of IR radiation
have been well described, it is clear that protection from IR radiation is necessary and
important and will be addressed further in the section on sunscreen [6, 53, 59, 60].
heaters, hot water bottles, and heated reclining chairs. Treatment is withdrawal of
the heat source, and if done, patients have a good prognosis [61].
Acne vulgaris has recently been shown to be successfully treated with light in the
visible range, as discussed above, but also with light sources in the infrared spectrum.
Diode lasers have been used to reduce acne lesions. The 810 and 1450 nm diode
lasers have been used successfully. The diode lasers work by inducing short-term
thermal alteration of sebaceous glands. When the 810 nm diode laser was
investigated, it was done following the administration of indocyanine green
chromophore. The indocyanine green concentrated in the sebaceous glands and was
subsequently targeted by the diode laser. The data for acne treatment with diode
lasers is promising; however as with acne treatment with visible light sources, more
research is necessary to elucidate the long-term efficacy and cost-effectiveness of
these treatment options [5, 62, 63].
Introduction to Lasers
Lasers can be classified by the wavelength they emit, as this is a very important
property of the laser. Examples of lasers that emit wavelengths in the visible light
spectrum are argon, KTP, copper bromide, APTD, krypton, PDL, ruby, and
alexandrite lasers. Table 1.2 lists some of the common lasers with wavelength in the
visible light spectrum and their respective wavelengths [5, 64].
There are many uses for lasers in dermatology. Some examples of what lasers
emitting wavelengths in the visible spectrum are used for include vascular lesions,
pigmented lesions, vitiligo, tattoo removal, hair removal, and keloids.
Common vascular lesions that have been successfully treated with lasers are port-
wine stains, hemangiomas, and telangiectasia. Vascular lesions contain oxygenated
hemoglobin, which is the molecule the laser targets for destruction when treating
vascular lesions. Oxyhemoglobin absorbs light strongly at wavelengths of 418, 542,
and 577 nm. PDL was specifically designed to treat vascular lesions based on the
14 K. Lawrence et al.
selective photothermolysis theory and is currently the first-line treatment for vascu-
lar lesions [5, 6466].
The Nd:YAG laser has also been used successfully for a variety of vascular
lesions such as port-wine stains, hemangiomas, and facial telangiectasia. Also, the
Nd:YAG and 800 nm diode lasers have been used successfully for varicose and spi-
der veins; however, sclerotherapy remains the gold standard for these lesions [5, 67].
Melanin has a broad absorption spectrum, from 504 to 750 nm. The wavelengths at
the shorter end of the range are more effective at removing pigmented lesions.
Longer wavelength lasers are useful for lesions with deeper pigment due to the
increased tissue penetration. The response of the tattoo to specific lasers is very
dependent on the color, depth, and nature of the tattoo pigment [5, 64, 68].
The pulsed lasers are also successful in removing tattoo pigment. The pigment is
altered by the lasers and then subsequently removed by tissue macrophages. For
black pigment, the Q-switched (QS) ruby, QS alexandrite, or QS Nd:YAG lasers are
most effective because black pigment absorbs throughout the red and infrared
spectrum. Blue and green pigments absorb best in the 600800 nm range and
therefore are best removed with ruby or alexandrite lasers. Yellow, orange, and red
pigments are removed most effectively with green light, making the 510 nm PDL or
532 nm QS Nd:YAG laser the best options for these pigments [5, 64].
The Nd:YAG laser has been found to be useful for pigmented lesions when the
pigment resides deeper in the dermis. Long-pulsed diode and long-pulsed Nd:YAG
lasers have been especially effective at eradicating pigmented lesions with terminal
hair growth, such as congenital melanocytic nevi and Beckers nevi [5, 64].
1 Clinical and Biological Relevance of Visible and Infrared Radiation 15
Light with wavelength between 600 and 1200 nm is best for hair removal because
the light can penetrate to the appropriate depth in the dermis and is able to target the
melanin in the hair shaft, hair follicle epithelium, and heavily pigmented matrix.
The energy is absorbed by the melanin-rich matrix and hair shaft, which then under-
goes a photothermal reaction and destroys the surrounding hair follicle [5, 64, 69].
Lasers currently approved for hair reduction include the long-pulsed ruby, long-
pulsed alexandrite, pulsed diode, and long-pulsed Nd:YAG [5, 64, 70]. Of note,
intense pulse light (IPL) with wavelength from 590 to 1200 nm can also be used for
hair removal and will be discussed in further detail below.
PDL has recently been used for the treatment of keloids and hypertrophic scars.
PDL has been shown to decrease erythema, increase pliability, and improve texture,
bulk, and dysesthesias [5, 64, 7173].
Ablative Lasers
Ablative lasers are used primarily for cutaneous facial resurfacing for severely
photodamaged skin, photoinduced facial rhytides, dyschromias, and atrophic scars.
High-energy, pulsed, and scanned CO2 and erbium:YAG lasers are the main ablative
lasers in use today, while the CO2 laser is currently the gold standard for facial
rejuvenation [5].
The short-pulsed erbium:YAG laser, 2940 nm, was designed to have the beneficial
effects of the CO2 laser while limiting the unwanted side effects. The erbium:YAG
has milder improvement than the CO2 laser but with also milder side effects and
faster recovery time [5].
Additionally, there are numerous other uses for the CO2 laser, which includes
removing a variety of epidermal and dermal lesions, treating premalignant and
malignant lesions, and excisional and incisional operations [5].
Low-level light therapy (LLLT) uses low-power light sources. LLLT can be
performed with either coherent light sources (lasers) or noncoherent light sources
(light-emitting diodes (LEDs)). LLLT is lower intensity and causes lower
temperature changes and less discomfort than other types of laser, while still being
effective [24].
LLLT works by absorption of red and near-infrared light by the protein
components of the respiratory chain in the mitochondria, mostly cytochrome c
oxidase. Absorption leads to dissociation of inhibitory nitric oxide from cytochrome
c oxidase and then increased enzyme activity, electron transport, and ATP production.
LLLT has also been shown to increase expression of genes related to cellular
migration and proliferation and also alters expression of growth factors and
cytokines [24].
Red LED LLLT has also been found to inhibit fibroblast proliferation in vitro
without affecting viability. Therefore, red LED LLLT could be a possible treatment
for scars or proliferative disorders in the future [75].
The helium-neon laser is a type of LLLT with wavelength of 632.8 nm. The
helium-neon laser has recently been shown to be another therapeutic option for
vitiligo, specifically segmental vitiligo. The mechanism by which this works is by
inducing melanocyte proliferation through the interaction with type IV collagen via
mitochondria-related pathways [76, 77].
The current uses of LLLT within the IR spectrum are to stimulate wound healing
and hair growth and for the treatment of herpes simplex. It has been shown that LLLT
stimulates wound healing by promoting contraction through the induction of fibroblast
to myofibroblast transition [78]. Recently, LLLT using a 1072 nm LED light source
has been found to be a potential treatment for herpes simplex labialis. Significantly
reduced healing times were experienced in patients treated with LLLT [79].
Photodynamic therapy (PDT) is a common way visible light is used clinically. PDT
is approved for the treatment of actinic keratosis in the United States; however, there
are many off-label uses which continue to expand [80]. PDT requires a
photosensitizer, a light source, and oxygen [81, 82].
1 Clinical and Biological Relevance of Visible and Infrared Radiation 17
Light Source
Any light source can be used for PDT, as long as the wavelength of light coincides
with the absorption spectrum of the photosensitizer, and the penetration depth of the
light is equal to the depth of the target cells or target tissue. Protoporphyrin IX has
important absorption peaks in the red and blue wavelength regions, from 404 to
420 nm and at 635 nm. Therefore, continuous red and blue light are very commonly
used in PDT [81].
Aminolevulinic acid (ALA) is only approved in North America for the treatment of
hypertrophic actinic keratosis on the face and scalp in combination with blue light.
Methyl aminolevulinate (MAL) is approved for non-hyperkeratotic actinic keratosis
of the face and scalp in the United States [81].
There are numerous off-label uses of PDT. PDT has been used to treat noninva-
sive, nonmelanoma skin cancers (NMSCs), mycosis fungoides, Kaposis sarcoma,
extramammary Pagets disease, cutaneous B-cell lymphoma, vascular malformations,
acne vulgaris, rosacea, hidradenitis suppurativa, morphea, actinic cheilitis,
cutaneous warts, condyloma acuminata, epidermodysplasia verruciformis,
molluscum contagiosum, herpes simplex virus, onychomycosis, cutaneous
leishmaniasis, erythrasma (Corynebacterium minutissimum infection), keloids, and
hypertrophic scars [81]. PDT has also been used for photorejuvenation.
causing the ZnO and TiO2 to appear white. The particle size determines the
absorption range. ZnO and TiO2 used in sunscreens are micronized (particle size of
less than 100 nm in diameter) because they are then less visible on the skin and
more cosmetically acceptable. Ferrous oxide, which is pigmented and opaque, has
recently been used and found to be effective in offering sun protection in the visible
light spectrum [84].
References
1. Brenner M, Hearing VJ (2008) The protective role of melanin against UV damage in human
skin. Photochem Photobiol 84(3):539549
2. Baron ED, Suggs AK (2014) Introduction to photobiology. Dermatol Clin 32(3):255266, vii
3. Barolet D (2008) Light-emitting diodes (LEDs) in dermatology. Semin Cutan Med Surg
27(4):227238
4. Rabe JH et al (2006) Photoaging: mechanisms and repair. J Am Acad Dermatol 55(1):119
5. Tanzi EL, Lupton JR, Alster TS (2003) Lasers in dermatology: four decades of progress. J Am
Acad Dermatol 49(1):131; quiz 3134
6. Sklar LR et al (2013) Effects of ultraviolet radiation, visible light, and infrared radiation on
erythema and pigmentation: a review. Photochem Photobiol Sci 12(1):5464
7. Honigsmann H (2002) Erythema and pigmentation. Photodermatol Photoimmunol Photomed
18(2):7581
1 Clinical and Biological Relevance of Visible and Infrared Radiation 19
8. Schmalwieser AW, Wallisch S, Diffey B (2012) A library of action spectra for erythema and
pigmentation. Photochem Photobiol Sci 11(2):251268
9. Mahmoud BH et al (2010) Impact of long-wavelength UVA and visible light on melanocom-
petent skin. J Invest Dermatol 130(8):20922097
10. Parrish JA, Zaynoun S, Anderson RR (1981) Cumulative effects of repeated subthreshold
doses of ultraviolet radiation. J Invest Dermatol 76(5):356358
11. Harmful effects of ultraviolet radiation. Council on Scientific Affairs (1989) JAMA
262(3):380384
12. Rottier PB, Van Der Leun JC (1960) Hyperaemia of the deeper cutaneous vessels after
irradiation of human skin with large doses of ultra-violet and visible light. Br J Dermatol 72:
256260
13. Porges SB, Kaidbey KH, Grove GL (1988) Quantification of visible light-induced melanogen-
esis in human skin. Photodermatol 5(5):197200
14. Lim HW, Honigsmann H, Hawk JLM (2007) Photodermatology. Informa Healthcare USA,
Inc., New York, pp 7589
15. Kollias N, Baqer A (1984) An experimental study of the changes in pigmentation in human
skin in vivo with visible and near infrared light. Photochem Photobiol 39(5):651659
16. Rosen CF et al (1990) Immediate pigment darkening: visual and reflectance spectrophotomet-
ric analysis of action spectrum. Photochem Photobiol 51(5):583588
17. Pathak MA, Riley FC, Fitzpatrick TB (1962) Melanogenesis in human skin following expo-
sure to long-wave ultraviolet and visible light. J Invest Dermatol 39:435443
18. Ramasubramaniam R et al (2011) Are there mechanistic differences between ultraviolet and
visible radiation induced skin pigmentation? Photochem Photobiol Sci 10(12):18871893
19. Duteil L et al (2014) Differences in visible light-induced pigmentation according to wave-
lengths: a clinical and histological study in comparison with UVB exposure. Pigment Cell
Melanoma Res 27(5):822826
20. Loertzer H et al (2006) Formation of ascorbate radicals as a measure of oxidative stress: an
in vitro electron spin resonance-study using 2,2-Azobis (2-amidinopropane) dihydrochloride
as a radical generator. Transplant Proc 38(3):674678
21. Halliwell B, Whiteman M (2004) Measuring reactive species and oxidative damage in vivo
and in cell culture: how should you do it and what do the results mean? Br J Pharmacol
142(2):231255
22. Buettner GR, Jurkiewicz BA (1993) Ascorbate free radical as a marker of oxidative stress: an
EPR study. Free Radic Biol Med 14(1):4955
23. Haywood R (2006) Relevance of sunscreen application method, visible light and sunlight
intensity to free-radical protection: a study of ex vivo human skin. Photochem Photobiol
82(4):11231131
24. Mahmoud BH et al (2008) Effects of visible light on the skin. Photochem Photobiol
84(2):450462
25. Liebel F et al (2012) Irradiation of skin with visible light induces reactive oxygen species and
matrix-degrading enzymes. J Invest Dermatol 132(7):19011907
26. Edstrom DW, Porwit A, Ros AM (2001) Effects on human skin of repetitive ultraviolet-A1
(UVA1) irradiation and visible light. Photodermatol Photoimmunol Photomed 17(2):6670
27. Kielbassa C, Roza L, Epe B (1997) Wavelength dependence of oxidative DNA damage induced
by UV and visible light. Carcinogenesis 18(4):811816
28. Hoffmann-Dorr S et al (2005) Visible light (>395 nm) causes micronuclei formation in mam-
malian cells without generation of cyclobutane pyrimidine dimers. Mutat Res
572(12):142149
29. Champion RH (1988) Urticaria: then and now. Br J Dermatol 119(4):427436
30. Stratigos AJ et al (2003) Spectrum of idiopathic photodermatoses in a Mediterranean country.
Int J Dermatol 42(6):449454
31. Uetsu N et al (2000) The clinical and photobiological characteristics of solar urticaria in 40
patients. Br J Dermatol 142(1):3238
20 K. Lawrence et al.
32. Ryckaert S, Roelandts R (1998) Solar urticaria. A report of 25 cases and difficulties in photo-
testing. Arch Dermatol 134(1):7174
33. Nitiyarom R, Wongpraparut C (2014) Hydroa vaccin iforme and solar urticaria. Dermatol Clin
32(3):345353, viii
34. Hasei K, Ichihashi M (1982) Solar urticaria. Determinations of action and inhibition spectra.
Arch Dermatol 118(5):346350
35. Leenutaphong V et al (1987) Plasmapheresis in solar urticaria. Photodermatol 4(6):308309
36. Dawe RS, Ferguson J (2003) Diagnosis and treatment of chronic actinic dermatitis. Dermatol
Ther 16(1):4551
37. Lim HW et al (1994) Chronic actinic dermatitis. An analysis of 51 patients evaluated in the
United States and Japan. Arch Dermatol 130(10):12841289
38. Paek SY, Lim HW (2014) Chronic actinic dermatitis. Dermatol Clin 32(3):355361, viiiix
39. Schulenburg-Brand D et al (2014) The cutaneous porphyrias. Dermatol Clin 32(3):369384, ix
40. Ennever JF (1990) Blue light, green light, white light, more light: treatment of neonatal jaun-
dice. Clin Perinatol 17(2):467481
41. American Academy of Pediatrics Subcommittee on Hyperbilirubinemia (2004) Management
of hyperbilirubinemia in the newborn infant 35 or more weeks of gestation. Pediatrics 114(1):
297316
42. Maisels MJ, Kring EA, DeRidder J (2007) Randomized controlled trial of light-emitting diode
phototherapy. J Perinatol 27(9):565567
43. Onishi S et al (1986) Metabolism of bilirubin and its photoisomers in newborn infants during
phototherapy. J Biochem 100(3):789795
44. Lightner DA, Linnane WP 3rd, Ahlfors CE (1984) Bilirubin photooxidation products in the
urine of jaundiced neonates receiving phototherapy. Pediatr Res 18(8):696700
45. Jori G, Rossi E, Rubaltelli FF (1980) Phototherapy-induced covalent binding of bilirubin to
serum albumin. Pediatr Res 14(12):13631366
46. Papageorgiou P, Katsambas A, Chu A (2000) Phototherapy with blue (415 nm) and red (660
nm) light in the treatment of acne vulgaris. Br J Dermatol 142(5):973978
47. Cunliffe WJ, Goulden V (2000) Phototherapy and acne vulgaris. Br J Dermatol 142(5):
855856
48. Rkein AM, Ozog DM (2014) Photodynamic therapy. Dermatol Clin 32(3):415425, x
49. Piazena H, Kelleher DK (2010) Effects of infrared-A irradiation on skin: discrepancies in
published data highlight the need for an exact consideration of physical and photobiological
laws and appropriate experimental settings. Photochem Photobiol 86(3):687705
50. Pujol JA, Lecha M (1992) Photoprotection in the infrared radiation range. Photodermatol
Photoimmunol Photomed 9(6):275278
51. Lee HS et al (2006) Minimal heating dose: a novel biological unit to measure infrared irradia-
tion. Photodermatol Photoimmunol Photomed 22(3):148152
52. Kligman LH (1982) Intensification of ultraviolet-induced dermal damage by infrared radia-
tion. Arch Dermatol Res 272(34):229238
53. Schroeder P et al (2008) Infrared radiation-induced matrix metalloproteinase in human skin:
implications for protection. J Invest Dermatol 128(10):24912497
54. Krutmann J, Schroeder P (2009) Role of mitochondria in photoaging of human skin: the defec-
tive powerhouse model. J Investig Dermatol Symp Proc 14(1):4449
55. Krutmann J, Morita A, Chung JH (2012) Sun exposure: what molecular photodermatology
tells us about its good and bad sides. J Invest Dermatol 132(3 Pt 2):976984
56. Shin MH et al (2012) Chronic heat treatment causes skin wrinkle formation and oxidative
damage in hairless mice. Mech Ageing Dev 133(23):9298
57. Menezes S et al (1998) Non-coherent near infrared radiation protects normal human dermal
fibroblasts from solar ultraviolet toxicity. J Invest Dermatol 111(4):629633
58. Jantschitsch C et al (2009) Infrared radiation confers resistance to UV-induced apoptosis via
reduction of DNA damage and upregulation of antiapoptotic proteins. J Invest Dermatol
129(5):12711279
59. Zastrow L et al (2009) The missing link light-induced (2801,600 nm) free radical formation
in human skin. Skin Pharmacol Physiol 22(1):3144
1 Clinical and Biological Relevance of Visible and Infrared Radiation 21
60. Jung T et al (2010) Effects of water-filtered infrared A irradiation on human fibroblasts. Free
Radic Biol Med 48(1):153160
61. Miller K et al (2011) Erythema ab igne. Dermatol Online J 17(10):28
62. Paithankar DY et al (2002) Acne treatment with a 1,450 nm wavelength laser and cryogen
spray cooling. Lasers Surg Med 31(2):106114
63. Lloyd JR, Mirkov M (2002) Selective photothermolysis of the sebaceous glands for acne treat-
ment. Lasers Surg Med 31(2):115120
64. Hruza GJ, Avram MM (2013) Lasers and lights: procedures in cosmetic dermatology, 3rd edn.
Elsevier
65. Anderson RR, Parrish JA (1983) Selective photothermolysis: precise microsurgery by selective
absorption of pulsed radiation. Science 220(4596):524527
66. Levine VJ, Geronemus RG (1995) Adverse effects associated with the 577- and 585-nanome-
ter pulsed dye laser in the treatment of cutaneous vascular lesions: a study of 500 patients.
J Am Acad Dermatol 32(4):613617
67. Weiss RA, Weiss MA (1999) Early clinical results with a multiple synchronized pulse 1064
NM laser for leg telangiectasias and reticular veins. Dermatol Surg 25(5):399402
68. Margolis RJ et al (1989) Visible action spectrum for melanin-specific selective photothermoly-
sis. Lasers Surg Med 9(4):389397
69. Lask G et al (1997) Laser-assisted hair removal by selective photothermolysis. Preliminary
results. Dermatol Surg 23(9):737739
70. McDaniel DH et al (1999) Laser hair removal: a review and report on the use of the long-
pulsed alexandrite laser for hair reduction of the upper lip, leg, back, and bikini region.
Dermatol Surg 25(6):425430
71. Alster TS, Williams CM (1995) Treatment of keloid sternotomy scars with 585 nm flashlamp-
pumped pulsed-dye laser. Lancet 345(8959):11981200
72. Lupton JR, Alster TS (2002) Laser scar revision. Dermatol Clin 20(1):5565
73. Manuskiatti W, Fitzpatrick RE (2002) Treatment response of keloidal and hypertrophic ster-
notomy scars: comparison among intralesional corticosteroid, 5-fluorouracil, and 585-nm
flashlamp-pumped pulsed-dye laser treatments. Arch Dermatol 138(9):11491155
74. Sadick NS et al (2000) Long-term photoepilation using a broad-spectrum intense pulsed light
source. Arch Dermatol 136(11):13361340
75. Lev-Tov H et al (2013) Inhibition of fibroblast proliferation in vitro using red light-emitting
diodes. Dermatol Surg 39(8):11671170
76. Lan CC et al (2009) Low-energy helium-neon laser induces melanocyte proliferation via inter-
action with type IV collagen: visible light as a therapeutic option for vitiligo. Br J Dermatol
161(2):273280
77. Yu HS et al (2003) Helium-neon laser irradiation stimulates migration and proliferation in
melanocytes and induces repigmentation in segmental-type vitiligo. J Invest Dermatol
120(1):5664
78. Demidova-Rice TN et al (2007) Low-level light stimulates excisional wound healing in mice.
Lasers Surg Med 39(9):706715
79. Dougal G, Lee SY (2013) Evaluation of the efficacy of low-level light therapy using 1072 nm
infrared light for the treatment of herpes simplex labialis. Clin Exp Dermatol 38(7):713718
80. Calzavara-Pinton P et al (2010) A critical reappraisal of off-label indications for topical photo-
dynamic therapy with aminolevulinic acid and methylaminolevulinate. Rev Recent Clin Trials
5(2):112116
81. Wan MT, Lin JY (2014) Current evidence and applications of photodynamic therapy in derma-
tology. Clin Cosmet Investig Dermatol 7:145163
82. Dolmans DE, Fukumura D, Jain RK (2003) Photodynamic therapy for cancer. Nat Rev Cancer
3(5):380387
83. Meinke MC et al (2013) Radical protection by differently composed creams in the UV/VIS
and IR spectral ranges. Photochem Photobiol 89(5):10791084
84. Castanedo-Cazares JP et al (2014) Near-visible light and UV photoprotection in the treatment
of melasma: a double-blind randomized trial. Photodermatol Photoimmunol Photomed
30(1):3542
22 K. Lawrence et al.
85. Rajadhyaksha M, Anderson RR, Webb RH (1999) Video-rate confocal scanning laser micro-
scope for imaging human tissues in vivo. Appl Opt 38(10):21052115
86. Rajadhyaksha M et al (1999) In vivo confocal scanning laser microscopy of human skin II:
advances in instrumentation and comparison with histology. J Invest Dermatol 113(3):
293303
87. Selkin B et al (2001) In vivo confocal microscopy in dermatology. Dermatol Clin 19(2):369
377, ixx
88. Welzel J (2001) Optical coherence tomography in dermatology: a review. Skin Res Technol
7(1):19
89. Welzel J et al (1997) Optical coherence tomography of the human skin. J Am Acad Dermatol
37(6):958963
90. van der Vorst JR et al (2013) Dose optimization for near-infrared fluorescence sentinel lymph
node mapping in patients with melanoma. Br J Dermatol 168(1):9398
91. Padilla-Medina JA et al (2014) Assessment technique for acne treatments based on statistical
parameters of skin thermal images. J Biomed Opt 19(4):046019
Chapter 2
Photoprotection and Skin Cancer Prevention
Key Points
Ultraviolet radiation is a major risk factor for the development of skin can-
cer, the most common form of cancer in the United States.
Ultraviolet radiation causes both direct and indirect damages to DNA,
leading to mutations and malignant transformation if the damage is not
repaired.
Skin cancer can be prevented by reducing intentional exposure to ultravio-
let radiation and using photoprotective strategies, including sunscreens.
Daily sunscreen application protects against the development of actinic
keratoses, squamous cell carcinoma, nevi formation, and melanoma.
2.1 Introduction
Environmental exposures to both natural and man-made substances are a major risk
factor for the development of many types of cancers. Skin serves as the interface
between the body and the environment and is frequently exposed to potentially haz-
ardous environmental elements. Viral and bacterial infections, smoking, radiother-
apy, immunosuppressant drugs, artificial ultraviolet sources for phototherapy and
tanning, and chemical carcinogens have all been shown to predispose individuals to
skin cancers [1].
Ultraviolet radiation (UVR) from sunlight is a major risk factor for melanoma and
non-melanoma skin cancer (NMSC) [2]. Epidermal cells accumulate UVR-induced
DNA damage which can lead to DNA base mutations and malignant transformation.
This chapter discusses the biologic impact of UVR on the skin and its role in the
development of both NMSC and melanoma. In addition, the role of photoprotection
to prevent cutaneous malignancies is reviewed.
2.2 UV Radiation
Solar radiation is divided into ultraviolet (200400 nm), visible light (400700 nm),
and infrared radiation (>700 nm) [3]. UVR plays a major role in the development of
photoaging and skin cancer [4]. Due to inherent differences in biologic effects, the
UV spectrum is further subdivided into UVC (200290 nm), UVB (290320 nm),
UVA2 (320340 nm), and UVA1 (340400 nm) [4, 5]. The subdivision of the UVA
range is due to a change in the slope of the action spectrum for erythema occurring
near 340 nm, with UVA2 having more erythemogenic activity than UVA1.
The different types of UVR vary in their intensity at the Earths surface and their
effects on human skin. Nearly all of the UVC radiation from the sun is absorbed by
the ozone layer, effectively negating its effects on the human body [6]. Approximately
95 % of the solar radiation reaching the Earths surface is UVA, with the remaining
5 % UVB [7]. The intensity of UVB has been shown to increase between 4 and
10 % with every 1000 ft of elevation and by approximately 3 % for every degree of
latitude as approaching the equator [8]. UVB, being a shorter wavelength compared
to UVA, is only capable of penetrating down to the basal layer of the epidermis and
superficial dermis, while UVA can penetrate deeper into the reticular dermis [9].
Compared to UVA, the erythemogenic potential of UVB is 1000 times greater [10].
UVA is generally more closely associated with tanning and photoaging changes
such as loss of skin elasticity and wrinkling, although UVB can also produce the
same effects [4, 11]. Both UVB and UVA have been implicated in the development
of skin cancers.
synthesis [14]. Failure to repair these defects can lead to a collapse in the replication
fork at the damaged site, causing a DNA double-strand breaks and ultimately cell
death. Furthermore, the presence of UV-induced photoproducts can interfere with
base pairing during DNA replication, leading to mutations.
Although normal cells maintain a high repair fidelity, errors in repair can lead to
cytosine (C) thymine (T) base substitution at dipyrimidine sites and CC TT
tandem base substitutions [14, 15]. These are known as UV signature mutations,
indicating damages from past UVR exposure [15]. While these mutations were once
known as UVB signature mutations, further studies have demonstrated that a high
proportion of C T transitions also occur with UVA-induced damage, but at a
lower frequency (65 % for UVA vs. 85 % for UVB) [16, 17]. The rates of repair for
6-4PP and CPD photoproducts are different. Nearly 90 % of the 6-4PP lesions are
repaired at 3 h post-UV exposure [12, 18]. In contrast, only 10 % of CPD lesions are
repaired at 3 h and 50 % at 24 h after exposure [18]. Repair capacity diminishes with
age, and there is a cumulative loss of 25 % in repair ability between the ages of 20
and 60 years; this difference may account for the increased risk of skin cancer that
begins in middle age [19]. Individuals with defective nuclear excision repair
pathways, such as patients with xeroderma pigmentosum, are exceptionally
vulnerable to UV-induced cutaneous malignancies.
UVA indirectly damages DNA via a free radical-mediated pathway [12]. UVA
reacts with chromophores and photosensitizers, such as porphyrins, cytochromes,
heme, riboflavin, and tryptophan, which generate free radicals [2024]. In addition,
UVA reacts with oxygen species and induces the formation of reactive oxygen
species (ROS) [20]. Within 20 min of UVA exposure, expression of NADPH oxidase
in human keratinocytes increases by 2-fold [25]. NADPH oxidase converts oxygen
molecules to superoxide anions, which are ultimately converted to ROS such as
hydrogen peroxide, superoxide anion (O2), peroxide (O22), hydroxyl radical
(OH), hydroxyl ion (OH), and singlet oxygen (1O2) [26]. These short-lived free
radicals damage DNA in a myriad of ways, including cross-linking DNA to proteins
and forming single-strand and double-strand breaks [12, 27]. It is important to note
that UVB can also trigger oxidative damage [20, 28, 29].
Aside from these forms of nonspecific DNA damage, UVA-induced oxidation
leads to specific DNA base mutations. The molecule 8-hydroxyguanine (8OH-G) is
a mutagenic base that results from ROS interaction with guanine [30]. 8OH-G
is preferentially generated with UV wavelengths greater than 350 nm and hence is
thought to be UVA signature mutation [28]. This particular lesion has been shown
to create G:C T:A transversions in DNA [31]. In addition, UVA generates CPD
mutations at nearly five times that of 8OH-G mutations [32]. However, compared to
CPD mutations from UVB, the overall number of UVA-generated DNA photoprod-
ucts is significantly lower [20].
26 B.P. Hibler et al.
Upon DNA damage, cells can either repair the mutation or, if the damage is beyond
repair, target the cell for apoptosis. The p53 tumor suppressor gene plays a major
role in regulation of cell cycle checkpoint activity, DNA repair, and apoptosis.
However, if the p53 gene becomes mutated, these protective cellular mechanisms
may fail, leading to carcinogenesis. Clones of cells with UV signature mutations
(e.g., C T and CC TT transitions) in the p53 tumor suppressor gene have been
found in sun-exposed skin, actinic keratoses, squamous cell carcinoma, basal cell
carcinoma, and melanoma, supporting its role in photocarcinogenesis [33, 34].
Under normal circumstances, p53 responds to DNA damage by blocking the
progression of the cell cycle. Immediately after UV irradiation, p53 transcription is
upregulated and DNA damage leads to the alteration of the p53 protein, allowing for
phosphorylation by other protein kinases [35]. Elevated levels of p53 that occur
after UV exposure lead to induction of p21 (also known as WAF1 or CIP1), which
is responsible for cell cycle arrest and inhibiting apoptosis [36]. The p21 protein is
capable of competitively forming a complex with cyclin-dependent kinase (CDK),
blocking its interaction with cyclin and effectively inhibiting cell entry to the S
phase where DNA replication takes place [37, 38]. Cell cycle arrest may also occur
at checkpoints during S phase or after G2 (before mitosis) to ensure DNA fidelity
[39]. By inhibiting progression of the cell cycle, the cell is providing itself time to
repair, to prevent passage of mutated DNA onto daughter cells.
Upon cell cycle arrest, DNA repair mechanisms are activated to correct the
UV-induced lesions. Two major mechanisms for DNA repair include base excision
repair (BER) and nucleotide excision repair (NER). BER is used to remove dam-
aged bases, such as the oxidized form of guanine (8OH-G) [40]. In this pathway,
DNA glycosylases remove specific damaged or inappropriate bases forming a
single-strand break, which is then repaired with small fragments of 112 nucleo-
tides [41]. NER is used to repair a variety of bulky DNA damages, including CPDs
and 6-4PPs, that commonly result from UVB exposure [42]. NER involves single-
strand incisions flanking the lesion, followed by DNA repair synthesis and ligation.
As mentioned earlier, 6-4PPs are repaired much more quickly than CPDs. This is
thought to be because 6-4PPs are more destabilizing and cause a greater degree of
unwinding in the DNA helix than CPDs [4, 43, 44]. Repair of these UV-specific
CPD and 6-4PP lesions significantly decreases the overall apoptotic response [45].
If the DNA damage is beyond repair, apoptotic pathways are activated to prevent
passage of daughter cells carrying those mutations. The molecule p53 can induce apop-
tosis through two major pathways, either the intrinsic mitochondrial pathway or the
extrinsic death receptor pathway [46]. In the mitochondrial pathway, p53 upregulates
pro-apoptotic genes, such as Bax and Bak, or p53 represses transcription of antiapop-
totic genes, such as survivin. Furthermore, p53 induces caspase activation and apopto-
sis [46, 47]. To a lesser extent, p53 activates the death receptor pathway by promoting
fas transcription and its cell-surface expression [48, 49]. Additionally, p53 induces
DDB2 (damaged-DNA binding protein 2) which promotes programmed cell death by
2 Photoprotection and Skin Cancer Prevention 27
Skin cancer is the most common form of cancer in the United States [5860]. Nearly
five million people in the United States are treated for skin cancer every year with
an estimated annual cost over $8 billion [6163]. Basal cell carcinoma (BCC) and
squamous cell carcinoma (SCC) account for nearly 95 % of skin cancers, and
melanoma makes up approximately 5 %. In the United States in 2014, it is estimated
that there would be over 76,000 new cases of invasive melanoma and 9710
melanoma-related deaths [64]. The risk of skin cancers is governed by both genetic
factors and exposure to UV radiation.
28 B.P. Hibler et al.
Incidence rates of BCC and SCC are 510 times greater in the Caucasians than in
darker-skinned individuals [58]. Individuals with blue or green eyes, red or blond
hair, and lighter skin type have higher risk for developing skin cancer [65, 66].
Those individuals tend to have MC1R mutation and generate more pheomelanin
than eumelanin [67]. Pheomelanin is less effective in absorbing UV, and furthermore,
upon UVA exposure, pheomelanin are pro-oxidative and generate free radicals that
can damage DNA and nearby cellular organelles. Other phenotypic traits associated
with increased risks for skin cancer include high nevus count, tendency to sunburn,
inability to tan, and a history of sunburn at a young age [6870]. Additionally,
individuals with a personal or family history of skin cancer are at an increased risk,
suggesting the presence of additional genetic factors increasing susceptibility that
have not yet been phenotypically identified.
The incidence of NMSC increases with higher exposure to ambient solar radiation
and is greater in individuals with higher mean daily UV radiation [71]. Ecologic
studies have shown that the incidence of skin cancer is higher in regions of low
latitude and high UV index [72, 73]. The Nurses Health Study reported an increased
risk of skin cancer in individuals who lived in areas with moderate or high UV index
(greater than or equal to 6), with more pronounced effects seen for women who
grew up in states with higher UV indices [74]. Further studies have demonstrated
that early childhood exposure to high UV radiation increases the risk of skin cancer.
A migration study from Western Australia demonstrated that immigrants from
Great Britain who arrived before the age of 10 years had similar rates of melanoma
compared with the native-born population, whereas the incidence was nearly a
quarter of the native rate in those who arrived after the age of 15 years [75]. Similar
findings in migrant populations have been documented in other countries, including
the United States [7678].
Although a strong relationship exists between ambient solar radiation and the
incidence of skin cancer, patterns of sun exposure appear to have an impact on the
type of skin cancers. Chronic UV exposure has been implicated in the development
of both precancerous actinic keratoses (AK) and SCCs [7981]. These lesions tend
to occur on sun-exposed sites, such as the head, neck, and dorsal hands. The
association between BCC and sun exposure is more complex, because a large
percentage of BCCs are located on non-sun-exposed sites [82, 83]. As a result, it is
postulated that BCC may result from intermittent UV exposure or exposure early in
life rather than cumulative UV exposure.
Likewise, the overall risk of melanoma appears to also be associated with
more intense and intermittent exposure to high levels of UVR, often stemming
2 Photoprotection and Skin Cancer Prevention 29
from recreational activities or exposures occurring during childhood [66, 84, 85].
Melanoma is not often found on chronically sun-exposed sites, but rather is more
common on locations that are sporadically exposed, such as the trunk in males
and the legs in females [86]. However, certain subtypes of melanoma, such as
lentigo maligna melanoma or desmoplastic melanoma, are more commonly
found on chronically sun-exposed sites with a predilection for the head, neck,
and upper extremities [8790]. These lesions are often found on sun-damaged
skin in older individuals [88, 91, 92]. These observations suggest that different
subtypes of melanoma may result from either cumulative or intermittent sunlight
exposure.
Aside from genetic traits, individuals with certain occupations and those who
carry out high-risk behaviors have an increased probability of developing skin
cancer. Outdoor workers tend to have extensive amounts of UV radiation. A sys-
tematic review and meta-analysis of 18 studies (6 cohort and 12 casecontrol)
reported that 16 of the 18 studies (89 %) showed an increased risk of SCC in indi-
viduals with occupational UV exposure compared against individuals without UV
exposure (OR = 1.77; 95 % CI = 1.402.22) [93]. As for BCC, a meta-analysis
including 23 epidemiologic studies found a weaker, but still significant, associa-
tion between occupational sun exposure and risk of BCC (OR = 1.43; 95 %
CI = 1.231.66) [94]. The data on melanomas is mixed. While some studies have
suggested that outdoor workers may not be at an increased risk of melanoma
[95, 96], others have shown an increased risk among workers in UV-intense areas
and a strong association between melanoma incidence and both intermittent and
total UVR exposures [4, 97, 98]. These observations further emphasize the need
for adequate protection for individuals who are exposed to the damaging effects of
UVR in the workplace.
Individuals, especially young women, seeking indoor tanning are at high risk for
developing skin cancer. A recent systematic review and meta-analysis concluded
that there are an estimated 400,000 NMSCs and 6000 cases of melanoma annually
in the United States attributable to indoor tanning [99]. In a study of tanning bed
users, any use of tanning devices was associated with an increased risk of SCC
(OR = 2.5; 95 % CI = 1.73.8) and BCC (OR = 1.5; 95 % CI = 1.12.1) [100]. A
separate meta-analysis concluded that individuals with any history of indoor tanning
had an increased risk of melanoma (OR = 1.16, 95 % CI = 1.051.28) [101]. The risk
of skin cancer has a strong doseresponse relationship with tanning, thought to be
due to the accumulation of UV exposure [102]. Indoor tanning exposes users to
elevated amounts of UV radiation, and in 2009, the World Health Organization
(WHO) classified indoor tanning devices as group I human carcinogens due to
numerous studies showing the link between tanning and increased cancer risk [103].
Furthermore, the FDA recently upgraded sunlamps to moderate-risk (class II)
30 B.P. Hibler et al.
devices, requiring enhanced product labeling detailing the potential health effects of
use [104]. As such, use of these devices should be strongly discouraged due to the
adverse effects they can have on the skin.
2.6 Photoprotection
Numerous studies have shown that skin cancers can be prevented by reducing
intentional exposure to UV radiation and improving photoprotective strategies.
Effective photoprotection involves seeking shade, wearing protective clothing, and
applying sunscreen properly. Although sunscreen is less effective than other
protective measures, it is by far the most widely used vehicle for sun protection. A
large body of clinical research has demonstrated that sunscreens, when used
appropriately, can prevent skin cancers and precursor lesions.
participants in the vehicle control group had an average increase of one AK, while
participants in the sunscreen group actually saw a decrease in the mean number of
AKs by 0.6.
The last study was a randomized controlled trial in the United States assessing
AK prevention by sunscreen use in 37 high-risk patients with a history of
precancerous lesions or NMSC over a 2-year period [106]. The subjects in the
sunscreen group were instructed to apply sunscreen (SPF 29) every day, while the
control group applied the vehicle cream without active ingredients. After controlling
for differences in risk factors, a 36 % decrease in the annual rate of new AKs was
seen in the sunscreen group compared with the placebo group. These three studies
demonstrate that daily use of sunscreen has protective benefits for AKs.
The same population of Australian adults from the Nambour Trial was also observed
over the same period from 1992 to 1996 to determine the effect of sunscreen use on
the development of NMSC [108]. At enrollment, participants completed a survey
and underwent a complete skin exam by a dermatologist. Any prevalent skin cancers
were removed. Those randomized to the treatment group were instructed to apply a
layer of SPF-16 sunscreen to all exposed sites on the head, neck, arms, and hands
every morning, with reapplication after heavy sweating, bathing, or long sun
exposure. The control group was permitted to use sunscreen at their discretion, and
no sunscreen was provided. Compliance for the sunscreen group was assessed by
weighing sunscreen bottles every 3 months. At follow-up clinics in 1994 and 1996,
dermatologists blinded to treatment allocation reexamined all participants, with
histologic confirmation of all clinically diagnosed skin cancers.
After 4.5 years of follow-up, the investigators observed that sunscreen use had no
effect on either the incidence of BCC or in the total number of BCC tumors.
However, the overall incidence of SCC, in terms of persons affected, was 12 %
lower in the sunscreen treatment group (n = 22) compared with the control group
(n = 25), but this difference was not statistically significant. The study found a 39 %
reduction in the total number of SCC tumors among participants assigned to the
daily sunscreen group, with 28 SCCs occurring in the sunscreen group compared
with 46 SCCs in the control group (95 % CI = 0.460.81).
A follow-up study was published in 2006 to assess for potential latency of sun-
screen use [109]. Participants were followed for an additional 8 years. There was a
rate reduction of 35 % (95 % CI = 0.430.98) in the incidence of SCC, and there was
a rate reduction of 38 % (95 % CI = 0.380.99) in the total number of SCCs diag-
nosed in the sunscreen group. When the analysis was limited to the late follow-up
period (20012004), there was a rate reduction of 51 % for both the incidence SCC
and total tumor number. In contrast, the prolonged follow-up failed to demonstrate
a statistically significant reduction in the incidence of BCC (persons affected) or
total number of BCCs occurring in the daily sunscreen group. However, there was a
32 B.P. Hibler et al.
25 % reduction in the total number of BCCs in the treatment group in the late fol-
low-up period (20012004), although this difference is not statistically significant
(rate ratio = 0.75, 95 % CI = 0.491.14).
2.6.3 Nevi
Having many nevi or having at least 1 atypical nevus is the strongest constitutional
risk factors for melanoma. Studies have shown that UVR promotes the growth of
nevi [110112]. Currently, there is only one randomized controlled trial conducted
in Vancouver, British Columbia, that demonstrated the protective effect of broad-
spectrum sunscreen in reducing the development of nevi in children [113].
Schoolchildren, ages 610, were randomized to either a sunscreen group and
provided with SPF 30 broad-spectrum sunscreen or control group which received
no sunscreen and were given no advice about sunscreen use. Each childs nevi were
counted at the beginning and end of the 3-year trial. Based on an initial questionnaire
and dermatologic examination, the authors found that factors such as hair color, skin
response to sun exposure, facial freckling, and sunburn score in the first 5 years of
life were all associated with nevus counts. Analysis revealed regular use of sunscreen
was associated with a significant reduction in new nevi (median counts 24 vs. 28;
p = 0.048). Additionally, a greater effect was seen for sunscreen used in individuals
with a higher degree of freckling, with models suggesting that freckled children
using sunscreen would develop 3040 % fewer new nevi than untreated freckled
children. These data demonstrate the importance of regular sunscreen use on
attenuating the development of new nevi which are a known risk factor for
melanoma.
2.6.4 Melanoma
There have been controversies regarding the protective role of sunscreens against
the development of melanoma. Some of the early casecontrol studies suggested an
increased risk of melanoma with sunscreen use [114116]. A meta-analysis of the
literature published between 1966 and 1999 found no association between sunscreen
use and increased risk of melanoma (relative risk = 1.01; 95 % CI = 0.46, 2.28)
[117]. A second review also found a similar result (odds ratio 1.0; 95 % CI = 0.8
1.2) [118]. However, these early casecontrol studies failed to account for skin
sensitivity. Specifically, individuals who are more susceptible to burning and
developing melanoma were more likely to use sunscreen, and hence there could be
uncontrolled confounding by indication. Other explanations are related to inappro-
priate application of sunscreen with low SPF and lack of UVA protection.
The controversy was largely put to rest with the results from Nambour Trial in
Queensland, Australia [119]. The participants were observed after long-term
2 Photoprotection and Skin Cancer Prevention 33
follow-up to assess whether application of sunscreen during the first 4.5 years had
an effect on their risk of primary cutaneous melanoma. At the end of 10-year
follow-up (nearly 15 years from the start of the trial), there were a total of 11 primary
melanomas (3 invasive) in participants randomized to the sunscreen group and 22
primary melanomas (11 invasive) in the discretionary use (control) group. The study
showed a 50 % reduction in the risk of overall melanomas in the sunscreen group
(p = 0.051) and a 73 % reduction in the risk of invasive melanomas among the daily
sunscreen group (p = 0.045). These results demonstrated that daily sunscreen use
over a 4.5-year period appears to reduce the long-term melanoma incidence over a
10-year period, with the most pronounced effect seen for invasive melanoma.
It is important to note that the control group in the Nambour Trial for the AK,
SCC, BCC, and melanoma studies was not given a placebo or inactive sunscreen,
but rather was allowed to continue discretionary use of sunscreen. The design of the
trial underestimates the full protective benefits of sunscreen against melanoma.
Furthermore, the sunscreen used in the trial was SPF 16 and not UVA stable.
Modern-day sunscreens have higher SPF values and are photostable, and
theoretically they should offer superior protection.
2.7 Conclusion
References
1. Maru GB et al (2014) The role of inflammation in skin cancer. Adv Exp Med Biol
816:437469
2. Young C (2009) Solar ultraviolet radiation and skin cancer. Occup Med 59(2):8288
3. Dupont E, Gomez J, Bilodeau D (2013) Beyond UV radiation: a skin under challenge. Int
J Cosmet Sci 35(3):224232
4. Kozma B, Eide MJ (2014) Photocarcinogenesis: an epidemiologic perspective on ultraviolet
light and skin cancer. Dermatol Clin 32(3):301313, viii
5. Diffey BL (2002) What is light? Photodermatol Photoimmunol Photomed 18(2):6874
6. Lautenschlager S, Wulf HC, Pittelkow MR (2007) Photoprotection. Lancet 370(9586):
528537
7. Miller SA et al (1998) An analysis of UVA emissions from sunlamps and the potential impor-
tance for melanoma. Photochem Photobiol 68(1):6370
34 B.P. Hibler et al.
8. Rigel DS, Rigel EG, Rigel AC (1999) Effects of altitude and latitude on ambient UVB radia-
tion. J Am Acad Dermatol 40(1):114116
9. de Laat A et al (1996) Cell cycle kinetics following UVA irradiation in comparison to UVB
and UVC irradiation. Photochem Photobiol 63(4):492497
10. Parrish JA, Zaynoun S, Anderson RR (1981) Cumulative effects of repeated subthreshold
doses of ultraviolet radiation. J Invest Dermatol 76(5):356358
11. Narayanan DL, Saladi RN, Fox JL (2010) Ultraviolet radiation and skin cancer. Int J Dermatol
49(9):978986
12. Ichihashi M et al (2003) UV-induced skin damage. Toxicology 189(1):2139
13. Kobayashi N et al (2001) Quantitation and visualization of ultraviolet-induced DNA damage
using specific antibodies: application to pigment cell biology. Pigment Cell Res 14(2):
94102
14. Ikehata H, Ono T (2011) The mechanisms of UV mutagenesis. J Radiat Res 52(2):115125
15. Brash DE et al (1991) A role for sunlight in skin cancer: UV-induced p53 mutations in squa-
mous cell carcinoma. Proc Natl Acad Sci U S A 88(22):1012410128
16. Ikehata H et al (2008) UVA1 genotoxicity is mediated not by oxidative damage but by
cyclobutane pyrimidine dimers in normal mouse skin. J Invest Dermatol 128(9):
22892296
17. Runger TM (2008) CT transition mutations are not solely UVB-signature mutations,
because they are also generated by UVA. J Invest Dermatol 128(9):21382140
18. Nakagawa A et al (1998) Three-dimensional visualization of ultraviolet-induced DNA dam-
age and its repair in human cell nuclei. J Invest Dermatol 110(2):143148
19. Wei Q et al (1993) DNA repair and aging in basal cell carcinoma: a molecular epidemiology
study. Proc Natl Acad Sci U S A 90(4):16141618
20. Runger TM, Kappes UP (2008) Mechanisms of mutation formation with long-wave ultravio-
let light (UVA). Photodermatol Photoimmunol Photomed 24(1):210
21. Wondrak GT, Jacobson MK, Jacobson EL (2006) Endogenous UVA-photosensitizers:
mediators of skin photodamage and novel targets for skin photoprotection. Photochem
Photobiol Sci 5(2):215237
22. Peak JG, Peak MJ, MacCoss M (1984) DNA breakage caused by 334-nm ultraviolet light is
enhanced by naturally occurring nucleic acid components and nucleotide coenzymes.
Photochem Photobiol 39(5):713716
23. Walrant P, Santus R (1974) N-formyl-kynurenine, a tryptophan photooxidation product, as a
photodynamic sensitizer. Photochem Photobiol 19(6):411417
24. McCormick JP et al (1976) Characterization of a cell-lethal product from the photooxidation
of tryptophan: hydrogen peroxide. Science 191(4226):468469
25. Fisher GJ et al (2002) Mechanisms of photoaging and chronological skin aging. Arch
Dermatol 138(11):14621470
26. Chen L, Hu JY, Wang SQ (2012) The role of antioxidants in photoprotection: A critical
review. J Am Acad Dermatol 67(5):10131024
27. Tran TT, Schulman J, Fisher DE (2008) UV and pigmentation: molecular mechanisms and
social controversies. Pigment Cell Melanoma Res 21(5):509516
28. Kielbassa C, Roza L, Epe B (1997) Wavelength dependence of oxidative DNA damage
induced by UV and visible light. Carcinogenesis 18(4):811816
29. Heck DE et al (2003) UVB light stimulates production of reactive oxygen species: unex-
pected role for catalase. J Biol Chem 278(25):2243222436
30. Epe B (1991) Genotoxicity of singlet oxygen. Chem Biol Interact 80(3):239260
31. Cheng KC et al (1992) 8-Hydroxyguanine, an abundant form of oxidative DNA damage,
causes G----T and A----C substitutions. J Biol Chem 267(1):166172
32. Cadet J et al (2012) Photoinduced damage to cellular DNA: direct and photosensitized reac-
tions. Photochem Photobiol 88(5):10481065
33. Daya-Grosjean L, Dumaz N, Sarasin A (1995) The specificity of p53 mutation spectra in
sunlight induced human cancers. J Photochem Photobiol B 28(2):115124
34. Hussein MR (2005) Ultraviolet radiation and skin cancer: molecular mechanisms. J Cutan
Pathol 32(3):191205
2 Photoprotection and Skin Cancer Prevention 35
62. Jemal A et al (2011) Recent trends in cutaneous melanoma incidence and death rates in the
United States, 1992-2006. J Am Acad Dermatol 65(5 Suppl 1):S17S25.e13
63. Lazovich D, Choi K, Vogel RI (2012) Time to get serious about skin cancer prevention.
Cancer Epidemiol Biomarkers Prev 21(11):18931901
64. Siegel R et al (2014) Cancer statistics, 2014. CA Cancer J Clin 64(1):929
65. Mitra D et al (2012) An ultraviolet-radiation-independent pathway to melanoma carcinogen-
esis in the red hair/fair skin background. Nature 491(7424):449453
66. Gandini S et al (2005) Meta-analysis of risk factors for cutaneous melanoma: II. Sun expo-
sure. Eur J Cancer 41(1):4560
67. Raimondi S et al (2008) MC1R variants, melanoma and red hair color phenotype: a meta-
analysis. Int J Cancer 122(12):27532760
68. Zanetti R et al (1996) The multicentre south European study Helios. I: skin characteristics
and sunburns in basal cell and squamous cell carcinomas of the skin. Br J Cancer 73(11):
14401446
69. Gallagher RP et al (1995) Sunlight exposure, pigmentary factors, and risk of nonmelanocytic
skin cancer. I. Basal cell carcinoma. Arch Dermatol 131(2):157163
70. Kricker A et al (1995) Does intermittent sun exposure cause basal cell carcinoma? a case-
control study in Western Australia. Int J Cancer 60(4):489494
71. Xiang F et al (2014) Incidence of nonmelanoma skin cancer in relation to ambient UV radia-
tion in white populations, 1978-2012: empirical relationships. JAMA Dermatol
150(10):10631071
72. Rivas M, Rojas E, Calaf GM (2012) Prediction of skin cancer occurrence by ultraviolet solar
index. Oncol Lett 3(4):893896
73. Eide MJ, Weinstock MA (2005) Association of UV index, latitude, and melanoma incidence
in nonwhite populations--US Surveillance, Epidemiology, and End Results (SEER) Program,
1992 to 2001. Arch Dermatol 141(4):477481
74. Qureshi AA et al (2008) Geographic variation and risk of skin cancer in US women.
Differences between melanoma, squamous cell carcinoma, and basal cell carcinoma. Arch
Intern Med 168(5):501507
75. Holman CD, Armstrong BK (1984) Cutaneous malignant melanoma and indicators of total
accumulated exposure to the sun: an analysis separating histogenetic types. J Natl Cancer Inst
73(1):7582
76. Mack TM, Floderus B (1991) Malignant melanoma risk by nativity, place of residence at
diagnosis, and age at migration. Cancer Causes Control 2(6):401411
77. Levine H et al (2013) Country of origin, age at migration and risk of cutaneous melanoma: a
migrant cohort study of 1,100,000 Israeli men. Int J Cancer 133(2):486494
78. Autier P et al (1997) Melanoma risk and residence in sunny areas. EORTC Melanoma
Co-operative Group. European Organization for Research and Treatment of Cancer. Br
J Cancer 76(11):15211524
79. Kennedy C et al (2003) The influence of painful sunburns and lifetime sun exposure on the
risk of actinic keratoses, seborrheic warts, melanocytic nevi, atypical nevi, and skin cancer.
J Invest Dermatol 120(6):10871093
80. Strickland PT et al (1989) Quantitative carcinogenesis in man: solar ultraviolet B dose
dependence of skin cancer in Maryland watermen. J Natl Cancer Inst 81(24):19101913
81. Woodhead AD, Setlow RB, Tanaka M (1999) Environmental factors in nonmelanoma and
melanoma skin cancer. J Epidemiol 9(6 Suppl):S102S114
82. Raasch B et al (1998) Body site specific incidence of basal and squamous cell carcinoma in
an exposed population, Townsville, Australia. Mutat Res 422(1):101106
83. Franceschi S et al (1996) Site distribution of different types of skin cancer: new aetiological
clues. Int J Cancer 67(1):2428
84. Jhappan C, Noonan FP, Merlino G (2003) Ultraviolet radiation and cutaneous malignant
melanoma. Oncogene 22(20):30993112
85. Gallagher RP et al (2010) Ultraviolet radiation. Chronic Dis Can 29(Suppl 1):5168
86. Bataille V (2013) Sun exposure, sunbeds and sunscreens and melanoma. What are the contro-
versies? Curr Oncol Rep 15(6):526532
2 Photoprotection and Skin Cancer Prevention 37
87. Kraft S, Granter SR (2014) Molecular pathology of skin neoplasms of the head and neck.
Arch Pathol Lab Med 138(6):759787
88. Clark WH Jr, Mihm MC Jr (1969) Lentigo maligna and lentigo-maligna melanoma. Am
J Pathol 55(1):3967
89. Feng Z et al (2011) Incidence and survival of desmoplastic melanoma in the United States,
1992-2007. J Cutan Pathol 38(8):616624
90. Wasif N, Gray RJ, Pockaj BA (2011) Desmoplastic melanoma - the step-child in the mela-
noma family? J Surg Oncol 103(2):158162
91. Jain S, Allen PW (1989) Desmoplastic malignant melanoma and its variants. A study of 45
cases. Am J Surg Pathol 13(5):358373
92. Chen LL et al (2013) Desmoplastic melanoma: a review. J Am Acad Dermatol 68(5):
825833
93. Schmitt J et al (2011) Occupational ultraviolet light exposure increases the risk for the devel-
opment of cutaneous squamous cell carcinoma: a systematic review and meta-analysis. Br
J Dermatol 164(2):291307
94. Bauer A, Diepgen TL, Schmitt J (2011) Is occupational solar ultraviolet irradiation a relevant
risk factor for basal cell carcinoma? A systematic review and meta-analysis of the
epidemiological literature. Br J Dermatol 165(3):612625
95. Gallagher RP, Elwood JM, Yang CP (1989) Is chronic sunlight exposure important in account-
ing for increases in melanoma incidence? Int J Cancer 44(5):813815
96. Garland FC et al (1990) Occupational sunlight exposure and melanoma in the U.S. Navy.
Arch Environ Health 45(5):261267
97. Whiteman DC et al (2006) Anatomic site, sun exposure, and risk of cutaneous melanoma.
J Clin Oncol 24(19):31723177
98. Chang YM et al (2009) Sun exposure and melanoma risk at different latitudes: a pooled
analysis of 5700 cases and 7216 controls. Int J Epidemiol 38(3):814830
99. Wehner MR et al (2014) International prevalence of indoor tanning: a systematic review and
meta-analysis. JAMA Dermatol 150(4):390400
100. Karagas MR et al (2002) Use of tanning devices and risk of basal cell and squamous cell skin
cancers. J Natl Cancer Inst 94(3):224226
101. Colantonio S, Bracken MB, Beecker J (2014) The association of indoor tanning and mela-
noma in adults: systematic review and meta-analysis. J Am Acad Dermatol 70(5):847857.
e118
102. Lazovich D et al (2010) Indoor tanning and risk of melanoma: a case-control study in a
highly exposed population. Cancer Epidemiol Biomarkers Prev 19(6):15571568
103. El Ghissassi F et al (2009) A review of human carcinogens Part D: radiation. Lancet Oncol
10(8):751752
104. US Food and Drug Administration. FDA to require warnings on sunlamp products. FDA
News Release 2014 May 29, 2014; Available from: http://www.fda.gov/NewsEvents/
Newsroom/PressAnnouncements/ucm399222.htm
105. Thompson SC, Jolley D, Marks R (1993) Reduction of solar keratoses by regular sunscreen
Use. N Engl J Med 329(16):11471151
106. Naylor MF et al (1995) High sun protection factor sunscreens in the suppression of actinic
neoplasia. Arch Dermatol 131(2):170175
107. Darlington S et al (2003) A randomized controlled trial to assess sunscreen application and beta
carotene supplementation in the prevention of solar keratoses. Arch Dermatol 139(4):451455
108. Green A et al (1999) Daily sunscreen application and betacarotene supplementation in pre-
vention of basal-cell and squamous-cell carcinomas of the skin: a randomised controlled trial.
Lancet 354(9180):723729
109. van der Pols JC et al (2006) Prolonged prevention of squamous cell carcinoma of the skin by
regular sunscreen use. Cancer Epidemiol Biomarkers Prev 15(12):25462548
110. Harrison SL, MacLennan R, Buettner PG (2008) Sun exposure and the incidence of melano-
cytic nevi in young Australian children. Cancer Epidemiol Biomarkers Prev 17(9):
23182324
38 B.P. Hibler et al.
111. Psaty EL et al (2010) Defining the patient at high risk for melanoma. Int J Dermatol
49(4):362376
112. Bauer J et al (2005) Risk factors of incident melanocytic nevi: a longitudinal study in a cohort
of 1,232 young German children. Int J Cancer 115(1):121126
113. Gallagher RP et al (2000) Broad-spectrum sunscreen use and the development of new nevi in
white children: a randomized controlled trial. JAMA 283(22):29552960
114. Westerdahl J et al (1995) Is the use of sunscreens a risk factor for malignant melanoma?
Melanoma Res 5(1):5965
115. Westerdahl J et al (2000) Sunscreen use and malignant melanoma. Int J Cancer 87(1):
145150
116. Wolf P et al (1998) Phenotypic markers, sunlight-related factors and sunscreen use in patients
with cutaneous melanoma: an Austrian case-control study. Melanoma Res 8(4):370378
117. Huncharek M, Kupelnick B (2002) Use of topical sunscreens and the risk of malignant mela-
noma: a meta-analysis of 9067 patients from 11 case-control studies. Am J Public Health
92(7):11731177
118. Dennis LK, Beane Freeman LE, VanBeek MJ (2003) Sunscreen use and the risk for mela-
noma: a quantitative review. Ann Intern Med 139(12):966978
119. Green AC et al (2011) Reduced melanoma after regular sunscreen use: randomized trial fol-
low-up. J Clin Oncol 29(3):257263
Chapter 3
Photoprotection for Photodermatoses
Key Points
Photodermatoses represent a broad and extensive group of disorders
caused by exposure to sunlight.
Elucidation of action spectrum of a disease should be performed at the
earliest convenience to assure most adequate management of the patient.
The cornerstone of photoprotection in all photodermatoses involves the use of
long-sleeve shirts, wide-brim hats, sunglasses, appropriate types and amounts
of sunscreens, and sun avoidance during peak hours of sun intensity.
If photosensitivity is due to administration of an exogenous agent or the
result of an accumulation or deficiency of an endogenous entity, removal
of the offending agent and correction of the deficiency are paramount to
treatment.
3.1 Background
D. Gutierrez, BS
Department of Dermatology, University Hospitals Case Medical Center,
11100 Euclid, Ave. Lakeside 3500, Cleveland, OH 44106-5028, USA
E.D. Baron, MD (*)
School of Medicine, Case Western Reserve University, Cleveland, OH, USA
Department of Dermatology, University Hospitals Case Medical Center,
11100 Euclid, Ave. Lakeside 3500, Cleveland, OH 44106-5028, USA
Louis Stokes Department of Veterans, Affairs Medical Center, Cleveland, OH, USA
e-mail: [email protected]
The group of IMPs consists of five distinct conditions: actinic prurigo, chronic
actinic dermatitis, hydroa vacciniforme, polymorphous light eruption (PMLE),
and solar urticaria [3]. The pathophysiology underlying each of these photoderma-
toses has not been fully characterized. However, it is hypothesized that these dis-
orders result from dysregulation of the immune system due to UVR in genetically
susceptible individuals. Each of these conditions will be discussed separately.
3 Photoprotection for Photodermatoses 41
treat pruritic lesions can also be considered, but should be used only as adjunct to
other therapies. Polypodium leucotomos, carotenoids, afamelanotide, and nicotin-
amide have been used as adjuncts to topical photoprotection. However, there lack of
sufficient evidence assessing their to warrant recommendation at this time in lieu of
other proven photoprotective. Above all, individuals should be counseled on the use
of physical barriers for photoprotection: sunglasses, wide-brimmed hats, and long-
sleeve shirts. Use of photoprotective clothing is a mainstay of preventing disease
exacerbation.
Solar urticaria is a very rare photodermatosis that results in wheal and flare
development within minutes of light exposure with resolution within 24 h. The
presumed pathophysiology is that of a type I hypersensitivity response in which
chromophore absorption of a photon causes the formation of neoantigens capable of
recognition by IgE antibodies [14]. These antibodies then bind to the Fc receptor on
mast cells and upon re-exposure to light cause degranulation and release of
inflammatory mediators. The action spectrum is vast among patients and extends
over UV to the visible light spectrum [15] so much so that it has been reported that
even infrared radiation causes exacerbation [16]. As the action spectrum is variable,
phototesting should first be performed to determine a patients action spectrum to
best ensure adequate photoprotective strategies are used [6]. It should be emphasized
that phototesting reading needs to be done immediately after exposure as the wheal
and flare response will resolve.
General photoprotective strategies including sun avoidance, use of broad-
spectrum sunscreen with high SPF, and use of tightly woven, thick, dark fabrics are
initial precautions that can be taken to prevent acute flares. The visible light
spectrum, in particular, is difficult to protect against. For topical visible
photoprotection to be effective, the topical agent must be opaque. As such, there are
no sunscreens currently available that provide coverage against the visible light
spectrum. With respect to protection against visible light, it should be made known
that the SPF of a sunscreen does not correlate to protection against the visible action
spectrum [17]. Sunscreens with a higher concentration of iron oxides, which are
pigmented, have been shown to be better at blocking visible light when compared to
the sunscreens only containing micronized zinc oxide and titanium dioxide [17, 18].
No clinical trials have been performed evaluating the efficacy of such pigmented
sunscreens in the idiopathic photodermatoses, there exists a likelihood of benefit
given results from artificial sensitization against visible light [19].
Phototherapy, with UVA being the most commonly used light source, to facilitate
hardening provides the next step of management for more severely affected patients.
Recent evidence suggests that using wavelengths outside the action spectrum of a
patient may induce tolerance [20, 21]. UVA rush-hardening protocols have reported,
suggesting this as a viable option for treatment in the future [22]. An interesting agent
3 Photoprotection for Photodermatoses 43
for systemic photoprotection in solar urticaria involves the use of the -melanocyte-
stimulating hormone analogue, afamelanotide. In a cohort of 5 individuals receiving a
single subcutaneous the urticarial dose necessary for eliciting wheal formation
increased [23]. Dihydroxyacetone followed by an application of naphthoquinone over
a period of 7 month yielded an SPF increase of 18 in 18 of the 30 patients tested [24].
Though quite promising, more trials are necessary in order to truly assess efficacy.
In the event of an exacerbation, topical corticosteroids and antihistamines can be
used for symptomatic treatment. Although systemic corticosteroids are more effec-
tive at controlling flares, adverse side effects prevent their long-term use. In addition,
IVIG [25, 26], plasmapheresis [27], and omalizumab [28, 29] have proven successful
in select cases, supporting the proposed antibody-mediated pathophysiology. Most
recently, a phase 3 multicenter study of omalizumab in 323 patients with chronic
idiopathic or spontaneous urticaria, diseases similar to solar urticaria, had reported
symptomatic relief in those where antihistamines had failed to alleviate symptoms of
urticaria [30]. Based on these initial findings, it appears that immunomodulatory ther-
apy should be more aggressively pursued as a treatment option for solar urticaria.
the viral infection. Successful treatment of EBV has resulted in increased ability to
spend time in sunlight without the development of new skin lesions while also pre-
venting any systemic manifestations associated with the infection [39].
True photoallergic responses are much less common than phototoxic reactions. This
type IV hypersensitivity response results from formation of a neoantigen due to any
amount of UV exposure. Photoallergic reactions are usually not observed for
13 days; most common offending agents are topical agents, usually sunscreens.
Following this period, the response manifests itself as an eczematous reaction
spreading to areas unexposed to sunlight. Spongiosis is seen histologically. For
classic photoallergic responses to develop, prior sensitization is necessary. Cross
reactivity of many molecules, however, permit the development of photoallergic
reactions from first exposure due to exposure of a structurally similar entity. The
prototypical example of this phenomenon is photoallergy due to cross reactivity
between thimerosal, a preservative used in skin antigen testing, and piroxicam. The
thiosalicylic moiety in thiomersal is highly antigenic and causes allergic responses
in some patients. Piroxicam, a nonsteroidal anti-inflammatory drug (NSAID), is
photodecomposed when exposed to UVA into molecule that is structurally similar
to the thiosalicylic moiety in thimerosal causing a photoallergic response upon the
first exposure to the NSAID [6971] Photoallergies can result from many topicals
and systemic medications.
Phototoxic reactions are significantly more common than photoallergic reaction and
are observed within minutes to hours. These reactions occur on first exposure to an
agent in the setting of sufficient amount of agent and UVA exposure. An exogenous
agent will topically absorb photons causing it to reach an excited state. To reach the
ground state, the agent becomes involved in a series of oxygen-dependent reactions
eventually causing the formation of free radicals causing cellular damage. There are
two types of phototoxic reactions. Type I phototoxic reactions involve a photosensi-
tizer combining with cellular components or transferring hydrogen or electrons to
other molecules, forming free radicals that cause cellular damage [72, 73]. Type II
phototoxicity, on the other hand, involves the excited agent transferring electrons to
oxygen directly, causing the formation of oxygen radicals [72, 73].
3 Photoprotection for Photodermatoses 47
Cutaneous
Location Heme biosynthesis Enzyme Pathology Inheritance Acute manifestations Treatment options
Glycine +
succinyl-CoA
Mitochondria Aminolevulinic acid X-linked sideroblastic X-linked No N/A N/A
synthase 2 anemia recessive
Delta aminolevulinic
acid
Aminolevulinic acid ALA dehydratase Autosomal Yes N/A Intravenous hematin
dehydratase porphyria recessive
Porphobilinogen
Hydroxymethylbilane Acute intermittent Autosomal Yes N/A Intravenous hematin
synthase porphyria dominant
Cytosol Hydroxymethylbilane
Uroporphyrinogen Congenital Autosomal No Bullae, vesicles, Hydroxyurea, red
synthase erythropoietic recessive skin thickening, blood transfusion,
porphyria (Gnther hypo- and activated charcoal,
disease) hyperpigmentation, bone marrow
hypertrichosis, transplant
scarring, loss of
nails, loss of digits
Uroporphyrinogen III
Uroporphyrinogen Porphyria cutanea Type I/ No PCT: vesicles, Hydroxychloroquine
dehydrogenase tarda (PCT)/ III - bullae, erosions heal [111], Phlebotomy
hepatoerythropoietic sporadic with [111]
porphyria (HEP) type II: hyperpigmentation HEP: same as PCT
autosomal or atrophy,
dominant periorbital
hypertrichosis, milia
D. Gutierrez and E.D. Baron
Coproporphyrinogen
III
Coproporphyrinogen Hereditary Autosomal Yes Skin lesions similar Intravenous hematin
oxidase coproporphyria dominant to PCT
Protoporphyrinogen
IX
Mitochondria Protoporphyrinogen Variegate porphyria Autosomal Yes Similar to PCT but Intravenous hematin,
oxidase dominant with milder dihydroxyacetone
symptoms [112]
Protoporphyrin IX
Ferrochelatase Erythropoietic Pseudo- No Pruritus, wheals, Afamelanotide [113],
protoporphyria dominant painful edema, cholestyramine,
Photoprotection for Photodermatoses
erythema, chenodeoxycholic
petechiae, purpura, acid [74], cysteine
lichenification, [114], beta carotene,
thick activated charcoal,
pseudovesicles blood transfusion,
intravenous hematin
Heme
All information adapted from Sassa [115] unless otherwise stated
Not shown in the above, nonenzymatic conversion of hydroxymethylbilane to uroporphyrinogen I and then to coproporphyrinogen I accounts for the photosensitivity
resulting from uroporphyrinogen synthase deficiency
49
50 D. Gutierrez and E.D. Baron
3.3.7 Pellagra
Photoaggravated disorders comprise the widest and least specific group of photoder-
matoses. The only unifying factor among the disorders involves exacerbation by UVR
exposure. The list of photoaggravated disorders is extensive: acne vulgaris, atopic
dermatitis, bullous pemphigoid, carcinoid syndrome, cutaneous T-cell lymphoma,
Dariers disease, dermatomyositis, disseminated superficial actinic porokeratosis, ery-
thema multiforme, Grovers disease, lichen planus, lupus erythematosus, pemphigus,
pityriasis rubra pilaris, psoriasis, reticular erythematous mucinosis, rosacea, sebor-
rheic dermatitis, and various viral infections [3]. We will be discussing photoprotec-
tive strategies primarily in regard to lupus erythematosus and dermatomyositis.
3 Photoprotection for Photodermatoses 51
3.4.2 Dermatomyositis
3.6 Conclusions
References
1. Brenner M, Hearing V (2008) The protective role of melanin against UV damage in human
skin. Photochem Photobiol 84(3):539549
2. Diffey B, Kochevar I (2007) Basic principles of photobiology. In: Lim H, Honigsmann H,
Hawk JL (eds) Photodermatology. Informa Healthcare, New York, pp 1527
3. Santoro FA, Lim HW (2011) Update on photodermatoses. Semin Cutan Med Surg
30(4):229238
4. Hlzle E, Plewig G, von Kries R, Lehmann P (1987) Polymorphous light eruption. J Invest
Dermatol 88(3 Suppl):32s38s
5. Norris PG, Morris J, McGibbon DM, Chu AC, Hawk JL (1989) Polymorphic light eruption:
an immunopathological study of evolving lesions. Br J Dermatol 120(2):173183
6. Lehmann P, Schwarz T (2011) Photodermatoses: diagnosis and treatment. Dtsch Arztebl Int
108(9):135141
7. Labeling and effectiveness testing; sunscreen drug products for over-the-counter human use.
21 CFR Parts 201 and 310. Sect. Vol. 76, No. 117 (17 June 2011)
8. Bech-Thomsen N, Wulf HC (1992) Sunbathers application of sunscreen is probably inade-
quate to obtain the sun protection factor assigned to the preparation. Photodermatol
Photoimmunol Photomed. 9(6):242244
9. Azurdia RM, Pagliaro JA, Diffey BL, Rhodes LE (1999) Sunscreen application by photosen-
sitive patients is inadequate for protection. Br J Dermatol 140(2):255258
10. Bissonnette R, Nigen S, Bolduc C (2012) Influence of the quantity of sunscreen applied on
the ability to protect against ultraviolet-induced polymorphous light eruption. Photodermatol
Photoimmunol Photomed 28(5):240243
11. Gruber-wackernagel A, Byrne SN, Wolf P (2008) Pathogenic mechanisms of polymorphic
light eruption. J Natl Cancer Inst Monogr 1:341354
3 Photoprotection for Photodermatoses 55
12. Man I, Dawe RS, Ferguson J (1999) Artificial hardening for polymorphic light eruption:
practical points from ten years experience. Photodermatol Photoimmunol Photomed
15:9699
13. Patel DC, Bellaney GJ, Seed PT, McGregor JM, Hawk JL (2000) Efficacy of short-course
oral prednisolone in polymorphic light eruption: a randomized controlled trial. Br J Dermatol
143(4):828831
14. Nitiyarom R, Wongpraparut C (2014) Hydroa vacciniforme and solar urticaria. Dermatol
Clin 32(3):345353, viii
15. Millard TP, Hawk JLM (2002) Photosensitivity disorders: cause, effect and management. Am
J Clin Dermatol 3(4):239246
16. Mekkes JR, de Vries HJ, Kammeyer A (2003) Solar urticaria induced by infrared radiation.
Clin Exp Dermatol 28(2):2223
17. Schalka S, SantAnna Addor FA, Agelune CM, de Melo C, Pereira V (2012) Sunscreen pro-
tection against visible light: a new proposal for evaluation. Surg Cosmet Dermatol
3(4):4552
18. Sayre R, Kollias N, Roberts R (1990) Physical sunscreens. J Soc Cosmet Chem 41(2):103109
19. Bissonnette R, Nigen S, Bolduc C, Mry S, Nocera T (2008) Protection afforded by sun-
screens containing inorganic sunscreening agents against blue light sensitivity induced by
aminolevulinic acid. Dermatol Surg 34(11):14691476
20. Wolf R, Herzinger T, Grahovac M, Prinz JC (2013) Solar urticaria: long-term rush hardening
by inhibition spectrum narrow-band UVB 311 nm. Clin Exp Dermatol 38(4):446447
21. Roelandts R (1985) Pre-PUVA UVA, desensitization for solar urticaria. Photodermatol
2(3):174176
22. Masuoka E (2012) Fukunaga a, Kishigami K, et al. Successful and long-lasting treatment of
solar urticaria with ultraviolet A rush hardening therapy. Br J Dermatol 167(1):198201
23. Haylett AK, Nie Z, Brownrigg M, Taylor R, Rhodes LE (2011) Systemic photoprotection in
solar urticaria with -melanocyte-stimulating hormone analogue [Nle4-D-Phe7]--MSH. Br
J Dermatol 164(2):407414
24. Fusaro RM, Rice EG (2005) The maillard reaction for sunlight protection. Ann N Y Acad Sci
1043:174183
25. Maksimovic L, Frmont G, Jeanmougin M, Dubertret L, Viguier M (2009) Solar urticaria
successfully treated with intravenous immunoglobulins. Dermatology 218(3):252254
26. Hughes R, Cusack C, Murphy GM, Kirby B (2009) Solar urticaria successfully treated with
intravenous immunoglobulin. Clin Exp Dermatol 34(8):e660e662
27. Collins P, Ahamat R, Green C, Ferguson J (1996) Plasma exchange therapy for solar urti-
caria. Br J Dermatol 134(6):10937
28. Gzelbey O, Ardelean E, Magerl M, Zuberbier T, Maurer M, Metz M (2008) Successful
treatment of solar urticaria with anti-immunoglobulin E therapy. Allergy 63(11):15635
29. Waibel KH, Reese DA, Hamilton RG, Devillez RL (2010) Partial improvement of solar urti-
caria after omalizumab. J Allergy Clin Immunol 125(2):490491
30. Maurer M, Rosn K, Hsieh HJ, Saini S, Grattan C, Gimenz-Arnau A, Agarwal S, Doyle R,
Canvin J, Kaplan A, Casale T (2013) Omalizumab for the treatment of chronic idiopathic or
spontaneous urticaria. N Engl J Med 368(10):924935
31. Iwatsuki K, Satoh M, Yamamoto T, Oono T, Morizane S, Ohtsuka M, Xu ZG, Suzuki D, Tsuji
K (2006) Pathogenic link between hydroa vacciniforme and Epstein-Barr virusassociated
hematologic disorders. Arch Dermatol 142(5):58795
32. Jacyk WK, Moosa Y (2010) Crusts and vacciniforme scars on sun-exposed skin. Clin Exp
Dermatol 35(1):9798
33. Halasz CL, Leach EE, Walther RR, Poh-Fitzpatrick MB (1983) Hydroa vacciniforme: induc-
tion of lesions with ultraviolet A. J Am Acad Dermatol 8(2):171176
34. Sunohara A, Mizuno N, Sakai M, Kawabe Y, Sakakibara S (1988) Action spectrum for UV
erythema and reproduction of the skin lesions in hydroa vacciniforme. Photodermatol
5(3):139145
56 D. Gutierrez and E.D. Baron
35. Iwatsuki K, Ohtsuka M, Akiba H, Kaneko F (1999) Atypical hydroa vacciniforme in child-
hood: from a smoldering stage to Epstein-Barr virus-associated lymphoid malignancy. J Am
Acad Dermatol 40(2 Pt 1):283284
36. Yamamoto T, Tsuji K, Suzuki D, Morizane S, Iwatsuki K (2007) A novel, noninvasive diag-
nostic probe for hydroa vacciniforme and related disorders: detection of latency-associated
Epstein-Barr virus transcripts in the crusts. J Microbiol Methods 68(2):403407
37. Huggins RH, Leithauser LA, Eide MJ, Hexsel CL, Jacobsen G, Lim HW (2009) Quality of
life assessment and disease experience of patient members of a web-based hydroa
vacciniforme support group. Photodermatol Photoimmunol Photomed 25(4):209215
38. Gupta G, Man I, Kemmett D (2000) Hydroa vacciniforme: a clinical and follow-up study of
17 cases. J Am Acad Dermatol 42(2 Pt 1):208213
39. Almeida HL, Kopp J, Jorge VM, Sartori DS, Velloso CD (2013) Extensive hydroa vaccin-
iforme. An Bras Dermatol 88(4):6202
40. Meola T, Sanchez M, Lim HW, Buchness MR, Soter NA (1997) Chronic actinic dermatitis
associated with human immunodeficiency virus infection. Br J Dermatol 137(3):431436
41. Kerr HA, Lim HW (2007) Photodermatoses in African Americans: a retrospective analysis of
135 patients over a 7-year period. J Am Acad Dermatol 57(4):638643
42. Lim HW, Buchness MR, Ashinoff R, Soter NA (1990) Chronic actinic dermatitis. Study of
the spectrum of chronic photosensitivity in 12 patients. Arch Dermatol 126(3):317323
43. Russell SC, Dawe RS, Collins P, Man I, Ferguson J (1998) The photosensitivity dermatitis
and actinic reticuloid syndrome (chronic actinic dermatitis) occurring in seven young atopic
dermatitis patients. Br J Dermatol 138(3):496501
44. Yap LM, Foley P, Crouch R, Baker C (2003) Chronic actinic dermatitis: a retrospective analy-
sis of 44 cases referred to an Australian photobiology clinic. Australas J Dermatol
44(4):256262
45. Robert S, Dawe JF (2003) Diagnosis and treatment of chronic actinic dermatitis. Dermatol
Ther 16(1):4551
46. Moseley H, Cameron H, MacLeod T, Clark C, Dawe R, Ferguson J (2001) New sunscreens
confer improved protection for photosensitive patients in the blue light region. Br J Dermatol
145(5):789794
47. Dawe R, Russell SFJ (1996) Borrowing from museums and industry: two photoprotective
devices. Br J Dermatol 135(6):10167
48. Paek SY, Lim HW (2014) Chronic actinic dermatitis. Dermatol Clin 32(3):355361, viiiix
49. Leigh IM, Hawk JL (1984) Treatment of chronic actinic dermatitis with azathioprine. Br
J Dermatol 110(6):691695
50. Murphy GM, Maurice PD, Norris PG, Morris RW, Hawk JL (1989) Azathioprine treatment
in chronic actinic dermatitis: a double-blind controlled trial with monitoring of exposure to
ultraviolet radiation. Br J Dermatol 121(5):639646
51. Ma Y, Lu Z (2010) Treatment with topical tacrolimus favors chronic actinic dermatitis: a
clinical and immunopathological study. J Dermatolog Treat 21(3):171177
52. Evans AV, Palmer RA, Hawk JLM (2004) Erythrodermic chronic actinic dermatitis respond-
ing only to topical tacrolimus. Photodermatol Photoimmunol Photomed 20(1):5961
53. Uetsu N, Okamoto H, Fujii K, Doi R, Horio T (2002) Treatment of chronic actinic dermatitis
with tacrolimus ointment. J Am Acad Dermatol 47(6):881884
54. Larangeira de Almeida H (2005) Successful treatment of chronic actinic dermatitis with topi-
cal pimecrolimus. Int J Dermatol 44(4):343344
55. Thomson MA, Stewart DG, Lewis HM (2005) Chronic actinic dermatitis treated with myco-
phenolate mofetil. Br J Dermatol 152(4):784786
56. Toonstra J, Henquet CJ, van Weelden H, van der Putte SC, van Vloten WA (1989) Actinic
reticuloid. A clinical photobiologic, histopathologic, and follow-u study of 16 patients. J Am
Acad Dermatol 21(2 Pt 1):205214
57. Norris PG, Camp RD, Hawk JL (1989) Actinic reticuloid: response to cyclosporine. J Am
Acad Dermatol 21(2 Pt 1):307309
3 Photoprotection for Photodermatoses 57
58. Wiseman MC, Orr PH, Macdonald SM, Schroeder ML, Toole JW (2001) Actinic prurigo:
clinical features and HLA associations in a Canadian Inuit population. J Am Acad Dermatol
44(6):952956
59. Birt AR, Davis RA (1971) Photodermatitis in North American Indians: familial actinic pru-
rigo. Int J Dermatol 10(2):107114
60. Surez A, Valbuena MC, Rey M, de Porras Quintana L (2006) Association of HLA subtype
DRB10407 in Colombian patients with actinic prurigo. Photodermatol Photoimmunol
Photomed 22(2):5558. doi:10.1111/j.1600-0781.2006.00205.x
61. Zuloaga-Salcedo S, Castillo-Vazquez M, Vega-Memije E et al (2007) Class I and class II
major histocompatibility complex genes in Mexican patients with actinic prurigo. Br
J Dermatol 156(5):10741075
62. Du P, Menage H, Vaughan RW, Baker CS et al (1996) HLA-DR4 May determine expression
of actinic prurigo in British patients. J Invest Dermatol 106(2):362364
63. Ferguson J, Ibbotson S (1999) The idiopathic photodermatoses. Semin Cutan Med Surg
18(4):257273
64. Ker KJ, Chong WS, Theng CTS (2013) Clinical characteristics of adult-onset actinic prurigo
in Asians: a case series. Indian J Dermatol Venereol Leprol 79(6):783788
65. Kerr AC, Ferguson J (2007) Actinic prurigo deterioration due to degradation of DermaGard
window film. Br J Dermatol 157(3):619620
66. Crouch R, Foley P, Baker C (2002) Actinic prurigo: a retrospective analysis of 21 cases
referred to an Australian photobiology clinic. Australas J Dermatol 43(2):128132
67. Daldon PE, Pascini M, Correa M (2010) Case for diagnosis. Actinic prurigo. An Bras
Dermatol 85(5):733735
68. Yong-Gee SA, Muir JB (2001) Long-term thalidomide for actinic prurigo. Australas
J Dermatol 42(4):281283
69. Trujillo MJ, de Barrio M, Rodrguez A, Moreno-Zazo M, Snchez I, Pelta R, Tornero P,
Herrero T (2001) Piroxicam-induced photodermatitis. Cross-reactivity among oxicams.
A case report. Allergol Immunopathol (Madr) 29(4):133136
70. Ikezawa Z, Kitamura K, Osawa J, Hariya T (1992) Photosensitivity to piroxicam is induced
by sensitization to thimerosal and thiosalicylate. J Invest Dermatol 98(6):918922
71. Gonalo M, Figueiredo A, Tavares P, Ribeiro CA, Teixeira F, Baptista AP (1992)
Photosensitivity to piroxicam: absence of cross-reaction with tenoxicam. Contact Dermatitis
27(5):287290
72. Kutlubay Z, Sevim A, Engin B, Tzn Y (2014) Photodermatoses, including phototoxic and
photoallergic reactions (internal and external). Clin Dermatol 32(1):7379
73. Bjrn LO, Huovinen P (2002) Phototoxicity. In: Bjrn LO (ed) Photobiology the science of
light and life, vol 18. Kluwer Academic Publishers, Dordrecht, pp 219238
74. Chantorn R, Lim HW, Shwayder TA (2012) J Am Acad Dermatol 67(6):1093.e1e18; quiz
11111112
75. Wan P, Moat S, Anstey A (2011) Pellagra: a review with emphasis on photosensitivity. Br
J Dermatol 164(6):11881200
76. Charman CR, Ryan A, Tyrrell RM, Pearse AD, Arlett CF, Kurwa HA (1998) Photosensitivity
associated with the SmithLemliOpitz syndrome. Br J Dermatol 138(5):885888
77. Anstey AV (1999) Photosensitivity in the Smith-Lemli-Opitz syndrome. Photodermatol
Photoimmunol Photomed 15(6):217218
78. Anstey AV, Ryan A, Rhodes LE, Charman CR, Arlett CF, Tyrrell RM, Taylor CR, Pearse AD
(1999) Characterization of photosensitivity in the Smith-Lemli-Opitz syndrome : a new
congenital photosensitivity syndrome. Br J Dermatol 141(3):406415
79. Nwokoro NA, Mulvihill JJ (1997) Cholesterol and bile acid replacement therapy in children
and adults with Smith-Lemli-Opitz (SLO/RSH) syndrome. Am J Med Genet 68(3):315321
80. OGorman SM, Murphy GM (2014) Photoaggravated disorders. Dermatol Clin 32(3):385398, ix
81. Zandman-Goddard G, Solomon M, Rosman Z, Peeva E, Shoenfeld Y (2012) Environment
and lupus-related diseases. Lupus 21(3):241250
58 D. Gutierrez and E.D. Baron
104. Fenton L, Ferguson J, Moseley H (2012) Analysis of energy saving lamps for use by photo-
sensitive individuals. Photochem Photobiol Sci 11(8):13461355
105. Procaccini EM, Riccio G, Casula L, Posteraro G, Monfrecola G (1997) Thirty hours evalu-
ation of UVB-induced erythema by chromometry and microflowmetry. Dermatology
195(4):317320
106. Khazova M, OHagan JB (2008) Optical radiation emissions from compact fluorescent
lamps. Radiat Prot Dosimetry 131(4):521525
107. Tamura D, DiGiovanna JJ, Khan SG, Kraemer KH (2014) Living with xeroderma pigmento-
sum: comprehensive photoprotection for highly photosensitive patients. Photodermatol
Photoimmunol Photomed 30(23):146152
108. Milota M, Jones DL, Cleaver J, Jamall IS (2011) Xeroderma pigmentosum family support
group: helping families and promoting clinical initiatives. DNA Repair (Amst) 10(7):792797
109. Webb S (2008) Xeroderma pigmentosum. BMJ 336(7641):444446
110. Yarosh D, Klein J, OConnor A, Hawk J, Rafal E, Wolf P (2001) Effect of topically applied
T4 endonuclease V in liposomes on skin cancer in xeroderma pigmentosum: a randomised
study. Xeroderma Pigmentosum Study Group. Lancet 357(9260):926929
111. Cainelli T, Di Padova C, Marchesi L, Gori G, Rovagnati P, Podenzani SA, Bessone E, Cantoni
L (1983) Hydroxychloroquine versus phlebotomy in the treatment of porphyria cutanea
tarda. Br J Dermatol 108(5):593600
112. Asawanonda P, Oberlender S, Taylor C (1999) The use of dihydroxyacetone for photoprotec-
tion in variegate porphyria. Int J Dermatol 38(12):9168
113. Harms J, Lautenschlager S, Minder CE, Minder EI (2009) An alpha-melanocyte-stimulating
hormone analogue in erythropoietic protoporphyria. N Engl J Med 360(3):306307
114. Mathews-Roth MM, Rosner B (2002) Long-term treatment of erythropoietic protoporphyria
with cysteine. Photodermatol Photoimmunol Photomed 18(6):307309
115. Sassa S (2006) Modern diagnosis and management of the porphyrias. Br J Haematol
135(3):281292
Chapter 4
Photoprotection and Photoaging
Key Points
Photoaging refers to the long-term effects of ultraviolet radiation on chron-
ically exposed skin.
Clinical manifestations of photoaging include wrinkling, pigmentary alter-
ations, and telangiectasias.
Characteristic histopathological abnormalities underlie these clinical
manifestations.
Despite advances in skin rejuvenation technologies, photoprotection
remains the most cost efficient and effective means of minimizing the clin-
ical effects of photoaging.
4.1 Introduction
Situated at the interface between the viscera and the physical world, the skin
provides protection from numerous environmental insults in real time. Although
skin possesses remarkable resiliency, it undergoes characteristic, often undesirable
B.J. Friedman
Department of Dermatology, Henry Ford Medical Center,
Detroit, MI, USA
H.W. Lim
Department of Dermatology, Henry Ford Medical Center New Center O,
Detroit, MI, USA
S.Q. Wang (*)
Division of Dermatology, Memorial Sloan-Kettering Cancer Center, Basking Ridge, NJ, USA
e-mail: [email protected]
functional and esthetic changes with time. One common paradigm in the study of
skin aging is to differentiate between intrinsic and extrinsic skin aging, of
which the first is a genetically influenced chronological process, while the latter is
driven primarily by environmental factors [1]. Ultimately, both forms of aging inter-
act in contributing to both a decline in skin structure and other cutaneous immuno-
logical, endocrinological, and neural functions. Recent work suggests that both
genetic and environmental factors in skin aging may actually share common path-
ways [2].
Of the various harmful environmental factors contributing to extrinsic aging,
ultraviolet (UV) light is considered to be the most significant and has also been the
most widely studied. Photoaging (or dermatoheliosis) refers to the effects of long-term
UV exposure and sun damage superimposed on intrinsically aging skin. Previous
work has suggested that up to 80 % of facial aging may be attributable to UV, although
other factors, such as cigarette smoking, may also promote premature facial wrin-
kling [3]. Photoaging is a universal phenomenon as the majority of light-skinned
individuals manifest some form of chronic sun damage before the age of 50 [4].
Some of the clinico-morphological manifestations of photoaging include fine
and coarse wrinkling, dyspigmentation, dry texture, increased laxity, telangiectasia,
and solar purpura. These adverse changes in skin appearance and integrity often
occur in parallel to the development of cutaneous malignances, a process referred to
as photocarcinogenesis. Persons of lighter skin tone, or low Fitzpatrick skin type,
and those with greater degrees of sun exposure (e.g., living in sunnier climates or
working outdoors) are preferentially affected by photoaging [57]. Within
individuals, sun-exposed areas such as the face, neck, extensor forearms and arms,
and dorsal hands are among the most susceptible to these changes.
Underlying the clinico-morphological features of photoaged skin are specific
histopathological alterations in epidermal and dermal structure. Ongoing research
advances in photobiology have helped illuminate various key molecular pathways
targeted by UV that induce these alterations. As the mechanisms of photoaging
continue to be better understood, newer therapeutic strategies for reversing these
processes and masking the photoaged phenotype continue to be developed. At the
current time, the most cost-effective therapy still remains primary prevention in the
form of sun avoidance, sun protective clothing, and use of sunscreens [8].
among the most prominent features seen with chronic UV exposure and are major
constituents of various photoaging scales aimed at quantifying a given individuals
degree of photodamage (Table 4.1) [1114]. Other clinical features of photoaging
include a dry leathery appearance, sallowness, vascular telangiectasia, sagging
appearance, and fragility (aka solar purpura). This is in contrast to sun-protected
skin, which ages in a more subtle fashion with increased laxity, fine wrinkling, and
the development of seborrheic keratosis. It notably lacks the pigmentary and
vascular changes characteristic of photoaging [15].
Individuals of Caucasian and Asian descent who sustain chronic UV exposure are
prone to developing solar lentigines (SLs). These benign lesions tend to present as
fixed tan to dark brown macules and patches on chronically sun-exposed skin and
are most commonly seen after the age of 50 (Fig. 4.1a) [16, 17]. SLs can be
contrasted from ephelides, which, despite also being induced by UV and having a
similar distribution and appearance to SLs, tend to be restricted to phototypes I and
II, are dynamic (i.e., become more pigmented during the summer months), develop
64 B.J. Friedman et al.
a b
Fig. 4.1 Clinical manifestations of photoaging. (a) Multiple scattered solar lentigines are seen on
this sun-damaged forearm (indicated by black arrows). (b) Coarse wrinkling is seen on the neck of
this elderly Caucasian patient
early on in childhood and adolescence, and partially disappear with age [18]. SLs
are actually seen more frequently in skin types III and IV, which has been thought
to be a result of more active melanocytes in those skin types [19, 20].
There are multiple lines of evidence that support the relationship of SLs to sun
exposure, beyond the observation that SLs have a predilection for sun-exposed skin.
In one large epidemiological study (n = 962), facial SLs were associated with
cumulative lifetime sun exposure, while SLs on the back were associated with
cumulative sun exposure and a sunburn history before the age of 20 [21]. Similar
findings were captured by Ezzidine et al. who showed that SLs were associated with
lifetime sun exposure in 523 French middle-age women [20]. A smaller case control
study failed to demonstrate a link of SLs to cumulative or occupational sun exposure
(n = 118), but did find an association between SLs and both frequent sunburns and
recreational sun exposure [19]. Additional indirect evidence supporting the link
between photoaging and the development of SLs comes from the phenomenon of
so-called PUVA lentigines. Patients treated with psoralens and ultraviolet A light
(PUVA) for inflammatory skin conditions tend to develop lentigines in otherwise
sun-protected areas [22, 23]. Although PUVA lentigines have definite
histopathological differences as compared with SLs (including more active
melanocytes with longer and more numerous dendrites and a higher frequency of
basal keratinocytes containing large, single melanosomes), these may be explained
by the higher potency and/or the pulse nature of PUVA treatment as compared with
natural sunlight [24].
4 Photoprotection and Photoaging 65
Hypermelanosis of the skin in the setting of chronic UV exposure may also mani-
fest as mottled or heterogeneous pigmentation, diffuse hyperpigmentation, pig-
mented actinic keratoses, and/or pigmented seborrheic keratosis [25]. Malignant
growths such as lentigo maligna and lentigo maligna melanoma represent less com-
mon causes of hypermelanosis in the skin, though they are almost exclusively seen in
the context of sun damage. Idiopathic guttate hypomelanosis (IGH) is the most well-
described yet poorly understood pattern of hypomelanosis seen in the context of skin
aging. Seen in over 80 % of those over the age of 70, IGH occurs in all phototypes,
though it is typically more apparent and striking in darker persons. IGH classically
presents as well-circumscribed and sharply defined whitish macules with a predilec-
tion for the forearms and shins. Although chronic UV exposure has been postulated
as a contributing factor to IGH, the cause may actually be multifactorial [2628].
4.2.2 Wrinkling
One of the telltale signs of skin aging is wrinkling. These rippled changes in the skin
surface presenting as variably sized creases and furrows may be most noticeable around
the forehead, eyes, cheeks, and neck (Fig. 4.1b). In some patients, wrinkles may form
interlacing patterns. Perhaps not surprisingly, studies have shown that there is a high
correlation between perceived age and the degree of facial wrinkling in persons [29].
Various forms of wrinkling have been described. Dynamic wrinkles are those that tem-
porarily result from contraction of underlying muscle fibers perpendicular to the direc-
tion of the visible skin lines. Over a period of time, these so-called facial expression
lines may further deepen and develop a static component. Static wrinkles develop in
thin stretched skin and are present even when underlying muscles are in a relaxed state.
The pathophysiology of wrinkling is complex, with likely contributions from intrinsic
skin aging, constant gravitational forces, intermittent positional pressures, repetitive facial
movements, pollution, and smoking [30]. Chronic UV exposure is thought to play a major
role in accelerating and accentuating skin lines and wrinkles, though the precise histo-
pathological correlate is still debated [31]. Furthermore, the presence of deep, coarser
wrinkles are thought to be a more prominent feature of photoaged skin as compared with
intrinsically aged skin [32]. In one recent cross-sectional study of a Mediterranean popu-
lation (n = 574), chronic sun exposure was found to be significantly associated with degree
of wrinkling (p <0.01), as assessed by the Daniell skin-wrinkling grading system [33].
Multiple other studies have demonstrated similar associations [3436].
There are a number of other phenotypic alterations that are seen with increasing age
and have a predilection for sun-exposed skin and in which chronic UV exposure is
thought to play a role. Sebaceous hyperplasia (SH) presents as small yellowish or
66 B.J. Friedman et al.
a b
sun-exposed skin as compared with sun-protected skin [43, 44]. Additional dermal
changes may include vasculature that is abnormally tortuous and dilated as well as
postcapillary venules displaying concentric wall thickening with deposits of base-
ment membrane-like material [45].
Epidermal thickness in photoaged skin can vary between individuals, demon-
strating either atrophy with effacement of the dermal-epidermal junction, hypertro-
phy with acanthosis, or no appreciable change [43]. Basement membrane thickening
can also be seen, which may signify chronic UV-mediated damage to the basal
keratinocytes. Melanocytes are irregularly distributed along the basal layer and can
vary widely in size, dendritic branching, and pigmentation [46, 47]. SL, when pres-
ent, demonstrates bulb-like elongation of the rete ridges with increased pigmenta-
tion of the basal keratinocytes located at the rete tips (Fig. 4.2b). Not infrequently,
biopsies of photoaged skin may display keratinocyte atypia, which presents as cel-
lular crowding of large and hyperchromatic-appearing cells. The atypia may extend
variably upward from the basal layer and is a marker of photo-carcinogenicity.
Various cultural pressures combined with increased age expectancy have created a
great demand for therapies which can restore a more youthful appearance. Crucial
in the pursuit and ultimately the development of new antiaging therapies is achieving
a more complete understanding of how chronic UV exposure initiates and propagates
the clinical effects of photoaging. Perhaps not surprisingly, this area of photobiology
has been and continues to be a subject of great interest and research. In recent years,
there have been many new and exciting advancements in the field, which have
begun to shed light on some of the key molecular pathways involved.
Both UVA (320400 nm) and UVB (290320) appear to contribute to photoaging,
although UVA is thought to play a greater role among the two. This is largely due to
the fact that UVA penetrates more deeply in the dermis and is at least 10 times more
abundant than UVB when it reaches the earths surface [48]. UVB is predominantly
absorbed by DNA in the epidermis and in turn induces various forms of cellular
damage through the formation of cyclobutane dimers. In this manner, UVB has been
shown to be a key component in the pathogenesis of sunburn, cutaneous immuno-
suppression, and photocarcinogenicity [49]. In contrast, much of the damage
inflicted by UVA is focused on dermal fibroblasts, extracellular matrix, and endo-
thelial cells through the generation of reactive oxygen species (ROS). UVA-induced
ROS have been shown to cause damage to various cellular compartments including
lipid membranes, DNA, and mitochondria. This damage may take the form of lipid
68 B.J. Friedman et al.
ROS
Collagen production
Collagen breakdown
Elastin accumulation
Extracellular
Matrix
Fig. 4.3 Schematic demonstrating the various key molecular steps thought to be involved in
photoaging (Adapted from Ref. [63])
factor (EGF), IL-1, and TNF-a are all activated in both keratinocytes and fibroblasts
[7]. Activation of these key proinflammatory receptors has been proposed to result
from ROS-induced oxidation and subsequent inhibition of protein-tyrosine phos-
phatases which normally function to downregulate these receptors [64, 66].
4.5 Photoprotection
As mentioned above, darker skin types tend to display the clinical signs of
photoaging much later in life as compared with lighter skin types [51]. This is
largely attributable to the increase in melanin in darker skin, which absorbs and
scatters detrimental UV rays [6]. An increase in melanin production occurs routinely
in response to UV irradiation and is one natural defense mechanism against
photocarcinogenesis and photoaging. This tanning response has previously been
shown to be induced by oligonucleotides containing thymine dinucleotides (pTpT),
which are formed as a result of UV damage [71]. Epidermal thickness is another
inherent property of the skin influencing the penetration of UV. A reactive increase
in skin thickness is a second intrinsic protective mechanism, with one study
demonstrating increased epidermal and dermal mitotic activity 2448 h after acute
UV exposure [72].
The presence of antioxidants may also blunt the chronic effects of UV. The skin is
equipped with a wide array of antioxidants, which normally serve to provide protec-
tion against oxidative stress developing in the context cellular respiration. A few
notable ones include vitamin E, coenzymeQ10 (CoQ10), ascorbate, carotenoids,
superoxide dismutase, catalase, and glutathione peroxidase [73]. There is indirect
evidence to suggest that these antioxidants may also be important in the skins defense
against UV exposure, which, as previously discussed, is known to generate harmful
ROS (see 6.4.1). UV irradiation has been shown to transiently deplete levels of cuta-
neous antioxidants [74]. It has been postulated that if such a depletion were to persist,
increased tissue damage and subsequent premature photoaging might occur [75].
4.5.2 Sunscreen
There have been only two randomized control studies performed to date investigating
the effects of sunscreen on photoaging. The first involved 53 patients with a history
of skin cancer who were randomly assigned to apply either sunscreen or placebo at
least twice a day. Skin biopsies were taken from preauricular skin at 0, 12, and
24 months, and epithelial thickness and dermal elastosis were assessed by blinded
4 Photoprotection and Photoaging 71
raters. Although 34 patients completed the study, complete data was only secured
from 16. At 24 months, there was significantly less elastosis among the sunscreen
group compared with the placebo group, but when repeated measurements were
accounted for in the analysis, no differences were observed between the treatment
groups [76].
The Nambour trial was a population-based intervention study conducted on a
community of adult residents living in Queensland, Australia, in the early 1990s
[77]. Subjects were categorized as daily sunscreen users versus discretionary
sunscreen users, depending on whether they were randomized to apply study-
provided sunscreen every day or to use their own discretion as to when/and if to
apply sunscreen. 903 adults under the age of 55 (mean age 39 years) had silicone-
based impressions taken on the dorsal left hand at baseline and then 4 years later.
The presence and severity of photoaging was assessed by blinded raters using the
Beagley and Gibson scale of microtopography grades, a previously validated
method for predicting the extent of dermal elastosis [78]. At the conclusion of the
follow-up period, the daily sunscreen group showed no detectable increase in skin
aging. Moreover, as compared with discretionary sunscreen users, daily sunscreen
users were 24 % less likely to show increased skin aging (relative odds = 0.76; 95 %
CI = 0.500.98) [79].
4.6 Conclusion
References
1. Kligman AM (1969) Early destructive effect of sunlight on human skin. JAMA 210(13):
23772380
2. Goukassian DA, Gilchrest BA (2004) The interdependence of skin aging, skin cancer, and
DNA repair capacity: a novel perspective with therapeutic implications. Rejuvenation Res
7(3):175185
3. Uitto J (1997) Understanding premature skin aging. N Engl J Med 337(20):14631465
4. Drake LA et al (1996) Guidelines of care for photoaging/photodamage. American Academy of
Dermatology. J Am Acad Dermatol 35(3 Pt 1):462464
72 B.J. Friedman et al.
32. Langton AK et al (2010) A new wrinkle on old skin: the role of elastic fibres in skin ageing.
Int J Cosmet Sci 32(5):330339
33. Ekiz O et al (2012) Factors influencing skin ageing in a Mediterranean population from Turkey.
Clin Exp Dermatol 37(5):492496
34. Chung JH et al (2001) Cutaneous photodamage in Koreans: influence of sex, sun exposure,
smoking, and skin color. Arch Dermatol 137(8):10431051
35. Rexbye H et al (2006) Influence of environmental factors on facial ageing. Age Ageing
35(2):110115
36. Raduan AP, Luiz RR, Manela-Azulay M (2008) Association between smoking and cutaneous
ageing in a Brazilian population. J Eur Acad Dermatol Venereol 22(11):13121318
37. Lesnik RH, Kligman LH, Kligman AM (1992) Agents that cause enlargement of sebaceous
glands in hairless mice. II. Ultraviolet radiation. Arch Dermatol Res 284(2):106108
38. Zouboulis CC, Boschnakow A (2001) Chronological ageing and photoageing of the human
sebaceous gland. Clin Exp Dermatol 26(7):600607
39. Innocenzi D et al (1993) UV-induced colloid milium. Clin Exp Dermatol 18(4):347350
40. Katoulis AC et al (2005) Poikiloderma of Civatte: a clinical and epidemiological study. J Eur
Acad Dermatol Venereol 19(4):444448
41. Mitchell RE (1967) A light and electron microscopical study of collagen degeneration. Aust N
Z J Surg 36(4):310318
42. Smith JG Jr et al (1962) Alterations in human dermal connective tissue with age and chronic
sun damage. J Invest Dermatol 39:347350
43. El-Domyati M et al (2002) Intrinsic aging vs. photoaging: a comparative histopathological,
immunohistochemical, and ultrastructural study of skin. Exp Dermatol 11(5):398405
44. Talwar HS et al (1995) Reduced type I and type III procollagens in photodamaged adult human
skin. J Invest Dermatol 105(2):285290
45. Lavker RM, Kligman AM (1988) Chronic heliodermatitis: a morphologic evaluation of
chronic actinic dermal damage with emphasis on the role of mast cells. J Invest Dermatol
90(3):325330
46. Gilchrest BA, Blog FB, Szabo G (1979) Effects of aging and chronic sun exposure on melano-
cytes in human skin. J Invest Dermatol 73(2):141143
47. Breathnach AS, Wyllie LM (1964) Electron microscopy of melanocytes and melanosomes in
freckled human epidermis. J Invest Dermatol 42:389394
48. Yaar M, Gilchrest BA (2007) Photoageing: mechanism, prevention and therapy. Br J Dermatol
157(5):874887
49. Benjamin CL et al (2008) p53 tumor suppressor gene: a critical molecular target for UV induc-
tion and prevention of skin cancer. Photochem Photobiol 84(1):5562
50. Han A, Chien AL, Kang S (2014) Photoaging. Dermatol Clin 32(3):291299, vii
51. Montagna W, Carlisle K (1991) The architecture of black and white facial skin. J Am Acad
Dermatol 24(6 Pt 1):929937
52. Sternlicht MD, Werb Z (2001) How matrix metalloproteinases regulate cell behavior. Annu
Rev Cell Dev Biol 17:463516
53. Scharffetter-Kochanek K (1997) Photoaging of the connective tissue of skin: its prevention and
therapy. Adv Pharmacol 38:639655
54. Fisher GJ et al (1997) Pathophysiology of premature skin aging induced by ultraviolet light. N
Engl J Med 337(20):14191428
55. Scharffetter K et al (1991) UVA irradiation induces collagenase in human dermal fibroblasts
in vitro and in vivo. Arch Dermatol Res 283(8):506511
56. Koivukangas V et al (1994) UV irradiation induces the expression of gelatinases in human skin
in vivo. Acta Derm Venereol 74(4):279282
57. Fisher GJ et al (1996) Molecular basis of sun-induced premature skin ageing and retinoid
antagonism. Nature 379(6563):335339
58. Sato H, Seiki M (1993) Regulatory mechanism of 92 kDa type IV collagenase gene expression
which is associated with invasiveness of tumor cells. Oncogene 8(2):395405
74 B.J. Friedman et al.
59. Angel P et al (1987) Phorbol ester-inducible genes contain a common cis element recognized
by a TPA-modulated trans-acting factor. Cell 49(6):729739
60. Fisher GJ, Voorhees JJ (1998) Molecular mechanisms of photoaging and its prevention by reti-
noic acid: ultraviolet irradiation induces MAP kinase signal transduction cascades that induce
Ap-1-regulated matrix metalloproteinases that degrade human skin in vivo. J Investig Dermatol
Symp Proc 3(1):6168
61. Fisher GJ et al (2000) c-Jun-dependent inhibition of cutaneous procollagen transcription fol-
lowing ultraviolet irradiation is reversed by all-trans retinoic acid. J Clin Invest 106(5):
663670
62. Quan T et al (2002) Ultraviolet irradiation alters transforming growth factor beta/smad path-
way in human skin in vivo. J Invest Dermatol 119(2):499506
63. Chen L, Hu JY, Wang SQ (2012) The role of antioxidants in photoprotection: a critical review.
J Am Acad Dermatol 67(5):10131024
64. Gross S et al (1999) Inactivation of protein-tyrosine phosphatases as mechanism of UV-induced
signal transduction. J Biol Chem 274(37):2637826386
65. Heck DE et al (2004) Solar ultraviolet radiation as a trigger of cell signal transduction. Toxicol
Appl Pharmacol 195(3):288297
66. Barford D, Jia Z, Tonks NK (1995) Protein tyrosine phosphatases take off. Nat Struct Biol
2(12):10431053
67. Kimlin MG, Guo Y (2012) Assessing the impacts of lifetime sun exposure on skin damage and
skin aging using a non-invasive method. Sci Total Environ 425:3541
68. Ichihashi M et al (2003) UV-induced skin damage. Toxicology 189(1-2):2139
69. Grob JJ et al (1993) Study of sunbathing habits in children and adolescents: application to the
prevention of melanoma. Dermatology 186(2):9498
70. Glaser DA (2003) Anti-aging products and cosmeceuticals. Facial Plast Surg Clin North Am
11(2):219227
71. Gilchrest BA, Eller MS (1999) DNA photodamage stimulates melanogenesis and other photo-
protective responses. J Investig Dermatol Symp Proc 4(1):3540
72. Epstein JH, Fukuyama K, Fye K (1970) Effects of ultraviolet radiation on the mitotic cycle and
DNA, RNA and protein synthesis in mammalian epidermis in vivo. Photochem Photobiol
12(1):5765
73. Shindo Y et al (1994) Enzymic and non-enzymic antioxidants in epidermis and dermis of
human skin. J Invest Dermatol 102(1):122124
74. Fuchs J (1998) Potentials and limitations of the natural antioxidants RRR-alpha-tocopherol,
L-ascorbic acid and beta-carotene in cutaneous photoprotection. Free Radic Biol Med
25(7):848873
75. Shindo Y, Hashimoto T (1997) Time course of changes in antioxidant enzymes in human skin
fibroblasts after UVA irradiation. J Dermatol Sci 14(3):225232
76. Boyd AS et al (1995) The effects of chronic sunscreen use on the histologic changes of derma-
toheliosis. J Am Acad Dermatol 33(6):941946
77. Green A et al (1994) The Nambour skin cancer and actinic eye disease prevention trial: design
and baseline characteristics of participants. Control Clin Trials 15(6):512522
78. Hughes MC et al (2012) Validation of skin surface microtopography as a measure of skin
photoaging in a subtropical population aged 40 and over. Photodermatol Photoimmunol
Photomed 28(3):153158
79. Hughes MC et al (2013) Sunscreen and prevention of skin aging: a randomized trial. Ann
Intern Med 158(11):781790
Chapter 5
Photoprotection, Photoimmunology
and Autoimmune Diseases
Key Points
There are several autoimmune diseases that are known to be aggravated by
sun exposure, such as lupus erythematosus and dermatomyositis.
Both UVB and UVA have been implicated in lupus erythematosus, and
UVB has been implicated in dermatomyositis.
Photoprotection is an important part of management in these patients.
5.1 Photoprotection
part include behaviour to avoid undue exposure to UVR, the use of UV protective
clothing and the use of sunscreens.
When UVR hits the skin, the photons are either absorbed, reflected back by the
stratum corneum, or scattered within the skin and subsequently absorbed by
chromophore(s). Keratin is very protective, so areas of the body with thick layers of
keratin such as palms and heels are well protected. Melanin is more complex;
dark-skinned people have mainly eumelanin as the protective pigment in skin,
whereas fair-skinned people have pigment comprised of eumelanin and
phaeomelanin, the latter being yellow/red in appearance. Eumelanin absorbs UVR
and dissipates energy harmlessly. Phaeomelanin is more photochemically active
and generates free radicals which lead to DNA damage. Over time direct DNA
damage and indirect oxidative stress, unless repaired, can give rise to the adverse
consequences of exposure to UV radiation which include photoaging, photoderma-
toses and photocarcinogenesis.
Photoprotection may be achieved by limiting ones personal UVR exposure. The
main ways to achieve this include sun avoidance at times of UVR increased intensity,
i.e., midday 3 h, awareness of the effects of latitude, altitude and prolonged
exposure especially in summer months, seeking of shade, wearing of UVR protective
clothing/broad-brimmed hat and the use of sun-protective products. The majority of
our personal exposure to UVR comes from sun exposure. Other additional sources
include artificial lighting, medical equipment, tanning booths, etc. The amount of
sunlight we receive depends on ambient UV, our surrounding environment and our
behaviour. Geographical location, cloud cover, terrain (snow, white sand, sparkling/
rippling water) and surrounding environment (urban/open space) all affect our total
exposure to UV radiation. Some of these considerations are beyond our control;
however, our behaviour while outdoors can have a significant impact on our personal
exposure to UV light. Our work environment; the time we spend outdoors, where
we take part in physical activities; and overseas holidays all affect our total UV
exposure.
Photoprotection can be achieved in four ways. The first three are essentially free
and the last, the more expensive way of protecting oneself from UV radiation, is the
most advertised method.
1. Sun avoidance considerable protection from UV exposure can be achieved by
avoiding the 3 h surrounding solar noon (for both temperate and tropical
latitudes). During this time approximately 50 % of the days total UV exposure
during summer occurs.
2. Shade this can be provided by natural objects such as trees and cliffs or by
man-made buildings, umbrellas, etc. At noon, 50 % of the UV reaching our skin
comes from direct sunlight and the other half is from scatter from the sky. Shade
will remove all of the direct sunlight that reaches our skin. Shade can also remove
part of the diffuse (scattered) sunlight.
3. Protective clothing/hats the level of UV protection is primarily based on the
tightness of the garments weave; hence tightly woven cotton offers good
protection. This has been extensively tested by the Australian Radiation
5 Photoprotection, Photoimmunology and Autoimmune Diseases 77
Protection and Nuclear Safety Agency (ARPANSA) [1] and other regulatory
agencies. It has been shown that regular clothing provides good sun protection;
hence specialized clothing is not necessary in order to be adequately protected
from the sun.
4. Sunscreens these are the most popular means of photoprotection. In order for
them to provide good protection from UV exposure, they should have safe and
effective ingredients, stability including photostability and a good physical
structure which feels comfortable on the skin so that compliance rates are high
and should be reliable (namely, realistic claims regarding their SPF/UVA
protection/water resistance).
Ideally one should practice sun avoidance during peak times, first and foremost,
followed by a combination of shade seeking, protective clothing/hat and the use of
sunscreens.
5.2 Photoimmunology
This is the study of the effects of solar radiation on the immune system. Solar
radiation is divided into different regions of the electromagnetic spectrum, UVC
(<280 nm), UVB (280315 nm) and UVA (315400 nm), which is CIE (Commission
Internationale lEclairage) [2] definition based on physiological effects by photons
in those regions. The concept that UV, which is a part of the external environment,
can affect the immune response, whose components are internal, arose from
experiments in the 1970s by Kripke et al. [3] aimed at discovering a role for the
immune response in rejecting UV radiation-induced skin cancer cells [4]. Their
work demonstrated that UV radiation exerted suppressive effects on immune
responses to UV-induced skin cancer cells and that the inhibition of tumour rejection
could be adoptively transferred by T lymphocytes in an antigen-specific manner.
Many laboratories have subsequently studied this topic and revealed details on how
UV radiation induced the suppression of immune responses (also known as immune
tolerance). This suppression was observed not only for skin cancers but also
infectious agents and chemical antigens. UVR affects the immune response by
affecting the cells and other components of the immune system to alter the balance
and functions of immune cells to infectious agents, chemicals and skin cancer. We
know sunlight is immunosuppressive, as UVB depletes Langerhans cells and
diminishes their function such that antigens are not presented efficiently to the
immune system [5, 6]. Alterations in delayed hypersensitivity also occur such that
if a universal allergen is applied to UV-irradiated skin, no response occurs; rather a
population of antigen-specific T cells displaying tolerance to the antigen emerges.
In mouse models, tolerance may be passively transferred to other mice conferring
tolerance to the antigen. These observations also apply to UV-induced skin tumours,
underwriting the importance of cell-based immunity in skin surveillance and the
prevention of skin cancer. Chronically UV-exposed skin is immunosuppressed and
78 G.M. Murphy and N. Ralph
more prone to develop viral warts. Acute exposure to UVR may also precipitate
herpes simplex infections. Many viral diseases become more manifest in
UV-damaged or the exposed skin. This has also been noted in many photoaggravated
diseases.
These are diseases in which impaired function and the destruction of tissue are
caused by autoantibodies formation. Autoimmune diseases include a wide variety
of disorders which can affect internal organs, muscles, joints and the skin. Some of
these diseases may be exacerbated by UV radiation; hence photoprotection is
imperative in the management of such diseases.
Photodermatoses may be classified into idiopathic photodermatoses, genoder-
matoses, photoaggravated dermatoses and photodermatoses which are secondary to
exogenous agents including photoallergy/phototoxicity. In this chapter, we will dis-
cuss photoaggravated disorders of the autoimmune category (Table 5.1).
Immunologically mediated photodermatoses with no or minimal systemic mani-
festations other than in the skin include polymorphic light eruption (PLE), juvenile
spring eruption, actinic prurigo, solar urticaria, chronic actinic dermatitis and
hydroa vacciniforme; these are covered in another chapter.
5 Photoprotection, Photoimmunology and Autoimmune Diseases 79
5.3.1.1 Introduction
5.3.1.2 History
In the mid-nineteenth century, Cazenave first coined the term lupus erythematosus
[7] and helped to differentiate it from lupus vulgaris, a cutaneous form of
tuberculosis. In the mid-twentieth century, Dubois described the spectrum of disease
seen with lupus ranging from cutaneous involvement to a multisystem disease [8].
5.3.1.3 Epidemiology
5.3.1.4 Pathogenesis
prolonged exposure of DNA and extractable nuclear antigen to the immune system,
generating anti-DNA and ENA antibodies [21, 22]. The mechanisms underlying
this process have been reviewed recently [23, 24]. UV light also up-regulates
adhesion molecules, such as ICAM-1 [25] in patients with LE. Nitric oxide synthase
capable of inducing cytokines (iNOS) is released from keratinocytes following UV
irradiation. In patients with LE this release is delayed but prolonged [26]. The
promoter polymorphism 308A of TNF- is seen with increased frequency in SCLE
[27] and transcription is photoregulated [28].
The development of the skin lesions after UV injury may be delayed (days to
3 weeks) and the lesions may persist for months. This may make it difficult to elicit
a history of photosensitivity from the patient, as the delay between UV exposure
and exacerbation means patients may not make this association. UVA and UVB
radiation are both implicated in the pathogenesis of LE in both in vitro [29] and
in vivo [30] studies. One study documented that photosensitivity in cutaneous LE
was UVB induced in 33 % of cases and UVA induced in 14 %, and in the majority
of cases (53 %), it was mediated by a combination of UVB and UVA [14]. Nonsolar
sources of UV, such as photocopiers [31], fluorescent and some energy-saving light
sources [32], can aggravate LE. Interestingly, while UVB consistently aggravates
LE, studies documented reduction in LE disease activity with UVA-1 (340400 nm)
irradiation [3335]. Subtypes of LE appear to have varying degrees of
photoaggravation, with lupus tumidus [36] and SCLE appearing to be the most
photosensitive of the LE subtypes [37], although one study of phototesting with
UVA, UVB and visible light in 100 patients (24 with SLE, 30 with SCLE and 46
with DLE) found no association between photosensitivity and LE subtype [38].
Phototesting is not routinely required in clinical practice to make a diagnosis of LE,
as clinical history, examination, serologic studies and skin biopsy for histology and
direct immunofluorescence suffice.
Polymorphous light eruption (PLE) is seen commonly in patients with LE
(49 %), and the onset of PLE precedes the onset of LE by over 7 years in half of
patients. This suggests that there may be features of pathogenesis common to both
entities and that PLE may predispose to LE in a subset of patients [39].
The clinical presentation across the subtypes of LE is very diverse. The underlying
disease process however is very similar. Patients with SLE can present acutely with
internal organ damage secondary to circulating autoantibodies. No organ is protected
from the immune-mediated destruction and SLE can present with arthralgia, central
nervous system involvement, nephritis, pleuritis and vasculitis. At the other end of
the spectrum, patients with other forms of LE may develop cutaneous lesions and
never progress to systemic involvement.
Cutaneous manifestations of LE are varied and can be divided into LE-nonspecific
and LE-specific lesions. LE-nonspecific mucocutaneous manifestations include oral
ulcers, Raynauds syndrome, scarring alopecia and vasculitis. The characteristic
5 Photoprotection, Photoimmunology and Autoimmune Diseases 81
5.3.1.6 Investigations
5.3.1.7 Serology
SLE is the LE subtype most strongly associated with positive antinuclear antibody
(ANA) and double-stranded DNA (ds-DNA) [10]. Given the propensity of SLE to
affect internal organs, a comprehensive systemic workup including complete blood
count, complement levels and renal function tests including urinalysis should be
undertaken. The presence of anti-Ro (SS-A) and anti-La (SS-B) is associated with
an abnormal photoprovocation reaction [44]. Anti-Ro (SS-A) is found in 72 % of
patients with SCLE, 47 % of patients with acute cutaneous LE and 22 % of patients
with chronic cutaneous LE. Anti-La antibodies are found in 36 % of patients with
82 G.M. Murphy and N. Ralph
5.3.1.8 Treatment
Essential lifestyle changes for patients with cutaneous lupus erythematosus are
photoprotection [46] and smoking cessation. Broad-spectrum sunscreen is required
[47] due to the implication of both UVA and UVB in the action spectrum of
LE. Vitamin D levels may be reduced in patients with cutaneous LE who practice
rigorous photoprotection; hence supplementation is often required [5]. Topical
corticosteroids and antimalarials [48] are first-line treatment options for cutaneous
LE. Second-line treatment options include dapsone [49], thalidomide [50], oral
retinoids [51] and immunosuppressant medications such as mycophenolate mofetil
[52, 53], azathioprine and methotrexate. Every patient with cutaneous and systemic
LE should be evaluated to ensure no causative drug is implicated.
5.3.2 Dermatomyositis
This is an autoimmune disease of the skin and striated muscle. It is associated with
an increased risk of malignancy, the risk being highest in the first 3 years after
diagnosis of myositis but increased for up to 5 years [54].
5.3.2.1 History
Bohan and Peter compiled generally accepted diagnostic criteria in 1975 [55, 56].
A more recent revision of the classification means that myositis is no longer required
for diagnosis [57].
5.3.2.2 Epidemiology
5.3.3 Pathogenesis
The skin and striated muscle may be affected to varying degrees. Cutaneous find-
ings accompany muscle involvement in 60 % of cases and precede it in 30 %.
Dermatomyositis sine myositis where there is no muscle involvement is described
in approximately 10 % of cases. Cutaneous findings include blue/violaceous or
erythematous plaques and patches which may be edematous affecting the face and
the V of the neck. The pathognomonic heliotrope rash refers to a lilac discoloration
of the eyelids with periorbital oedema. The dorsum of the hands may demonstrate
mauve linear plaques along the back of the fingers and dusky erythematous papules
with atrophy, termed Gottrons papules, over the joints. They tend to spare the skin
in between the joints. These Gottrons papules represent a hyperkeratotic response
to inflammation. The nail folds may be hyperkeratotic with haemorrhage. Capillary
microscopy of the nail fold vessels shows coiling and enlargement. Photosensitivity
is seen with poikilodermatous change (atrophy, hyper/hypopigmentation,
telangiectasia).
Proximal muscles are those most affected; the quadriceps and triceps are
symmetrically involved with a slow onset of weakness and myalgia. Patients may
report difficulty getting up from a seated position. Distal muscles are involved in the
advanced disease state. Involvement of the pharyngeal muscles may occur and
manifests as dyspnoea or dysphagia. Complications include myocarditis, pulmonary
fibrosis and vasculitis.
Dermatomyositis may be chronic or can spontaneously remit in 23 years. If an
underlying malignancy is present, removal can result in rapid resolution of the
symptoms.
5.3.3.2 Investigations
5.3.3.3 Serology
5.3.3.4 Phototesting
5.3.3.5 Treatment
described. The condition is much more common in females and the diagnosis is usu-
ally made in those older than 40 years of age. People with this condition may also
experience joint pains, fatigue, salivary gland swelling and xerosis. This condition
may also have systemic involvement affecting organs such as the thyroid, lung, liver
and kidneys. Due to this the cutaneous manifestations are often minimized, albeit
relatively common. Cutaneous manifestations associated with primary SS include
photosensitive rashes in the context of positive anti-Ro antibodies, alopecia, annular
erythema, B cell lymphoma, vasculitis and vitiligo [67].
5.3.6 Pemphigus
5.3.7 Psoriasis
In the majority of patients with psoriasis, UV therapies are used to control the
disease; however there is a defined subset of patients, 520 %, whom have an
exacerbation of their psoriasis with UV exposure [7274]. They tend to be older
females with a positive family history of psoriasis and early onset of disease;
patients are more likely to have psoriasis affecting their hands [75, 76]. There is a
strong association with HLA-Cw*0602 [74]. Approximately 50 % develop PLE
following sun exposure with subsequent development of psoriasis in the lesions of
PLE, while the other 50 % have photoaggravated psoriasis with no associated PLE
[73]. In the former group, symptoms of PLE followed by psoriasis were more easily
provoked by UVA, while in the second group, UVB was the more effective
photoaggravating spectrum [76]. If there is a question regarding photoaggravated
psoriasis, patients should undergo confirmatory phototesting. Photoprotection is
advisable in these patients.
photoaggravated atopic dermatitis; if they are elderly, they may fall into another
cohort of patients with chronic actinic dermatitis. The action spectrum is usually
similar to the erythema action spectrum, in which case the patient may be labelled
chronic actinic dermatitis. If the patient has UVA photosensitivity, then
photosensitivity caused by a drug should be suspected.
This is a rare disease which mainly affects elderly men (~10 % female). It is also
rarely found in HIV. They may give a history of previous atopy or allergic contact
dermatitis to compositate [77]. They have severely lichenified, xerotic skin affecting
the face, neck and dorsal hands most commonly with sparing behind the ears and
inferior chin. Hypopigmentation may also be seen in CAD, and this seems to be
post-inflammatory hypopigmentation. Skin biopsy from the affected areas may
show an eczematous eruption particularly if biopsied following monochromator
testing, but if severe it may also resemble a cutaneous T-cell lymphoma/reticulosis;
hence, the other name it is also known by actinic reticuloid is sometimes used.
There is a dense dermal lymphocytic dermatitis with atypical cerebriform
lymphocytic cells identical to those in cutaneous T-cell lymphoma.
The eruption can be provoked by very minimal UV exposure. It is usually worse
in the summer months and may improve in some individuals in the winter months.
It is therefore more easily diagnosed in those with severe, year-round photosensitivity
and disabling effects. If left untreated it may progress to erythroderma. Phototesting
(monochromator) is essential to make the diagnosis of CAD. In the absence or
abnormal tests, the diagnosis cannot be made. The pattern of the action spectrum in
90 % of patients with CAD implicates DNA as the chromophore, as the action
spectrum is identical to the erythema action spectrum, i.e., UVB, but occurring at a
lower dose [78]. The minority of patients exhibit UVA photosensitivity, but this is
more commonly the pattern of drug-induced photosensitivity; thus drugs should be
excluded in such cases [79].
Treatment includes UV protection (clothing, hat, broad-spectrum sunscreens)
and sun avoidance during peak hours, topical tacrolimus, and, in more severe cases,
azathioprine, cyclosporine, or mycophenolate mofetil. For patients who are
extremely photosensitive (UVB, UVA extending into visible light), this condition
can be an extremely debilitating disease.
5.3.10 Rosacea
This is a common inflammatory skin condition mainly affecting the face of middle-
aged adults. Four major clinical subtypes have been identified: erythematotelangi-
ectatic, papulopustular, phymatous and ocular. Rosacea is characterized by facial
88 G.M. Murphy and N. Ralph
flushing, erythema, chronic inflammation in the form of papules and pustules and
fibrosis. Some aspects of the pathophysiology of rosacea has been characterized in
more detail in recent years; however the interplay of these dysregulated systems is
still poorly understood. UVR effects on rosacea have yet to be fully understood as
many trials to date show conflicting reports. Does the UVR induce damage to the
dermal connective tissue permitting vasodilatation and vascular pooling which
leads to erythema? Is it also that in the majority of rosacea patients who describe
photoaggravated rosacea, there is failure to downgrade the immune response to
UVR? Or does UVR actually have a beneficial effect by decreasing inflammation
resulting in skin clearance as seen in some patients undergoing phototherapy for
coexisting conditions?
This is a rare, acquired, chronic subepidermal bullous disease of the skin and mucosa
due to autoantibodies to type VII collagen structures which are a major component
of anchoring fibrils which attach the dermis to the epidermis. The autoantibodies
which are either bound or circulating attack type VII collagen resulting in a reduc-
tion or change of normally functioning anchoring fibrils. Patients present with skin
fragility, blistering, erosions, scarring, milia formation and sometimes loss of nails.
There is not one satisfactory treatment for this condition but some therapeutic suc-
cess has been achieved with the use of colchicine, dapsone, infliximab, intravenous
immunoglobulins and plasmapheresis [80]. There have been rare case reports of this
condition with sensitivity to both UVA and UVB radiation [81].
This is a type of lymphoma which presents in the skin but may evolve, often over
many years to have systemic involvement. Mycosis fungoides, first described by a
French physician Jean Luis Marc Alibert in 1806, is the most common type of
CTCL. Patients present most commonly from the 4th decade onward with patches
or plaques which can sometimes be misdiagnosed as eczema initially. Children may
be affected but it is much more common in adults and also in males more so than
females. Patients may also present with erythroderma and some progress to tumour
stage with lymphadenopathy. Pruritus is commonly associated with this condition.
Unfortunately there is no cure for this disease and the treatment is about control of
skin eruption and symptomatic relief. One of the first-line treatments is phototherapy,
both NB-UVB and PUVA. There are case reports of photosensitive mycosis
fungoides [82], whereby patients may present with actinic reticuloid-type pattern
of facial features, also known as chronic actinic dermatitis; however biopsies of the
skin combined with T-cell gene rearrangement studies confirm it to be CTCL. These
5 Photoprotection, Photoimmunology and Autoimmune Diseases 89
patients are not photosensitive and usually respond very well to PUVA despite the
eruption clinically mimicking CAD. This disease may progress over a very short
period of time to tumour stage or may smoulder for many decades.
Some of the photoaggravated autoimmune diseases have a known action
spectrum; however, as the exact action spectra are not fully defined for all of the
photoaggravated autoimmune diseases, a key component of their management
includes photoprotection with behavioural change, UV protective clothing and
broad-spectrum sunscreen.
References
18. Golan TD, Elkon KB, Gharavi AE, Krueger JG (1992) Enhanced membrane binding of
autoantibodies to cultured keratinocytes of systemic lupus erythematosus patients after
ultraviolet B/ultraviolet A irradiation. J Clin Invest 90(3):10671076
19. Furukawa F, Kashihara-Sawami M, Lyons MB, Norris DA (1990) Binding of antibodies to the
extractable nuclear antigens SS-A/Ro and SS-B/La is induced on the surface of human
keratinocytes by ultraviolet light (UVL): implications for the pathogenesis of photosensitive
cutaneous lupus. J Invest Dermatol 94(1):7785
20. Bennion SD, Ferris C, Lieu TS, Reimer CB, Lee LA (1990) IgG subclasses in the serum and
skin in subacute cutaneous lupus erythematosus and neonatal lupus erythematosus. J Invest
Dermatol 95(6):643646
21. Caricchio R, McPhie L, Cohen PL (2003) Ultraviolet B radiation-induced cell death: critical
role of ultraviolet dose in inflammation and lupus autoantigen redistribution. J Immunol
171(11):57785786
22. Majai G, Kiss E, Tarr T et al (2014) Decreased apopto-phagocytic gene expression in the mac-
rophages of systemic lupus erythematosus patients. Lupus 23:133145
23. Janko C, Schorn C, Grossmayer GE et al (2008) Inflammatory clearance of apoptotic remnants
in systemic lupus erythematosus (SLE). Autoimmun Rev 8(1):912
24. Yu C, Chang C, Zhang J (2013) Immunologic and genetic considerations of cutaneous lupus
erythematosus: a comprehensive review. J Autoimmun 41:3445
25. Norris DA, Lyons MB, Middleton MH, Yohn JJ, Kashihara-Sawami M (1990) Ultraviolet
radiation can either suppress or induce expression of intercellular adhesion molecule 1 (ICAM-
1) on the surface of cultured human keratinocytes. J Invest Dermatol 95(2):132138
26. Kuhn A, Fehsel K, Lehmann P, Krutmann J, Ruzicka T, Kolb-Bachofen V (1998) Aberrant
timing in epidermal expression of inducible nitric oxide synthase after UV irradiation in
cutaneous lupus erythematosus. J Invest Dermatol 111(1):149153
27. Millard TP, Kondeatis E, Cox A et al (2001) A candidate gene analysis of three related photo-
sensitivity disorders: cutaneous lupus erythematosus, polymorphic light eruption and actinic
prurigo. Br J Dermatol 145(2):229236
28. Werth VP, Zhang W, Dortzbach K, Sullivan K (2000) Association of a promoter polymorphism
of tumor necrosis factor-alpha with subacute cutaneous lupus erythematosus and distinct
photoregulation of transcription. J Invest Dermatol 115(4):726730
29. Golan TD, Foltyn V, Roueff A (1991) Increased susceptibility to in vitro ultraviolet B radiation
in fibroblasts and lymphocytes cultured from systemic lupus erythematosus patients. Clin
Immunol Immunopathol 58(2):289304
30. Nived O, Johansen PB, Sturfelt G (1993) Standardized ultraviolet-A exposure provokes skin
reaction in systemic lupus erythematosus. Lupus 2(4):247250
31. Klein LR, Elmets CA, Callen JP (1995) Photoexacerbation of cutaneous lupus erythematosus
due to ultraviolet A emissions from a photocopier. Arthritis Rheum 38(8):11521156
32. Rihner M, McGrath H Jr (1992) Fluorescent light photosensitivity in patients with systemic
lupus erythematosus. Arthritis Rheum 35(8):949952
33. McGrath H Jr (1999) Ultraviolet A1 (340-400 nm) irradiation and systemic lupus erythemato-
sus. J Investig Dermatol Symp Proc 4(1):7984
34. Molina JF, McGrath H Jr (1997) Long term ultraviolet-A1 irradiation therapy in systemic
lupus erythematosus. J Rheumatol 24(6):10721074
35. McGrath H, Martinez-Osuna P, Lee FA (1996) Ultraviolet-A1 (340-400 nm) irradiation ther-
apy in systemic lupus erythematosus. Lupus 5(4):269274
36. Kuhn A, Sonntag M, Richter-Hintz D et al (2001) Phototesting in lupus erythematosus tumi-
dusreview of 60 patients. Photochem Photobiol 73(5):532536
37. Chlebus E, Wolska H, Blaszczyk M, Jablonska S (1998) Subacute cutaneous lupus erythema-
tosus versus systemic lupus erythematosus: diagnostic criteria and therapeutic implications.
J Am Acad Dermatol 38(3):405412
38. Sanders CJ, Van Weelden H, Kazzaz GA, Sigurdsson V, Toonstra J, Bruijnzeel-Koomen CA
(2003) Photosensitivity in patients with lupus erythematosus: a clinical and photobiological
study of 100 patients using a prolonged phototest protocol. Br J Dermatol 149(1):131137
5 Photoprotection, Photoimmunology and Autoimmune Diseases 91
39. Nyberg F, Hasan T, Puska P et al (1997) Occurrence of polymorphous light eruption in lupus
erythematosus. Br J Dermatol 136(2):217221
40. Tan EM, Cohen AS, Fries JF et al (1982) The 1982 revised criteria for the classification of
systemic lupus erythematosus. Arthritis Rheum 25(11):12711277
41. Hochberg MC (1997) Updating the American College of Rheumatology revised criteria for the
classification of systemic lupus erythematosus. Arthritis Rheum 40(9):1725
42. Duarte-Garcia A, Fang H, To CH, Magder LS, Petri M (2012) Seasonal variation in the activity
of systemic lupus erythematosus. J Rheumatol 39(7):13921398
43. Szeto CC, Mok HY, Chow KM et al (2008) Climatic influence on the prevalence of noncutane-
ous disease flare in systemic lupus erythematosus in Hong Kong. J Rheumatol
35(6):10311037
44. Hasan T, Nyberg F, Stephansson E et al (1997) Photosensitivity in lupus erythematosus, UV
photoprovocation results compared with history of photosensitivity and clinical findings. Br
J Dermatol 136(5):699705
45. Tsukazaki N, Watanabe M, Shimizu K, Hamasaki Y, Katayama I (2002) Photoprovocation test
and immunohistochemical analysis of inducible nitric oxide synthase expression in patients
with Sjogrens syndrome associated with photosensitivity. Br J Dermatol 147(6):
11021108
46. Stege H, Budde MA, Grether-Beck S, Krutmann J (2000) Evaluation of the capacity of sun-
screens to photoprotect lupus erythematosus patients by employing the photoprovocation test.
Photodermatol Photoimmunol Photomed 16(6):256259
47. Kuhn A, Gensch K, Haust M et al (2011) Photoprotective effects of a broad-spectrum sun-
screen in ultraviolet-induced cutaneous lupus erythematosus: a randomized, vehicle-con-
trolled, double-blind study. J Am Acad Dermatol 64(1):3748
48. Seiger E, Roland S, Goldman S (1991) Cutaneous lupus treated with topical tretinoin: a case
report. Cutis 47(5):351355
49. Neri R, Mosca M, Bernacchi E, Bombardieri S (1999) A case of SLE with acute, subacute and
chronic cutaneous lesions successfully treated with Dapsone. Lupus 8(3):240243
50. Duong DJ, Spigel GT, Moxley RT 3rd, Gaspari AA (1999) American experience with low-
dose thalidomide therapy for severe cutaneous lupus erythematosus. Arch Dermatol 135(9):
10791087
51. Newton RC, Jorizzo JL, Solomon AR Jr et al (1986) Mechanism-oriented assessment of
isotretinoin in chronic or subacute cutaneous lupus erythematosus. Arch Dermatol 122(2):
170176
52. Schanz S, Ulmer A, Rassner G, Fierlbeck G (2002) Successful treatment of subacute cutane-
ous lupus erythematosus with mycophenolate mofetil. Br J Dermatol 147(1):174178
53. Kreuter A, Tomi NS, Weiner SM, Huger M, Altmeyer P, Gambichler T (2007) Mycophenolate
sodium for subacute cutaneous lupus erythematosus resistant to standard therapy. Br
J Dermatol 156(6):13211327
54. Buchbinder R, Forbes A, Hall S, Dennett X, Giles G (2001) Incidence of malignant disease in
biopsy-proven inflammatory myopathy. A population-based cohort study. Ann Intern Med
134(12):10871095
55. Bohan A, Peter JB (1975) Polymyositis and dermatomyositis (first of two parts). N Engl J Med
292(7):344347
56. Bohan A, Peter JB (1975) Polymyositis and dermatomyositis (second of two parts). N Engl
J Med 292(8):403407
57. Sontheimer RD (1999) Cutaneous features of classic dermatomyositis and amyopathic derma-
tomyositis. Curr Opin Rheumatol 11(6):475482
58. Bendewald MJ, Wetter DA, Li X, Davis MD (2010) Incidence of dermatomyositis and clini-
cally amyopathic dermatomyositis: a population-based study in Olmsted County. Minnesota
Arch Dermatol 146(1):2630
59. Hengstman GJ, van Venrooij WJ, Vencovsky J, Moutsopoulos HM, van Engelen BG (2000)
The relative prevalence of dermatomyositis and polymyositis in Europe exhibits a latitudinal
gradient. Ann Rheum Dis 59(2):141142
92 G.M. Murphy and N. Ralph
60. Okada S, Weatherhead E, Targoff IN, Wesley R, Miller FW (2003) Global surface ultraviolet
radiation intensity may modulate the clinical and immunologic expression of autoimmune
muscle disease. Arthritis Rheum 48(8):22852293
61. Cheong WK, Hughes GR, Norris PG, Hawk JL (1994) Cutaneous photosensitivity in dermato-
myositis. Br J Dermatol 131(2):205208
62. Dourmishev L, Meffert H, Piazena H (2004) Dermatomyositis: comparative studies of cutane-
ous photosensitivity in lupus erythematosus and normal subjects. Photodermatol Photoimmunol
Photomed 20(5):230234
63. Werth VP, Callen JP, Ang G, Sullivan KE (2002) Associations of tumor necrosis factor alpha
and HLA polymorphisms with adult dermatomyositis: implications for a unique pathogenesis.
J Invest Dermatol 119(3):617620
64. Pablos JL, Santiago B, Galindo M, Carreira PE, Ballestin C, Gomez-Reino JJ (1999)
Keratinocyte apoptosis and p53 expression in cutaneous lupus and dermatomyositis. J Pathol
188(1):6368
65. Woo TY, Callen JP, Voorhees JJ, Bickers DR, Hanno R, Hawkins C (1984) Cutaneous lesions
of dermatomyositis are improved by hydroxychloroquine. J Am Acad Dermatol 10(4):
592600
66. Dalakas MC, Illa I, Dambrosia JM et al (1993) A controlled trial of high-dose intravenous
immune globulin infusions as treatment for dermatomyositis. N Engl J Med 329(27):
19932000
67. Nishiyama S, Maywaki S, Hashimoto T (1995) Cutaneous manifestations of primary Sjogren
syndrome. Nihon Rinsho 53(10):25512556
68. Yoshida K, Inoue H, Komai K, Yamane T, Hashiramoto A, Shiozawa K et al (2013) Mixed
connective tissue disease is distinct from systemic lupus erythematosus: study of major
histocompatibility complex class I polypeptide-related sequence A and HLA gene
polymorphisms. Tissue Antigens 81(1):4445
69. Igawa K, Matsunaga T, Nishioka K (2004) Involvement of UV-irradiation in pemphigus folia-
ceus. J Eur Acad Dermatol Venereol 18(2):216217
70. Kano Y, Shimosegawa M, Mizukawa Y, Shiohara T (2000) Pemphigus foliaceus induced by
exposure to sunlight. Report of a case and analysis of photochallenge-induced lesions.
Dermatology 201(2):132138
71. Reis VM, Toledo RP, Lopez A, Diaz LA, Martins JE (2000) UVB-induced acantholysis in
endemic Pemphigus foliaceus (Fogo selvagem) and Pemphigus vulgaris. J Am Acad Dermatol
42(4):571576
72. Farber EM, Bright RD, Nall ML (1968) Psoriasis. A questionnaire survey of 2,144 patients.
Arch Dermatol 98(3):248259
73. Ros AM, Eklund G (1987) Photosensitive psoriasis. An epidemiologic study. J Am Acad
Dermatol 17(5 Pt 1):752758
74. Rutter KJ, Watson RE, Cotterell LF, Brenn T, Griffiths CE, Rhodes LE (2009) Severely photo-
sensitive psoriasis: a phenotypically defined patient subset. J Invest Dermatol 129(12):
28612867
75. Nalluri R, Arun B, Rhodes LE (2010) Photoaggravated hand and foot psoriasis. Photodermatol
Photoimmunol Photomed 26(5):261262
76. Ros AM, Wennersten G (1986) Photosensitive psoriasisclinical findings and phototest results.
Photodermatol 3(6):317326
77. Murphy GH, White IR, Hawk JL (1990) Allergic airborne contact dermatitis to compositae
with photosensitivity chronic actinic dermatitis in evolution. Photodermatol Photoimmunol
Photomed 7:3839
78. Hd M, Harrison GI, Potten CS, Young AR, Hawk JL (1995) The action spectrum for induction
of chronic actinic dermatitis is similar to that for sunburn inflammation. Photochem Photobiol
62:976997
79. OReilly FM, McKenna D, Murphy GM (1999) Is monochromatic irradiation testing useful in
the differentiation of drug-induced photosensitivity from: chronic actinic dermatitis? Clin Exp
Dermatol 24:118121
5 Photoprotection, Photoimmunology and Autoimmune Diseases 93
Key Points
Vitamin D is unquestionably important in the development and mainte-
nance of skeletal health but may have broader implications beyond bone
health.
There are varying definitions of vitamin D deficiency and recommended
daily requirements. Current recommendation on vitamin D screening is
that it should be done only for at-risk populations.
Real-world application of sunscreen does not impact vitamin D status,
although rigorous photoprotection practice does.
Given the low cost, broad availability, and safety profile of oral vitamin D
supplementation, replenishing vitamin D by solar and artificial ultraviolet
radiation is not advised.
6.1 Introduction
Vitamin D is a fat-soluble hormone obtained from sun exposure, diet, and oral sup-
plements with many biologic effects. It is primarily known for its crucial role in the
optimization of bone mass via its effects on parathyroid hormone and calcium and
phosphorous homeostasis. However, it also may have extra-skeletal associations,
such as cardiovascular disease, immune susceptibility, and cancers [1]. Thus, con-
cern has developed whether photoprotection would affect vitamin D status. This
chapter will discuss how humans acquire vitamin D, vitamin Ds physiologic
effects, associations with disease, impact of photoprotection, and recommendations
to patients. Terms and conversions used in the discussion of vitamin D can be
viewed in Table 6.1.
plasma membrane and transported to the liver by serum vitamin D binding protein
for conversion to 25(OH)D by 25-hydroxylase. 25(OH)D undergoes further hydrox-
ylation to 1,25(OH)2D by primarily renal 1-alpha-hydroxylase. However, keratino-
cytes, macrophages, T-lymphocytes, dendritic cells, bone, prostate, and placental
cells also can convert 25(OH)D to 1,25(OH)2D.
As cutaneous vitamin D synthesis requires UVB radiation, factors that attenuate
or absorb UVB and the duration of exposure influence vitamin D production.
Atmospheric ozone absorbs UVB radiation. However, its absolute and relative thick-
ness in relation to the sun varies. The intensity of UVB radiation is greatly reduced
early/late in the day, at higher latitude, at lower altitudes, and in the winter when the
tilt of the earth is at its greatest. Analysis of published serum 25(OH)D levels in the
northern hemisphere indicates there is insufficient ambient UVB for adequate vita-
min D production during the winter at latitudes above 33 [3]. Additionally, UVB is
reduced further exogenously by high nitrogen dioxide and ozone levels in polluted
urban environments, such as Los Angeles, California, and Mexico City, Mexico [4].
Endogenously, UVBs effects on 7-DHC vary by skin phototype, as melanin
mitigates UVBs penetration. A 1991 study of skin pigmentation and serum vitamin
UVB
Epithelium
Lumisterol
7-DHC Pre-Vitamin D Tachysterol
(Heat) (Inactive)
Supersterol I & II
Circulation Vitamin D3 5, 6-trans-Vitamin D3
(Cholecaliferol) (Inactive)
Vitamin D
Binding Diet/Supplementation:
Protein
Vitamin D3 Vitamin D2
&
(Cholecaliferol) (Ergocaliferol)
Liver
25-hydroxylase
25(OH)D3
(Calcidiol)
Kidneys
1-hydroxylase
24-hydroxylase
1, 25(OH)2D3 24, 25(OH)2D3
(Calcitriol) (Inactive)
(Active)
D3 levels found significantly higher levels in Caucasian and Asian subjects com-
pared to those of African American and East Indian descent [5]. Similar inverse
relationships between skin pigmentation and serum vitamin D3 levels were also
reported in subsequent 2004, 2010, and 2014 publications [68].
Artificial sources of UVB radiation can rapidly and effectively raise serum
vitamin D levels at suberythemogenic levels via the same mechanisms as solar
radiation without exogenous, atmospheric attenuators. However, it should be noted
that ultraviolet exposure from artificial devices or natural sunlight can increase
ones risk of skin cancers, and tanning booths are a poor source of artificial UVB for
vitamin D as they primarily emit ultraviolet A (340400 nm).
Exogenous vitamin D can be obtained through dietary intake, but only a few foods,
such as cod liver, specific fish (mackerel, sockeye salmon, tuna), beef, egg yolks,
shiitake mushrooms, and cheese, naturally contain high levels of vitamin
D. Therefore, many countries, including the United States, fortify milk, orange
juice, yogurt, cereal, and other foods to enhance the dietary sources of vitamin
D. Additionally, vitamin D can be obtained through over-the-counter multivitamins
and vitamin D supplements or prescription supplementation. Although fungal-/
yeast-derived vitamin D2 (ergocalciferol) is available over the counter and by
prescription, commercially synthesized vitamin D3 (cholecalciferol) is primarily
utilized in fortification and supplementation. Following ingestion, both forms of
exogenous vitamin D are metabolized by the liver to 25(OH)D. While the
bioequivalence of D2 and D3 remains a controversial topic [9], a 2012 meta-analysis
of randomized clinical trials found D3 superior in raising 25(OH)D levels [10].
was observed with less than 400 IU [12]. Interestingly, a meta-analysis of eight dou-
ble-blind RCTs reported supplementation of greater than 700 IU of vitamin D reduced
the fall risk of elderly patients by 19 % compared to those less than 700 IU [13].
These, as well as a myriad of other studies, conclusively demonstrated sufficient evi-
dence for a dose-response relationship between vitamin D and bone health to merit a
Dietary Reference Intake recommendation by the Institute of Medicine [2]. However,
it should be noted that despite a 2 to 9 times higher prevalence of low vitamin D in the
African American population compared to Caucasians, the fracture risk in the African
Americans is half that of the Caucasian population [14]. The reported lower levels of
vitamin D binding protein in African American, compared to Caucasian, hence result-
ing in similar levels of bioavailable vitamin D, may account for this finding [15].
Extra-skeletally, vitamin D appears to modulate the immune system, limit malig-
nant potential, and mitigate vascular morbidity. Binding to T cells, B cells, natural
killer, and monocyte vitamin D receptors, active vitamin D stimulates the innate
immune system and repress of the adaptive system.
Clinically, this has implications in a variety of dermatologic and non-dermato-
logic conditions. Due to vitamin Ds essential role in the containment and destruc-
tion of Mycobacterium tuberculosis, incorporation of vitamin D supplementation to
the standard tuberculoid regimen in those with vitamin D deficiency induces acceler-
ated clinical and radiographic improvement compared to the standard regimen alone
[16]. Interestingly, this beneficial effect of vitamin D also has implications prior to
clinical disease by reducing ones susceptibility and risk of progression from infec-
tion to disease [16]. Similarly, an inverse relationship between gastrointestinal can-
cer, breast cancer, all cancer mortality, and total life cancer incidence to vitamin D
levels has been indicated [16]. Metabolites of 7-DHC and 1, 25 (OH)2D3 also have
pro-differentiation and apoptotic effects [17, 18]. Dermatologists take advantage of
vitamin Ds regulation of cell differentiation and T-cell activity through the topical
management of psoriasis, atopic dermatitis, pityriasis alba, and other cutaneous con-
ditions with vitamin D analogues [19]. However, the usefulness of oral vitamin D for
these dermatologic conditions, excluding psoriasis, remains conflicted [1923].
Lastly, vitamin D has receptors on vascular smooth muscle that appear to influence
cardiovascular, cerebrovascular, and pregnancy-vasculature morbidity [24, 25].
Despite these and other published findings, the Institute of Medicine concluded
that there were insufficient prospective trials to provide adequate evidence warrant-
ing any Dietary Reference Intake recommendations for extra-skeletal medical sys-
tems. [2] The United States Preventative Services Task Force (USPSTF) was also
unable to find sufficient extra-skeletal evidence for recommendations [14].
Therefore, all daily vitamin D recommendations are based on date on skeletal health.
Individuals with limited sun exposure have an increased risk for vitamin D insuffi-
ciency and deficiency. A retrospective review of 165 patients with photosensitizing
conditions found those practicing strict photoprotection (e.g., xeroderma pigmentosa)
or developing symptoms within one hour of sun exposure (e.g., solar urticaria)
100 J.L. Griffith et al.
6.5 Recommendations
Society and American Geriatric Society for individuals at risk for vitamin D
deficiency due to an underlying condition or behavior [33, 34]. Once tested, deciding
the threshold for initiation of treatment varies based upon the organization focus:
nutrition repletion, treatment of vitamin deficiencies in asymptomatic individuals,
or prevention of a specific negative health outcome regardless of vitamin deficiency
[14]. To add more confusion to this matter, serum levels of vitamin D can vary
1020 % depending upon the assay method and laboratory performing the
assessment [14]. Thus, while most practitioners follow the Institute of Medicines
published guidelines, which are discussed below, vitamin D level remains a heated
debate.
In 2011, the Institute of Medicine published their evidence-based review and
suggested recommendations on vitamin D deficiency and insufficiency for skeletal
health. As serum 1,25(OH)D is under tight endocrine control, serum 25(OH)D is
used for determining vitamin D status. Serum levels can be reported as either ng/mL
or nmol/L (2.5 ng/mL = nmol/L). According to the IOM Committee, 25(OH)D
levels of 1620 ng/mL are sufficient for 97.5 % of the population. However,
approximately 50 % of the population only requires 1216 ng/mL of 25(OH)D to
cover their requirements. Therefore, levels below 20 ng/mL suggest risk for
insufficiency, and levels below 12 ng/mL indicate risk of deficiency. Levels above
20 ng/mL should not raise significant concern regarding potential adverse effects
until levels exceed 50 ng/mL [2].
While these guidelines for vitamin D status are relevant for most conditions,
special consideration should be noted for certain conditions, such as sarcoidosis.
Patients with sarcoidosis may have falsely low 25(OH)D with sufficient 1,
25(OH)2D due to macrophage conversion of 25(OH)D to 1, 25(OH)2D. Thus,
vitamin D supplementation in sarcoidosis may inadvertently cause hypervitamin-
osis D. Therefore, 1, 25(OH)2D and parathyroid hormone should be evaluated in
these patients. Low 1,25(OH)2D with normal PTH merits 400800 International
Units (IU) per day of cholecalciferol, while normal vitamin D with elevated PTH
levels warrants a consultation by endocrinology for concerns of hyperparathy-
roidism [35].
Based upon the IOMs recommended dietary allowances (RDAs) for adults and
allowable intake (AI) for infants less than 1 year of age, the required daily nutrition
to meet the skeletal requirements of vitamin D for 97.5 % of the population and
ensure adequate nutrition (RDA and AI, respectively) is listed by age in Table 6.2.
The upper daily intake limit unlikely to pose risk of hypervitaminosis D is 2500 IU/
day for 13 years old, 3000 IU/day for 48 year olds, and 4000 IU/day for those
greater than 9 years of age [36].
However, both adequate vitamin D levels and daily requirements of vitamin D
are highly debated (Table 6.3).
102 J.L. Griffith et al.
6.6 Conclusion
Vitamin D possesses a beneficial role in skeletal health and may have broader impli-
cations beyond calcemic health. While strict photoprotective measures can reduce
serum vitamin D levels, frequent sunscreen use in the real world does not appear to
impact vitamin D status. Instead, standard risk factors of low sun exposure, darker
skin types, older age with history of falls and non-traumatic fractures, obesity, mal-
absorption syndromes, severe liver or renal disease, solely breastfed infants, granu-
loma-forming disorders, and specific medications (glucocorticoids, antiepileptic,
antifungal, and autoimmune deficiency syndrome medications) may place individu-
als at increased risk for vitamin D deficiency or insufficiency [37]. While laboratory
assessment and vitamin D supplementation should be considered in these at-risk
groups, widespread testing of all individuals is not recommended. While there is
much debate about vitamin D screening, definition of deficiency, and daily require-
ments, most medical bodies agree that replacing vitamin D by solar and artificial
UV radiation or recommending against photoprotective practices is ill-advised,
given the low cost, broad availability, and safety profile of oral vitamin D
supplementation.
References
3. Diffey BL (2010) Modelling the seasonal variation of vitamin D due to sun exposure. Br
J Dermatol 162(6):13421348
4. Kannan S, Lim HW (2014) Photoprotection and vitamin D: a review. Photodermatol
Photoimmunol Photomed 30(2-3):137145
5. Matsuoka LY, Wortsman J, Haddad JG, Kolm P, Hollis BW (1991) Racial pigmentation and
the cutaneous synthesis of vitamin D. Arch Dermatol 127(4):536538
6. Bischoff-Ferrari HA, Dietrich T, Orav EJ, Dawson-Hughes B (2004) Positive association
between 25-hydroxyvitamin D levels and bone mineral density: a population-based study of
younger and older adults. Am J Med 116(9):634639
7. Gozdzik A, Barta JL, Weir A, Cole DE, Vieth R, Whiting SJ, Parra EJ (2010) Serum
25-hydroxyvitamin D concentrations fluctuate seasonally in young adults of diverse ancestry
living in Toronto. J Nutr 140(12):22132220
8. Au LE, Harris SS, Dwyer JT, Jacques PF, Sacheck JM (2014) Association of serum
25-hydroxyvitamin D with race/ethnicity and constitutive skin color in urban school children.
J Pediatr Endocrinol Metab 27(1112):10951100
9. Institute of Medicine (US) Committee to Review Dietary Reference Intakes for Vitamin D and
Calcium; Ross AC, Taylor CL, Yaktine AL et al (eds) (2011) Chapter 3: Overview of vitamin
D. In: Dietary reference intakes for calcium and vitamin D. National Academies Press (US),
Washington (DC). Available from: http://www.ncbi.nlm.nih.gov/books/NBK56061/
10. Tripkovic L, Lambert H, Hart K, Smith CP, Bucca G, Penson S, Chope G, Hypponen E, Berry
J, Vieth R, Lanham-New S (2012) Comparison of vitamin D2 and vitamin D3 supplementation
in raising serum 25-hydroxyvitamin D status: a systematic review and meta-analysis. Am
J Clin Nutr 95(6):13571364
11. Pike JW, Shevde NK, Hollis BW, Cooke NE, Zella LA (2008) Vitamin D-binding protein
influences total circulating levels of 1,25-dihydroxyvitamin D-3 but does not directly modulate
the bioactive levels of the hormone in vivo. Endocrinology 149(7):36563667
12. Bischoff-Ferrari HA, Willett WC, Wong JB, Stuck AE, Staehelin HB, Orav EJ, Thoma A, Kiel
DP, Henschkowski J (2009) Prevention of nonvertebral fractures with oral vitamin D and dose
dependency: a meta-analysis of randomized controlled trials. Arch Intern Med 169:551561
13. Bischoff-Ferrari HA, Dawson-Hughes B, Staehelin HB, Oravek JE, Stuck AE, Theiler R, Wong
JB, Egli A, Kiel DP, Henschkowski J (2009) Fall prevention with supplemental and alpha-
hydroxylated vitamin D: a meta-analysis of randomized controlled trials. BMJ 339:36923703
14. LeFevre ML, on behalf of the U.S. Preventive Services Task Force (2015) Screening for vita-
min D deficiency in adults: U.S. Preventive Services Task Force recommendation statement.
Ann Intern Med 162:133140
15. Powe CE, Evans MK, Wenger J, Zonderman AB, Berg AH, Nalls M, Tamez H, Zhang D, Bhan
I, Karumanchi SA, Powe NR, Thadhani R (2013) Vitamin D-binding protein and vitamin D
status of black Americans and white Americans. N Engl J Med 369(21):19912000
16. Salahuddin N, Ali F, Hasan Z, Rao N, Ageel M, Mahmood F (2013) Vitamin D accelerates
clinical recovery from tuberculosis: results of the SUCCINCT Study [Supplementary
Cholecalciferol in recovery from tuberculosis]. A randomized, placebo-controlled, clinical trial
of vitamin D supplementation in patients with pulmonary tuberculosis. BMC Infect Dis 13:22
17. Slominski AT, Kim TK, Janjetovic Z, Tuckey RC, Bieniek R, Yue J, Li W, Chen J, Nguyen
MN, Tang EK, Miller D, Chen TC, Holick M (2011) 20-Hydroxyvitamin D2 is a noncalcemic
analog of vitamin D with potent antiproliferative and prodifferentiation activities in normal
and malignant cells. Am J Physiol Cell Physiol 300(3):C526C541
18. Slominski AT, Janjetovic Z, Fuller B, Zmijewski MA, Tuckey RC, Nguyen MN, Sweatman T,
Li W, Zjawiony J, Miller D, Chen TC, Lozanski G, Holick MF (2010) Products of vitamin D3
or 7-dehydrocholesterol metabolism by cytochrome P4850scc show anti-leukemia effects,
having low or absent calcemic activity. PLoS One 5(3):e9907
19. Wat H, Dytoc M (2014) Off-label uses of topical vitamin D in dermatology: a systematic
review. J Cutan Med Surg 18(2):91108
20. Soleymani T, Hung T, Soung J (2015) The role of vitamin D in psoriasis: a review. Int
J Dermatol 54(4):38392
104 J.L. Griffith et al.
21. Kamangar F, Koo J, Heller M, Lee E, Bhutani T (2013) Oral vitamin D, still a viable treatment
option for psoriasis. J Dermatolog Treat 24(4):261267
22. Camargo CA, Ganmaa D, Sidbury R, Erdenedelger K, Radnaakhand N, Khandsuren B (2014)
Randomized trial of vitamin D supplementation for winter-related atopic dermatitis in chil-
dren. J Allergy Clin Immunol 134(4):831835
23. Hata TR, Audish D, Kotol P, Coda A, Kabigting F, Miller J, Alexandrescu D, Boguniewicz M,
Taylor P, Aertker L, Kesler K, Hanifin JM, Leung DY, Gallo RL (2014) A randomized con-
trolled double-blind investigation of the effects of vitamin D dietary supplementation in sub-
jects with atopic dermatitis. J Eur Acad Dermatol Venereol 28(6):781789
24. LoPiccolo MC, Lim HW (2010) Vitamin D in health and disease. Photodermatol Photoimmunol
Photomed 26:224229
25. Sablok A, Batra A, Thariani K, Batra A, Bharti R, Aggarwal AR, Kabi BC, Chellani H (2015)
Supplementation of Vitamin D in pregnancy and its correlation with feto-maternal outcome.
Clin Endocrinol (Oxf) 83(4):536541. [Epub ahead of print]
26. Holme SA, Ansley AV, Badminton MN, Elder GH (2008) Serum 25-hydroxyvitamin D in
erythropoietic protoporphyria. Br J Dermatol 159:211213
27. Cusack C, Danby C, Fallon JC (2008) Photoprotective behavior and sunscreen use: impact on
vitamin D levels in cutaneous lupus erythematosus. Photodermatol Photoimmunol Photomed
24:260267
28. Haney EM, Stadler D, Bliziotes MM (2005) Vitamin D insufficiency in internal medicine resi-
dents. Calcif Tissue Int 76:1116
29. Vu LH, Whiteman DC, van der Pols JC, Kimlin MG, Neale RE (2011) Serum vitamin D levels
in office workers in a subtropical climate. Photochem Photobiol 87:714720
30. Reid SM, Robinson M, Kerr AC, Ibbotson SH (2012) Prevalence and predictors of low vitamin
D status in patients referred to a tertiary photodiagnostic service: a retrospective study.
Photodermatol Photoimmunol Photomed 28:9196
31. Norval M, Wulf HC (2009) Does chronic sunscreen use reduce vitamin D production to insuf-
ficient levels? Br J Dermatol 161(4):732736
32. Osmancevic A, Sandstrom K, Gillstedt M, Landin-Wilhelmsen K, Larko O, Wennberg
Larko AM, F Holick M, Krogstad AL (2015) Vitamin D production after UVB exposure a
comparison of exposed skin regions. J Photochem Photobiol B 143:3843
33. Holick MF, Binkley NC, Bischoff-Ferrari HA, Gordon CM, Hanley DA, Heaney RP et al;
Endocrine Society (2011) Evaluation, treatment, and prevention of vitamin D deficiency: an
Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 96:19111930
34. American Geriatrics Society Workgroup on Vitamin D Supplementation for Older Adults
(2014) Recommendations Abstracted from the American Geriatrics Society Consensus
Statement on Vitamin D for Prevention of Falls and Their Consequences. J Am Geriatr Soc
62:147152
35. Sage RJ, Rao DS, Burke RR, Lim HW (2011) Preventing vitamin D toxicity in patients with
sarcoidosis. J Am Acad Dermatol 64(4):795796
36. Ross AC, Manson JE, Abrams SA (2011) The 2011 report on dietary reference intakes for
calcium and vitamin D from the Institute of Medicine: what clinicians need to know. J Clin
Endocrinol Metab 96(1):5358
37. Pramyothin P, Holick M (2012) Vitamin D supplementation: guidelines and evidence for sub-
clinical deficiency. Curr Opin Gastroenterol 28(2):139150
Chapter 7
Photoprotection and Skin of Color
Key Points
UVR exposure is a risk factor for skin cancer in POC; however, melanins
photoprotective properties likely reduce this risk. Despite decreased skin
cancer incidence, POC often have increased skin cancer morbidity and
mortality.
Darker constitutive pigmentation exhibits an inverse relationship with
degree of photoaging; thus darker POC manifest photoaging much later in
life.
There is a lack of sufficient data regarding ideal photoprotection practices
for POC.
Race is a poorly defined term that is a political and social construct more than
a biologic phenomenon [12, 87]. Ethnicity is a somewhat broader term referring to
. . .large groups of people classed according to common racial, national, tribal,
religious, linguistic, or cultural origin or background [77]. Each of these terms is
Various systems have been developed to make the classification of skin color more
phenotypically objective. Fitzpatrick skin-type scale, originally developed for
Caucasian skin, is a frequently used and valid tool for categorizing skin according to
ultraviolet radiation (UVR) sensitivity; however, it has been identified as less useful in
POC including blacks, Asians, and likely other POC [31, 36, 92, 111, 127]. In an
evaluation of various methods to measure skin color, Daniel et al. [27] found that a
simple seven-point Likert scale for self-reported natural skin color (very fair/light to
very dark) better correlated with spectrophotometry than did the Fitzpatrick skin-type
scale. The study participants self-identified as Caucasian, Asian American/Pacific
Islander, African American, Hispanic/Latino, or other ethnicity. Similarly, the Skin
Color Chart is a tool developed by LOreal that has been validated in Caucasian,
Asian, African American, and Indian skin ([30]; Del Bino S, 2015, personal commu-
nication). It allows for evaluation of skin color on any body surface based on a fan
deck of 52 cards each with a three-centimeter hole through which skin color can be
7 Photoprotection and Skin of Color 107
55
10
tan
50
Brown
41 < ITA< 55 Light
45
40
28 < ITA < 41 Intermediate
brown
35 -30
Dark
10 < ITA < 28 Tan
30
25
-30 < ITA < 10 Brown dark
20
0 5 10 15 20 25 30 35 40
ITA < -30 Dark
b* (yellow chrom.)
Fig. 7.1 (a) Skin colour volume on the L*b* plane (CIELAB 1976 system). The vertical axis L*
is the luminance; the horizontal axis b* is the yellowblue component. (b) The individual typology
angle (ITA) allows skin colour classification into six groups, from very light to dark skin. (c)
FontanaMasson staining of melanin granules shows a good correlation between skin colour clas-
sification and melanin quantity and distribution (Reprinted with permission from Del Bino and
Bernerd [31])
compared to the card [30]. In addition, colorimetric parameters have been used to cre-
ate an individual typology angle (ITA)-based skin color classification system com-
posed of 6 skin tones from very light to dark (Fig. 7.1) [23, 33]. ITA is both quantifiable
and objective [33]. Visual phenotype/ethnically defined Caucasian skin had ITA val-
ues classifying it as light, intermediate, and tan [31]. Likewise defined Hispanic skin
ITA values ranged from light to brown; and African skin ITA values ranged from
intermediate to dark. Asian ITA scores showed a geographic split with northeast Asian
skin ITA values of light, intermediate, and tan, and southeast Asian skin ITA values
had a broad range from light to dark. Thus ITA-based skin color classification allows
for precise evaluation of in vivo constitutive pigmentation. Furthermore, a correlation
exists between ITA-determined skin color and DNA damage with greater levels of
UVR-induced DNA damage correlating with lighter skin color [33]. These findings
reveal that a spectrum of phenotypes exists in Caucasians and POC; thus objective
measures of constitutive skin pigmentation could serve as concrete, consistent, and
biophysiologically relevant criteria for defining skin color. Such phenotypically ger-
mane terminology is needed in dermatologic research as an alternative means of clas-
sification separate from race/ethnicity for greater clarity and consistency in medical
literature. The authors and other researchers believe that such objective measurements
could be used by dermatologists to make personalized photoprotection recommenda-
tions (e.g., maximum daily UVR exposure time, ideal sunscreens, skin cancer preven-
tion strategies, etc.) ([31]; Del Bino S, 2015, personal communication).
Research has shown that epidermal melanin largely determines constitutive
pigmentation [3, 4, 9, 110]. Though the beneficial effects of photoprotection in
108 K.J. Buster and J.J. Ledet
7.3.1 Photocarcinogenesis
UVR exposure is a risk factor for skin cancer in POC including Hispanics, blacks,
and Asians [25, 37, 48, 49, 52, 59, 61, 90]; however, due to the photoprotective
effect of melanin, UVR may play a smaller role in skin cancer development in
darkly pigmented skin [37]. Basal cell carcinoma (BCC) is most common on sun-
exposed skin across the spectrum of skin types and ethnic backgrounds (Fig. 7.2a)
[35, 37, 74, 82]. Though squamous cell carcinoma (SCC) is most common on non-
sun-exposed skin in blacks [35, 48, 75, 106], several studies have shown increased
nonmelanoma skin cancer (NMSC) in sun-exposed skin of blacks [83, 90]. Risk
factors for malignant melanoma (MM) in darker POC are unclear and data are con-
flicting (see Fig. 7.2b for images). An evaluation of the Surveillance, Epidemiology,
and End Results Program (SEER) data revealed no UVR index/lower latitude asso-
ciation with MM incidence in blacks or Hispanics [34]. However, several studies
have identified an association between MM and UVR exposure in blacks (the United
States), Hispanics (the United States), and Asians (India) [52, 61, 90, 98].
Despite decreased incidence, POC often have increased skin cancer morbidity
and mortality [37, 49, 121, 124]. Groups with poorer skin cancer outcomesinclud-
ing POCmore commonly have misperceptions regarding skin cancer (including
expectation of symptoms, discounting importance of skin exams, and confusion on
prevention strategies), and these may contribute to skin cancer disparities [19].
NMSC occurs with increased frequency in geographic areas with greater UVB
exposure (SCC more than BCC) [101]. In the United States, this means areas of
decreased latitude. During the 1980s and 1990s, the Earth experienced alarming
loss of the ozone layer over midlatitudes of the Northern Hemisphere due to
increased use of ozone-depleting substances over the previous decades [85]. With
continued depletion, predictably, all humans, including POC, would have a progres-
sively increased risk of NMSC due to increased UVB reaching the Earths surface.
A US National Cancer Institute study found that for each 1 % relative increase in
UVB, there could be a 2 % increase in incidence of NMSC [101]. Similarly, a 2013
study found that NMSC incidence would increase by more than 1000 cases (26.9 %)
yearly in Korea secondary to UVB from a constant 10 % decrease in ozone
7 Photoprotection and Skin of Color 109
a a b c d
b a b c
d e f
Fig. 7.2 (a) Nonmelanoma skin cancers in people of color. Pigmented basal cell carcinoma in elderly
Hispanic man (right lateral orbital rim) (a); middle-aged Asian woman (right cheek) (b); middle-aged
Hispanic man (right forehead) (c); middle-aged Hispanic man (left nasal ala) (d). (b) Melanomas in
people of color. Lentigo maligna in middle-aged Hispanic woman (vermilion upper and lower lips) (a);
melanoma in middle-aged black woman (right fourth toe) (b); Hispanic woman (left fifth toe) (c);
middle-aged Hispanic man (left plantar foot) (d); elderly Hispanic man (right cheek) (e); and Asian
woman (side of left leg) (f) (Reprinted with permission from Agbai et al. [1])
to tan skin develops cyclobutane pyrimidine dimers (CPD) in all epidermal layers,
whereas brown and dark brown skin only form CPD in the suprabasal layers [31].
CPD develop secondary to UVR absorption and are subsequently found in skin
cancers [112]. Immediately after UVR exposure, 79100 % of melanocytes in light
skin are CPD positive, whereas 17 % in brown skin and 15 % in dark skin are CPD
positive [31]. Even at suberythemal doses, light, medium, and dark skin types incur
DNA damage [105, 109], suggesting that photoprotection can be beneficial in all
skin types. However, as noted above, the increased melanin content of darker skin
does help protect it from photodamage [54]. Numerous studies have identified an
inverse relationship between constitutive skin pigmentation and DNA damage [31
33, 97, 109]. Given this intrinsic photoprotection, the necessary level of external
photoprotection for prevention of DNA damage and subsequent skin cancer in POC
of different levels of skin pigmentation may vary.
7.3.2 Photoaging
Photoaging has been defined as the combination of intrinsic aging and photodamage
[116]. In contrast to intrinsic (chronological) aging, photoaging is associated with
significant changes in skin composition including undesirable changes in texture,
wrinkling, increased pigmentation, greater vascularity, laxity, and cutaneous malig-
nancy [41, 103, 104, 116]. Compared to Caucasians, darker POC manifest photoag-
ing much later in life, in approximately the 5th and 6th decade [50]. In one study
comparing facial skin of black and white women from ages 20 to 50, blacks had no
obvious wrinkles, while most white women 4550 years old had wrinkles of the
lateral canthi (crows feet) and oral commissures [81]. Visual assessment has shown
that African American skin exhibits photoaging changes of hyperpigmentation and
uneven skin tone and white skin shows more severe fine lines, wrinkles, laxity, and
overall photodamage [44].
Goh [38] observed photoaging as both hyperpigmentation and wrinkles in Asian
(Singaporean, Malaysian, and Indonesian) skin. Similarly, Korean photoaging is
7 Photoprotection and Skin of Color 111
In an analysis of ethnic variation in melanin content, Alaluf et al. [3] found that
the quantity of epidermal melanin in heavily pigmented (i.e., African and Indian)
skin is about double that is seen in relatively lightly pigmented (Mexican, Chinese,
and European) skin. Prior to that, Yohn et al. [126] found that melanocytes of blacks
have significantly more melanin than whites. The dispersion, size, and number of
melanosomes are also on a spectrum with darker skin exhibiting greater dispersion,
larger size, and increased numbers of melanosomes than lighter skin [3, 81, 114].
Melanosomes are largest in African skin and progressively decrease in size in
Indian, Mexican, Chinese, and European skin [3]. Increased melanin correlates with
higher constitutive pigmentation, which as aforementioned typically exhibits an
inverse relationship with degree of photoaging.
Avoidance of sun exposure and use of sunscreen are widely accepted photoprotective
practices as they limit or eliminate UVR-induced DNA and collagen damage that
lead to photoaging [10]. UVR also induces oxidative stress (which eventually leads
to matrix metalloproteinase degradation of collagen), and antioxidants have been
shown to inhibit the UVR cascade that leads to photoaging [56]. Some antioxidants
(e.g., ferulic acid, vitamin C combined with vitamin E) also serve as photoprotectants
[66, 67].
7.3.3.1 Melasma
UV protection is a core element in the treatment of melasma, PIH, and other disorders
of increased skin pigmentation. However, physicians are less likely to prescribe sun-
screen for treatment of dyschromias in POC than whites. In an analysis of more than
five million patient visits for the sole diagnosis of dyschromia, Kang et al. [57] found
114 K.J. Buster and J.J. Ledet
that sunscreen use was prescribed for 32 % of whites (3rd most common treatment
prescribed for this population), 17 % of blacks (6th most common treatment), and 7 %
for Asians (10th most common treatment). Though reasons for this discrepancy are
unclear, authors speculate that it may be it is due to dermatologists recognition of the
photoprotective effect of melanin and decreased risk of sunburning in darker skin.
However, they note that sunscreen is key in treating hyperpigmentation in all skin types.
Broad-spectrum sunscreen with good UVA protection plays a pivotal role in the
treatment of melasma as it may help minimize melasma relapses. Lakhdar et al. [63]
found that vigilant sunscreen use as the sole treatment in women (Fitzpatrick skin
types IIV) during and after pregnancy led to fewer cases of melasma. With use of
broad-spectrum (SPF 50, UVA protective factor 28) sunscreen every 2 h, nearly 80 %
of women had lighter skin or the same skin tone at the end of the study. These results
are encouraging, but outside a clinical study, compliance with sunscreen application
every 2 h is likely to be poor. This is especially likely in POC since they, as noted
earlier in this section, use sunscreen and other forms of sun protection less often.
Recently, the utility of Polypodium leucotomos extract (PLE) as a treatment for
disorders of pigmentation has been evaluated. In a randomized double-blinded pla-
cebo-controlled trial (RCT) of 40 Hispanic women with moderate to severe facial
melasma, Ahmed et al. [2] found that 240 mg of oral PLE three times daily plus
once daily (morning) application of broad-spectrum sunscreen was not significantly
better than sunscreen application alone. However, a smaller RCT (n = 21) revealed
significant improvement in women with epidermal melasma treated with twice
daily PLE and broad-spectrum (SPF 45) sunscreen compared to sunscreen alone
[72]. The skin types of the participants were not revealed in this study. Though, to
date, there is no direct research implicating PLE as a useful agent in treatment of
other common forms of hyperpigmentation in POC, this is an area worth exploring.
A 2004 study in subjects of skin phototypes II and III revealed that PLE (7.5 mg/kg
the night prior to exposure) decreased PUVA-induced acute phototoxicity as well as
PUVA-induced hyperpigmentation [79]. A 2014 article on dermatologic applica-
tions for PLE reviewed research showing promising results for photodermatoses,
pigmentary disorders, photoaging, and other dermatologic conditions [24]. Data
from such studies indicate that PLE may have additional utility in prevention and
treatment of pigmentary disorders in people of all skin types.
7.4 Conclusion
Table 7.1 Risks and benefits of sun exposure: position statement by Cancer Council Australia, the
Australian and New Zealand Bone and Mineral Society, Osteoporosis Australia, and the
Australasian College of Dermatologists [93]
1. For most people sun protection to prevent skin cancer is required when the UV index is
moderate or above (i.e., UV index is 3 or higher). At such times sensible sun protection
behavior is warranted and is unlikely to put people at risk of vitamin D deficiency
2. Most people probably achieve adequate vitamin D levels through the UVB exposure they
receive during typical day to day outdoor activities. For example, it has been estimated that
fair-skinned people can achieve adequate vitamin D levels (>50 nmol/L) in summer by
exposing the face, arms, and hands or the equivalent area of skin to a few minutes of sunlight
on either side of the peak UV periods on most days of the week. In winter, in the southern
regions of Australia where UV radiation levels are less intense, maintenance of vitamin D
levels may require 23 h of sunlight exposure to the face, arms, and hands or equivalent area
of skin over a week
3. Some people are at high risk of skin cancer. They include people who have had skin cancer,
have received an organ transplant, or are highly sun sensitive. These people need to have more
sun protection and therefore should discuss their vitamin D requirements with their medical
practitioner to determine whether dietary supplementation with vitamin D would be
preferable to sun exposure
4. Some groups in the community are at increased risk of vitamin D deficiency. They include
naturally dark-skinned people, those who cover their skin for religious or cultural reasons, the
elderly, babies of vitamin D-deficient mothers, and people who are housebound or are in
institutional care. Naturally dark-skinned people (Fitzpatrick skin types 5 and 6 rarely or
never burns) are relatively protected from skin cancer by the pigment in their skin; they could
safely increase their sun exposure. Others on this list should discuss their vitamin D status with
their medical practitioner as some might benefit from dietary supplementation with vitamin D
Table 7.2 AAD recommendations for photoprotection and early detection of skin cancer in people
of color [1]
Seek shade whenever possible
Wear sun-protective clothing
Wear a wide-brimmed hat to shade the face and neck as well as shoes that cover the entire foot
Wear sunglasses with UV-absorbing lenses
Apply broad-spectrum sunscreen with an SPF 30 or greater. Sunscreens without inorganic filters
(titanium dioxide and zinc oxide) are generally better accepted by people of color due to their
better cosmesis on dark skin
Apply sunscreen to dry skin 1530 min before going outdoors. When outdoors, reapply every 2
h to all exposed skin and after perspiring or swimming
Avoid exposure to indoor tanning beds/lamps
Take vitamin D supplement
Perform monthly self-skin examinations, paying close attention to subungual skin, palms,
soles, mucous membranes, groin, and perianal area
statement with recommendations that attempt to strike a balance (See Table 7.1). In
2010 the British Association of Dermatologists (BAD) made a similar consensus
statement with other national organizations advising minutes of regular midday sun
exposure without sunscreen (avoiding burning) to promote vitamin D formation
without unduly increasing skin cancer risk [15]. The AAD recommendations for
photoprotection in POC are listed in Table 7.2.
7 Photoprotection and Skin of Color 117
Acknowledgments The authors thank Sandra Del Bino, MsD, and LOreal Research and
Innovation for provision of Fig. 7.1.
References
1. Agbai ON, Buster K, Sanchez M, Hernandez C, Kundu RV, Chiu M, Roberts WE, Draelos
ZD, Bhushan R, Taylor SC, Lim HW (2014) Skin cancer and photoprotection in people of
color: a review and recommendations for physicians and the public. J Am Acad Dermatol
70(4):748762. doi:10.1016/j.jaad.2013.11.038
2. Ahmed AM, Lopez I, Perese F, Vasquez R, Hynan LS, Chong B, Pandya AG (2013) A ran-
domized, double-blinded, placebo-controlled trial of oral Polypodium leucotomos extract as
an adjunct to sunscreen in the treatment of melasma. JAMA Dermatol 149(8):981983.
doi:10.1001/jamadermatol.2013.4294
3. Alaluf S, Atkins D, Barrett K, Blount M, Carter N, Heath A (2002) Ethnic variation in mela-
nin content and composition in photoexposed and photoprotected human skin. Pigment Cell
Res (Sponsored by the European Society for Pigment Cell Research and the International
Pigment Cell Society) 15(2):112118
118 K.J. Buster and J.J. Ledet
4. Alaluf S, Atkins D, Barrett K, Blount M, Carter N, Heath A (2002) The impact of epidermal
melanin on objective measurements of human skin colour. Pigment Cell Res (Sponsored by
the European Society for Pigment Cell Research and the International Pigment Cell Society)
15(2):119126
5. Alexis AF, Sergay AB, Taylor SC (2007) Common dermatologic disorders in skin of color: a
comparative practice survey. Cutis 80(5):387394
6. Association AM (2007) AMA manual of style: a guide for authors and editors, 10th edn.
Oxford University Press, Oxford/New York
7. Autier P, Gandini S (2007) Vitamin D supplementation and total mortality: a meta-analysis of
randomized controlled trials. Arch Intern Med 167(16):17301737. doi:10.1001/
archinte.167.16.1730
8. Balkrishnan R, McMichael AJ, Camacho FT, Saltzberg F, Housman TS, Grummer S, Feldman
SR, Chren MM (2003) Development and validation of a health-related quality of life instru-
ment for women with melasma. Br J Dermatol 149(3):572577
9. Battie C, Jitsukawa S, Bernerd F, Del Bino S, Marionnet C, Verschoore M (2014) New
insights in photoaging. UVA induced damage and skin types. Exp Dermatol 23(Suppl 1):
712. doi:10.1111/exd.12388
10. Baumann L (2005) How to prevent photoaging? J Investig Dermatol 125(4):xiixiii.
doi:10.1111/j.0022-202X.2005.23810.x
11. Bernerd F, Asselineau D (1998) UVA exposure of human skin reconstructed in vitro induces
apoptosis of dermal fibroblasts: subsequent connective tissue repair and implications in
photoaging. Cell Death Differ 5(9):792802. doi:10.1038/sj.cdd.4400413
12. Bigby M (2009) Epidemiology of cutaneous disease. In: Alexis AF, Taylor S (eds)
Dermatology for skin of color, 1st edn. McGraw Hill Medical, New York
13. Binkley N, Novotny R, Krueger D, Kawahara T, Daida YG, Lensmeyer G, Hollis BW,
Drezner MK (2007) Low vitamin D status despite abundant sun exposure. J Clin Endocrinol
Metabol 92(6):21302135. doi:10.1210/jc.2006-2250
14. Briley JJ Jr, Lynfield YL, Chavda K (2007) Sunscreen use and usefulness in African-
Americans. J Drugs Dermatol JDD 6(1):1922
15. British Association of Dermatologists (2010) Vitamin D and the sun: consensus statement.
http://www.bad.org.uk/for-the-public/skin-cancer/vitamin-d. Jan 2015
16. British Association of Dermatologists (2011) World UV App. v1.3.2 edn. 22 Design
17. Bureau USC (2012) U.S. Census Bureau Projections Show a Slower Growing, Older, More
Diverse Nation a Half Century from Now. https://www.census.gov/newsroom/releases/
archives/population/cb12-243.html. Accessed 10 Jan 2015
18. Buster KJ, Stevens EI, Elmets CA (2012a) Dermatologic health disparities. Dermatol Clin
30(1):5359, viii. doi:10.1016/j.det.2011.08.002
19. Buster KJ, You Z, Fouad M, Elmets C (2012) Skin cancer risk perceptions: a comparison
across ethnicity, age, education, gender, and income. J Am Acad Dermatol 66(5):771779.
doi:10.1016/j.jaad.2011.05.021
20. Centers for Disease Control and Prevention (2007) Sunburn prevalence among adults--United
States, 1999, 2003, and 2004. MMWR Morb Mortal Wkly Rep 56(21):524528
21. Centers for Disease Control and Prevention (2012) Sunburn and sun protective behaviors
among adults aged 1829 years--United States, 20002010. MMWR Morb Mortal Wkly Rep
61(18):317322
22. Chan JL, Ehrlich A, Lawrence RC, Moshell AN, Turner ML, Kimball AB (2005) Assessing
the role of race in quantitative measures of skin pigmentation and clinical assessments of
photosensitivity. J Am Acad Dermatol 52(4):609615. doi:10.1016/j.jaad.2004.03.051
23. Chardon A, Cretois I, Hourseau C (1991) Skin colour typology and suntanning pathways. Int
J Cosmet Sci 13(4):191208. doi:10.1111/j.1467-2494.1991.tb00561.x
24. Choudhry SZ, Bhatia N, Ceilley R, Hougeir F, Lieberman R, Hamzavi I, Lim HW (2014)
Role of oral polypodium leucotomos extract in dermatologic diseases: a review of the
literature. J Drugs Dermatol 13(2):148153
7 Photoprotection and Skin of Color 119
25. Chuang TY, Reizner GT, Elpern DJ, Stone JL, Farmer ER (1995) Nonmelanoma skin cancer
in Japanese ethnic Hawaiians in Kauai, Hawaii: an incidence report. J Am Acad Dermatol
33(3):422426
26. Chung JH, Lee SH, Youn CS, Park BJ, Kim KH, Park KC, Cho KH, Eun HC (2001) Cutaneous
photodamage in Koreans: influence of sex, sun exposure, smoking, and skin color. Arch
Dermatol 137(8):10431051
27. Daniel LC, Heckman CJ, Kloss JD, Manne SL (2009) Comparing alternative methods of
measuring skin color and damage. Cancer Causes Control CCC 20(3):313321. doi:10.1007/
s10552-008-9245-3
28. Darlington S, Williams G, Neale R, Frost C, Green A (2003) A randomized controlled trial to
assess sunscreen application and beta carotene supplementation in the prevention of solar
keratoses. Arch Dermatol 139(4):451455. doi:10.1001/archderm.139.4.451
29. Davis EC, Callender VD (2010) Postinflammatory hyperpigmentation: a review of the epide-
miology, clinical features, and treatment options in skin of color. J Clin Aesthet Dermatol
3(7):2031
30. de Rigal J, Abella ML, Giron F, Caisey L, Lefebvre MA (2007) Development and validation
of a new Skin Color Chart. Skin Res Technol Off J Int Soc Bioeng Skin 13(1):101109.
doi:10.1111/j.1600-0846.2007.00223.x
31. Del Bino S, Bernerd F (2013) Variations in skin colour and the biological consequences of
ultraviolet radiation exposure. Br J Dermatol 169(Suppl 3):3340. doi:10.1111/bjd.12529
32. Del Bino S, Sok J, Bernerd F (2013) Assessment of ultraviolet-radiation-induced DNA dam-
age within melanocytes in skin of different constitutive pigmentation. Br J Dermatol
168(5):11201123. doi:10.1111/bjd.12201
33. Del Bino S, Sok J, Bessac E, Bernerd F (2006) Relationship between skin response to ultra-
violet exposure and skin color type. Pigment Cell Res (Sponsored by the European Society
for Pigment Cell Research and the International Pigment Cell Society) 19(6):606614.
doi:10.1111/j.1600-0749.2006.00338.x
34. Eide MJ, Weinstock MA (2005) Association of UV index, latitude, and melanoma incidence
in nonwhite populationsUS Surveillance, Epidemiology, and End Results (SEER) Program,
1992 to 2001. Arch Dermatol 141(4):477481. doi:10.1001/archderm.141.4.477
35. Fleming ID, Barnawell JR, Burlison PE, Rankin JS (1975) Skin cancer in black patients.
Cancer 35(3):600605
36. Galindo GR, Mayer JA, Slymen D, Almaguer DD, Clapp E, Pichon LC, Hoerster K, Elder JP
(2007) Sun sensitivity in 5 US ethnoracial groups. Cutis 80(1):2530
37. Gloster HM Jr, Neal K (2006) Skin cancer in skin of color. J Am Acad Dermatol 55(5):741
760, quiz 761744. doi:10.1016/j.jaad.2005.08.063
38. Goh SH (1990) The treatment of visible signs of senescence: the Asian experience. Br
J Dermatol 122(Suppl 35):105109
39. Grant WB (2002) An estimate of premature cancer mortality in the U.S. due to inadequate
doses of solar ultraviolet-B radiation. Cancer 94(6):18671875
40. Grant WB, Garland CF (2006) The association of solar ultraviolet B (UVB) with reducing
risk of cancer: multifactorial ecologic analysis of geographic variation in age-adjusted cancer
mortality rates. Anticancer Res 26(4A):26872699
41. Gray J, Hawk JLM (eds) (1998) The benefit of lifetime photoprotection. International
Congress & Symposium Royal Society of Medicine Press, London
42. Green A, Williams G, Neale R, Hart V, Leslie D, Parsons P, Marks GC, Gaffney P,
Battistutta D, Frost C, Lang C, Russell A (1999) Daily sunscreen application and beta
carotene supplementation in prevention of basal-cell and squamous-cell carcinomas of the
skin: a randomised controlled trial. Lancet 354(9180):723729. doi:10.1016/
S0140-6736(98)12168-2
43. Green AC, Williams GM, Logan V, Strutton GM (2011) Reduced melanoma after regular
sunscreen use: randomized trial follow-up. J Clin Oncol Off J Am Soc Clin Oncol 29(3):
257263. doi:10.1200/JCO.2010.28.7078
120 K.J. Buster and J.J. Ledet
44. Grimes P, Edison BL, Green BA, Wildnauer RH (2004) Evaluation of inherent differences
between African American and white skin surface properties using subjective and objective
measures. Cutis 73(6):392396
45. Grimes PE (1995) Melasma. Etiologic and therapeutic considerations. Arch Dermatol
131(12):14531457
46. Grimes PE (2009) Management of hyperpigmentation in darker racial ethnic groups. Semin
Cutan Med Surg 28(2):7785. doi:10.1016/j.sder.2009.04.001
47. Grimes PE, Yamada N, Bhawan J (2005) Light microscopic, immunohistochemical, and
ultrastructural alterations in patients with melasma. Am J Dermatopathol 27(2):96101
48. Halder RM, Bang KM (1988) Skin cancer in blacks in the United States. Dermatol Clin
6(3):397405
49. Halder RM, Bridgeman-Shah S (1995) Skin cancer in African Americans. Cancer 75(2 Suppl):
667673
50. Halder RM, Nootheti PK (2003) Ethnic skin disorders overview. J Am Acad Dermatol
48(6 Suppl):S143S148. doi:10.1067/mjd.2003.274
51. Hernandez-Perez E, Ibiett EV (2002) Gross and microscopic findings in patients submitted to
nonablative full-face resurfacing using intense pulsed light: a preliminary study. Dermatol
Surg Off Publ Am Soc Dermatol Surg [et al] 28(8):651655
52. Hu S, Ma F, Collado-Mesa F, Kirsner RS (2004) UV radiation, latitude, and melanoma in US
Hispanics and blacks. Arch Dermatol 140(7):819824. doi:10.1001/archderm.140.7.819
53. Jablonski NG (2014) Is there a golden mean for sun exposure? J Intern Med 276(1):7173.
doi:10.1111/joim.12248
54. Kaidbey KH, Agin PP, Sayre RM, Kligman AM (1979) Photoprotection by melanin--a com-
parison of black and Caucasian skin. J Am Acad Dermatol 1(3):249260
55. Kang HY, Suzuki I, Lee DJ, Ha J, Reiniche P, Aubert J, Deret S, Zugaj D, Voegel JJ, Ortonne
JP (2011) Transcriptional profiling shows altered expression of wnt pathway- and lipid
metabolism-related genes as well as melanogenesis-related genes in melasma. J Investig
Dermatol 131(8):16921700. doi:10.1038/jid.2011.109
56. Kang S, Chung JH, Lee JH, Fisher GJ, Wan YS, Duell EA, Voorhees JJ (2003) Topical N-acetyl
cysteine and genistein prevent ultraviolet-light-induced signaling that leads to photoaging in
human skin in vivo. J Investig Dermatol 120(5):835841. doi:10.1046/j.1523-1747.2003.12122.x
57. Kang SJ, Davis SA, Feldman SR, McMichael AJ (2014) Dyschromia in skin of color. J Drugs
Dermatol JDD 13(4):401406
58. Kim EH, Kim YC, Lee ES, Kang HY (2007) The vascular characteristics of melasma.
J Dermatol Sci 46(2):111116. doi:10.1016/j.jdermsci.2007.01.009
59. Koh D, Wang H, Lee J, Chia KS, Lee HP, Goh CL (2003) Basal cell carcinoma, squamous
cell carcinoma and melanoma of the skin: analysis of the Singapore Cancer Registry data
196897. Br J Dermatol 148(6):11611166
60. Kotrajaras R, Kligman AM (1993) The effect of topical tretinoin on photodamaged facial
skin: the Thai experience. Br J Dermatol 129(3):302309
61. Krishnamurthy S (1992) The geography of non-ocular malignant melanoma in India: its asso-
ciation with latitude, ozone levels and UV light exposure. Int J Cancer 51(2):169172
62. Lacz NL, Vafaie J, Kihiczak NI, Schwartz RA (2004) Postinflammatory hyperpigmentation:
a common but troubling condition. Int J Dermatol 43(5):362365. doi:10.1111/j.1365-4632.
2004.02267.x
63. Lakhdar H, Zouhair K, Khadir K, Essari A, Richard A, Seite S, Rougier A (2007) Evaluation of
the effectiveness of a broad-spectrum sunscreen in the prevention of chloasma in pregnant women.
J Eur Acad Dermat Venereol JEADV 21(6):738742. doi:10.1111/j.1468-3083.2007.02185.x
64. Lee DJ, Lee J, Ha J, Park KC, Ortonne JP, Kang HY (2012) Defective barrier function in melasma
skin.JEurAcadDermatVenereolJEADV26(12):15331537.doi:10.1111/j.1468-3083.2011.04337.x
65. Lee SG, Ko NY, Son SW, Bae HJ, Ha JS, Pak HY, Shin YS (2013) The impact of ozone deple-
tion on skin cancer incidence in Korea. Br J Dermatol 169(5):11641165. doi:10.1111/
bjd.12472
7 Photoprotection and Skin of Color 121
66. Lin FH, Lin JY, Gupta RD, Tournas JA, Burch JA, Selim MA, Monteiro-Riviere NA, Grichnik
JM, Zielinski J, Pinnell SR (2005) Ferulic acid stabilizes a solution of vitamins C and E and
doubles its photoprotection of skin. J Investig Dermatol 125(4):826832. doi:10.1111/
j.0022-202X.2005.23768.x
67. Lin JY, Selim MA, Shea CR, Grichnik JM, Omar MM, Monteiro-Riviere NA, Pinnell SR
(2003) UV photoprotection by combination topical antioxidants vitamin C and vitamin E. J
Am Acad Dermatol 48(6):866874. doi:10.1067/mjd.2003.425
68. Lindqvist PG, Epstein E, Landin-Olsson M, Ingvar C, Nielsen K, Stenbeck M, Olsson H
(2014) Avoidance of sun exposure is a risk factor for all-cause mortality: results from the
Melanoma in Southern Sweden cohort. J Intern Med 276(1):7786. doi:10.1111/joim.12251
69. Mahler HI (2014) Reasons for using and failing to use sunscreen: comparison among whites,
Hispanics, and Asian/Pacific Islanders in Southern California. JAMA dermatology
150(1):9091. doi:10.1001/jamadermatol.2013.4992
70. Mahmoud BH, Ruvolo E, Hexsel CL, Liu Y, Owen MR, Kollias N, Lim HW, Hamzavi IH
(2010) Impact of long-wavelength UVA and visible light on melanocompetent skin. J Investig
Dermatol 130(8):20922097. doi:10.1038/jid.2010.95
71. Marionnet C, Grether-Beck S, Seite S, Marini A, Jaenicke T, Lejeune F, Bastien P, Rougier
A, Bernerd F, Krutmann J (2011) A broad-spectrum sunscreen prevents UVA radiation-
induced gene expression in reconstructed skin in vitro and in human skin in vivo. Exp
Dermatol 20(6):477482. doi:10.1111/j.1600-0625.2011.01265.x
72. Martin L, Caperton C, Wollert-Lloyd H, Avashia N (2012) A randomized double-blind pla-
cebo controlled study evaluating the effectiveness and tolerability of oral Polypodium leu-
cotomos in patients with melasma. J Am Acad Dermatol 66(4, Suppl 1):AB21
73. Masu S, Seiji M (1983) Pigmentary incontinence in fixed drug eruptions. Histologic and
electron microscopic findings. J Am Acad Dermatol 8(4):525532
74. Matsuoka LY, Schauer PK, Sordillo PP (1981) Basal cell carcinoma in black patients. J Am
Acad Dermatol 4(6):670672
75. McCall CO, Chen SC (2002) Squamous cell carcinoma of the legs in African Americans.
J Am Acad Dermatol 47(4):524529
76. Medicine Io (2011) Dietary reference intakes for adequacy: calcium and vitamin D; implica-
tions and special concerns. Dietary reference intakes for calcium and vitamin D. National
Academies Press, Washington, DC
77. Merriam-Webster Ethnicity. www.Merriam-Webster.com
78. Mersha TB, Abebe T (2015) Self-reported race/ethnicity in the age of genomic research: its
potential impact on understanding health disparities. Hum Genomics 9(1):1. doi:10.1186/
PREACCEPT-2695828013752627
79. Middelkamp-Hup MA, Pathak MA, Parrado C, Garcia-Caballero T, Rius-Diaz F, Fitzpatrick
TB, Gonzalez S (2004) Orally administered Polypodium leucotomos extract decreases
psoralen-UVA-induced phototoxicity, pigmentation, and damage of human skin. J Am Acad
Dermatol 50(1):4149. doi:10.1016/S0190
80. Moin A, Jabery Z, Fallah N (2006) Prevalence and awareness of melasma during pregnancy.
Int J Dermatol 45(3):285288. doi:10.1111/j.1365-4632.2004.02470.x
81. Montagna W, Carlisle K (1991) The architecture of black and white facial skin. J Am Acad
Dermatol 24(6 Pt 1):929937
82. Mora RG, Burris R (1981) Cancer of the skin in blacks: a review of 128 patients with basal-
cell carcinoma. Cancer 47(6):14361438
83. Mora RG, Perniciaro C (1981) Cancer of the skin in blacks. I. A review of 163 black patients
with cutaneous squamous cell carcinoma. J Am Acad Dermatol 5(5):535543
84. Moyal D (2012) Need for a well-balanced sunscreen to protect human skin from both
Ultraviolet A and Ultraviolet B damage. Indian J Dermatol Venereol Leprol 78(Suppl 1):S24
S30. doi:10.4103/0378-6323.97352
85. Newman P, Herman J (2009) Ozone and UV: Where are we now? Skin Cancer Found J
27:3842
122 K.J. Buster and J.J. Ledet
86. Obaidi J, Musallam E, Al-Ghzawi HM, Azzeghaiby SN, Alzoghaibi IN (2015) Vitamin d and
its relationship with breast cancer: an evidence based practice paper. Glob J Health Sci
7(1):35687. doi:10.5539/gjhs.v7n1p261
87. Oppenheimer GM (2001) Paradigm lost: race, ethnicity, and the search for a new population
taxonomy. Am J Public Health 91(7):10491055
88. Ortonne JP (1992) Retinoic acid and pigment cells: a review of in-vitro and in-vivo studies.
Br J Dermatol 127(Suppl 41):4347
89. Pandeya N, Purdie DM, Green A, Williams G (2005) Repeated occurrence of basal cell car-
cinoma of the skin and multifailure survival analysis: follow-up data from the Nambour Skin
Cancer Prevention Trial. Am J Epidemiol 161(8):748754. doi:10.1093/aje/kwi098
90. Pennello G, Devesa S, Gail M (2000) Association of surface ultraviolet B radiation levels
with melanoma and nonmelanoma skin cancer in United States blacks. Cancer Epidemiol
Biomarkers Prev Publ Am Assoc Cancer Res (Cosponsored by the American Society of
Preventive Oncology) 9(3):291297
91. Pichon LC, Corral I, Landrine H, Mayer JA, Norman GJ (2010) Sun-protection behaviors
among African Americans. Am J Prev Med 38(3):288295. doi:10.1016/j.amepre.2009.10.041
92. Pichon LC, Landrine H, Corral I, Hao Y, Mayer JA, Hoerster KD (2010) Measuring skin
cancer risk in African Americans: is the Fitzpatrick Skin Type Classification Scale culturally
sensitive? Ethn Dis 20(2):174179
93. Position statement developed in 2006. http://www.dermcoll.edu.au/wp-content/uploads/2014/05/
Risks_and_Benefits_of_Sun_Exposure03May07.pdf. Accessed Jan 2015
94. Powe CE, Evans MK, Wenger J, Zonderman AB, Berg AH, Nalls M, Tamez H, Zhang D,
Bhan I, Karumanchi SA, Powe NR, Thadhani R (2013) Vitamin D-binding protein and vita-
min D status of black Americans and white Americans. N Engl J Med 369(21):19912000
95. Resnik S (1967) Melasma induced by oral contraceptive drugs. JAMA 199(9):601605
96. Rigopoulos D, Gregoriou S, Katsambas A (2007) Hyperpigmentation and melasma. J Cosmet
Dermatol 6(3):195202. doi:10.1111/j.1473-2165.2007.00321.x
97. Rijken F, Bruijnzeel PL, van Weelden H, Kiekens RC (2004) Responses of black and white
skin to solar-simulating radiation: differences in DNA photodamage, infiltrating neutrophils,
proteolytic enzymes induced, keratinocyte activation, and IL-10 expression. J Investig
Dermatol 122(6):14481455. doi:10.1111/j.0022-202X.2004.22609.x
98. Rouhani P, Pinheiro PS, Sherman R, Arheart K, Fleming LE, Mackinnon J, Kirsner RS
(2010) Increasing rates of melanoma among nonwhites in Florida compared with the United
States. Arch Dermatol 146(7):741746. doi:10.1001/archdermatol.2010.133
99. Sanchez MR (2003) Cutaneous diseases in Latinos. Dermatol Clin 21(4):689697
100. Sanchez NP, Pathak MA, Sato S, Fitzpatrick TB, Sanchez JL, Mihm MC Jr (1981) Melasma:
a clinical, light microscopic, ultrastructural, and immunofluorescence study. J Am Acad
Dermatol 4(6):698710
101. Scotto J, Fears T, Fraumeni JJ (1983) Incidence of non-melanoma skin cancer in the United
States. http://www.ciesin.org/docs/001-526/001-526.html. Accessed Jan 2015
102. Scragg R, Sowers M, Bell C, Third National H, Nutrition Examination S (2004) Serum
25-hydroxyvitamin D, diabetes, and ethnicity in the Third National Health and Nutrition
Examination Survey. Diabetes Care 27(12):28132818
103. Seite S, Fourtanier A, Moyal D, Young AR (2010) Photodamage to human skin by suberythe-
mal exposure to solar ultraviolet radiation can be attenuated by sunscreens: a review. Br
J Dermatol 163(5):903914. doi:10.1111/j.1365-2133.2010.10018.x
104. Seite S, Fourtanier AM (2008) The benefit of daily photoprotection. J Am Acad Dermatol
58(5 Suppl 2):S160S166. doi:10.1016/j.jaad.2007.04.036
105. Sheehan JM, Cragg N, Chadwick CA, Potten CS, Young AR (2002) Repeated ultraviolet
exposure affords the same protection against DNA photodamage and erythema in human skin
types II and IV but is associated with faster DNA repair in skin type IV. J Investig Dermatol
118(5):825829. doi:10.1046/j.1523-1747.2002.01681.x
7 Photoprotection and Skin of Color 123
106. Singh B, Bhaya M, Shaha A, Har-El G, Lucente FE (1998) Presentation, course, and outcome
of head and neck skin cancer in African Americans: a case-control study. Laryngoscope
108(8 Pt 1):11591163
107. Skaaby T (2015) The relationship of vitamin D status to risk of cardiovascular disease and
mortality. Danish Med J 61(2)
108. Smit NP, Vink AA, Kolb RM, Steenwinkel MJ, van den Berg PT, van Nieuwpoort F, Roza L,
Pavel S (2001) Melanin offers protection against induction of cyclobutane pyrimidine dimers
and 64 photoproducts by UVB in cultured human melanocytes. Photochem Photobiol
74(3):424430
109. Tadokoro T, Kobayashi N, Zmudzka BZ, Ito S, Wakamatsu K, Yamaguchi Y, Korossy KS,
Miller SA, Beer JZ, Hearing VJ (2003) UV-induced DNA damage and melanin content in
human skin differing in racial/ethnic origin. FASEB J Off Publ Fed Am Soc Exp Biol
17(9):11771179. doi:10.1096/fj.02-0865fje
110. Tadokoro T, Yamaguchi Y, Batzer J, Coelho SG, Zmudzka BZ, Miller SA, Wolber R, Beer JZ,
Hearing VJ (2005) Mechanisms of skin tanning in different racial/ethnic groups in response
to ultraviolet radiation. J Investig Dermatol 124(6):13261332. doi:10.1111/j.0022-202X.
2005.23760.x
111. Taylor SC, Cook-Bolden F (2002) Defining skin of color. Cutis 69(6):435437
112. Tewari A, Sarkany RP, Young AR (2012) UVA1 induces cyclobutane pyrimidine dimers but
not 64 photoproducts in human skin in vivo. J Investig Dermatol 132(2):394400.
doi:10.1038/jid.2011.283
113. Thompson SC, Jolley D, Marks R (1993) Reduction of solar keratoses by regular sunscreen
use. N Engl J Med 329(16):11471151. doi:10.1056/NEJM199310143291602
114. Toda K, Pathak MA, Parrish JA, Fitzpatrick TB, Quevedo WC Jr (1972) Alteration of racial
differences in melanosome distribution in human epidermis after exposure to ultraviolet light.
Nature New Biol 236(66):143145
115. Tomita Y, Maeda K, Tagami H (1992) Melanocyte-stimulating properties of arachidonic acid
metabolites: possible role in postinflammatory pigmentation. Pigment Cell Res (Sponsored
by the European Society for Pigment Cell Research and the International Pigment Cell
Society) 5(5 Pt 2):357361
116. Tsoureli-Nikita E, Watson RE, Griffiths CE (2006) Photoageing: the darker side of the sun.
Photochem Photobiol Sci Off J Eur Photochem Assoc Eur Soc Photobiol 5(2):160164.
doi:10.1039/b507492d
117. van der Pols JC, Williams GM, Pandeya N, Logan V, Green AC (2006) Prolonged prevention
of squamous cell carcinoma of the skin by regular sunscreen use. Cancer Epidemiol
Biomarkers Prev Publ Am Assoc Cancer Res (Cosponsored by the American Society of
Preventive Oncology) 15(12):25462548. doi:10.1158/1055-9965.EPI-06-0352
118. Victor FC, Gelber J, Rao B (2004) Melasma: a review. J Cutan Med Surg 8(2):97102.
doi:10.1007/s10227-004-0158-9
119. Vidal-Ortiz S (2008) People of Color. In: Schaefer RT (ed) Encyclopedia of race, ethnicity,
and society. SAGE Publications, Los Angeles, pp 10371039
120. Warren R, Gartstein V, Kligman AM, Montagna W, Allendorf RA, Ridder GM (1991) Age,
sunlight, and facial skin: a histologic and quantitative study. J Am Acad Dermatol 25(5 Pt 1):
751760
121. Weinstock MA (1993) Nonmelanoma skin cancer mortality in the United States, 1969
through 1988. Arch Dermatol 129(10):12861290
122. Weinstock MA, Moses AM (2009) Skin cancer meets vitamin D: the way forward for derma-
tology and public health. J Am Acad Dermatol 61(4):720724. doi:10.1016/j.jaad.2009.04.016
123. World Health Organization (2002) Global solar UV index: a practical guide. http://www.who.
int/uv/publications/en/UVIGuide.pdf. Accessed 21 Jan 2015
124. Wingo PA, Bolden S, Tong T, Parker SL, Martin LM, Heath CW Jr (1996) Cancer statistics
for African Americans, 1996. CA Cancer J Clinicians 46(2):113125
124 K.J. Buster and J.J. Ledet
125. Yang L, Lof M, Veierod MB, Sandin S, Adami HO, Weiderpass E (2011) Ultraviolet exposure
and mortality among women in Sweden. Cancer Epidemiol Biomarkers Prev Publ Am Assoc
Cancer Res (Cosponsored by the American Society of Preventive Oncology) 20(4):683690.
doi:10.1158/1055-9965.EPI-10-0982
126. Yohn JJ, Lyons MB, Norris DA (1992) Cultured human melanocytes from black and white
donors have different sunlight and ultraviolet A radiation sensitivities. J Investig Dermatol
99(4):454459
127. Youn JI, Oh JK, Kim BK, Suh DH, Chung JH, Oh SJ, Kim JJ, Kang SH (1997) Relationship
between skin phototype and MED in Korean, brown skin. Photodermatol Photoimmunol
Photomed 13(56):208211
128. Zeeb H, Greinert R (2010) The role of vitamin D in cancer prevention: does UV protection
conflict with the need to raise low levels of vitamin D? Deutsches Arzteblatt Int 107(37):
638643. doi:10.3238/arztebl.2010.0638
129. Zheng Y, Zhu J, Zhou M, Cui L, Yao W, Liu Y (2013) Meta-analysis of long-term vitamin D
supplementation on overall mortality. PLoS One 8(12):e82109. doi:10.1371/journal.
pone.0082109
Chapter 8
The Controversy of Sunscreen Product
Exposure: Too Little, Too Much, or Just Right
Key Points
Sunscreen products have a controversial history. Fundamental to the con-
troversies surrounding sunscreens is product use or exposure.
It is alleged too little sunscreen product is applied reducing their effective-
ness or, conversely, that too much product is used resulting in unfavorable
health effects.
The weight of evidence is supportive of daily use of sunscreens as part of
a safe sun strategy including wearing protective clothing and seeking
shade. Importantly, a consistent, simple public health message is required
and supported broadly by all stakeholders.
8.1 Introduction
Exposure to sunlight, which is crucial for human survival, can have detrimental
effects on our skin. The absence of hair covering our bodies makes human skin
vulnerable to the effects of ultraviolet radiation (UVR) in sunlight. Acute overexpo-
sure to sunlight results in erythema, i.e., sunburn, and in more extreme cases edema,
which are thought to be a manifestation of complex molecular events, including
DNA damage and the release of cytokines [10, 22, 57]. Exposure to UVR also trig-
gers melanogenesis or tanning, a protective mechanism but only to the extent that
damage is the initiating biological event [32, 49].
Beyond the acute effects of sunlight overexposure, the prevailing view is that
UVR-induced skin damage is cumulative [24, 26]. Such damage, over decades of
life, may lead directly or significantly contribute to nonmelanoma and melanoma
skin cancers [12, 68, 90] and photoaging, characterized by wrinkles, pigmentary
unevenness, and telangiectasia [25, 67].
Public health education campaigns seem to be having the desired effect since
there is general knowledge among teenagers and adults in the USA that exposure
to UVR from sunlight can cause skin cancers and photoaging [21]. The use of
sun-protective behaviors has held steady and actually increased from 2000 to
2010, although the percentage remains relatively low, i.e., less than 35 % for
women and men [15]. Nevertheless, there still are many cases of skin cancers in
the USA.1 It is possible that there is a lag between widespread sun-protective
activities and prevalence of skin cancers. However, it is equally likely that behav-
iors including indoor tanning and sunbathing contribute disproportionately to
unfavorable long-term health effects. Thus, despite gains in awareness regarding
the detrimental effects of sunlight, it would seem cosmetic/appearance benefits
make it difficult for individuals to change behavior even when they know it is
harmful [48, 78]. Further complicating the desire to intentional expose oneself to
UVR is the evidence that such behavior may have some addictive components
[27, 89].
Given the unequivocal cause-effect relationship between sunlight exposure
and skin damage as well as the health-related messages advocating active out-
door lifestyles, it would seem products whose singular purpose is to reduce the
dose of solar UVR might be of unquestionable benefit. In the simplest of terms,
sunscreens are such products. The ultraviolet (UV) filters are active ingredients
applied to sun-exposed areas of the skin with the sole purpose of reducing the
number of photons reaching areas where damage might occur. After absorbing
energy, the UV filters dissipate it in the form of heat or phosphorescence
[43, 72]. In some cases the photon energy is reflected or scattered, again reducing
the energy reaching vital cells in the skin. Thus, when shade, clothing, or hats are
not options in high-intensity exposure scenarios, sunscreens serve as the best
alternative to protect against sunlight. As well, for daily incidental exposure,
such products are, quite arguably, the most effective agents to reduce the signs of
aging.
Despite this elementary proposition and decades of use, a host of controversies
follow sunscreens. There are numerous issues, many of which have been reviewed
elsewhere [14, 47, 50, 69, 86]. What these controversies share, to a large extent, is
linked by exposure, generally too little or too much. Thus, the purpose herein is
not to repeat the arguments made by others but to consider the principle, underpinning
the controversies regarding sunscreen use.
1
www.skincancer.org.
8 The Controversy of Sunscreen Product Exposure: Too Little, Too Much, or Just Right 127
For sunscreen products, there are many concerns related to efficacy and safety that
might be classified as controversies. Importantly, in both cases these concerns have
a shared origin, namely, exposure. For the purposes of this paper, exposure is a
borrowed term from risk assessment where the magnitude, frequency, and duration
of use are measured or estimated [45]. In toxicology, exposure is coupled with
hazard, i.e., adverse effect, and dose-response data to determine risk. In the context
of efficacy, magnitude, or dose, frequency or reapplication and duration of use will
be discussed and how they are controversial relative to human health.
Exposure to sunlight is also part of the consideration. As stated, sunlight damages
skin. Sunscreens or more accurately UV filters are without an endogenous biological
target, i.e., lacking pharmacological activity. As such, application of such products to
the skin has no effect in the absence of sunlight. Thus, for sunscreens, the product expo-
sure is coupled inextricably to sunlight and serves as the basis for all the controversies.
Arguably, the most contentious issue involving sunscreens is the widely held view,
supported by numerous studies, that they are under dosed, i.e., not enough is
applied, under ad-lib conditions. This view is tied inseparably to the SPF test which
has for decades been conducted using a dose of 2 mg/cm2 [23, 29, 71]. The reason
for using 2 mg/cm2 in SPF test has little to do with consumer use. As with any
procedure that may be used to support a product claim, reproducibility is
paramount to widespread acceptance. To have a universally applied laboratory
result, the inter- and intralaboratory variability must be low; otherwise test results
become untrustworthy. One of the primary sources of variability in the SPF test and
known for many years is product application [71]. As the SPF test was being
developed into a uniform, international method, largely led by the cosmetic trade
association in Europe, Cosmetic Europe or CE, formerly COLIPA, it was agreed
that 2 mg/cm2 application dose was reproducible. There was never the intention of
this efficacy test to mirror how a consumer used the product. It is, in fact, an
unreasonable expectation given habits and practices differences and diverse product
forms, e.g., water-resistant recreational products vs. moisturizers or lipsticks. As a
result, the SPF test is conducted as a means for product comparison and not an
absolute efficacy value. Unfortunately, too many professional/nonprofessionals
interpret SPF as an in-use, absolute quantitative value of efficacy. As stated and
generally speaking, it is not.
Finally, it is worth noting that there exists a standard method for determining
in vivo SPF, ISO 24444, which has been adopted, worldwide, except for the USA,
although in all methods, the application density is the same, i.e., 2 mg/cm2.
128 J.F. Nash and P.R. Tanner
Forgetting for the moment UV filters and film-forming characteristics which are
critical for sunscreen product efficacy, attributes like the scent, feel, and optical
appearance on the skin would be expected to impact how much and how often, i.e.,
frequency, it is used. If consumers do not like the feel, e.g., sticky and greasy, scent,
e.g., chemical base odor, or on-skin appearance, e.g., shine and whiteness of the
product, they will use less and perhaps avoid reapplication. Thus, formulating better
sunscreen products is about much more than just performing well in SPF clinical
tests or having new UV filters. What dose and reapplication translate into is com-
pliance, which is rarely discussed when considering sunscreens.
Another characteristic that impacts sunscreen dose is film-forming properties
and thickness. From a technical standpoint, the ability of a sunscreen product to
form a uniform film on the skin is closely tied to efficacy [63, 74]. In fact, film
formation/thickness is likely the key reason that product application is one of the
primary sources of variability in SPF testing, as mentioned earlier. As well, the film
formation/thickness has implications related to reapplication. To understand this,
one needs to consider the topography of the skin (Fig. 8.1). Macroscopically, the
surface of the skin is made up of hills and valleys. A thin layer applied over such
topography may result in uneven coverage where valleys are filled/covered, but
peaks are not. The analogy Diffey uses is that of painting a wall with an uneven
surface [18]. The first coat/application doesnt provide adequate coverage, and
therefore two coats (reapplication) are required. However, one goal of sunscreen
8 The Controversy of Sunscreen Product Exposure: Too Little, Too Much, or Just Right 131
c Uniform film
product development is to create products that have uniform film formation, high
efficacy, under ad-lib conditions of use. In this regard, uniform coverage may be
obtained at less than 2 mg/cm2. Certainly, the more product applied, the more likely
coverage will be achieved and in lock-step the more negative attributes such as
greasiness and product remaining on the skin, i.e., not absorbed,2 come into play.
In general, the combination of product attributes, efficacy, and experience drives
how much product is applied and/or reapplied.
Studies of acute sunscreen product failure under ad-lib use conditions are limited or
a secondary objective. Some examples which have reported erythema/sunburn in
people using sunscreen include McCarthy et al. [53] and Wright et al. [88]. Again,
in these examples and other such studies, it is difficult to know if sunburn was due
to inadequate dose or missed area on the body or overexposure to sunlight or
combinations of these. For example, intentional misuse of sunscreen to prolong
time spent in the sun for tanning purposes can result in sunburn suggestive of
product failure [24]. What is not factored into product failure are the millions if
not billions of product applications where sunburn has been prevented. Finally, the
work of Green et al. in the Nambour Skin Cancer and Actinic Eye Disease Prevention
2
Absorption in this context is a term used to describe whether a consumer feels product
remaining on the skin after application. It is not used in the context of pharmacokinetics, i.e.,
absorption into the skin, but rather an aesthetic attribute.
132 J.F. Nash and P.R. Tanner
Trial [34, 35, 41], Thompson et al. [80], Naylor et al. [59], and Gallagher et al. [28]
are suggestive that repeated, regular application of sunscreen under ad-lib use
prevent precursors as well as actual long-term skin damage supportive of the view
that ad-lib sunscreen use is efficacious.
The use of sunscreens is not limited to amount of product applied. The frequency of
application or reapplication and duration of use are critical in understanding
exposure and efficacy. In an experimental context, these have not received as much
attention as the amount of product applied. As such, the number of prospective,
stand-alone studies is less compared to those where amount of product applied has
been investigated. Nonetheless, there are some studies that have investigated
sunscreen product reapplication [13, 16, 64, 65, 82].
The duration of use has been studied in a limited number of prospective studies.
The most important of such studies is that of Adele Green and colleagues [34, 35,
41]. Others, as mentioned above include the work of Thompson et al. [80], Naylor
et al. [59], and Gallagher et al. [28].
Consumers do not apply enough sunscreen product to achieve the labeled SPF,
but this does not mean the product failed or there is no efficacy. On the contrary, the
preponderance of data supports the view that protection from harmful UVR is
achieved under normal use conditions. There are numerous opportunities to
reinforce behaviors including reapplication and daily use, which have been shown
to have real benefits (see above) and, in theory, if started early in life would have the
greatest impact [19, 76]. Unfortunately, the controversies related to amount/dose
applied and lack of short-/long-term benefits obscure the benefits and public health
message.
modeled after National Research Council [54], is used frequently to assess many
different chemicals used by humans and by design is a key part of toxicology/
product safety [37]. Many authoritative, e.g., Scientific Committee Cosmetic Safety,
and regulatory agencies, e.g., Environmental Protection Agency, around the world
use QRA as part of their approach toward ensuring consumer safety.
In the USA, there are nine UV filters commonly used in sunscreen products [83].
It is beyond the scope of this paper to review the safety of each UV filter in any
depth, and the interested reader may consider [30, 50, 58] for more information.
However, among the human safety concerns related to these UV filters and sunscreen
product exposure are: (1) spray products and inhalation, (2) endocrine disruption,
and (3) systemic absorption from lifetime exposure, i.e., cradle to grave, including
subpopulations, e.g., geriatric.
A complexity associated with safety controversies and sunscreens is what drives
the concern? Is it the product, one of the UV filters, or a combination? The attempt
here will be to outline the controversial concern and provide general comments with
support by specific examples knowing that this will be limited by design.
Sunscreen spray products became more widely available in the decade of 2000
as a convenient means of product application particularly for children. Whereas
pump sprays had been available for some time, the propellant-based continuous
sprays represent a new form that has grown and by some estimates represents up
to 50 % of recreational sunscreen product market in the USA [1, 20]. Spray products
are thought to improve coverage, dosage, and drive compliance (see Novick et al.
Table 8.1). The concern, however, is inhalation particularly among children. The
toxicological profile of UV filters following the inhalation route of administration
has not been systematically investigated. However, human exposure is intermittent,
indirect, and restricted to nasal passages and to a lesser extent the upper respiratory
tract based on the size of droplets [20]. Beyond the local effects in these tissues, i.e.,
nasal/upper part of the lung, systemic effects would be dependent on the exact UV
filter and the availability of repeat exposure data perhaps generated from another
route of administration, e.g., oral or diet. Whereas each marketed sunscreen spray
product would need a safety evaluation based on specific properties and UV filters,
in general, exposure to ingredients would be limited if not negligible.
UV filters have been shown to have endocrine effects in screening-type toxico-
logical studies with benzophenone-3/oxybenzone [84], 4-methybenzylidene-
camphor/4-MBC [56], and octyl methoxycinnamate/OMC [5], receiving the most
attention [87]. Clearly, in vitro screening studies and findings in animals are sugges-
tive of weak endocrine effects of select UV filters. The limited human studies have
found internal concentrations of select UV filters in ng/ml range with no impact on
measures of endocrine function, i.e., basal concentrations of hormones [42]. Greim,
discussing endocrine disruption, made the following observation: Overall, the sci-
ence-based knowledge on the robustness of the endocrine system, the well-under-
stood principles of substrate-receptor interactions, and the generally low exposure of
humans to potentially endocrine-disrupting chemicals make it unlikely that the latter
play a causative role in diseases and abnormalities observed in children and in the
human population in general [36]. That is not to dismiss the notion of subtle endo-
134 J.F. Nash and P.R. Tanner
crine effects attributed to UV filters but in the context of systemic exposure follow-
ing topical application, the risk is consider by many to be minimal if not negligible.
In the context of endocrine disruption, it is worth pointing out vitamin D might
be considered as having endocrine properties and most notably is activated by
sunlight in the skin. Hence, by definition, sunscreens disrupt vitamin D by
reducing photochemical activation. So much has been written on this controversy,
and still the debate continues. Suffice it to say that the argument persists with
staunch supporters of sunscreens not affecting vitamin D under ad-lib use and
equally dedicated opponents suggesting sunscreen use has an unfavorable effect on
serum vitamin D and the risk outweighs any possible benefit [11, 31, 33, 39, 40, 44,
73]. Perhaps the only undisputable facts are that sunscreens, by design, have the
potential to reduce photochemical conversation of vitamin D, while systemic
endocrine effects mediated directly by UV filters is, at best, weak.
Systemic absorption and lifetime exposure to UV filters after topical application
came to attention of the scientific community in the late 1990s following the Lancet
publication by Hayden et al. [38]. Although studies preceding this exist, most had
minimized the idea of systemic bioavailability of UV filters, perhaps in a dismissive
manner. UV filters may penetrate into/through the skin [52, 55, 81, 85], to cite a few
examples. Most studies have found limited penetration, but as analytical detection
methods improve, it is quite likely to see more examples demonstrating systemic
absorption from clinical investigations and in biomonitoring, e.g., NHANES.
The concern related to the potential topical bioavailability of UV filters is
systemic toxicity. In years past, many risk assessments focused only on local effects,
e.g., skin irritation, sensitization, phototoxicity, or photoallergy. With evidence of
absorption following topical application, there are reasons to consider systemic
toxicity. This, again, comes down to the data for individual UV filters supportive of
repeat exposure. Importantly, the presence of a substance is not evidence of toxicity.
This seems to be a common misconception.
The examples of human safety concerns, inhalation, endocrine effects, and sys-
temic toxicity from topically applied sunscreens have been presented in a very
superficial manner. The point of these examples is that there are fears of too much
exposure to sunscreen/UV filters. This is largely independent of any benefits that
use of such products might offer. The controversy regarding too much exposure
to sunscreen products remains an active area of interest.
8.5 Conclusions
eventually discover Goldilocks who in her fright flees never to be seen by the bears
again. Sunscreens are considered to be used not enough or relied upon too frequently
or by some just right. Yet, in the end, the user is confused and possibly frightened
away by all the controversy and divergent opinions.
Of course, this analogy isnt nearly as black and white for sunscreens. For years,
the same controversies find their way into articles or on websites generally coin-
ciding with the advent of summer. People dont apply enough, and the ingredients
are unsafe, i.e., too much/toxic. The controversies will not go away, but that
shouldnt be a cause to abandon efforts.
In the end, the responsibility to public health begs for a consistent message that
can be applied for all to follow. A single message to limit sun exposure and follow
the guidelines that have proven effective in Australia, namely, wear protective
clothing, use a sunscreen product on skin exposure to sunlight and seek shade/
shelter, may be the means of improving public health particularly as humans extend
the life span and therefore cumulative exposure to sunlight. Rather than confuse
people with messages of use an SPF 30 or SPF 15 daily, it should be agreed that a
consistent message would benefit the public.
Beyond consistence in messaging, it would be of value to consider practical
advice. For example, sunscreens in the USA and elsewhere are largely viewed as
recreational products to be used as needed. So the idea of daily application hardly
resonates with a typical user of such products. This is just common sense: Why
apply a recreational product on days I will have little exposure to sunlight. Yet those
days are thought to account for much of the cumulative damage [61]. Additionally,
in a heterogeneous society like the USA, significant segments of the population
including African-/Asian-Americans and Latinos may not use sunscreen products
because they believe it is unnecessary, even though photodamage occurs in all skin
types. Again, with regard to sunscreens, the idea of an SPF 30 applied daily in the
context of recreational products, i.e., those applied to large surface areas, is unlikely
and for the average person not affordable. Unfortunately, even well-meaning
conscientious advocates may be missing an opportunity.
The controversy is around just right. It is the responsible act to promote sun
safety of which sunscreens are a key part. The single message might be: wear sun-
screen, SPF 15 broad-spectrum or greater on exposed skin. Reapply as needed. If this
encourages people to try sunscreen, it is quite possible that such trail will lead to retrial
and rather than using high SPF, e.g., 3050+, a new user can begin with a SPF 15.
References
1. Amber KT, Bloom R, Staropoli P, Dhiman S, Hu S (2014) Assessing the current market of
sunscreen: a cross-sectional study of sunscreen availability in three metropolitan counties in
the United States. J Skin Cancer 2014:285357
2. Autier P (2009) Sunscreen abuse for intentional sun exposure. Br J Dermatol 161(Suppl 3):
4045
136 J.F. Nash and P.R. Tanner
3. Autier P, Dore JF, Negrier S, Lienard D, Panizzon R, Lejeune FJ, Guggisberg D, Eggermont
AM (1999) Sunscreen use and duration of sun exposure: a double-blind, randomized trial.
J Natl Cancer Inst 91:13041309
4. Autier P, Dore JF, Reis AC, Grivegnee A, Ollivaud L, Truchetet F, Chamoun E, Rotmensz N,
Severi G, Cesarini JP (2000) Sunscreen use and intentional exposure to ultraviolet A and B radia-
tion: a double blind randomized trial using personal dosimeters. Br J Cancer 83:12431248
5. Axelstad M, Boberg J, Hougaard KS, Christiansen S, Jacobsen PR, Mandrup KR, Nellemann
C, Lund SP, Hass U (2011) Effects of pre- and postnatal exposure to the UV-filter octyl
methoxycinnamate (OMC) on the reproductive, auditory and neurological development of rat
offspring. Toxicol Appl Pharmacol 250:278290
6. Azurdia RM, Pagliaro JA, Diffey BL, Rhodes LE (1999) Sunscreen application by photosensi-
tive patients is inadequate for protection. Br J Dermatol 140:255258
7. Azurdia RM, Pagliaro JA, Rhodes LE (2000) Sunscreen application technique in photosensi-
tive patients: a quantitative assessment of the effect of education. Photodermatol Photoimmunol
Photomed 16:5356
8. Bauer U, O'BRIEN DS, KIMLIN MG (2010) A new method to quantify the application thick-
ness of sunscreen on skin. Photochem Photobiol 86:13971403
9. Bech-Thomsen N, Wulf HC (1992) Sunbathers application of sunscreen is probably inade-
quate to obtain the sun protection factor assigned to the preparation. Photodermatol
Photoimmunol Photomed 9:242244
10. Bernard JJ, Cowing-Zitron C, Nakatsuji T, Muehleisen B, Muto J, Borkowski AW, Martinez L,
Greidinger EL, Yu BD, Gallo RL (2012) Ultraviolet radiation damages self noncoding RNA
and is detected by TLR3. Nat Med 18:12861290
11. Berwick M (2011) The good, the bad, and the ugly of sunscreens. Clin Pharmacol Ther 89:3133
12. Brash DE (2015) UV signature mutations. Photochem Photobiol 91:1526
13. Buller DB, Andersen PA, Walkosz BJ, Scott MD, Maloy JA, Dignan MB, Cutter GR (2012)
Compliance with sunscreen advice in a survey of adults engaged in outdoor winter recreation
at high-elevation ski areas. J Am Acad Dermatol 66:6370
14. Burnett ME, Wang SQ (2011) Current sunscreen controversies: a critical review. Photodermatol
Photoimmunol Photomed 27:5867
15. CDC (2012) Sunburn and sun protective behaviors among adults aged 1829 years United
States, 20002010. In: Moolenaar RL (ed). Office of Surveillance, Epidemiology, and
Laboratory Services, Centers for Disease Control and Prevention (CDC), U.S. Department of
Health and Human Service, Atlanta
16. De Villa D, Nagatomi AR, Paese K, Guterres S, Cestari TF (2011) Reapplication improves the
amount of sunscreen, not its regularity, under real life conditions. Photochem Photobiol
87:457460
17. Diaz A, Neale RE, Kimlin MG, Jones L, Janda M (2012) The children and sunscreen study: a
crossover trial investigating children's sunscreen application thickness and the influence of age
and dispenser type. Arch Dermatol 148:606612
18. Diffey B (2001) Sunscreen isnt enough. J Photochem Photobiol B 64:105108
19. Diffey BL (2011) The impact of topical photoprotectants intended for daily use on lifetime
ultraviolet exposure. J Cosmet Dermatol 10:245250
20. Durand L, Habran N, Denis C, Meulders L, Henschel V, Amighi K (2007) Influence of differ-
ent parameters on droplet size and size distribution of sprayable sunscreen emulsions with high
concentration of UV-filters. Int J Cosmet Sci 29:461471
21. Eide MJ, Weinstock MA (2006) Public health challenges in sun protection. Dermatol Clin
24:119124
22. Faustin B, Reed JC (2008) Sunburned skin activates inflammasomes. Trends Cell Biol 18:48
23. Ferguson J, Brown M, Alert D, Bielfeldt S, Brown J, Chardon A, Hourseau C, Mazilier C,
Cuthbert J, DArcy-Burt K, Jolley J, Murdoch M, Finkel P, Masson P, Merot F, Maclennan A,
Poret J, Siladgi S (1996) Collaborative development of a sun protection factor test method: a
proposed European Standard. COLIPA Task Force Sun Protection Measurement, Europe. Int
J Cosmet Sci 18:203218
8 The Controversy of Sunscreen Product Exposure: Too Little, Too Much, or Just Right 137
24. Fisher GJ, Kang S, Varani J, Bata-Csorgo Z, Wan Y, Datta S, Voorhees JJ (2002) Mechanisms
of photoaging and chronological skin aging. Arch Dermatol 138:14621470
25. Fisher GJ, Talwar HS, Lin J, Voorhees JJ (1999) Molecular mechanisms of photoaging in
human skin in vivo and their prevention by all-trans retinoic acid. Photochem Photobiol
69:154157
26. Fisher GJ, Wang ZQ, Datta SC, Varani J, Kang S, Voorhees JJ (1997) Pathophysiology of
premature skin aging induced by ultraviolet light. N Engl J Med 337:14191428
27. Flores KG, Erdei E, Luo L, White KA, Leng S, Berwick M, Lazovich D (2013) A pilot study
of genetic variants in dopamine regulators with indoor tanning and melanoma. Exp Dermatol
22:576581
28. Gallagher RP, Rivers JK, Lee TK, Bajdik CD, Mclean DI, Coldman AJ (2000) Broad-spectrum
sunscreen use and the development of new nevi in white children: a randomized controlled
trial. JAMA 283:29552960
29. Garzarella K, Caswell M (2013) Disparate SPF testing methodologies generate similar SPFs.
J Cosmet Sci 64:297307
30. Gasparro FP, Mitchnick M, Nash JF (1998) A review of sunscreen safety and efficacy.
Photochem Photobiol 68:243256
31. Gilchrest BA (2008) Sun exposure and vitamin D sufficiency. Am J Clin Nutr 88:570S577S
32. Gilchrest BA, Park HY, Eller MS, Yaar M (1996) Mechanisms of ultraviolet light-induced
pigmentation. Photochem Photobiol 63:110
33. Grant WB, Strange RC, Garland CF (2003) Sunshine is good medicine. The health benefits of
ultraviolet-B induced vitamin D production. J Cosmet Dermatol 2:8698
34. Green A, Williams G, Neale R, Hart V, Leslie D, Parsons P, Marks GC, Gaffney P, Battistutta D,
Frost C, Lang C, Russell A (1999) Daily sunscreen application and betacarotene supplementa-
tion in prevention of basal-cell and squamous-cell carcinomas of the skin: a randomised con-
trolled trial. Lancet 354:723729
35. Green AC, Williams GM, Logan V, Strutton GM (2011) Reduced melanoma after regular sun-
screen use: randomized trial follow-up. J Clin Oncol 29:257263
36. Greim H (2005) Chemicals with endocrine-disrupting potential: a threat to human health?
Angew Chem Int Ed Engl 44:55685574
37. Hartung T (2011) From alternative methods to a new toxicology. Eur J Pharm Biopharm
77:338349
38. Hayden CG, Roberts MS, Benson HA (1997) Systemic absorption of sunscreen after topical
application. Lancet 350:863864
39. Holick M (2002) Does sunscreen block the skins ability to make vitamin D? If so, how can I
get enough of this vitamin without raising my risk of skin cancer? Health News 8:12
40. Holick MF, Matsuoka LY, Wortsman J (1995) Regular use of sunscreen on vitamin D levels.
Arch Dermatol 131:13371339
41. Iannacone MR, Hughes MC, Green AC (2014) Effects of sunscreen on skin cancer and photo-
aging. Photodermatol Photoimmunol Photomed 30:5561
42. Janjua NR, Kongshoj B, Andersson AM, Wulf HC (2008) Sunscreens in human plasma and urine
after repeated whole-body topical application. J Eur Acad Dermatol Venereol 22:456461
43. Jansen R, Osterwalder U, Wang SQ, Burnett M, Lim HW (2013) Photoprotection: part II. Sunscreen:
development, efficacy, and controversies. J Am Acad Dermatol 69:867 e114; quiz 8812
44. Kannan S, Lim HW (2014) Photoprotection and vitamin D: a review. Photodermatol
Photoimmunol Photomed 30:137145
45. Kleinjans JC (2003) Principles in toxicological risk analysis. Toxicol Lett 140141:311315
46. Lademann J, Schanzer S, Richter H, Pelchrzim RV, Zastrow L, Golz K, Sterry W (2004)
Sunscreen application at the beach. J Cosmet Dermatol 3:6268
47. Lautenschlager S, Wulf HC, Pittelkow MR (2007) Photoprotection. Lancet 370:528537
48. Lazovich D, Forster J (2005) Indoor tanning by adolescents: prevalence, practices and policies.
Eur J Cancer 41:2027
49. Liu JJ, Fisher DE (2010) Lighting a path to pigmentation: mechanisms of MITF induction by UV.
Pigment Cell Melanoma Res 23:741745
138 J.F. Nash and P.R. Tanner
72. Shaath NA (1997) Evolution of modern sunscreen chemicals. Marcel Dekker, New York
73. Skolnick AA (1991) Sunscreen protection controversy heats up. JAMA 265:32183220
74. Sohn M, Heche A, Herzog B, Imanidis G (2014) Film thickness frequency distribution of dif-
ferent vehicles determines sunscreen efficacy. J Biomed Opt 19:115005
75. Stenberg C, Larko O (1985) Sunscreen application and its importance for the sun protection
factor. Arch Dermatol 121:14001402
76. Stern RS, Weinstein MC, Baker SG (1986) Risk reduction for nonmelanoma skin cancer with
childhood sunscreen use. Arch Dermatol 122:537545
77. Szepietowski JC, Nowicka D, Reich A, Melon M (2004) Application of sunscreen preparations
among young Polish people. J Cosmet Dermatol 3:6972
78. Taylor MF, Westbrook D, Chang P (2015) Using UV photoaged photography to better
understand Western Australian teenagers attitudes towards adopting sun-protective behaviors.
Int J Adolesc Med Health. ISSN (Online) 2191-0278, ISSN (Print) 0334-0139, DOI: 10.1515/
ijamh-2014-0071, March 2015
79. Thieden E, Philipsen PA, Sandby-Moller J, Wulf HC (2005) Sunscreen use related to UV
exposure, age, sex, and occupation based on personal dosimeter readings and sun-exposure
behavior diaries. Arch Dermatol 141:967973
80. Thompson SC, Jolley D, Marks R (1993) Reduction of solar keratoses by regular sunscreen
use. N Engl J Med 329:11471151
81. Walters KA, Brain KR, Howes D, James VJ, Kraus AL, Teetsel NM, Toulon M, Watkinson AC,
Gettings SD (1997) Percutaneous penetration of octyl salicylate from representative sunscreen
formulations through human skin in vitro. Food Chem Toxicol 35:12191225
82. Wang SQ, Dusza SW (2009) Assessment of sunscreen knowledge: a pilot survey. Br J Dermatol
161(Suppl 3):2832
83. Wang SQ, Tanner PR, Lim HW, Nash JF (2013) The evolution of sunscreen products in the
United States a 12-year cross sectional study. Photochem Photobiol Sci 12:197202
84. Watanabe Y, Kojima H, Takeuchi S, Uramaru N, Sanoh S, Sugihara K, Kitamura S, Ohta S
(2015) Metabolism of UV-filter benzophenone-3 by rat and human liver microsomes and its
effect on endocrine-disrupting activity. Toxicol Appl Pharmacol 282:119128
85. Watkinson AC, Brain KR, Walters KA, Hadgraft J (1992) Prediction of the percutaneous pen-
etration of ultra-violet filters used in sunscreen formulations. Int J Cosmet Sci 14:265275
86. Weinstock MA (1996) Controversies in the role of sunlight in the pathogenesis of cutaneous
melanoma. Photochem Photobiol 63:406410
87. Witorsch RJ, Thomas JA (2010) Personal care products an endocrine disruption: a critical
review of the literature. Crit Rev Toxicol 40:130
88. Wright MW, Wright ST, Wagner RF (2001) Mechanisms of sunscreen failure. J Am Acad
Dermatol 44:781784
89. Zeller S, Lazovich D, Forster J, Widome R (2006) Do adolescent indoor tanners exhibit depen-
dency? J Am Acad Dermatol 54:589596
90. Ziegler A, Jonason AS, Leffell DJ, Simon JA, Sharma HW, Kimmelman J, Remington L, Jacks
T, Brash DE (1994) Sunburn and p53 in the onset of skin cancer. Nature 372:773776
Part II
Chapter 9
The Chemistry of Ultraviolet Filters
Nadim A. Shaath
Key Points
This chapter describes the mechanism of action for both Inorganic
particulates and organic ultraviolet filters. It classifies all ultraviolet filters
in commerce today and lists their physical, chemical, and spectroscopic
properties as well as their regulatory status.
Synthetic approaches for the design of the current and future UV filters are
discussed, and the photostability of ultraviolet filters is addressed.
It concludes with an analysis of the future direction in designing new,
safer, and more effective ultraviolet filters.
9.1 Introduction
With the inclusion of any of those UV filters, protection from skin cancers
was considered possible, and US companies could claim that sunscreens reduce
the risk of skin cancer and early skin aging when used as directed, if the final
sunscreen product has SPF >15 and critical wavelength 370 nm. Despite the
increased use of sun care products since then, incidences of skin cancer have
quadrupled with no sign of abatement. Are people lulled into a false sense of
security when they use sunscreens? All this sun damage begs the question: do
sun care products provide enough protection? The search for the ultimate UV
filter goes on, and protocols for superior protection are still underway with lim-
ited success.
In this chapter, I will review the approaches for designing the current UV filters
that have been approved and are available for use worldwide. Understanding how
filters work can help us to determine if they offer consumers adequate protection
from the sun.
9 The Chemistry of Ultraviolet Filters 145
HOMO
Excited Singlet
State
Intersystem
Excited
Vibrational Relaxation
Crossing
Triplet
State
Intersystem Crossing
Flourescence
Absorption
Phosphorescence
Photoproduct
Photoproduct
Ground State
Fig. 9.3 The band gap energy in inorganic particulates Conduction band
between valence and conduction bands
e- band gap
valence band
Ultraviolet filters can be broadly classified into two types: UV absorbers and
inorganic particulates. There are only two inorganic particulates approved: zinc
oxide and titanium dioxide. Both ingredients are considered broad spectrum since
they absorb, scatter, and reflect UVB and UVA rays depending on their particle size.
The remaining UV-absorbing molecules are classified as either UVB or UVA filters
or both.
There are about 55 ultraviolet filters that are approved for use in sunscreen prod-
ucts globally, but only 10 of them are approved uniformly for international con-
sumption [3, 4]. Table 9.2 lists their UV absorbance maxima (max) and their
specific extinction E (1 %, 1 cm), namely, the nominal absorbance at the absorption
maximum of a 1 % solution of the filter in a 1 cm optical pathway cuvette, the molar
absorption coefficient (mol1 cm1), along with the countries or regions where
they are approved. Each filter is approved or rejected according to regional require-
ments. Note that currently there are only ten UV filters that are approved uniformly
worldwide and are marked with an XXX in Table 9.2 under category country/region.
Benzophenone-8 US, AZ, CA, SA UVA/B 284 380 13,270 327 300 10,440
Benzophenone-9 JN, SA UVA/B 284 260 331 175
3-Benzylidene Camphor EU, SA UVB 289 890 21,360
Benzylidene camphor sulfonic acid EU, AZ, JN, SA UVB 294 860 27,600
Beta, 2-glucopyranoxy propyl JN, SA UVA/B
hydroxy Benzophenone
Bis-ethylhexyloxyphenol EU, AZ, SA UVA/B 310 745 46,800 343 820 51,900
methoxyphenyl triazine
Butyl methoxydibenzoylmethane XXX ALL UVA 357 1110 34,140
Camphor benzalkonium methosulfate EU, AZ, SA UVB 284 590 24,500
Cinoxate US, AZ, CA, JN, UVB 308 825 20,650
SA
DEA methoxycinnamate CA, SA UVB 290 880 24,930
Diethylamino hydroxybenzoyl hexyl EU, AZ, JN, SA UVA 354 925 35,900
benzoate
Diethylhexyl butamido triazone EU, SA UVB 311 1460 111,700
(continued)
147
Table 9.2 (continued)
148
Menthyl anthranilate US, AZ, CA, JN, UVA 336 190 5230
SA
4-Methylbenzylidene camphor EU, AZ, CA, SA UVB 300 930 23,655
Methylene bis-benzotriazolyl EU, AZ, JN, SA UVA/B 305 400 26,600 360 495 33,000
tetramethylbutylphenol
Octocrylene XXX ALL UVB 303 340 12,290
PABA XXX ALL UVB 283 640 15,300
PEG-25 PABA EU, AZ, JN UVB 309 180
Pentyl dimethyl PABA JN UVB 310 310
Phenyl benzimidazole sulfonic ALL UVB 302 920 26,060
acid XXX
The Chemistry of Ultraviolet Filters
OR OR
+
R2N C R2N C
O O
OR OR
C C
O O
H + H
R O R O
filters in the world today. Numerous similar examples to illustrate the forces at play
in UV molecules are available. For instance, compare the parabens (para-disubsti-
tuted) to the salicylate (ortho-disubstituted molecules) in Fig 9.5.
Again, as predicted, the parabens would have a low UV absorbance of about
260 nm (that would not be considered a UVB filter) but with a considerable extinc-
tion coefficient, whereas the salicylates (homosalate or octisalate) have a higher UV
absorbance of 306 nm (UVB filter) but with a lower extinction coefficient of 180
due to its ortho-through-space hydrogen bonding as shown in Fig. 9.6.
For a detailed review of the mechanism of all the other approved UV filters (cin-
namates, benzopheones, dibenzoylmethanes, camphor, and triazone derivatives),
consult other references [2].
A series of molecules have recently been designed in Europe with high molecular
weights (over 500 Da) to diminish their penetration into the skin. These molecules
possess multiple chromophores that yield high extinction coefficients and also
broad-spectrum protection [5]. They are, unfortunately, not yet approved in the
USA. They are listed in Table 9.3.
In the USA, there are eight applications pending under the process termed TEA
(Time and Extent Application) that, when approved, will undoubtedly enhance the
UV protection of American consumers from the cancer-causing rays [6]. Two of the
eight TEA ingredients, bemotrizinol and bisoctrizole, when approved for use in the
USA, can be used to impart more photostable sunscreen formulations. See Table 9.4
below illustrating the properties of these two UVA ingredients.
152
Table 9.3 EU-approved UV filters for sunscreens designed with Daltons of 500 or higher
/dm3 Mol. weight
Filter type UV filter INCI name COLIPA# Trademark (supplier) mol1 cm1 max (nm) (Dalton)
UVB Benzylidene malonate S74 BMP Parsol SLX (DSM) 108,000 314 ~6000
polysiloxane
Dioctyl butamido triazone S78 DBT Uvasorb HEB (3V Sigma) 111,170 312 766
Ethylhexyl triazone S69 EHT Uvinul T150 (BASF) 119,500 312 823
UVA Disodium phenyl dibenzimidazole S80 DPDT Neo Heliopan AP (Symrise) 52,400 334 675
tetrasulfonate
Terephthalylidene dicamphor S71 TDSA Mexoryl SX (LOreal) 47,100 345 607
sulfonic acid
UVA/UVB Bis-ethylhexyloxyphenol S81 BEMT Tinosorb S (BASF/Ashland) 42,800/47,500 310/343 629
methoxyphenyltriazine
Drometrizole trisiloxane S73 DTS Mexoryl XL (LOreal) 15,900/15,500 303/341 501
Methylene bis-benzotriazolyl S79 MBBT Tinosorb M (BASF) 32,000/38,000 305/360 659
tetramethylbutylphenol
N.A. Shaath
9
N CH2 N
N N
OH N N OH
O O
The Chemistry of Ultraviolet Filters
These ingredients are chemicals that reflect, scatter, and absorb the UV radiation.
They include titanium dioxide and zinc oxide. They are available in micronized and
nanosized forms that enhance sun protection without imparting the traditional
opaqueness that was aesthetically unappealing in cosmetic formulations. These
metal oxides are reactive and insoluble in cosmetic formulations without chemical
treatment. This treatment includes coating of the metal core and dispersion and
suspension of the particles with oils, solubilizers, and emollients [8, 9]. Many users
falsely believe that natural claims are admissible if only inorganic particulates are
used in sunscreen products. Unfortunately, most of these chemical treatments ren-
der the inorganic particulates synthetic and unnatural.
There has been a shift to zinc oxide from titanium dioxide recently, mostly due
to its broad-spectrum and higher UVA protection. It is also popular since it has a
lower refractive index of 1.92.0 compared to titanium dioxides 2.52.7, which
leads to superior transparency. Recently, ZnO was also approved in Europe. In the
USA, combinations of ZnO and TiO2 with avobenzone are still not allowed.
Titanium is the ninth most common element on the Earths crust. In nature, it
exists only in combinations with other elements such as iron and oxygen. Three
titanium ores are of commercial importance: ilmenite, rutile, and anatase. Ilmenite
is a composite of oxides of iron and titanium. Rutile and anatase are also never pure
and contain various amounts of metal including those that may pose health hazards
to humans. Therefore, commercial TiO2 is always synthetic [8]. Rutile and anatase
have different crystalline structure and different physical and chemical properties.
Of the three forms of TiO2, rutile is the most thermally stable.
Zinc ranks 24th in abundance on the Earths crust but never occurs free in nature.
It is widespread around the world with important deposits located in North America
and Australia. ZnO is produced by oxidizing vapors of Zn in burners. Pure ZnO is
typically a white or yellow-white powder.
The optical behavior of ZnO and TiO2 consists mainly of scattering or absorbing the
light. The scattering from molecules and very tiny particles is predominantly Rayleigh
scattering. When the particle size is at the same magnitude as the wavelength, Mie scat-
tering predominates. The absorption, on the other hand, is a function of the number of
atoms that interact with the light in its pathway. Light with a wavelength below 420 mm
has enough energy to excite electrons in the valence band and can be absorbed by the
inorganic particulate (see Fig. 9.3). Since the bandgap wavelength of ZnO is longer
than that of TiO2, ZnO absorbs a broader-spectrum range of UV light than TiO2. TiO2
is not considered an efficient UVA absorber; rather, it is an efficient UVB absorber. The
attenuation of UVA by TiO2, therefore, mainly takes place via scattering.
When using inorganic particulates, the following parameters need to be carefully
evaluated:
(i) The type of metal
(ii) The particle size
(iii) The coating
9 The Chemistry of Ultraviolet Filters 155
H
O O O O
O O
Enol Keto
O O O O
C
+ CH2
OCH3 OCH3
9.9 Conclusions
Protection from the burning (erythemal) UVB rays is a basic requirement. Protection
from the UVA rays is paramount and so is protection from the damaging heat rays
and the longer wavelength radiation of the infrared [20, 21]. In my opinion, our
ingredients are woefully inadequate, especially the currently US-approved filters.
We can no longer ignore the facts: sunscreen ingredients in cosmetics are not
adequately preventing cancer incidence in the USA. We have lulled ourselves into a
false sense of security. A cream or a lotion alone cannot, at this date, guard you
entirely from the effects of the powerful sun. Heed all practical advice: wear
9 The Chemistry of Ultraviolet Filters 157
protective clothing, seek shade, avoid noon sun exposure, and do use adequate and
properly applied sunscreens. Until advanced ingredients are developed and
approved, use all available measures to mitigate the effects of the total spectrum of
the solar radiation.
References
Key Points
Sun filters can be classified as organic, organic particulates, polymeric, and
inorganic particulates.
The mechanism of action of all types of sun filters is primarily UV
absorption.
A global overview of sun filter approval levels, chemical structures, and
absorbance properties is included in this chapter.
Formulators must select the right combination of filters to deliver
photostable, broad-spectrum protection, with high SPF, and optimal
aesthetics to drive consumer compliance.
Regulatory approvals, the breadth and height of a sun filters UV
absorbance, and the sun filter solubility or dispersibility are key parameters
that formulators should consider during sunscreen design.
10.1 Introduction
Human skin is exposed daily to sunlight, which contains a significant amount of ultra-
violet (UV) radiation. It is well known that UV radiation can be harmful and that UV
exposure can play a significant role in development of skin damage [23, 27]. Various
compounds have been used to protect skin from the harmful rays of the sun over the
centuries. It is only over the last 100 years, however, that synthetic UV filters have been
developed to protect individuals from sunburn and UV-induced skin cancer [35].
For practical and historical purposes, the UV spectrum has been divided into UVA1
(340400 nm), UVA2 (320340 nm), UVB (290320 nm), and UVC (100290 nm).
UVC and some of the shorter UVB wavelengths emitted from the sun are filtered out
by the ozone before they reach the Earths surface. Both UVA and UVB rays can dam-
age DNA, lipids, and proteins; produce inflammation; and ultimately result in burns,
premature aging, and carcinogenesis [27, 30, 35]. An ideal sunscreen must protect the
user from UV radiation across the light wavelength spectrum associated with harmful
effects [24, 27].
Sunscreens protect skin from these harmful rays by forming a protective barrier on skin
surface. Most sunscreen active ingredients are organic molecules with conjugated, aro-
matic chemical structures. The mode of action of these sunscreen active ingredients is
primarily UV absorption [24]. By residing on skin surface as a film, these organic
molecules effectively transform the harmful UV energy to harmless forms of energy
and prevent the UV photons from entering into the skin [25, 30]. The electrons in these
chemical structures are active because they are capable of energy transfer when hit
by UV. Quantum mechanical calculations show that the energy of radiation quanta
present in UVB and UVA lies in the same order of magnitude as the resonance energy
of electron delocalization in aromatic compounds [35].
The electrons of sunscreen UV filters can accept the energy from UV photons and
move to higher electronic energy states. This energy can then be quickly converted
to heat by non-radiation energy dissipation or to other forms of light such as fluores-
cence, phosphorescence, or infrared rays [25]. The electrons will return back to
the ground state during the energy transfer, ready to receive the next UV photon. The
lifetime of excited states of these molecules is very short; therefore, as long as the
chemical structure of the sunscreen is stable at excited states, the process of excita-
tion and returning to ground states can occur continuously and repetitively without
any loss of efficacy.
A few sunscreen active ingredients are not photostable. The chemical structures of
these non-photostable molecules can change while the chemical is in the excited state
(photochemical reactions). When that happens, the original molecules are broken down
and not capable of repeating the excitation process and more importantly cannot absorb
the next UV photons. With the degradation of the original active ingredients, free radi-
cals (including singlet oxygen) may be generated that may then react with nearby mol-
ecules to form photobyproducts. Thus, the efficacy of the sunscreen decreases because
less active ingredients remain to absorb more incoming photons.
Sun filters do not need to penetrate into the skin in order to be effective. As
soon as the sunscreen film is present on skin surface, there will be at least some
level of protection because of its inherent absorption properties. The final protec-
tion level may be enhanced as the product dries on the skin and the film structure
is optimized [32].
10 Chemistry of Sunscreens 161
There are a number of different sun filters approved for the use in sunscreen products
around the globe. Currently, 16 sun filters are approved for sunscreen products in the
United States (Food and Drug Administration and Department of Health and Human
Services [14, 15, 39]), 20 in Canada [18], 28 in the European Union [12, 22], 28 in the
Association of Southeast Asian Nations [37], and 33 approved by MERCOSUR
(Southern Common Market, consisting of Argentina, Brazil, Paraguay, Uruguay, and
Venezuela) [37]. The complete listing of approved sun filters in these locations, along
with the approved concentrations, is shown in Table 10.1.
Sun filter actives can be classified into the following categories: organic (traditional
molecules or polymeric) or particulate (organic particulates or inorganic particulate), as
described in subsequent sections 3.1 and 3.2, respectively.
Organic filters are often referred to as chemical filters, but this can be misleading
because it suggests that it is possible to have a sun filter that is nonchemical. Strictly
speaking, all active sun filter compounds, both organic and inorganic, are made up of
chemical molecules originating from the periodic table, and all function primarily by
absorbing light [26].
Traditional organic sun filters are aromatic, small molecules, with molecular weight
values <900 g/mol. Today, the most widely used organic filters include avobenzone,
oxybenzone, octocrylene, salicylate derivatives (homosalate and ethylhexyl salicylate),
cinnamate derivatives (octyl-methoxycinnamate [OMC]), triazone derivatives (Uvinul
T150 [ethylhexyl triazone]; UVASorb HEB [diethylhexyl butamido triazone]; Tinosorb
S [bis-ethylhexyloxyphenol methoxyphenyl triazine]), benzoate derivatives (Uvinul A
Plus [diethylamino hydroxybenzoyl hexyl benzoate]), benzotriazole derivatives
(Mexoryl XL [drometrizole trisiloxane]), and camphor derivatives (Mexoryl SX
[ecamsule]; terephthalylidene dicamphor sulfonic acid). Anthranilate derivatives (like
meradimate) are less commonly used filters because of low efficacy.
Avobenzone (a dibenzoylmethane derivative) is one of the most efficient UVA-
absorbing filters used around the globe, and it is the only UVA-absorbing organic sun
filter approved in the USA. However, avobenzone is prone to photo instability because
of an enol-to-keto tautomerization as shown in Fig. 10.1 [25]. The enol form of avoben-
zone absorbs in the UVA (315400 nm), while the diketo form absorbs in the UVC
(200280 nm) and is prone to degradation [25]. Other photostabilizing ingredients
must be used in combination with avobenzone to prevent light-induced degradation [7].
In order to achieve photostability of avobenzone, it must be combined with ingredients
Table 10.1 List of sun filters approved in the USA, Canada, European Union, ASEAN, and MERCOSUR; alternate names; and approved usage levels per
162
region
Filter name Other names Coverage US Canada EU MERCOSUR Australia ASEAN
Maximum allowed concentration (%)
Benzophenone-3 Oxybenzone or UVA/B 6 6 10 10 10 10
2-hydroxy-4-methoxybenzophenone
Benzophenone-4 Sulisobenzone or 2-hydroxy-4- UVA/B 10 10 5** 10 10 5**
methoxybenzophenone-5-sulfonic acid and
its trihydrate
Benzophenone-5 2-Hydroxy-4-methoxybenzophenone-5-- UVA/B * 5 10 *
sulfonic acid (benzophenone-5) and its
sodium salt
Sulisobenzone sodium
Sodium hydroxymethoxybenzophenone
sulfonate
Benzophenone-8 Dioxybenzone or UVA/B 3 3 3 3
2,2-dihydroxy-4-methoxybenzophenone
Dioxybenzone
(2-hydroxy-4-methoxyphenyl)
(2-hydroxyphenyl)methanone
Methanone (2-hydroxy-4-methoxyphenyl)
(2-hydroxyphenyl)
3-Benzylidene camphor 3-Benzylidene camphor UVB 2 2 2
Bis-ethylhexyloxyphenol Tinosorb S or (1,3,5)-triazine-2,4-bis{[4-(2- UVA/B 10 10 10 10
methoxyphenyl triazine ethyl-hexyloxy)-2-hydroxy]-phenyl}-6-(4-
methoxyphenyl) or anisotriazine
Butyl methoxydibenzoyl Avobenzone or 1-(4-tert-butylphenyl)-3-(4- UVA 3 3 5 5 5 5
methane methoxyphenyl) propane-1,3-dione
S. Daly et al.
10
O O O OH
hv
O O
Diketo tautomer Enol tautomer
Fig. 10.1 The keto-to-enol tautomerization of avobenzone (Scheme 2 was reproduced with
permission from Kockler et al. [25])
that are efficient in both triplet quenching and singlet quenching. Examples of triplet
quenchers are the following UV filters: octocrylene, 4-methylbenzylidene camphor
(ex-US), Tinosorb S (ex-US), or emollients such as diethylhexyl-2,6-naphthalate [7].
In addition, higher levels of oxybenzone are known to stabilize avobenzone by the
singlet quenching mechanism [7]. A combination of singlet and triplet quenchers is
most efficient in stabilizing avobenzone.
Cinnamates are very efficient UVB absorbers but also have issues with photosta-
bility. OMC is a member of the cinnamate class that is known to react with avoben-
zone to produce non-UV light-absorbing photoproducts. Hence, combinations of
avobenzone and OMC are unfavorable and should be avoided because of enhanced
photo instability [7, 33].
Salicylate derivatives are photostable, UVB-absorbing filters that have a long
history of usage. They are excellent solubilizers for crystalline UV filters, including
oxybenzone and avobenzone, however the absorption efficiency of these filters is
quite low.
Oxybenzone (a benzophenone derivative) is used in many US sunscreen formula-
tions with absorbance in the UVB (290320 nm) and the UVA2 region (320340 nm).
Padimate O is a derivative of para-aminobenzoic acid that is a liquid and is oil soluble.
It is a very effective UVB filter with one of the highest molar extinction coefficients of
the approved filters. It is not widely used in products over concern that the parent mol-
ecule, para-aminobenzoic acid, has been associated with allergic reactions. Octocrylene
is another oil-soluble UVB filter that has been widely used to provide increased sun
protection factor (SPF) values and to also boost the photostability of avobenzone when
used in combination. Ensulizole (phenylbenzimidazole sulfonic acid) is a water-soluble
filter and is used in products formulated to feel lighter and less oily, such as daily use
cosmetic moisturizers. Currently, it is not permitted to be combined with avobenzone
in the USA and must be used in combination with on other UVA absorbers (such as
zinc oxide) to provide broad-spectrum protection.
Table 10.2 Relative lipophilicity of sunscreen chemicals based upon their calculated partition
coefficients between octanol and water
CTFA name Other names Log P at 25 C
Glyceryl PABA 1,2,3-Propanetriol,1-(4-aminobenzoate) 0.02
Benzophenone-4 Sulisobenzone 1.51
PABA p-Aminobenzoic acid 0.74
Benzophenone-8 Dioxybenzone 2.15
Cinoxate Ethoxyethyl methoxy cinnamate 2.55
Benzophenone-3 Oxybenzone 2.63
Ethyl dihydroxypropyl PABA Ethyl-4-bis(2-hydroxypropyl-aminobenzoate) 2.84
Amyl dimethyl PABA Amyl dimethyl PABA 4.53
Butylmethoxy dibenzoylmethane Butylmethoxy dibenzoylmethane 4.86
Menthyl anthranilate Methyl-O-aminobenzoate 5.05
Octyl salicylate 2-Ethylhexyl salicylate 5.30
Homosalate Homomenthyl salicylate 5.61
Octyl methoxy cinnamate Ethylhexyl-p-methoxycinnamate 5.65
Octocrylene Octyl cyanodiphenylacrylate 5.69
Octyl dimethyl PABA 2-Ethylhexyl-p-dimethyl aminobenzoate 6.08
Modified with permission from Agrapidis-Paloympis et al. [1]
CTFA Cosmetic, Toiletry, and Fragrance Association; PABA para-aminobenzoic acid
mild applications. The polysiloxane backbone not only links the chromophores
together, but it also provides a pleasant aesthetic to skin or hair [29]. Unfortunately,
this polymeric filter only absorbs in the UVB (max = 312 nm) part of the spectrum
and needs to be combined with UVA filters to achieve broad-spectrum protection.
While most organic filters must be dissolved into either the oil or water phases of a
formulation to be effective, particulate sunscreens are not dissolved in either phase, and
they exist in particle suspensions. Particulate filters are commonly used in mild and
baby sunscreen products, and they have been demonstrated in several studies to stay on
the surface of the skin [8, 16]. There are two types of particulate sunscreen filters:
organic and inorganic.
The inorganic particulate sunscreen class includes titanium dioxide (TiO2) and zinc
oxide (ZnO). It is important to point out that these particulate sunscreen active ingredi-
ents also absorb UV, with very little reflection and scattering in the UV portion of the
spectrum [4], so it is not appropriate to call them physical sunscreens. While the UV
absorption action of Tinosorb M is not very different from other organic molecules, for
TiO2 and ZnO, the electrons in the crystals can freely move from the valence band to
the conductance band when exposed to UV. This is because the energy band gap in
TiO2 or ZnO is lower than the energy conveyed by UV photons, allowing UV to excite
the free electrons in these semiconductor-like materials.
Particulate inorganic sunscreen active ingredients also protect skin from harmful
UV by absorbing, reflecting, and scattering; however, recent findings indicate that the
primary means of protection is by absorption (roughly 95 %) and the remaining 5 % by
scattering and reflecting. Incident light that is absorbed or backscattered by the particle
sunscreens does not enter into the skin. Scattering of reflected photons increases the
actual optical length of the UV photons as they pass through the absorbing sunscreen
10 Chemistry of Sunscreens 169
layer. The scattering by sunscreen particles depends on factors that include the volume
concentration of the particles, the relative refractive index of the particle to the medium
and/or coating, the particle size, and the scattering wavelength [11].
For the UV wavelength range, the absorption and scattering power of single TiO2 or
ZnO particles generally increases with the size of the particle, up to about 100 m. We
generally recognize, however, that absorption power increases monotonically when the
particle size is smaller. This is because the number of particles has to increase with
smaller and smaller particle size when evaluated for a fixed volume fraction (weight
percentage). Therefore, the overall absorption power for the system becomes greater
with smaller particle sizes. Based on both theoretical calculation and experimental
measurement, the light scattering of particulate sunscreen ingredients (TiO2, ZnO, and
Tinosorb M) does not contribute significantly to the attenuation of UV (290370 nm)
when compared absorption. For long UVA and visible light wavelength range (370
760 nm), however, reflection contributes much more to the protective effects of TiO2
and ZnO particles when applied on skin surface because of very limited absorption of
these ingredients within the visible wavelength range. Since absorption and scattering
of UV light depend on both the volume fraction of particles in the medium and also the
uniformity of the particles, dispersion of particles in sunscreen formulation plays a
critical role in the efficacy of UV attenuation. It is also critical to make sure the inor-
ganic particles are photostable and do not lead to generation of free radicals. Effective
surface treatment of inorganic particles ensures photostability of these inorganic sun-
screens. Examples of surface treatments include alkoxy silane, dimethicone, methi-
cone, polyhydroxystearic acid and aluminum stearate, silica, alumina, etc. Photostability
also depends on the type of the inorganic crystal. For example, antase is known to be
less stable than rutile grade TiO2.
ZnO has gained popularity as a mild, safe, and effective sun filter in the past 10 years.
It is the only other effective UVA1 filter besides avobenzone that is approved in the
USA. TiO2 has high UVB efficacy, but does not provide significant UVA protection.
On the other hand, ZnO provides very uniform UVB and UVA protection across the
whole spectrum, providing a flat spectral absorption curve [36]. Figure 10.2a shows a
comparison between absorbance of TiO2 and ZnO. It is desirable to maximize light
attenuation while limiting the scattering in the visible region, as consumers do not like
to see a white/blue haze on their skin. Formulators need to balance the particle size,
dispersion, solvent, and volume fraction to achieve an aesthetically acceptable and
effective inorganic sunscreen product.
A key performance metric for sun filters is absorbance intensity and breadth of cover-
age. Dilute solution UV spectroscopy is used to determine filter efficacy and is com-
monly reported as a specific extinction, E(1 %, 1 cm), value. E(1,1) corresponds to the
absorbance at the peak wavelength (max) for a 1 % solution in a cuvette with a 1 cm
170 S. Daly et al.
a 0.7
0.6
0.5
Absorbance
0.4
0.3 1% TiO2
1% ZnO
0.2
0.1
0.0
290 310 330 350 370 390
Wavelength, nm
b 0.7
0.6
0.5 Avobenzone
Absorbance
0.4 Tinosorb M
Fig. 10.2 The absorbance spectra for various sunscreen agents at 1 %; (a) TiO2 and ZnO, and (b)
key global UVA-absorbing filters
path length [35]. Table 10.3 shows the wavelength of absorbance maximum and spe-
cific extinction value for common organic filters, along with the molecular structures
and molecular weight values [35].
Avobenzone is the most efficient UVA-absorbing filter with an E(1,1) value of
1,110 (357 nm), followed by Uvinul A plus (E[1,1] is 925 [354 nm]), Mexoryl SX
(E[1,1] is 750 [345 nm]), and Tinosorb S (E[1,1] is 750 and 820 [310 and 343 nm,
respectively]). Figure 10.2b shows the absorbance spectral overlay for key UVA
filters (each at 1 %).
Although UVA protection is getting quite a bit of attention in recent years, UVB
protection is critical to appropriate protection from the sun, as the action spectra for
erythema, basal cell carcinoma, and squamous cell carcinoma are all known to be
driven by UVB [6, 9]. Uvinul T150 (ethylhexyl triazone) and Uvinul HEB (diethyl-
hexyl butamido triazone) are the two most efficient UVB filters with E(1,1) values of
1550 (at 314 nm) and 1460 (at 311 nm), respectively. Ethylhexyl diaminobenzoate,
phenylbenzimidazole sulfonic acid, and several cinnamate derivatives are also very
strong UVB absorbers. Benzophenone derivatives are modest UVB absorbers, and
salicylate derivatives are typically relatively weak UVB absorbers.
10
Table 10.3 List of sun filters, chemical structures, molecular weight, lambda max values, and specific extinction values E(1 %, 1 cm)
Molecular MAX MAX E1 E2
Filter name Chemical structure weight (g/mol) Coverage 1 2 (1 %, 1 cm) (1 %, 1 cm)
OH O
Benzophenone-3 228 UVA/B 286 324 630 400
O
O OH
Benzophenone-4 308 UVA/B 286 324 440 360
O
O S O
OH
OH O
Chemistry of Sunscreens
O
O
Bis-ethylhexyloxyphenol methoxyphenyl HO
628 UVA/B 310 343 745 820
N N
triazine N
H3CO HO O
H
O O O O
Butyl methoxydibenzoylmethane O O
310 UVA 357 1,110
CH3 CH3
CH3SO4
Camphor benzalkonium methosulfate N+ 410 UVB 284 590
O O
OH O
Diethylamino hydroxybenzoyl hexyl 398 UVA 354 925
N
benzoate
O
N
Diethylhexyl butamido triazone H 766 UVB 311 1,460
O HN O
N N O
O
N N
N H
H
ONa+
O
S O O
H
171
(continued)
Table 10.3 (continued)
172
O
Ethoxyethyl methoxycinnamate CH3O
250 UVB
O
CH3
N
H3C CH3
O
Ethylhexyl methoxycinnamate O
290 UVB 311 850
O
Ethylhexyl triazone O
823 UVB 314 1,550
O
O HN
O N N
N N NH
H
O O
O
Homosalate 262 UVB 306 180
O
HO
O O
Isoamyl p-methoxycinnamate 248 UVB 308 980
O
NH2
S. Daly et al.
Methylene bis-benzotriazolyl N N
659 UVA/B 305 360 400 495
N N
tetramethylbutylphenol
N
Octocrylene O
362 UVB 303 340
O
COOH
Para-aminobenzoic acid 137 UVB 283 640
NH2
Chemistry of Sunscreens
O
PEG-25 para-aminobenzoic acid O H2N
1,265 UVB 309 180
O
N
Phenylbenzimidazole sulfonic acid O 274 UVB 302 920
S N
HO H
O
n
Polyacrylamido methylbenzylidene camphor [323.44]n UVB 297 610
approx. 6% O O O
R=
O
O
approx. 1.5%
O O O
n 60
COOH-N(CH2CH2OH)3
Triethanolamine salicylate 287 UVB 298 120
OH
O O
Terephthalylidene dicamphor sulfonic acid O S
607 UVA 345 750
O OH
O S
O
HO
SO3H
Benzylidene camphor sulfonic acid 320 294 860
O
173
There is no single sun filter available today that on its own can provide high-SPF and
broad-spectrum protection without aesthetic drawbacks. With the current state of UV
filter technology, sunscreen products today require the right combination of filters in
the formulation to obtain both high efficacy in UV protection and optimal aesthetics to
enhance compliance. Formulations containing oil-soluble filters may feel occlusive
and or greasy [30]. Combinations of different filters may be used to improve the sen-
sory profile, as well as provide broad-spectrum protection. In the USA, broad spec-
trum can be claimed if the in vitro determined critical wavelength value is 370 nm
[15]. In Europe, products must achieve a 1:3 ratio of PFA (protection factor UVA):SPF
[21]. Although many sunscreen products in the market claim broad spectrum, it is hard
to differentiate between their UVA efficacies. Not all broad-spectrum sunscreens are
created equal because they may have different degrees of UVA protection (amplitude
of absorbance curve in UVA) with different filter combinations [5].
provide strong UVB protection, oxybenzone provides broad-spectrum UVB and UVA2
protection, and avobenzone provides the longer-wavelength UVA1 protection. In addi-
tion, both octocrylene and oxybenzone enhance the photostability of avobenzone by
singlet and triplet quenching.
The inorganic filters TiO2 and ZnO are often used together. ZnO is typically used
to achieve breadth of protection, while TiO2 brings higher SPF. The combination of
avobenzone and ZnO is currently not permitted in the USA [14]. The agency did not
approve the combination of ZnO with avobenzone in the latest monograph
publications.
In Europe and Latin America, many more filters are approved for combination use,
such as Tinosorb S, Tinosorb M, Uvinul T150, Uvinul A Plus, Mexoryl SX, or
Mexoryl XL. In Europe, it is common to omit oxybenzone. In Latin America, many
formulations include a combination of traditional organic filters and a small amount
of TiO2. In Japan, very light and fluid textures are preferred, and mildness is very
important; TiO2, ZnO, OMC, and Tinosorb S are widely used ingredients.
Beyond the filter combinations selected for a sunscreen product formulation, formula-
tion excipients, emulsion structure, and the sunscreen film structure are also important
for determining the final sunscreen performance. The presence of film formers or
emollients in the formulation [31, 34], the sunscreen rheological properties [2, 17], and
the structures of the dried down sunscreen film [13, 38] have all been linked to sun-
screen performance. Figure 10.3 illustrates how surface roughness plays a role in creat-
ing holes in a sunscreen film, and that the thickness of the sunscreen film above the skin
peaks may be quite small [32]. It can be envisioned that the physical properties of the
sunscreen film may act to increase the film thickness above the peaks and reduce set-
tling into the valleys to create a more ideal film structure as in Fig. 10.3a [32].
10.6 Conclusion
A variety of organic sun filters are available for use with different properties, and it is
important for formulators to understand their chemistry to maximize efficacy and cre-
ate sunscreen products with an acceptable level of SPF and broad-spectrum protection.
With the current state of sunscreen technology, it is necessary for formulators to select
176 S. Daly et al.
a CREAM
SUBSTRATE
b m
5
0 CREAM
-5
-10
-15
-20
-25 SUBSTRATE
-30
46.9 47.0 47.1 47.2 47.3 47.4 47.5 47.6 47.7 47.8 47.9 48.0 48.1 48.2 48.3 48.4 48.5 48.6 48.7mm
Fig. 10.3 Sunscreen distribution on a surface (a) ideal distribution (b) real distribution
(Reproduced with permission from Osterwalder et al. [32])
Acknowledgment The authors would like to thank Alex Loeb, PhD, CMPP, of Evidence Scientific
Solutions (Philadelphia, PA) for editorial support that was funded by JOHNSON & JOHNSON
Consumer Companies, Inc. (Skillman, NJ).
References
1. Agrapidis-Paloympis L, Nash RA, Shaath NA (1987) The effect of solvents on the ultraviolet
absorbance of sunscreens. J Soc Cosmet Chem 38:209221
2. Anderson MW, Hewitt JP, Spruce SR (1997) Broad-spectrum physical sunscreens: titanium diox-
ide and zinc oxide. In: Lowe NJ, Shaath NA, Pathak MA (eds) Sunscreens: development, evalua-
tion and regulatory aspects, vol 15, Cosmetic Science and Technology, vol 18. Marcel Dekker,
Inc., New York, pp 353398
3. Castanedo-Cazares JP, Hernandez-Blanco D, Carlos-Ortega B, Fuentes-Ahumada C, Torres-
lvarez B (2014) Near-visible light and UV photoprotection in the treatment of melasma: a dou-
ble-blind randomized trial. Photodermatol Photoimmunol Photomed 30(1):3542
4. Cole C, Shyr T, Ou-Yang H (2016) Metal oxide sunscreens protect skin by absorption not by
reflection or scattering. Photodermatol Photoimmunol Photomed 32(1): 510
10 Chemistry of Sunscreens 177
5. Cole CA (2014) Critical concepts about sunscreens. Pract Derm. (Suppl 2):14. http://practicalde-
rmatology.com/pdfs/0414_insert2.pdf
6. Cole CA, Forbes PD, Davies RE (1986) An action spectrum for UV photocarcinogenesis.
Photochem Photobiol 43(3):275284
7. Cole CA, Vollhardt J, Mendrok C (2009) Formulation and stability of sunscreen products. In: Lim
HW, Draelos ZD (eds) Clinical guide to sunscreens and photoprotection. Informa Healthcare,
New York, pp 3951
8. Cross SE, Innes B, Roberts MS, Tsuzuki T, Robertson TA, McCormick P (2007) Human skin
penetration of sunscreen nanoparticles: in-vitro assessment of a novel micronized zinc oxide for-
mulation. Skin Pharmacol Physiol 20(3):148154
9. de Gruijl FR, van der Leun JC (1991) Development of skin tumors in hairless mice after discon-
tinuation of ultraviolet irradiation. Cancer Res 51(3):979984
10. DSM nutritional products AG (2013) PARSOL SLX factsheet. http://www.respharma.com/
Brochure/DSM/PARSOL%20SLX_Factsheet.pdf. Accessed 30 Oct, 2014
11. Egerton TA, Tooley IR (2012) UV absorption and scattering properties of inorganic-based sun-
screens. Int J Cosmet Sci 34(2):117122
12. European Union (2009) Regulation (EC) No 1223/2009 of the European Parliament and of the
Council of 30 Nov 2009 on cosmetic products. Off J Eur Union L 342:59209
13. Ferrero L, Pissavini M, Marguerie S, Zastrow L (2003) Efficiency of a continuous height distribu-
tion model of sunscreen film geometry to predict a realistic sun protection factor. J Cosmet Sci
54(5):463481
14. Food and Drug Administration, Department of Health and Human Services (1999) Sunscreen
drug products for over-the-counter human use; final monograph. Fed Regist 64(98):
2766627693
15. Food and Drug Administration, HHS (2012) Over-the-counter sunscreen drug products; required
labeling based on effectiveness testing. Fed Regist 76:95107
16. Gamer AO, Leibold E, van Ravenzwaay B (2006) The in vitro absorption of microfine zinc oxide
and titanium dioxide through porcine skin. Toxicol In Vitro 20(3):301307
17. Gaspar LR, Maia Campos PM (2003) Rheological behavior and the SPF of sunscreens. Int
J Pharm 250(1):3544
18. Health Canada (2012) Draft: guidance document Sunscreen monograph. http://www.hc-sc.
gc.ca/dhp-mps/alt_formats/pdf/consultation/natur/sunscreen-ecransolaire-eng.pdf. Accessed 31
Oct 2014
19. Herzog B, Quass K, Schmidt E, Mller S, Luther H (2004) Physical properties of organic particu-
late UV absorbers used in sunscreens II. UV-attenuating efficiency as function of particle size.
J Colloid Interface Sci 276(2):354363
20. Household and Personal Products Industry (2010) DSM presents latest data on parsol SLX. http://
www.happi.com/contents/view_breaking-news/2010-11-01/dsm-presents-latest-data-on-parsol-
slx/. Accessed 30 Oct 2014
21. International Organization for Standardization (2012) ISO 24443:2012 determination of sun-
screen UVA photoprotection in vitro. http://www.iso.org/iso/catalogue_detail?csnumber=46522.
Accessed 11 Nov 2014
22. Janousek A (1983) The current status of sunscreens in the EEC. Cosmet Toiletries 98(3):7980
23. Jansen R, Osterwalder U, Wang SQ, Burnett M, Lim HW (2013) Photoprotection: part
II. Sunscreen: development, efficacy, and controversies. J Am Acad Dermatol 69(6):867.
e861814
24. Jansen R, Wang SQ, Burnett M, Osterwalder U, Lim HW (2013) Photoprotection: part
I. Photoprotection by naturally occurring, physical, and systemic agents. J Am Acad Dermatol
69(6):853.e851812
25. Kockler J, Oelgemller M, Robertson S, Glass BD (2012) Photostability of sunscreens.
J Photochem Photobiol C Photochem Rev 13(1):91110
26. Kollias N (1999) The absorption properties of physical sunscreens. Arch Dermatol 135(2):
209210
178 S. Daly et al.
27. Krutmann J, Morita A, Chung JH (2012) Sun exposure: what molecular photodermatology tells us
about its good and bad sides. J Invest Dermatol 132(3 Pt 2):976984
28. Mahmoud BH, Ruvolo E, Hexsel CL, Liu Y, Owen MR, Kollias N, Lim HW, Hamzavi IH (2010)
Impact of long-wavelength UVA and visible light on melanocompetent skin. J Invest Dermatol
130(8):20922097
29. Maillan P, Gripp A, Sit F, Jermann R, Westenfelder H (2005) Protecting against UV: induced
degradation and enhancing shine. Cosmet Toiletries 120(3):6571
30. Mancebo SE, Hu JY, Wang SQ (2014) Sunscreens: a review of health benefits, regulations, and
controversies. Dermatol Clin 32(3):427438
31. OLenick TO, Lott D (2011) The effects of solvents on sunscreens: a new ester to improve effi-
ciency. Cosmet Toiletries 126(6):431436
32. Osterwalder U, Sohn M, Herzog B (2014) Global state of sunscreens. Photodermatol
Photoimmunol Photomed 30(23):6280
33. Ou-Yang H, Stanfield JW, Cole C, Appa Y (2010) An evaluation of ultraviolet A protection and
photo-stability of sunscreens marketed in Australia and New Zealand. Photodermatol
Photoimmunol Photomed 26(6):336337
34. Prettypaul D, Fares H (2012) Microscopic evaluation of polymeric film properties of anhydrous
sunscreen compositions and their relation to absorption and water resistance. J Cosmet Sci
63(3):213221
35. Saath NA (2007) The encyclopedia of ultraviolet filters. Allured Publishing Corp, Carol Stream
36. Smijs TG, Pavel S (2011) Titanium dioxide and zinc oxide nanoparticles in sunscreens: focus on
their safety and effectiveness. Nanotechnol Sci Appl 4:95112
37. Stiefel C, Schwack W (2015) Photoprotection in changing times UV filter efficacy and safety,
sensitization processes and regulatory aspects. Int J Cosmet Sci 37(1):230. http://onlinelibrary.
wiley.com/doi/10.1111/ics.12165/abstract
38. Teichmann A, Pissavini M, Ferrero L, Dehais A, Zastrow L, Richter H, Lademann J (2006)
Investigation of the homogeneity of the distribution of sunscreen formulations on the human skin:
characterization and comparison of two different methods. J Biomed Opt 11(6):064005
39. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug
Evaluation and Research (2011) Guidance for industry enforcement policy OTC sunscreen
drug products marketed without an approved application (Draft guidance). http://www.fda.gov/
downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm259001.pdf.
Accessed 30 Oct 2012
40. Woodruff J (2001) Factors affecting the formulation of sunscreen products. http://www.creative-
developments.co.uk/papers/Factors%20affecting%20sun%20care%20formulations.html.
Accessed 30 Oct 2014
Chapter 11
Global UV Filters: Current Technologies
and Future Innovations
Key Points
Tremendous progress has been made in sunscreen technology over the last
two decades.
Unfortunately, in the USA, UV filter technology is lagging 15 years behind
compared to the rest of the world.
In Europe and the rest of the world, development goes on, but it is slower
than 20 years ago.
The major weakness of sunscreen and photoprotection remains the lack of
compliance by the user.
Innovation in photoprotection education, including behavior modification
and sunscreen use, is required.
11.1 Introduction
Sunscreens are used worldwide, especially by people with fairer skin phototypes
in geographic areas with high sun exposure but also by people with darker skin
to keep a uniform complexion, mainly of the face. This chapter focuses on global
sunscreens, i.e., sunscreens that contain UV filters that are available and in use
worldwide. Sunscreen is part of sun protection strategies consisting of seeking
U. Osterwalder (*)
Personal Care, BASF PCN GmbH, Monheim, Germany
e-mail: [email protected]
L. Hareng
Regulatory Toxicology, BASF SE, Ludwigshafen, Germany
shade, covering up with clothing and hats, and wearing sunglasses. Use of sun-
screens has now been demonstrated to slow down the photoaging process and to
decrease the development of squamous cell carcinoma, basal cell carcinoma, and
melanoma [13].
A recent study confirmed the preventative role of sunscreen for malignant mela-
noma in mice, but the control sites that were covered by cloth had less melanoma,
indicating that protection by sunscreen alone could not completely prevent skin
cancer [4]. This chapter reviews what still can be done to improve sunscreens to
become a yet more effective means of photoprotection.
The basic requirements for UV filters in sunscreens are (1) efficacy, (2) safety, (3)
registration, and (4) freedom to operate with respect to the status of intellectual
property [5]. Efficient UV absorber molecules are the basis of all sunscreens.
Efficacy indicates good UV absorbance in the spectral range between 290 and
400 nm. Good UV filters must also have the property of being able to be incorporated
in sufficient amounts into cosmetic formulations. They may be dissolved in the oil
phase or the water phase of sunscreen formulations, and thus the respective solubility
must be high enough. Alternately, filters can be used as dispersions of fine particles
of the absorbing substances.
100 %
Transmission 60 %
40 %
20 %
0%
290 320 340 370 400
Wavelength (nm)
PABA 8 % EHMC 7.5 % BMBM 3 %
BEMT 5 % MBBT 5 %
Fig. 11.1 Transmission of organic filters (Data obtained from the BASF sunscreen simulator, with
the % applied as single UV filter in a sunscreen). BEMT bis-ethylhexyloxyphenol methoxyphenyl
triazine, BMBM butyl methoxydibenzoylmethane, EHMC ethylhexyl methoxycinnamate, MBBT
methylene bis-benzotriazolyl tetramethylbutylphenol (nano), PABA ethylhexyl dimethyl para-
aminobenzoic acid
Sunscreens, especially those with a high sun protection factor (SPF), contain a consid-
erable amount of UV filters. Therefore, solubility of the active substance can be a
significant problem [5]. For this reason, particulate organic UV filters were developed
that allow high-SPF products to have relatively low concentrations of UV filters.
Examples of these UV filters include bisoctrizole and tris-biphenyl triazine [14, 15];
the former is under consideration for approval through the time and extent application
(TEA) process of the US Food and Drug Administration (FDA). These filters have
extremely low solubility in oil and in water but can thus be micronized in an aqueous
phase [1618]. Particulate bisoctrizole shows a broad absorption up to 380 nm
(Fig. 11.2). The UV absorbance spectrum of particulate bisoctrizole has a characteris-
tic shape [19]. The spectrum of the particles extends toward longer UVA1 wavelengths
with an additional shoulder around 320 and 380 nm caused by intermolecular interac-
tions of the -electrons inside the particles. Similar to small inorganic particle UV
filters, the contribution to protection by scattering or reflectance is 5 % or less [20].
Any inorganic material that absorbs in the UV range could potentially be used in sun-
screens. Figure 11.3 shows transmission curves in the UV and also visible range of a few
inorganic materials: titanium dioxide (TiO2), zinc oxide (ZnO), cerium dioxide (CeO2),
CeO2-doped ZnO, and various iron oxides [2123]. Doping refers to the addition of
182 U. Osterwalder and L. Hareng
500
0.16 m
400
0.22 m
300 0.34 m
E(1.1) 0.78 m
200 4.4 m
100
0
280 300 320 340 360 380 400
Wavelength/nm
Fig. 11.2 Influence of particle size on efficacy of particulate UV filters (bisoctrizole, MBBT)
60 %
40 %
20 %
0%
280 320 340 400 450 500 550 600 650
Wavelength (nm)
Fig. 11.3 Transmission of various inorganic UV filters in UV and VIS range (280650 nm)
small amounts of foreign atoms altering the lattice properties. TiO2, ZnO, and CeO2
show good absorption in the UV range; the relatively low absorption in the visible range
makes these materials colorless, hence qualifying them to be used in sunscreens. Cerium
oxide is not listed in any countrys positive list of sunscreen actives and is also slightly
yellowish colored. The iron oxides are colored materials absorbing in the visible range
in addition to the UV spectrum, which disqualifies them for use in sunscreens; however,
they are used in other forms, such as BB creams (Blemish Balm all-in-one facial cos-
metic product) or makeup and can contribute to photoprotection [24].
Innovation differs from improvement in that innovation refers to the notion of doing
something different rather than doing the same thing better. In the following paragraphs,
three different approaches are outlined assessing future innovations in suncare.
11 Global UV Filters: Current Technologies and Future Innovations 183
Innovations in sunscreens and UV filters over the last century were driven by
changes in society, most importantly that tanned skin became fashionable
(Table 11.1).
The development of UV filters started with the UVB filters salicylates and PABA
[30]. The first UVA filter, avobenzone (BMBM) [30], was patented in 1973 and
approved in Europe in 1978. Ten years later, it was available in the USA through the
New Drug Application (NDA) route; another 10 years later, it was considered
generally recognized as safe and effective (GRAS/E) and added to the FDA
sunscreen monograph. The fact that avobenzone is not photostable triggered the
search for alternatives. These were developed in the 1990s and brought into the
market around 2000 [30]. They are all mentioned in Table 11.1 as UVA and broad-
spectrum UV filters. In parallel to the development of new UV filters, there were
successful attempts to improve the photostability of avobenzone. Indeed it is now
184 U. Osterwalder and L. Hareng
In a foreseeable future, UV filter technology will bring UV coverage closer to ideal, i.e.,
covering the entire spectrum of UVB and UVA. But the use of sunscreen and the prac-
tice of photoprotection are still far from ideal [41]; this topic is covered in Chap. 11.3.
Fritz Zwicky, a Swiss astrophysicist and aerospace scientist based at the California
Institute of Technology (Caltech), called the morphological approach totality
research which in an unbiased way attempts to derive all the solutions of any
11 Global UV Filters: Current Technologies and Future Innovations 185
given problem [42]. Zwicky applied this method to such diverse fields as the clas-
sification of astrophysical objects, the development of jet and rocket propulsion
systems, and the legal aspects of space travel and colonization. He founded the
Society for Morphological Research and advanced the morphological approach
for some 30 years, between the 1940s and his death in 1974.
This approach may also help us discover new relationships or configurations,
which are not so evident or which we might have overlooked by other less system-
atic methods. Importantly, it encourages the identification and investigation of
boundary conditions, i.e., the limits and extremes of different contexts and
factors.
The three steps of a systematic general morphological analysis (GMA) are, first,
setting up the whole morphological box (x parameters with n values each); second,
cross-consistency assessment in order to excluding impossible combinations and
arriving at a manageable number of internally consistent configurations; and third,
choosing single or multiple drivers, i.e., fixing one or more values of certain param-
eters in order to arrive at a handful of combinations.
The generic sunscreen in Fig. 11.4 shows the possible sunscreen variations. The
core are always the UV filters, but not every type of UV filter is suitable for every
kind of sunscreen, e.g., the particulate UV filters, inorganic or organic filters, are not
suited for clear (transparent) formulations because a dispersion is always opaque, or
certain UVA requirements can only be fulfilled with sufficient UVA or broad-
spectrum UV filters.
The influencing parameters determining a sunscreen can be grouped into three
categories: technology, marketing/society, and regulation/standards (Fig. 11.4).
186 U. Osterwalder and L. Hareng
Table 11.2 Morphological box with 3 parameters and 3 values (27 combinations)
Selected parameters
Target Product
UV filter segment regulation
Organic and
3 inorganic Sport Natural product
Use of driver (natural sunscreen) to narrow down the sunscreen product, e.g., from 27 (3 3 3)
to 3 (1 3 1)
Since each parameter can assume many values, theoretically large numbers of
combinations, representing new sunscreens, can be envisaged. To illustrate the
morphological analysis, Table 11.2 shows an example of just one parameter of
each category with three values. This gives already a theoretical total of 3 3 3 = 27
variations of sunscreens. This simplified example illustrates how these countless
combinations can be reduced. If one was to choose only natural sunscreen, e.g.,
as defined by European COSMOS (cosmetic organic standard) trade standards
[43], then only inorganic UV filters (TiO2 and ZnO) could be used and thus only 3
out of the 27 product variations are left to choose from. The fixed value of a param-
eter, in this case natural sunscreen, is called a driver in the GMA nomencla-
ture. Table 11.3 lists systematically parameters and values of sunscreens. Such a
list is of course never exhaustive; there is always room for new ideas, but the sys-
tematic approach is also a checklist that helps in considering all aspects of the
sunscreen product.
Innovation has to be new but must also have an impact on the market place. From
this morphological approach, it becomes apparent that the three categories,
technology, marketing, and regulatory, all play an important role. Without UV filter
technology, no progress in more efficient and broader UV coverage as well as yet
higher safety could be achieved, but if the advantage is not perceived in public, the
best technology cannot make an impact on the market [41].
11 Global UV Filters: Current Technologies and Future Innovations 187
A third approach to learn more about future innovations in suncare is asking the
opinion of experts. The Delphi method has been developed by the RAND
Corporation [44]. Delphi is based on the principle that forecasts (or decisions) from
a structured group of individuals are more accurate than those from unstructured
groups [45, 46], based on the assumption that a group of experts can more accurately
predict the future.
188 U. Osterwalder and L. Hareng
The following question was asked to about 40 experts from all over the world
where sunscreens play an important role in sun protection: What Innovations in
sunscreens do you see happening in the next: (a) 12, (b) 35, and (c) 1015 years?
Regarding technology innovations, the experts confirm the trend of better UVA
protection toward spectral homeostasis. Furthermore, better sunscreen formulations
are expected based on new UVB and broad-spectrum UV filters (liquid UV filters or
polymers). Some also predict the trend away from nanoparticles to continue.
Regarding marketing and performance innovations, the extension of UV
protection claims beyond just SPF, but into protection in visible light and infrared
range is predicted. Antioxidant claims are also anticipated. Furthermore a trend
toward natural sunscreens and more public education are predicted.
Regarding regulatory/standards innovation, the pending issues at the US FDA
(TEA UV filters, spray, SPF cap at 50+, etc.) are predicted to be resolved in the next
few years. A worldwide ban of animal testing in cosmetics is anticipated as well as
a ban of SPF in vivo testing on humans.
This 3rd approach is especially valuable in the context of the two previous ones. It
confirms the extrapolation of the past (1st approach), but it also brings up some new ideas
that reach outside the morphological box (2nd approach), e.g., a ban of human testing.
Table 11.4 UV filters, globally approved and pending in the USA (TEA)
UV range INCI Type/
(nm) USAN Trademark Abbr.a stateb Max. concentration limit (%)
USA (year of TEA filing) EU
290340 Global Oxybenzone Uvinul M40 BP3 o/p 6 10
Sulisobenzone Uvinul MS40 BP4 o/p 10 5
Octinoxate Uvinul MC 80 EHMC o/l 7.5 10
Octisalate Neo Heliopan OS EHS o/l 5 5
Homosalate Eusolex HMS HMS o/l 15 10
Octocrylene Uvinul N 539 T OCR o/l 10 10
Ensulizole Eusolex 232 PBSA o/p 4 8
Titanium dioxide Eusolex T2000 TiO2 i/p, d 25 25
TEA (USA) Iscotrizinol (Uvasorb HEB) DBT o/p 10 (2005) 10
Octyltriazone Uvinul T150 EHT o/p 5 (2003) 5
Amiloxate Neo Heliopan E1000 IMC o/l 10 (2003) 10
Enzacamene Eusolex 6300 MBC o/p 4 (2002) 4
290400 Global Avobenzone Parsol 1789 BMBM o/p 3 5
Zinc oxidec Z-Cote HP1 ZnO i/p, d 25 25
TEA (USA) Bemotrizinol Tinosorb S BEMT o/p 10 (2005) 10
Bisoctrizole Tinosorb M (active) MBBT o/d 10 (2005) 10
Global UV Filters: Current Technologies and Future Innovations
Table 11.5 Comparison of available US State of the Art Sunscreen with sunscreens modified with
TEA ingredients, and ideal sunscreen (cloth)
Modified US sunscreen Ideal sunscreen:
Best US Sunscreena with TEA ingredientsa cloth [4]
SPFcalculated 37 (labeled up to 100) 35 (labeled up to 50+) >>100
UVA-protect.: 372 381 389
CW (nm) 0.67 0.81 1
UVA/UVB 0.29 (<0.33; fail) 0.47 (>0.33; pass) 1
UVA-PF/SPF
Composition: 6 % BP3, 5 % EHS, 15 % 5 % EHS, 5 % HMS, Cloth (black)
UVB/UVA2 HMS, 10 % 1 % EHT
UVA1, OCR
Broad- 3 % BMBM 3 % BEMT, 5 %
Spectrum MBBT
Total UV 39 % 19 % n.a.
filters
NTUV dose at 3.2 1.6 1.0
1 MED actual actual actual
ideal ideal
(Calculated) ideal
0.0 1.0 2.0 3.0 4.0 0.0 1.0 2.0 3.0 4.0 0.0 1.0 2.0 3.0 4.0
a
Abbreviation of the INCI Name (International Nomenclature of Cosmetic Ingredients): BEMT
bis-ethylhexyloxyphenol methoxyphenyl triazine, BMBM butyl methoxydibenzoylmethane, BP3
benzophenone-3, EHS ethylhexyl salicylate, EHT ethylhexyl triazone, HMS homomenthyl
salicylate, MBBT methylene bis-benzotriazolyl tetramethylbutylphenol (nano), OCR octocrylene
MED 1 minimal erythema dose passes through sunscreen onto skin, NTUV normalized transmitted
UV dose
wavelength. The difference of sunscreens at equal SPF is mainly in the UVA1 region
(340400 nm). We know now that not only the erythemally weighted UV radiation is
the cause of damage but also the extent of UVA1 radiation [5154].
As shown in Table 11.5 and Fig. 11.5, much higher protection in the UVA1 spec-
trum can be achieved by the incorporation of TEA broad-spectrum filters; further-
more, a better protection can be achieved with lower total amounts of UV filter
(19 % compared 39 %).
11 Global UV Filters: Current Technologies and Future Innovations 191
11.4 Safety
Sunscreen products are used widely, often daily across the whole population,
which leads to high safety requirements irrespective of the regulatory environ-
ment. Therefore a UV filter-specific safety assessment is mandatory for its regu-
latory approval. In contrast to drugs, the deposition of UV filters on the skin is
a prerequisite for their effectiveness and the uptake into the body is not intended.
The protection against the known carcinogenic effect of UV light is to be
emphasized as a health benefit of the UV filter besides its specific safety
profile.
The safety assessment approach combines all relevant toxicological data to
determine the UV filter intrinsic hazard profile that is to be compared to the expo-
sure situation under conventional use of the sunscreen product. Such a hazard pro-
file is initially determined by a basic set of studies addressing acute and topical
toxicity of the UV filter such as skin/eye irritation, skin sensitization, and photo-
induced toxicity. Furthermore, genotoxicity tests provide the basis for an adequate
assessment of a potential mutagenic or cytogenetic effect of the UV filter and the
absence of a genotoxic potential during the intended use in sunscreens is addressed
by photogenotoxicity tests.
Repeated administration of the UV filter to animals in subacute, (sub-)chronic,
or reproductive/developmental studies allows a thorough assessment of the sys-
temic or reproductive toxicity potential. These studies help to identify target organs
and are used for the determination of a UV filter-specific no observed adverse effect
level (NOAEL). If no tissue changes indicate the onset of a tumor formation after
repeated dosing, no genotoxic effects are observed, and no evident systemic uptake
of the UV filter is found, a carcinogenic potential can be excluded in a weight of
evidence without performing a definitive carcinogenicity test. However, if
carcinogenic alerts exist, animal carcinogenicity studies are considered as a last
resort to fully elucidate this endpoint [55].
Dermal penetration data [supported by studies on absorption, distribution,
metabolization, and excretion if available] represent an important pillar to
estimate the potential systemic human exposure with the UV filter during use.
Based on standard exposure parameters for the use of sunscreen products [56]
and UV filter-specific dermal penetration data, a systemic exposure dose of the
UV filter for humans can be determined. This exposure dose takes into account
the usual daily amount of sunscreen applied, the maximum concentration of the
UV filter in sunscreens, and the dermal absorption of the UV filter as
determined in the safety studies. In order to cover uncertainties due to variances
in toxicological susceptibility between animals and humans and within the
human population, the estimated human exposure dose needs to be at least
100-fold below the no observed adverse effect level identified in the relevant
animal toxicity study in order to demonstrate the safe use of the UV filter in
sunscreens [56].
192 U. Osterwalder and L. Hareng
Since safety is a prerequisite for the use of any consumer or medicinal product and
since sunscreen products are used widely, often daily and on young children, high
safety requirements apply. In Europe, UV filters must qualify for the positive list
(Annex VI) of the European Cosmetics Regulation [57, 58], and in the USA, UV
filters have to be listed in the FDA Over-the-Counter Sunscreen Monograph as
active ingredients [29]. Similar requirements exist in most countries, e.g., Australia,
Japan, China, and Brazil [5961].
For UV filters, the FDA sunscreen monograph [29] dictates the use of UV active
ingredients in sunscreens. All ingredients listed on the sunscreen monograph may
be combined into different sunscreens, with some important restrictions, e.g., no
avobenzone/TiO2 or avobenzone/ZnO combination is allowed. These restrictions go
back to concerns about chemical interactions leading to photoinstability. The issue
is still awaiting a final decision by the FDA [62].
Over the last two decades, new UV filters have been developed that are photostable and
cover a broad range of the UV spectrum [5, 48, 49]. The USA is the only country that has
not benefited from these innovations, because the US FDA has not added any new UV
filters to the sunscreen monograph since the addition of avobenzone in 1997 [63]. In 2002
the time and extent application (TEA) process was enacted to extend the approval process
to UV filters from abroad [64]; however, none of the eight UV filters that have been filed
through the TEA process has yet been approved (Table 11.5). There is only one UV filter
(avobenzone) for efficient UVA1 protection currently available in the USA. ZnO is much
less efficient and offers little protection beyond 370 nm and not permitted in combination
with avobenzone. On the other hand, there are a number of new UVA1 and broad-
spectrum UV filters in the TEA pipeline that have all been available in the rest of the world
for 15 years or more (bemotrizinol, bisoctrizole, ecamsule, and drometrizole).
Due to the lack of progress of the approval process, the Sunscreen Innovation
Act (SIA) was signed into law by the President of the USA on November 29, 2014,
stating defined time limits for the different steps of the approval process [65]. The
SIA had been initiated by the multi-stakeholder PASS coalition [66] and was rated
among the top health initiatives by the TIME magazine in 2014 [67]. In response,
the FDA held a meeting of its advisory committee on nonprescriptive drugs in
September 2014 [6872], where the industry had an opportunity to explain their
safety assessment approach (see Sect. 11.5). Early 2014, the FDA had already
started to send response letters to the TEA applicants requesting more data [7379].
Early 2015 it became apparent that there will not be any new UV filters on the US
market any time soon [80]. The FDA maintains that there is currently not enough
data to determine that any of the ingredients under review are generally recognized
as safe and effective [81].
11 Global UV Filters: Current Technologies and Future Innovations 193
An evaluation at the launched products over the last few years in the high and very
high SPF category done by the market research company MINTEL [24] shows that
only a limited number of UV filters is used in the USA and Canada (North America)
compared with the rest of the world (Fig. 11.6). US sunscreens that provide the
required UVA protection are mainly composed of the five UV filters: avobenzone,
homosalate (HMS), octisalate (EHS), octocrylene (OCR), and oxybenzone (BP3).
Only avobenzone provides UVA1 protection (beyond 360 nm). The globally most
frequently used UVB filter octinoxate (EHMC) is practically not used in sunscreens
in the USA because it destabilizes avobenzone. In the rest of the world, it is used
together with other UVA/broad-spectrum UV filters. Its slight photoinstability,
partly due to internal cis/trans conversion, does not significantly affect its efficacy.
TiO2 and ZnO are less frequently used because they are not allowed in combination
with avobenzone due to FDA monograph restrictions. US manufacturers have thus
to make the decision to use either avobenzone or ZnO as UVA protection platform.
Figure 11.6 shows that in the rest of the world many more UV filters are used,
besides the global filters, mainly the TEA filters for UVA, bemotrizinol (BEMT),
bisoctrizole (MBBT), drometrizole (DBT), and ecamsule (TDSA) and the UVB
filters octyl triazone (EHT) and iscotrizinol (DBT). It should be noted that a global
filter oxybenzone (BP3) is virtually not been used anymore in Europe, having been
replaced by the new UVA filters. One reason is the mandatory declaration contains
oxybenzone because of its allergy potential. Such phasing out of oxybenzone is not
yet being possible in the US market for lack of available alternatives.
11.6 Conclusion
Tremendous progress has been made in sunscreen technology over the last two
decades, with the development of new photostable filters that covers a broad range
of UV radiation, including UVA1. Unfortunately, in the USA, UV filter technology
is lagging 15 years behind compared to the rest of the world. Until FDA provides
approval of new UV filters that have been submitted through the TEA process, it is
difficult for the sunscreen industry to introduce truly innovative new sunscreen
products in the USA.
In Europe and the rest of the world, development goes on, but it is slower than 20
years ago. The animal test ban in the European Cosmetics regulation may further
slow down technology innovations, but it could also trigger a regulatory shift away
from cosmetics toward medical device.
The major weakness of sunscreen and photoprotection remains the lack of com-
pliance by the user. To solve this problem, innovation in photoprotection education,
including behavior modification and sunscreen use, is required.
194 U. Osterwalder and L. Hareng
80 %
UV filters in % of SPF products launched
70 %
60 %
50 %
40 %
30 %
20 %
10 %
0%
EHMC
TiO2
OCR
EHS
BP3
HMS
EHT
PBSA
DBT
IMC
BMBM
BEMT
MBBT
DTS
TDSA
ZnO
UVB-UVA 2 Broad-spectrum/UVA 1
USA Europe
Fig. 11.6 UV filters used in sunscreens launched (After MINTEL [24]). Please see footnote in
Table 11.4 for complete list of abbreviations
Acknowledgement Thanks go to those who have contributed to the Delphi survey, although they
will remain anonymous to the reader; further to Prof. Brian Diffey for all his scientific contributions
to the topic over the last three decades and the valuable discussions; furthermore to our BASF
colleagues Drs. Bernd Herzog, Mechtild Petersen-Thiery, and Myriam Sohn for valuable
discussions; and last but not the least to the intern Felix Presencio who helped compiling and
designing tables and figures.
References
1. Hughes MCB, Williams GM, Baker P, Green AC (2013) Sunscreen and maximum penetration
is only 0.6 % of the applied prevention of skin aging. A randomized trial. Ann Intern Med
158:781790
2. Green A, Williams G, Neale R et al (1999) Daily sunscreen application and beta-carotene
supplementation in prevention of basal cell and squamous-cell carcinomas of the skin: a
randomised controlled trial. Lancet 354:723729
3. Green AC, Williams GM, Logan V, Strutton GM (2011) Reduced melanoma after regular sun-
screen use: randomized trial follow-up. J Clin Oncol 29:257263
4. Viros A, Sanchez-Laorden B, Pedersen M, Furney SJ, Rae J, Hogan K, Ejiama S, Romina
Girotti M, Cook M, Dhomen N, Marais R (2014) Ultraviolet radiation accelerates BRAF-
driven melanomagenesis by targeting TP53. Nature 511:478482
5. Herzog B, Hueglin D, Osterwalder U (2005) New sunscreen actives. In: Shaath NA (ed)
Sunscreens regulations and commercial development, cosmetic science and technology, vol
28, 3rd edn. Taylor & Francis, Boca Raton, pp 291320
6. Meyers T, Agin P (2015) Chapter 11: Chemistry of sunscreens understanding properties of
organic and inorganic UV filters. In: Lim H, Wang S (eds) Principles and practices of
photoprotection. Springer, Heidelberg
7. Herzog B (2012) Photoprotection of human skin. In: Albini A (ed) Photochemistry, vol 40.
RSC Publishing/The Royal Society of Chemistry, Cambridge, UK, pp 250259
8. Anonymous. BASF sunscreen simulator, BASF SE, Ludwigshafen. 2010. Available at http://
www.basf.com/sunscreen-simulator. Accessed 18 Jan 2015.
11 Global UV Filters: Current Technologies and Future Innovations 195
lex.europa.eu/LexUriServ/LexUriServ.do?uri=COM:2008:0049:FIN:EN:PDF. Accessed 13
Feb 2015.
58. Rogiers V, Pauwels M (2008) Safety assessment of cosmetics in Europe, current problems in
dermatology. S Karger AG, Basel
59. Therapeutic Goods Administration. Australian regulatory guidelines for sunscreens, Version
1.0. 2012. https://www.tga.gov.au/publication/australian-regulatory-guidelines-sunscreens-
args, Accessed 31 Jan 2016.
60. Xia J. Review of cosmetics regulations in Asia. http://www.skin-care-forum.basf.com/en/
author-articles/review-of-cosmetics-regulations-in-asia/2012/12/07?id=a016034e-663a-4159-
bf20-2cdae216763e&mode=Detail. Accessed 5 Apr 2015.
61. ANVISA Brazil. http://portal.anvisa.gov.br/wps/content/Anvisa+Portal/Anvisa/Inicio/Cosmeticos.
Accessed 5 Apr 2015.
62. Hubbard WK, Sunscreen drug products for over-the-counter human use; Amendment to the ten-
tative final monograph; Enforcement Policy Federal Register 63(204)/Thursday, October 22,
1998/Proposed Rules, p. 56587.
63. Food and Drug Administration. Sunscreen drug products for over-the-counter human use; mar-
keting status of products containing Avobenzone, enforcement policy. Fed Regist.
62(83):23350. Wednesday, Apr 30, 1997. Rules and regulations.
64. Food and Drug Administration. Additional criteria and procedures for classifying over-the-
counter drugs as generally recognized as safe and effective and not misbranded, final rule. Fed
Regist. 67(15):3060. Wednesday, Jan 23 2002.
65. Reed J. Sunscreen Innovation Act (SIA), Public Law No. 113195, 26 Nov. 2014.
66. Anonymous, On the Brink of Year Anniversary of the Sunscreen Innovation Act Becoming
Law, Nothing Has Changed Except Rising Melanoma Rates, http://www.passcoalition.com/.
Accessed 14 Feb 2014.
67. Sifferlin A. 11 remarkable health advances from 2014. Time. http://time.com/3640238/11-
remarkable-health-advances-from-2014/. Accessed 5 Apr 2015.
68. Michele TM. FDA briefing document for a meeting of the Nonprescription Drugs Advisory
Committee Sunscreen Active Ingredients Topic: sunscreen active ingredient safety testing 4, 5
Sept, 2014, and 6 Aug 2014.
69. FDA briefing document for a meeting of the Nonprescription Drugs Advisory Committee
(NDAC). Sunscreen actives, topic: sunscreen active ingredient safety testing, 4, 5 Sept 2014.
70. Hong C, Neill R. FDA, summary minutes of the Nonprescription Drugs Advisory Committee
(NDAC) meeting 4, 5 Sept 2014.
71. Michele TM. FDA, slides of the Nonprescription Drugs Advisory Committee (NDAC) meeting
4, 5 Sept 2014.
72. Hong C. FDA, transcripts of the Nonprescription Drugs Advisory Committee (NDAC) Meeting
4, 5 Sept 2014.
73. Kweder SL. FDA, feedback letter on safety and effectiveness data of Amiloxate submitted by
Symrise Inc. and Ego Pharmaceuticals Pty. Ltd. 2014.
74. Kweder SL. FDA, feedback letter on safety and effectiveness data of Diethylhexyl Butamido
Triazone submitted by 3V Inc. 2014.
75. Kweder SL. FDA, feedback letter on safety and effectiveness data of Octyl Triazone submitted
by BASF AG. 2014.
76. Kweder SL. FDA, feedback letter on safety and effectiveness data of Drometrizole Trisiloxane
submitted by LOREAL USA. 2014.
77. Kweder SL. FDA, feedback letter on safety and effectiveness data of Bemotrizinol submitted
by Ciba Specialty Corp. 2014.
78. Ecamsule, FDA proposed order https://federalregister.gov/a/2015-03883
79. Enzacamene, FDA proposed order. https://federalregister.gov/a/2015-03884
80. Editorial, Washingtons Skin Cancer Hostages The FDA is defying Congress and Obama on better
sunscreens, WSJ. 2015. http://www.wsj.com/articles/washingtons-skin-cancer-hostages-1426287044.
Accessed 5 Apr 2015.
81. Michele TM. FDA is systematically screening for safe sunscreens, letters. WSJ. 2015. http://
www.wsj.com/articles/fda-is-systematically-screening-for-safe-sunscreens-letters-to-the-edi-
tor-1427649129
Chapter 12
Organotypic Models for Evaluating
Sunscreens
Key Points
The development of reconstructed skins has made possible in vitro assess-
ment of the effects of different types of UV exposure (UVB, UVA, or solar
simulation) in a three-dimensional context and in a cutaneous structure,
including different types of skin cells.
Reconstructed skin shows numerous biological endpoints which are pre-
dictive of in vivo response, hence allowing a better understanding of the
precise biological processes involved.
Reconstructed skin can be used to evaluate the photoprotection afforded by
sunscreens in vitro, providing additional biological data on sunscreen effi-
cacy to correlate with protection factors assessed in vivo.
The combination of 3-D skin models and new biological approaches such
as transcriptomic or proteomic will indisputably increase the added value
of such systems for evaluating sunscreen performance.
Skin, the largest organ of the human body, represents the main barrier ensuring a key
function of protection against external/environmental harm. Among this, solar and
especially ultraviolet (UV) rays can be considered as one of the major contributors.
The protective properties of the skin are supported by the whole skin structure in a
coordinate manner between the different compartments. The most superficial cuta-
neous layer, the epidermis, mainly composed of keratinocytes (approx. 90 %),
undergoes a stratification process and a specific and fine-tuned program of keratino-
cyte differentiation that leads to the formation of a compact stratum corneum. The
latter ultimately constitutes the first line of defense of the skin (for review, [67, 99]).
The epidermis is also the place of residence for the following: (1) melanocytes, the
pigmentary cells responsible for melanin synthesis [73], and (2) Langerhans cells, a
member of the antigen-presenting cell family involved in immune function [27]. The
underlying dermal compartment is mostly composed of extracellular matrix proteins
(ECM) synthesized by dermal fibroblasts and provides a mechanical and thermal
protective layer. It also hosts blood vessels as source of nutriments, nerve endings
and various appendages such as hair follicles, sebaceous and sweat glands [56].
Beside some beneficial effects of sunlight, such as vitamin D production, acute or
repetitive solar UV exposure can lead to harmful clinical consequences such as sun-
burn reaction associated with erythema and epidermal sunburn cells (SBC) forma-
tion but also middle- and long-term effects such as photoimmunosuppression,
photoaging mostly characterized by dermal alterations and the development of solar
elastosis and photocarcinogenesis, especially epidermal basal and squamous cell
carcinomas [65]. It is also known that hyperpigmentation, including the physiologi-
cal tanning response but also the appearance of hyperpigmented lesions such as
actinic lentigines, is directly related to sun exposure. Considering all these phenom-
ena, both compartments of the skin, dermis and epidermis, are affected. In addition,
it is now proven that all UV rays that reach the Earth surface are involved. UVB rays
(290320 nm), the most energetic wavelengths, can directly induce DNA lesions
such as cyclobutane pyrimidine dimers (CPDs) and 6,4-photoproducts. Most of the
direct UVB effects are located within the epidermis due to low penetration of these
wavelengths. Short (UVA2) or long (UVA1) UVA radiation (320340 nm and 340
400 nm, respectively) are less energetic than UVB but show progressively higher
penetration properties with increasing wavelength and can therefore reach the der-
mal compartment and its cells. Their major mode of action is the generation of reac-
tive oxygen species (ROS) that, in turn, lead to activate various signaling pathways.
For both designing and evaluating the most effective photoprotection strategies, it is
crucial to understand and characterize the early biological events that occur
following UV exposure. For practical and ethical reasons in vivo studies in human
volunteers are often difficult to perform. In contrast, classical two-dimensional
(2-D) skin cell cultures poorly reproduce physiological conditions and tissue
organization, such as epidermal differentiation and cell-cell and cell-matrix
interactions. Moreover, they cannot take into account the penetration of UV rays
through the different skin compartments.
In vitro 3-D engineered skin models have been developed during the last 30 years
on the basis of human skin cell culture and organotypic reconstruction techniques and
12 Organotypic Models for Evaluating Sunscreens 201
Fig. 12.1 Main steps of in vitro reconstruction of the full-thickness skin model. First, a human skin
sample is trypsinized to separate dermis from epidermis. From both compartments, epidermal kerati-
nocytes and dermal fibroblasts are isolated and amplified in their respective culture medium. The
reconstruction starts with the production of the living dermal equivalent by mixing a collagen type I
solution with medium and dermal fibroblasts. This gel is poured into a Petri dish and left in the incuba-
tor at 37 C for 34 days to allow the contraction to proceed. Then the epidermis reconstruction starts
by seeding epidermal keratinocytes on the top of the dermal equivalent. The ring allows for control of
cell density. The culture is left for the immersion phase corresponding to the formation of a simple
epithelium covering the dermal substrate. Afterward, the whole culture is raised to the air-liquid inter-
face, fed by capillarity to promote the stratification and epidermal differentiation process. At the end
of this phase (usually 7 days), a stratum corneum is formed ( LOral Research and Innovation)
know-how. From the first reconstructed epidermis on a cell-free dermal substrate [100],
the in vitro skin models have been perfected over the past decades by adding different
cell types, improving the dermal equivalent and increasing the functionality of the
models [5, 37, 42, 75]. Reconstruction of in vitro skin models usually follows similar
key step process ([33, 40], and Fig. 12.1): (1) extraction of keratinocytes from the
epidermis of skin biopsies and amplification and (2) seeding of keratinocytes on the top
of a dermal equivalent which can be either a de-epidermized dermis (DED), an acel-
lular collagen matrix, a polycarbonate membrane, or a living dermal equivalent com-
posed of ECM and dermal fibroblasts. The keratinocytes are allowed to proliferate onto
the surface of the support by being submerged by the culture medium. During this step,
other epidermal cell types may be added melanocytes or precursors for Langerhans
cells (CD34+ cells) depending on the model to be produced. In the last step, the
whole culture system is placed in contact with air, corresponding to the air-liquid inter-
face culture period. During that phase, the culture medium is added underneath the
dermal support, and the system is fed by capillarity. This air-exposed phase period is
mandatory for the stratification and full differentiation of the epidermal structure.
Figure 12.2 illustrates different skin models.
202 C. Marionnet and F. Bernerd
a b c
d e f
Full thickness (lattice model) Reconstructed epidermis + LC (DED) Full thickness model + MC
Fig. 12.2 Examples of organotypic skin models. (a) Normal human skin, bar = 50 m. (b)
Reconstructed epidermis on an acellular dead de-epidermized dermis (DED). (c) Reconstructed
epidermis on an acellular collagen matrix Episkin model. (d) Full-thickness skin model
composed of fibroblast-populated collagen matrix as dermal support and differentiated epidermis.
(ad) Hematoxylin-eosin staining. (e) Reconstructed epidermis on DED support; Langerhans cells
(brown) stained with Langerin antibody. (f) Pigmented full-thickness skin model. Melanocytes
(MD; green) are visualized using anti-tyrosinase-related protein (TRP)-1 antibody. Nuclei are
counterstained with propidium iodide
Fig. 12.3 Schematic representation of the main biological effects induced by UVB, UVA, or UV solar-stimulated radiation (SSR) exposure in a reconstructed
skin model. CPDs cyclobutane pyrimidine dimers, DCFH-DA 2, 7 dichlorofluorescin diacetate, ECM extracellular matrix, F fibroblasts, K keratinocytes, LC
Langerhans cells, M melanocytes, MMP1 matrix metalloproteinase 1, ROS reactive oxygen species, SBC sunburn cell
203
204 C. Marionnet and F. Bernerd
This points out the indirect impact of UVB radiation on fibroblasts and ECM, via its
direct action on epidermal keratinocytes and their release of diffusible IL1 and IL6
cytokines [45, 46]. UVB also increased the expression and activity of epidermal
MMPs, such as MMP-2 and MMP-9 in a full-thickness skin model [2]. Reinforcing
the involvement of UVB in photoaging process, Kurdykowski et al. showed the
UVB modulation of hyaluronidases expression in reconstructed epidermis [66].
The use of reconstructed pigmented skin, including a mix of melanocytes and
keratinocytes seeded onto a dermal equivalent or a de-epidermized dermis, enabled
the impact of UVB exposure on pigmentation to be investigated. Sequences of
repeated UVB exposures led to an increase in proliferation, dendricity, and activity
of melanocytes and an increase in melanin production and in melanosome transfer
from melanocytes to keratinocytes, resulting in a noticeable tanning of the
reconstructed epidermis [20, 21, 38, 52, 69, 90, 116, 117].
Studies of the impact of UVA in reconstructed human skin revealed that this 3-D
skin model enabled to reproduce main features of UVA effects observed in human
skin in vivo.
The immediate damage following UVA exposure in human skin in vivo is the gen-
eration of reactive oxygen species (ROS) leading to oxidative stress, as well as DNA
damage, especially pyrimidine dimers and 8-oxo-7,8-dihydro-2-deoxyguanosine
(8-OHdG) accumulating in basal keratinocytes that may lead in the long term to DNA
mutations [64, 114].
In reconstructed skin, UVA exposure also led to ROS formation, as visualized
using DCFH-DA probe, in a UVA dose-dependent manner in both fibroblasts and
keratinocytes. Increasing doses of UVA induced ROS deeply in the epidermal basal
layer but also in the deepest dermal fibroblasts illustrating the high penetration prop-
erties of UVA wavelengths [80, 118]. Six hours after UVA exposure, cells of recon-
structed skin responded to this oxidative stress with the upregulation of the expression
of genes involved in oxidative stress management such as genes of the Nrf2 pathway.
For example, a strong increase in HMOX1 and TXNRD1 mRNA could be observed
in fibroblasts, together with a strong increase in TXNRD1, NQO1, and FTL mRNA
in keratinocytes. A UVA induction of ferritin protein was detected in basal keratino-
cytes of reconstructed epidermis [109]. The expression of genes involved in the
redox status of glutathione was also modulated [86]. ROS may lead to cell compo-
nents alterations in reconstructed skin, such as lipid peroxidation and protein oxida-
tion [54]. Lipid peroxidation can in turn lead to cell membrane damage and can also
act as cell signal mediators since particular oxidized phospholipids could induce
HMOX1 expression in reconstructed skin [54]. Protein oxidation phenomenon can
be amplified by pheomelanin, acting as a photosensitizing agent, as shown by
Maresca et al., in a reconstructed model including melanocytes [34, 77, 80].
206 C. Marionnet and F. Bernerd
As found in human skin in vivo, thymine dimers were also detected after UVA
exposure, in basal keratinocytes of reconstructed skin using immunostaining, albeit
in a much lower amount than post UVB exposure [13, 80, 114]. Immunostainings
also revealed increased levels of 8-OHdG oxidative DNA lesion in epidermal and
dermal cells nuclei following UVA exposure of living skin equivalents [36].
Furthermore, exposure to repeated low doses of UVA induced p53 mutations in
basal keratinocytes of the epidermis [63].
Regarding histological alterations, in contrast to UVB, major features of UVA
effects were located in the dermal compartment of reconstructed skin in vitro, in
correlation with previous human in vivo studies showing that repetitive exposures to
low UVA doses induced early morphological and biochemical alterations in the
dermis [11, 68, 74].
Forty-eight hours after UVA exposure of reconstructed skin, the dermal fibro-
blasts localized in the superficial portion of the dermal equivalent disappeared,
underlining the significant biological impact of UVA in deeper layers of skin and
confirming that dermal fibroblasts were more sensitive to UVA-induced oxidative
stress than keratinocytes [3, 88]. UVA cytotoxicity toward fibroblast was direct
and mostly due to apoptosis, accompanied by an upregulation of the expression of
genes related to cell death and apoptosis, such as DDIT3, IER3, BIRC3, and
NR4A1, NR4A2, and NR4A3 [13, 43, 80]. This particular impact on dermis was
emphasized by the upregulation of several MMP gene and protein expression
(e.g., MMP-1, MMP-9, MMP-3) [78, 86]. It was shown that UVA exposure, in
contrast to UVB exposure, induced the production of MMP1 by fibroblasts in a
direct manner, since the removal of epidermis immediately after UVA exposure
did not alter this effect [118]. The expression of COL1A1 gene was downregu-
lated in fibroblasts of reconstructed skin exposed to UVA rays [79, 80, 86]. The
epidermal structure and organization were to a lesser extent impacted by exposure
to UVA, with a slight impact on the upper layers and parakeratosis [13]. Higher
doses of UVA could lead to disorganization of the living epidermis together with
a reduced skin barrier function, increase in phospholipid, and decrease of ceramide
levels [103].
In vivo, UVA exposure also impacts skin immunity, with inflammatory effects and
with immune suppression [28, 57, 58]. In line with these in vivo clinical features, UVA
exposure of reconstructed skin leads to the upregulation of proinflammatory genes
and/or proteins such as IL1, IL6, IL8, GM-CSF, COX-2, or PGE2 [32, 78, 80, 92]. In
contrast, numerous genes encoding proteins involved in antiviral defense were strongly
downregulated following UVA exposure in fibroblasts and keratinocytes of recon-
structed skin, possibly related to photoimmune suppression observed in vivo [80].
Concerning pigmentation process, Duval et al., using a reconstructed epidermis
including melanocytes, showed that UVA exposure led to the production and
transfer of melanin to the neighboring keratinocytes and resulted in tanning of the
reconstructed epidermis, like UVB exposure, and as observed in vivo [38, 97].
Altogether these results illustrated the penetration properties of UVA rays as
attested by the direct UVA-induced biological damage in dermis and the particular
vulnerability to UVA rays of the deepest epidermal layer, location of epidermal
stem cells, proliferative keratinocytes, and melanocytes [59, 113]. The particular
12 Organotypic Models for Evaluating Sunscreens 207
impact of UVA on the dermal compartment observed in vivo and in 3-D models
in vitro may be involved in early events occurring during photoaging leading to
drastic alterations of dermal structure and formation of the solar elastosis, classically
observed in photoaged skin [25].
The effects of solar-simulated radiation (SSR), including UVA and UVB, have been
studied in reconstructed human skin models. Today, two types of solar simulation
can be distinguished: UV solar-simulated radiation (UV-SSR) and daily UV
radiation (DUVR). Both include UVA and UVB rays, but in different proportion,
the DUVR spectrum including a higher UVA proportion than the UV-SSR spectrum,
in order to simulate two distinct types of sun exposure. UV-SSR spectrum mimics a
condition of exposure under a summer zenithal sunlight (i.e., sunbathing on a beach
in summer under a clear sky) and may rapidly lead to erythema in human skin
in vivo therefore maximizing UVB impact. In turn, DUVR spectrum simulates a
non-extreme condition of sun exposure corresponding to a western spring or autumn
sunlight, with a solar elevation angle lower than 45, which does not give rise to any
visible immediate clinical damage [26, 53, 83, 106].
12.3.3.1 UV-SSR
It has been shown that UV-SSR induce DNA damage in keratinocytes of recon-
structed skin, such as pyrimidine dimers, (6-4) photoproducts, photooxidative dam-
age, and single-strand breaks [19, 22, 85, 98]. This was followed by an accumulation
of p53 and an upregulation of genes controlled by p53 involved in DNA repair and
in cell cycle regulation, such as p21, MDM2, and GADD45 genes. In addition,
genes of the Nrf2 pathway were upregulated post UV-SSR exposure in keratino-
cytes [85]. The levels of HSP27, MnSOD, and PDX-2 proteins, also involved in
oxidative stress response, were upregulated after UV-SSR exposure as revealed by
proteomic profiling of reconstructed epidermis exposed to UV-SSR [62].
Histologically, changes induced by UV-SSR exposure of reconstructed skin
closely resemble to those observed in vivo and those observed following pure UVB
exposure. UV-SSR clearly impacted epidermis, with the induction of epidermal
SBC formation 24 h after exposure, as well as an absence of laminin deposition at
the basement membrane. Exposure to higher doses of UV-SSR led to an epidermal
disorganization, a thickened stratum corneum and a reduction in the number of epi-
dermal cell layers [19, 22, 46].
Repeated UV-SSR exposures also impacted the morphology of melanocytes in
reconstructed skin: they became more dendritic, as observed in vivo. Exposure of
pigmented reconstructed skin to UV-SSR induced an increase in melanin content
and tanning of the 3-D model [4, 16, 39].
208 C. Marionnet and F. Bernerd
12.3.3.2 DUVR
Reconstructed skins, including different types of skin cells, present a tissue organiza-
tion close to that of in vivo human skin, with a correct epidermal differentiation, a
dermal epidermal junction as well as cell-cell and cell-matrix interactions. Due to
their 3-D architecture, they are useful tools to take into account the penetration of UV
rays through the different skin compartments. Moreover major UV-induced damage
observed in vivo can be reproduced in these 3-D skin models, with wavelength spe-
cific and common skin targets within epidermal and dermal compartments (Fig. 12.3).
Another great advantage of such 3-D skin models is the possibility to apply cos-
metic/dermatologic formulations directly on the skin surface as it can be done in real-
life situations. This aspect becomes paramount when dealing with sunscreen products
that are only topically applied, forming a barrier between solar UV rays and skin cells.
By using classical endpoints and new ones such as gene expression profiling,
reconstructed skins can be used to evaluate skin photoprotection incurred by
chemical (organic) or physical (inorganic) sunscreens. Single absorbers or complex
sunscreen formulations composed of a combination of filters, to provide the largest
absorption profile comprising UVB+UVA wavelengths domains, have been tested.
Table 12.1 summarizes the different studies found in the literature.
The first approach using reconstructed skin for the evaluation of photoprotective
efficacy determined the global cellular viability, by performing the MTT
(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) tetrazolium reduc-
tion assay [6, 92]. Augustin et al. tested photoprotection afforded by sunscreen
ingredient, Eusolex 6300 (Merck, USA), a UVB blocking benzylidene camphor
derivative (absorption peak at 300 nm), or Eusolex 8020 (Merck, USA), a UVA
blocking dibenzoylmethane derivative (absorption peak at 350 nm), both diluted at
3 % and applied topically onto skin equivalents. After exposure to UVB or UVA,
respectively, the residual cellular viability was found higher in photoprotected skin
versus control, demonstrating the photoprotective effect of the filters [6].
Commercially available sunscreen products were also tested using the same proto-
col. A sunscreen product with a SPF (sunburn protection factor) 15 containing
organic UVB or UVA filters and a sunscreen product, with ultra-high protection
including mineral filters, showed both good efficacy regarding viability of skin
equivalents following UVB or UVA exposure [6]. In an attempt to rank sunscreens
Table 12.1 Summary of studies using organotypic models for the evaluation of sunscreens
210
Protection
Sunscreena factorsb Organotypic Model Biological endpoints UV source Reference
Single absorbers (concentrations when available)
Homosalate (8 %) SPF 4.24 Full-thickness skin model MTT UV-SSR [92]
Photoplex (with PABA ester + BMDM) PFA 1.8 (LSE ) MTT UVA
Padimate O (7 %) PFA 1.31 MTT UVA
MTT UVA
Benzophenone 3 (2 %, 5 %) PFA 2.33, 3.97
4-Methylbenzylidene camphor (3 %) NA Dermal equivalent MTT UVB [6]
Isopropyl dibenzoylmethane (3 %) NA Full-thickness skin model MTT IL1 UVA
2-EHMC (5 %) SPF 5.8 Full-thickness skin model SBC, CPDs UVB [18]
Terephthalylidene camphorsulfonic SPF 5.1 Dermal fibroblasts, histology, UVA
acid (5 %) vimentin, MMP1
Drometrizole trisiloxane NA Full-thickness skin model SBC, CPDs UVB [14]
Dermal fibroblasts, histology, UVA
vimentin
Cinnamate SPF 12 Reconstructed epidermis SBC, CPDs, protein oxidation, UVB [24]
(DED model) +/ lipoperoxidation, vit E, SOD/ UVA
melanocytes Catalase ratio UVA + UVB
TiO2 (16 %) SPF 28 Full-thickness skin model 6.4 PP, CPD, photooxidative UV-SSR [104]
damage
Terephthalylidene camphorsulfonic SPF 5.1 Pigmented epidermis Pigmentation (visual, UV-SSR [39]
acid (5 %) (DED model) colorimetric L*)
Reconstructed epidermis Langerhans cells number and UV-SSR
(DED model) + morphology (dendricity)
Langerhans cells
C. Marionnet and F. Bernerd
12
Terephthalylidene camphorsulfonic acid NA Reconstructed epidermis Langerhans cells number and UV-SSR [44]
(1 % or 4 %) (Episkin model) + morphology (dendricity), SBC
Langerhans cells
Terephthalylidene camphorsulfonic NA Reconstructed epidermis DNA lesions (comet assay) UV-SSR [49]
acid (0.01 %) (Episkin)
Complex formulations (concentrations of sunscreens when available)
Chemical sunscreensc SPF 4, 8, 12, 15 Full-thickness skin model IL1 release UV-SSR [102]
Physical sunscreenc SPF 15 (Skinc)
4-Methylbenzylidene camphor (6 %), SPF 15 Full-thickness skin model MTT, IL1 UVA [6]
terephthalylidene camphorsulfonic acid MTT UVB
(1 %), BMDM (2 %)
TiO + TiO2 NA
Terephthalylidene camphorsulfonic SPF 25 PPD 5 Reconstructed epidermis MTT (phototoxicity induced by UVA [29]
acid + TiO2 (Episkin) chlorpromazine)
Terephthalylidene camphorsulfonic acid, SPF 60 PPD 12
Organotypic Models for Evaluating Sunscreens
Protection
Sunscreena factorsb Organotypic Model Biological endpoints UV source Reference
TiO2 (11 %), ZnO (4 %) NA Reconstructed epidermis Histology, SBC, viability, p53 UV-SSR [51]
Octyl methoxycinnamate (4 %), NA (SkinEthic RHE) protein expression
methylene bis-benzothiazolyl
tetramethylbutylphenol (4 %), TiO2
(1 %), ZnO (0.5 %)
BMDM (1.5 %), ethylhexyl triazone SPF 25,2 Full-thickness skin model Dermal fibroblasts, histology, UVA- [84]
(2 %), ethylbenzilidene camphor (5 %) in vimentin, MMP1 UV-SSR
15 % Miglyol
BMDM (1.5 %), ethylhexyl triazone SPF 22,8
(2 %), octyl methoxycinnamate (5 %) in
15 % vaseline oil
BMDM (2 %), octocrylene (10 %), SPF 15 Full-thickness skin model Dermal fibroblasts, histology, DUVR [71]
terephthalylidene camphorsulfonic vimentin, MMP1
acid (2 %)
Octyl methoxycinnamate (6 %), ZnO SPF 15
(3 %)
BMDM (3 %), terephthalylidene SPF 18
camphorsulfonic acid (3 %), octocrylene
(5 %), drometrizole trisiloxane (1 %)
Octocrylene (2.5 %), ethylhexyl SPF 27
methoxycinnamate (7.5 %), ethylhexyl
salicylate (2.5 %), ZnO (6 %)
2-EHMC, BEMT, methylene bis- NA Reconstructed epidermis Cell viability (LDH, ERK2 UV-SSR [8]
benzotriazolyl, tetramethylbutylphenol (SkinEthic RHE) release), DNA damage (thymin
dimers, DNA fragmentation),
apoptosis (TUNEL, caspase 3
activation), SBC
C. Marionnet and F. Bernerd
12
BMDM (3 %), homosalate (15 %), SPF 45 Full-thickness skin model ROS (DCFH-DA), IL1, UV-SSR [101]
ethylhexyl salicylate (5 %), octocrylene (StrataTest) IL1-RA, CPD
(4.5 %), benzophenone 3 (6 %)
BMDM (4 %), octocrylene (2.5 %), SPF 67.5- PPD Full-thickness skin model Gene expression/Protein UVA [78]
ethylhexyl salicylate (5 %), 31.1 expression
terephthalylidene dicamphor sulfonic
acid (1 %), drometrizole trisiloxane
(0.5 %), TiO2 (4.5 %), ethylhexyl
triazone (1 %), BEMT (3 %)
BMDM (3 %), octocrylene (5 %), SPF 13- PPD Full-thickness skin model Gene expression/Protein DUVR [82]
ethylhexyl salicylate (5 %), 10.5 expression
terephthalylidene dicamphor sulfonic
acid (3 %)
Commercial sunscreens SPF >20+/UVA SPF 2050 Full-thickness skin model DMPO protein-radical adduct UV-SSR [61]
protection (EpiDermTM Full
BMDM (5 %), 2-EHMC (10 %) SPF 15 Thickness)
Organotypic Models for Evaluating Sunscreens
on their photoprotection efficacy, Nelson and Gay compared three UVA filters and
one placebo having different UVA protection factors established in human skin by
scoring delayed erythema or tanning 24 h after UVA exposure [30]. Placebo, 7 %
padimate O, 2 % oxybenzones, and 5 % oxybenzones exhibited in vivo UVA protec-
tion factors of 1.15, 1.31, 2.33, and 3.97, respectively. Cytotoxicity measurements
in Living Skin Equivalent (LSE), a full-thickness skin model, exposed to UVA
allowed the authors to calculate in vitro photoprotection values of sunscreens, by
dividing the UVA50 in the sunscreen-applied skin equivalents by the UVA50 in the
unprotected samples. All the tested filters exhibited a higher photoprotection effi-
cacy compared to untreated skin. However, in vitro photoprotection values did not
fully rank sunscreens as in vivo protection factors. The authors suggested that such
discrepancy may be related to the different UVA light sources used in in vivo and
in vitro studies, due to the use of different cut-off filters (2 mm WG-345 filter
in vitro vs 3 mm WG-335 filter in vivo) [92].
Beyond cytotoxicity and because UVB and UVA induced specific damage in
human skin, endpoints related to wavelength range have been used in photoprotec-
tion studies.
In vivo, one of the first approaches to evaluate protection of sunscreens is the
determination of the SPF based on the prevention of cutaneous erythema, an end-
point mostly induced by UVB radiation. The clinical appearance of erythema has
been correlated with the formation of epidermal SBC, whose apoptotic process is
due to high levels of unrepaired DNA lesions. Moreover it has been shown in vivo
that sunscreens with appropriate SPF values are efficient in preventing UV-induced
DNA lesions [50, 72]. For these reasons, SBC and thymine dimer formation
appeared to be relevant to evaluate photoprotection against UVB or UV-SSR-induced
damage. Using these endpoints, the photoprotective efficiency of 2-ethylhexyl-p-
methoxycinnamate (2-EHMC, ParsolTM MCX), a UVB absorber, with an in vivo
SPF of 5.8, was tested in reconstructed skin exposed to UVB rays. By analyzing
thymine dimers and SBC formation after application of 2-EHMC, the authors
showed that the highest dose able to prevent SBC formation corresponded to five
times the biological efficient dose previously determined in the in vitro model [18],
in line with the in vivo SPF value (Fig. 12.4). Studying the protection afforded by
the UVB filter cinnamate, or complex formulations including cinnamate, Cario-
Andr et al. confirmed the relevance of using the prevention of SBC and CPD
formation as endpoints after UVB or UVA+UVB exposure of reconstructed epider-
mis on dead de-epidermized dermis [24].
The quantification of UV-induced lesions, i.e., pyrimidine dimers, 6,4-photo-
products, and photooxidative damage, in the epidermis of reconstructed skin by
alkaline gel electrophoresis and radioimmunoassay methods, was also used to eval-
uate chemical or physical sunscreens. This allowed the determination of a DNA
protection factor (DNA-PF) defined as the frequency of lesions induced in unpro-
tected reconstructed skin divided by the frequency of lesions induced in sunscreen-
protected samples. Results suggested that, using this method, a 1-2 DNA-PF would
correspond to an SPF 30 sunscreen [22, 104].
12 Organotypic Models for Evaluating Sunscreens 215
Fig. 12.4 Thymine dimers DNA lesions used for the evaluation of UVB photoprotection. Without
prior sunscreen application, thymine dimers are immediately formed after exposure to pure UVB
(50 mJ/cm2) as revealed by the H3 antibody. When a sunscreen absorbing in the UVB range is
applied prior to UVB exposure, the DNA lesions can be prevented. Note that the highest dose able
to prevent thymine dimer formation is fivefold the biologically efficient dose, in line with the SPF
value of 5.8 (Adapted from Bernerd et al. [18])
agreement with their similar SPF values. In contrast, following UVA or UV-SSR
exposure, dose-response experiments showed that the sunscreen with the highest
UVA-PF provided a better protection with regard to dermal damage, as compared to
the other one (Fig. 12.5) [19]. These results pointed out that the SPF value is, per se,
not sufficient to reflect the efficiency of sunscreens over the entire solar UV spec-
trum and against the major biological damage induced by sun exposure. These
in vitro results were also in agreement with an in vivo study using the same sun-
screen products showing a higher efficacy of the product having a UVB-UVA bal-
anced absorption profile [108].
Because of the major contribution of UVA rays in daily UV radiation exposure
[79], the importance of UVA absorption by sunscreens was also demonstrated under
a non-zenithal UV exposure condition. Two commercial sunscreens with similar
SPF values (approx. 15) but with different absorption profiles in the UVA range
were tested on reconstructed skins exposed to DUVR. The sunscreen formulation
with the highest UVA-PF afforded a better protection of dermal damage such as
fibroblast disappearance and MMP1 release than the other one. To test if a highest
SPF could compensate a low UVA-PF, the protection against DUVR-induced dam-
age of two other sunscreen products were compared: one product having an SPF of
27 and a low UVA absorption and the other having an SPF of 18 and a well-balanced
UVB-UVA absorption profile. The study of prevention against dermal alterations
indicated that a higher SPF value did not compensate for low UVA filtration, the
SPF18 product with well-balanced UVA-UVB absorption being more effective than
the SPF27 product [71].
Endpoints related to oxidative stress have been used to study UVA or UVB+UVA
photoprotection, such as protein and lipid oxidation and antioxidant depletion.
However, protective effects of sunscreens were not fully evidenced using these
endpoints, partly due to the difficulty of spreading the cream onto reconstructed
skin samples [24].
The importance of sunscreen photostability in photoprotection has also been
addressed using a full-thickness model of reconstructed skin. A photostable
sunscreen formulation was compared to a photounstable formulation after topical
application on reconstructed skin further exposed to UVA or to UV-SSR. The results
evidenced that only the photostable product ensured an efficient photoprotection
against UVA or UV-SSR dermal damage and MMP1 production [84].
In order to assess some biological endpoints that could be related to other
clinical consequences of UV exposure such as pigmentation or photoimmuno-
suppression, photoprotection against UV-induced pigmentation or UV-induced
alterations of Langerhans cells can be tested in adapted 3-D models (Fig. 12.6).
For example, the application of the broad-spectrum absorber MexorylTM SX on
a model of reconstructed pigmented epidermis was able to prevent UV-induced
pigmentation as visually assessed and by measuring the luminance L* factor
[39]. In addition, in 3-D models comprising Langerhans cells, the same filter
preserved the morphology and the number of Langerhans cells under UV-SSR
exposure [39, 44].
12 Organotypic Models for Evaluating Sunscreens 217
Fig. 12.5 Dermal fibroblast alterations and disappearance used for evaluating UVA photoprotection.
Sunscreen A or sunscreen B was applied onto full-thickness skins prior to exposure to the same
dose of UV-SSR. Whereas sunscreen A, having the higher UVA protection factor (UVA-PF), is
able to protect the skin against dermal damage, sunscreen B is not efficient in preventing the
dermal fibroblasts disappearance (oval). No epidermal changes were observed in both cases under
such a UV-SSR exposure due to the same SPF value of both sunscreens products (Adapted from
Bernerd et al. [19])
Fig. 12.6 Reconstructed epidermis containing Langerhans cells or melanocytes for the evaluation
of photoprotection. (a) Epidermal sheets stained with anti-Langerin antibody. In control (Ctrl)
sample, the Langerhans cells are randomly distributed throughout the epidermis while in the
UV-SSR-exposed sample, Langerhans cells are sparse, and the remaining cells display a round and
non-dendritic morphology. Application of the sunscreen before UV-SSR exposure prevented these
alterations. (b) Reconstructed epidermis (DED model) containing melanocytes. The pigmentation
is increased after exposure to UV-SSR, but this increase can be limited when the sunscreen is
applied onto the sample prior to UV-SSR exposure. Pigmentation is assessed macroscopically and
by measuring the luminance L*parameter (Adapted from Facy et al. [44] and Duval et al. [39]).
MSX MexorylTM SX
modulation of gene expression in both cell types (32 modulated genes in fibroblasts
and 44 in keratinocytes). The modulated genes were involved in ECM homeostasis,
oxidative stress, heat shock response, cell growth, inflammation, and epidermal dif-
ferentiation. Application of sunscreen on reconstructed skin before UV exposure
mitigated these effects, with a reduction in the number of modulated genes (4 modu-
lated genes in fibroblasts and 11 in keratinocytes) and in the intensity of modulation
of the residual modulated genes (Fig. 12.7). The UVA-induced release of MMP1
protein and proinflammatory cytokines in the culture medium was also alleviated by
using the sunscreen (Fig. 12.7). Prevention of gene expression modulation incurred
by the sunscreen was confirmed in human skin in vivo by quantifying the expression
of five genes involved in oxidative stress response and photoaging (HO-1, SOD2,
GPX, CAT and MMP1), reinforcing the relevance of using the 3-D model to test
photoprotection on such endpoints [78].
Gene expression analysis in fibroblasts and keratinocytes of reconstructed skin
was also used to assess photoprotection efficiency of a broad-spectrum sunscreen
(SPF 13, UVA-PF (PPD) 10.5) against DUVR. Again, this method demonstrated the
protection afforded by the sunscreen, with very close gene expression profiles
between unexposed samples and DUVR exposed but protected samples, as shown
by hierarchical clustering, a decreased number of modulated genes, and a decrease
in intensity of gene modulation for the residual modulated genes [82]. Thus, gene
expression profiling constitutes a complementary approach to histological and bio-
chemical studies for assessing of photoprotection in 3-D skin models.
12 Organotypic Models for Evaluating Sunscreens 219
Fig. 12.7 The use of gene and protein expression for the evaluation of sunscreen efficacy. (a)
Modulation levels of gene expression in fibroblasts of reconstructed skin exposed to UVA. Bars
represent modulation ratios after UVA exposure for each studied transcript. Positive and negative
values denote up- and downregulation of gene expression, respectively. Note the flatter aspect of
the modulation profile when the broad-spectrum sunscreen was used (lower panel), as compared
to unprotected samples (upper panel). (b) Examples of gene and soluble-protein expression after
UVA exposure of reconstructed skin in the presence or absence of broad-spectrum sunscreen
(Adapted from Marionnet et al. [78])
12.5 Conclusion
The development of organotypic skin models has made possible the in vitro assess-
ment of the effects of different types of UV exposure (UVB, UVA, or solar simulation)
in a three-dimensional context and in a cutaneous structure including different types of
skin cells. Reconstructed skin appeared to be useful, showing predictive responses
with numerous biological endpoints closely related to in vivo clinical data. It also
allowed to increase the knowledge on the precise biological processes involved and
can therefore be used to study the photoprotection afforded by sunscreens in vitro,
providing additional biological data on sunscreen efficacy, complementing the in vivo
protection factors (SPF or UVA-PF). Although these 3-D models did not follow a strict
validation process for protection factor determination, they evidenced important con-
cepts in photoprotection, such as the need of using a well-balanced photostable sun-
screen absorbing over the entire UV spectrum of solar radiation for preserving essential
biological functions. They also revealed from a biological point of view the limits of
the SPF value for predicting the level of protection in the UVA range. The combination
of 3-D skin models and new biological approaches such as transcriptomic or proteomic
will indisputably increase the added value of such systems for evaluating sunscreen.
220 C. Marionnet and F. Bernerd
References
20. Bessou S, Surlve-Bazeille JE, Pain C et al (1996) Ex vivo study of skin phototypes. J Invest
Dermatol 107:684688
21. Bessou S, Surlve-Bazeille JE, Sorbier E et al (1995) Ex vivo reconstruction of the epidermis
with melanocytes and the influence of UVB. Pigment Cell Res 8:241249
22. Bissonauth V, Drouin R, Mitchell DL et al (2000) The efficacy of a broad-spectrum sunscreen
to protect engineered human skin from tissue and DNA damage induced by solar ultraviolet
exposure. Clin Cancer Res 6:41284135
23. Blumenberg M (2006) DNA microarrays in dermatology and skin biology. OMICS 10:
243260
24. Cario-Andr M, Briganti S, Picardo M et al (2002) Epidermal reconstructs: a new tool to
study topical and systemic photoprotective molecules. J Photochem Photobiol B 68:7987
25. Chen VL, Fleischmajer R, Schwartz E et al (1986) Immunochemistry of elastotic material in
sun-damaged skin. J Invest Dermatol 87:334337
26. Christiaens FJ, Chardon A, Fourtanier A et al (2005) Standard ultraviolet daylight for nonex-
treme exposure conditions. Photochem Photobiol 81:874878
27. Clausen BE, Kel JM (2010) Langerhans cells: critical regulators of skin immunity? Immunol
Cell Biol 88:351360
28. Clydesdale GJ, Dandie GW, Muller HK (2001) Ultraviolet light induced injury: immunologi-
cal and inflammatory effects. Immunol Cell Biol 79:547568
29. Cohen C, Roguet R, Cottin M (1998) The use of a reconstructed epidermis in the evaluation
of protective effect of sunscreens against chemical phototoxicity by UVA. In: Rougier A,
Schaefer H (eds) Protection of the skin against utraviolet radiations. John Libbey Eurotext,
Paris, pp 195199
30. Cole C, VanFossen R (1992) Measurement of sunscreen UVA protection: an unsensitized
human model. J Am Acad Dermatol 26:178184
31. Cooper KD, Oberhelman L, Hamilton TA et al (1992) UV exposure reduces immunization
rates and promotes tolerance to epicutaneous antigens in humans: relationship to dose,
CD1a-DR+ epidermal macrophage induction, and Langerhans cell depletion. Proc Natl Acad
Sci U S A 89:84978501
32. Corsini E, Sangha N, Feldman SR (1997) Epidermal stratification reduces the effects of UVB
(but not UVA) on keratinocyte cytokine production and cytotoxicity. Photodermatol
Photoimmunol Photomed 13:147152
33. Cotovio J, Pellevoisin C, Parsad D (2014) In vitro skin models. In: Srinivas CR, Verschoore
M (eds) Basic science for modern cosmetic dermatology. Jaypee Brothers Medical Publishers,
New Delhi, pp 2546
34. Coulomb B, Lebreton C, Mathieu N et al (1996) UVA-induced oxidative damage in fibro-
blasts cultured in a 3-dimensional collagen matrix. Exp Dermatol 5:161167
35. Dehaven C, Hayden PJ, Armento A, Oldach J (2014) DNA photoprotection conveyed by
sunscreen. J Cosmet Dermatol 13:99102
36. Dekker P, Parish WE, Green MR (2005) Protection by food-derived antioxidants from
UV-A1-induced photodamage, measured using living skin equivalents. Photochem Photobiol
81:837842
37. Duval C, Chagnoleau C, Pouradier F et al (2012) Human skin model containing melanocytes:
essential role of keratinocyte growth factor for constitutive pigmentation-functional response
to alpha-melanocyte stimulating hormone and forskolin. Tissue Eng Part C Methods 18:
947957
38. Duval C, Regnier M, Schmidt R (2001) Distinct melanogenic response of human melano-
cytes in mono-culture, in co-culture with keratinocytes and in reconstructed epidermis, to UV
exposure. Pigment Cell Res 14:348355
39. Duval C, Schmidt R, Regnier M et al (2003) The use of reconstructed human skin to evaluate
UV-induced modifications and sunscreen efficacy. Exp Dermatol 12 Suppl 2:6470
40. Egles C, Garlick JA, Shamis Y (2010) Three-dimensional human tissue models of wounded
skin. Methods Mol Biol 585:345359
222 C. Marionnet and F. Bernerd
41. Enk CD, Shahar I, Amariglio N et al (2004) Gene expression profiling of in vivo UVB-
irradiated human epidermis. Photodermatol Photoimmunol Photomed 20:129137
42. Eungdamrong NJ, Higgins C, Guo Z et al (2014) Challenges and promises in modeling
dermatologic disorders with bioengineered skin. Exp Biol Med (Maywood)
239:12151224
43. Evans-Johnson JA, Garlick JA, Johnson EJ et al (2013) A pilot study of the photoprotective
effect of almond phytochemicals in a 3D human skin equivalent. J Photochem Photobiol B
126:1725
44. Facy V, Flouret V, Rgnier M et al (2005) Reactivity of Langerhans cells in human recon-
structed epidermis to known allergens and UV radiation. Toxicol In Vitro 19:787795
45. Fagot D, Asselineau D, Bernerd F (2002) Direct role of human dermal fibroblasts and indirect
participation of epidermal keratinocytes in MMP-1 production after UV-B irradiation. Arch
Dermatol Res 293:576583
46. Fagot D, Asselineau D, Bernerd F (2004) Matrix metalloproteinase-1 production observed
after solar-simulated radiation exposure is assumed by dermal fibroblasts but involves a
paracrine activation through epidermal keratinocytes. Photochem Photobiol 79:499505
47. Fernandez TL, Van Lonkhuyzen DR, Dawson RA et al (2014) Characterization of a human
skin equivalent model to study the effects of ultraviolet B radiation on keratinocytes. Tissue
Eng Part C Methods 20:588598
48. Fisher GJ, Datta SC, Talwar HS et al (1996) Molecular basis of sun-induced premature skin
ageing and retinoid antagonism. Nature 379:335339
49. Flamand N, Marrot L, Belaidi JP et al (2006) Development of genotoxicity test procedures
with Episkin, a reconstructed human skin model: towards new tools for in vitro risk
assessment of dermally applied compounds? Mutat Res 606:3951
50. Freeman SE, Ley RD, Ley KD (1988) Sunscreen protection against UV-induced pyrimidine
dimers in DNA of human skin in situ. Photodermatol 5:243247
51. Gelis C, Mavon A, Vicendo P (2003) Evaluation of biological effects of ultraviolet radiations
and of photoprotection on human reconstructed epidermis. Nouvelles Dermatol 22:351
52. Gibbs S, Murli S, De Boer G et al (2000) Melanosome capping of keratinocytes in pigmented
reconstructed epidermiseffect of ultraviolet radiation and 3-isobutyl-1-methyl-xanthine on
melanogenesis. Pigment Cell Res 13:458466
53. Grothmann K, Kaase H (1993) Testung von Lichtschutzmitteln: Vorschlag zur Definition
einer Referenz-Spektralverteilung fr UV-Sonnensimulatoren (Sun protection measurement:
proposal for a definition of a UV solar simulator standard spectrum). Dermatol Monatsschr
179:108111
54. Gruber F, Oskolkova O, Leitner A et al (2007) Photooxidation generates biologically active
phospholipids that induce heme oxygenase-1 in skin cells. J Biol Chem 282:1693416941
55. Haake AR, Polakowska RR (1995) UV-induced apoptosis in skin equivalents: inhibition by
phorbol ester and Bcl-2 overexpression. Cell Death Differ 2:183193
56. Haake A, Scott GA, Holbrook KA (2001) Structure and function of the skin: overview of the
epidermis and dermis. In: Freinkel RK, Woodley DT (eds) The biology of the skin. Parthenon
Publishing Group Limited, Lancs, pp 1945
57. Halliday GM (2005) Inflammation, gene mutation and photoimmunosuppression in response to
UVR-induced oxidative damage contributes to photocarcinogenesis. Mutat Res 571:107120
58. Halliday GM, Byrne SN, Damian DL (2011) Ultraviolet A radiation: its role in immunosup-
pression and carcinogenesis. Semin Cutan Med Surg 30:214221
59. Halliday GM, Cadet J (2012) Its all about position: the basal layer of human epidermis is
particularly susceptible to different types of sunlight-induced DNA damage. J Invest Dermatol
132:265267
60. Harriger MD, Hull BE (1994) Characterization of ultraviolet radiation-induced damage to
keratinocytes in a skin equivalent in vitro. Arch Dermatol Res 286:319324
61. Haywood R, Volkov A, Andrady C et al (2012) Measuring sunscreen protection against solar-
simulated radiation-induced structural radical damage to skin using ESR/spin trapping:
development of an ex vivo test method. Free Radic Res 46:265275
12 Organotypic Models for Evaluating Sunscreens 223
62. Hensbergen PJ, Alewijnse AE, Kempenaar J et al (2005) Proteomic profiling identifies an
UV-induced activation of cofilin-1 and destrin in human epidermis. J Invest Dermatol
124:818824
63. Huang XX, Bernerd F, Halliday GM (2009) Ultraviolet A within sunlight induces mutations
in the epidermal basal layer of engineered human skin. Am J Pathol 174:15341543
64. Javeri A, Huang XX, Bernerd F et al (2008) Human 8-oxoguanine-DNA glycosylase 1 pro-
tein and gene are expressed more abundantly in the superficial than basal layer of human
epidermis. DNA Repair (Amst) 7:15421550
65. Kochevar I, Taylor R, Krutmann J (2008) Fundamentals of cutaneous photobiology and pho-
toimmunology. In: Wolff K, Goldsmith LA, Katz SI (eds) Fitzpatricks dermatology in gen-
eral medicine, 7th edn. McGraw-Hill, New York, pp 797808
66. Kurdykowski S, Mine S, Bardey V et al (2011) Ultraviolet-B irradiation induces differential
regulations of hyaluronidase expression and activity in normal human keratinocytes.
Photochem Photobiol 87:11051112
67. Kypriotou M, Huber M, Hohl D (2012) The human epidermal differentiation complex: cornified
envelope precursors, S100 proteins and the fused genes family. Exp Dermatol 21:643649
68. Lavker RM, Veres DA, Irwin CJ et al (1995) Quantitative assessment of cumulative damage
from repetitive exposures to suberythemogenic doses of UVA in human skin. Photochem
Photobiol 62:348352
69. Lee SJ, Jung TH, Kim H et al (2014) Inhibition of c-Kit signaling by diosmetin isolated from
Chrysanthemum morifolium. Arch Pharm Res 37:175185
70. Lehmann B, Genehr T, Knuschke P et al (2001) UVB-induced conversion of 7-dehydrocho-
lesterol to 1, 25- dihydroxyvitamin D3 in an in vitro human skin equivalent model. J Invest
Dermatol 117:11791185
71. Lejeune F, Christiaens F, Bernerd F (2008) Evaluation of sunscreen products using a recon-
structed skin model exposed to simulated daily ultraviolet radiation: relevance of filtration
profile and SPF value for daily photoprotection. Photodermatol Photoimmunol Photomed
24:249255
72. Ley RD, Fourtanier A (1997) Sunscreen protection against ultraviolet radiation-induced
pyrimidine dimers in mouse epidermal DNA. Photochem Photobiol 65:10071011
73. Lin JY, Fisher DE (2007) Melanocyte biology and skin pigmentation. Nature 445:843850
74. Lowe NJ, Meyers DP, Wieder JM et al (1995) Low doses of repetitive ultraviolet A induce
morphologic changes in human skin. J Invest Dermatol 105:739743
75. MacNeil S (2007) Progress and opportunities for tissue-engineered skin. Nature 445:
874880
76. Mahns A, Wolber R, Stab F et al (2004) Contribution of UVB and UVA to UV-dependent
stimulation of cyclooxygenase-2 expression in artificial epidermis. Photochem Photobiol Sci
3:257262
77. Maresca V, Flori E, Briganti S et al (2006) UVA-induced modification of catalase charge
properties in the epidermis is correlated with the skin phototype. J Invest Dermatol
126:182190
78. Marionnet C, Grether-Beck S, Seite S et al (2011) A broad-spectrum sunscreen prevents UVA
radiation-induced gene expression in reconstructed skin in vitro and in human skin in vivo.
Exp Dermatol 20:477482
79. Marionnet C, Lejeune F, Pierrard C et al (2012) Biological contribution of UVA wavelengths
in non extreme daily UV exposure. J Dermatol Sci 66:238240
80. Marionnet C, Pierrard C, Golebiewski C et al (2014) Diversity of biological effects induced
by Longwave UVA rays (UVA1) in reconstructed skin. PLoS One 9:e105263
81. Marionnet C, Pierrard C, Lejeune F et al (2010) Different oxidative stress response in kerati-
nocytes and fibroblasts of reconstructed skin exposed to non extreme daily-ultraviolet radia-
tion. PLoS One 5:e12059
82. Marionnet C, Pierrard C, Lejeune F et al (2012) Modulations of gene expression induced by
daily ultraviolet light can be prevented by a broad spectrum sunscreen. J Photochem Photobiol
B 116:3747
224 C. Marionnet and F. Bernerd
83. Marionnet C, Tricaud C, Bernerd F (2014) Exposure to non-extreme solar UV daylight: spec-
tral characterization, effects on skin and photoprotection. Int J Mol Sci 16:6890
84. Marrot L, Beladi JP, Lejeune F et al (2004) Photostability of sunscreen products influences
the efficiency of protection with regard to UV-induced genotoxic or photoageing-related
endpoints. Br J Dermatol 151:12341244
85. Marrot L, Planel E, Ginestet AC et al (2010) In vitro tools for photobiological testing: molec-
ular responses to simulated solar UV of keratinocytes growing as monolayers or as part of
reconstructed skin. Photochem Photobiol Sci 9:448458
86. Meloni M, Farina A, de Servi B (2010) Molecular modifications of dermal and epidermal
biomarkers following UVA exposures on reconstructed full-thickness human skin. Photochem
Photobiol Sci 9:439447
87. Moyal D, Chardon A, Kollias N (2000) Determination of UVA protection factors using the
persistent pigment darkening (PPD) as the end point. (Part 1). Calibration of the method.
Photodermatol Photoimmunol Photomed 16:245249
88. Moysan A, Clement-Lacroix P, Michel L et al (1996) Effects of ultraviolet A and antioxidant
defense in cultured fibroblasts and keratinocytes. Photodermatol Photoimmunol Photomed
11:192197
89. Muller HK, Woods GM (2013) Ultraviolet radiation effects on the proteome of skin cells.
Adv Exp Med Biol 990:111119
90. Nakazawa K, Nakazawa H, Sahuc F et al (1997) Pigmented human skin equivalent: new
method of reconstitution by grafting an epithelial sheet onto a non-contractile dermal
equivalent. Pigment Cell Res 10:382390
91. Neale R, Russell A, Muller HK et al (1997) Sun exposure, sunscreen and their effects on
epidermal Langerhans cells. Photochem Photobiol 66:260264
92. Nelson D, Gay RJ (1993) Effects of UV irradiation on a living skin equivalent. Photochem
Photobiol 57:830837
93. Nix TE Jr (1967) Ultraviolet-induced changes in epidermis. In: Zelickson AS (ed)
Ultrastructure of normal and abnormal skin. Lea & Febiger, Philadelphia, pp 304319
94. Nix TE Jr, Nordquist RE, Scott JR et al (1965) Ultrastructural changes induced by ultraviolet
light in human epidermis: basal and spinous layers. J Invest Dermatol 45:5264
95. Obi-Tabot ET, Tian XQ, Chen TC et al (2000) A human skin equivalent model that mimics
the photoproduction of vitamin D3 in human skin. In Vitro Cell Dev Biol 36:201204
96. Olsen WM (1988) Early cell kinetic effects of a single dose of monochromatic ultraviolet B
irradiation on hairless mouse epidermis. J Invest Dermatol 91:585589
97. Pathak MA, Fanselow DL (1983) Photobiology of melanin pigmentation: dose/response of
skin to sunlight and its contents. J Am Acad Dermatol 9:724733
98. Podda M, Traber MG, Weber C et al (1998) UV-irradiation depletes antioxidants and causes
oxidative damage in a model of human skin. Free Radic Biol Med 24:5565
99. Proksch E, Brandner JM, Jensen JM (2008) The skin: an indispensable barrier. Exp Dermatol
17:10631072
100. Prunieras M (1979) Epidermal cell cultures as models for living epidermis. J Invest Dermatol
73:135137
101. Rasmussen C, Gratz K, Liebel F et al (2010) The StrataTest(R) human skin model, a consis-
tent in vitro alternative for toxicological testing. Toxicol In Vitro 24:20212029
102. Reece BT, Deeds D, Rozen M (1992) As an in vitro method for screening sunscreen formula-
tions for sun protection factor using a full-thickness skin model. J Soc Cosmet Chem
43:307312
103. Robert M, Bissonauth V, Ross G et al (1999) Harmful effects of UVA on the structure and
barrier function of engineered human cutaneous tissues. Int J Radiat Biol 75:317326
104. Rouabhia M, Mitchell DL, Rhainds M et al (2002) A physical sunscreen protects engineered
human skin against artificial solar ultraviolet radiation-induced tissue and DNA damage.
Photochem Photobiol Sci 1:471477
12 Organotypic Models for Evaluating Sunscreens 225
M.Sohn
Key Points
The paradox of achieving great SPF values while using small amounts of
UV filters explains the high interest in boosting the performance of the UV
filter combination. As a prerequisite for performance, the UV filters must
be uniformly dispersed and/or solved at first in the emulsion and then in the
applied sunscreen film on the skin.
Boosting the photoprotection is possible either by optimizing the efficacy
of the UV-absorbing system or by improving the film-forming properties
of the product during spreading.
The optimization of the performance of the UV filtering system includes
the combination of UVB- and UVA-absorbing molecules, the consider-
ation of the photostability of the UV filters individually and in combina-
tion, as well as the synergy of water- and oil-dispersed UV filters. The
addition of scattering particles was also shown to increase the efficiency of
the UV filter system by increasing the optical path length.
The improvement of film-forming properties and distribution of the UV
molecules on the skin can be achieved by the addition of film formers, the
choice of the sunscreen vehicle, and its viscosity.
M. Sohn
Global Technical Center Sun Care, BASF Grenzach GmbH,
Grenzach-Whylen, Germany
School of Life Sciences, Institute of Pharmaceutical Technology, University of Applied
Sciences and Arts Northwestern Switzerland, Muttenz/Basel, Switzerland
e-mail: [email protected]
13.1 Introduction
1.00
0.80
Arbitrary units
0.60
0.40
Erythemal action spectrum
Solar spectral irradiance
0.20
Erythemal effectiveness spectrum
0.00
290 300 310 320 330 340 350 360 370 380 390 400
Wavelength (nm)
Fig. 13.1 Erythemal action spectrum [6], solar spectral irradiance [7], and erythema effectiveness
spectrum, in arbitrary units. Approximately 85% of erythema originates from UVB radiation, a
non-negligible part also from UVA radiation
benzoate (DHHB)
calculated using the BASF
6
sunscreen simulator [10, 11]
0
0 % DHHB 5 % DHHB
The addition of UVA filters are, therefore, a prerequisite to increase SPF values
[8]. Indeed, a mere UVB sunscreen that would solely block radiations from 290
to 320nm and transmit UVA radiation would reach in principle a maximum SPF of
11 only [9], because continuous level of erythemally active UVA2 radiation is trans-
mitted. As a consequence, the addition of UVA filters is a requisite to obtain a sub-
stantial rise of the SPF value.
Figure 13.2 gives the SPF in silico value of an oil-in-water (O/W) sunscreen
combination containing 2% of the UVB filter octyl triazone without and with the
UVA filter having the INCI diethylamino hydroxybenzoyl hexyl benzoate (DHHB).
Accordingly, the basic for sunscreen development and achievement of higher SPFs
is the judicious combination of UVB and UVA filters. The presence of UVA filters is
230 M. Sohn
nowadays generally the case since UVA filters are incorporated to reach the minimal
UVA protection that is required in most regions [7]. Therefore, an appropriate UV filter
system should combine UVB and UVA filters to achieve optimized UV shield [12].
Besides their individual absorbance profile and extinction properties, UV filters are
characterized also by their intrinsic photostability and photocompatibility with
other UV filters. The two worldwide accepted UVA filter avobenzone and UVB
filter octinoxate are known to be very photounstable under UV exposure [13],
resulting in a loss of approximately 70% and 40% after ten Minimal Erythemal
Dose (MED) for avobenzone and octinoxate, respectively [14]. Moreover, their
combination leads to an increased photochemical instability due to a 2+2-hetero-
photocycloaddition [15] producing non-UV-absorbing cyclobutylketone
photoproducts. This issue often obliged sunscreen manufacturers to use either the
one or the other filter in their sunscreen development.
Regarding octinoxate, it undergoes at first a trans-cis photoisomerization that
equilibrates rapidly after UV irradiation [16]. Upon further UV irradiation, the
molecule undergoes a irreversible 2+2-homo-photocycloaddition resulting in non-
UV-absorbing cinnamate dimers [15, 17]. In the case of avobenzone, an equilibrium
mixture between the two tautomeric enol and keto forms of the molecule is present
[15, 18, 19], the enol tautomer being involved in the irreversible photocycloaddition
with octinoxate when the two filters are combined. Further, upon UV irradiation the
enol form is photoisomerized into the photoreactive keto isomer that achieves a
triplet excited state. This state is responsible for the irreversible photodegradation of
the molecule via a Norrish type I cleavage of the CO-C bond, resulting in the
formation of two radicals that can further react and form photoproducts [20]. The
complete photodegradation process of avobenzone was proposed elsewhere [15, 18].
To slow down the formation of the keto form and subsequent consequences, the
simple addition of other UV filters may compete with avobenzone for absorbing
light, thus delaying the formation of the excited keto triplet state [21]. This is,
however, only a partial protection of avobenzone and cannot avoid the generation of
excited state molecules. To overcome this limitation in photoinstability issue, some
ingredients were found to show quenching properties of the excited state to prevent
from photodegradation of the excited molecule.
For photostable UV filters, the dissipation of absorbed energy occurs through
internal conversion, and the absorbed energy is then released in the form of heat due
to an intramolecular hydrogen transfer [1]. However, in the case of the photounstable
UV filter avobenzone, the molecule can perform an intersystem crossing from the
singlet exited state to the triplet excited state, the latter showing a longer lifetime
and therefore promoting photodegradation as mentioned above. As a consequence,
the stabilization of photounstable UV filters such as avobenzone is possible either
by quenching the excited singlet state to avoid the formation of the triplet excited
13 UV Booster andPhotoprotection 231
Octocrylene
400
Octisalate
Polyester-8
200 Butyloctyl salicylate
150
100
50
0
290 310 330 350 370 390
Wavelength (nm)
state or by quenching the formed triplet excited state. Triplet-triplet energy transfer
from the photounstable molecule to the quencher molecule is the most common
energy transfer mechanism for photostabilization. To make this process work, the
quencher molecule must show a similar energy level to that of the photoexcited state
of the photounstable molecule to absorb the excitation energy. The quencher mole-
cule should then ideally return intact to its ground state without self-degradation. As
an example, avobenzone shows a triplet energy level close to 60kcal/mol [22].
Efficient triplet quenchers of avobenzone include other UV filters, particularly
bemotrizinol [13] and octocrylene [14], the latter showing a triplet energy level of
5560kcal/mol [23] close to that of avobenzone. The two bemotrizinol [13] and
octocrylene [14] UV filters were shown to raise significantly the photostability of
avobenzone; the recovery of 1% avobenzone equals 80% after 10MED when com-
bined with 3% bemotrizinol compared to a recovery of 25% only without bemotriz-
inol [14]. In addition to UV filters, some emollients are promoted to show triplet
quenching efficacy as well. Examples of such compounds are diethylhexyl 2,6-naph-
thalate [24], butyloctyl salicylate, tridecyl salicylate, polyester-8 [25], diethylhexyl
syringylidene malonate [26], benzotriazolyl dodecyl p-cresol, and undecylcrylene
dimethicone. Recently, the compound ethyhexyl methoxycrylene [27, 28] was
introduced for its ability to quench the singlet excited state to avoid the transfer of
the molecule from the singlet to its triplet state. To be an effective excited state
quencher of avobenzone, these ingredients most often show an inherent UV
absorbance as depicted in Fig.13.3 in terms of specific extinction E1,1. E1,1 is the
232 M. Sohn
Emulsions are the main formulation type for sunscreen products counting for more
than 80% of the launched sunscreens in 20122013 worldwide [33]. Most of the
registered UV filters are oil-soluble and will, thus, be formulated into the oil phase
of the emulsion. In case of a water-in-oil (W/O) emulsion, the UV filters are distrib-
uted in the continuous oil phase directly in contact with the skin during spreading,
forming a good coverage and subsequently a uniform protective film. This may
explain why W/O sunscreens produce greater SPF values [8, 34]. In the contrary, in
oil-in-water (O/W) emulsions, the most popular emulsion type, the filters are distrib-
uted in the internal oil phase that hinders the achievement of a uniform distribution
of the UV filters after spreading. To visualize this phenomenon, an O/W emulsion
containing the oil-dispersed dye Sudan red III pigment (color index 26100) and the
water-dispersed blue pigment with the color index 42090 was observed under light
microscopy, before (Fig.13.4a), during (Fig.13.4b), and after spreading (Fig.13.4c).
Before spreading, the oil phase is contained inside the droplets (Fig.13.4a).
During spreading, the droplets merge, and the oil phase is released (Fig.13.4b);
finally, after spreading, the water evaporated, and the oil phase is predominant.
Nevertheless, a nonvolatile part of the water phase remains in the film, shown as
blue spots in Fig.13.4c. The residual water parts lack UV filters and offer no
protection, resulting in unprotected area or holes in the protection film and, thus,
in reduced UV performance. To overcome this drawback, water- and oil-soluble UV
filters may be incorporated in the two phases of the emulsion. This leads to an
enhanced efficacy as the nonvolatile water part remaining after water evaporation is
protected with the water-dispersed UV filter. The resulting sunscreen film will not
13 UV Booster andPhotoprotection 233
a b
Fig. 13.4 Microscopic evaluation (Olympus CKX41) of an O/W emulsion containing a red oil-
and a blue water-dispersed pigment, (a) before spreading, (b) during spreading, and (c) after
spreading. Not protecting the water part (in blue) with a water-dispersed UV filter would result in
unprotected area or holes in the sunscreen film and reduced UV efficacy
show any unprotected area and will end up in a better coverage and optimized UV
protection.
Further, the effect of the UV filter distribution in the oil and in the water phase
on the UV performance was investigated by Neuenschwander and Herzog [35]. The
Colipa P3 standard formulation containing a mixture of oil- and water-dispersed
UV filters was used [6]; the ratio between the water filter ensulizole and the oil filter
octinoxate was varied to cover a Relative Erythema Active Extinction in the oil
phase (REAE) between almost 0 and 1. A REAE of 1 corresponds to a UV filter
system based on oil-soluble UV filters exclusively, and to the opposite, a REAE of
0 corresponds to a UV filter system based on water-dispersed UV filters solely. The
SPF invitro of each formulation variant was measured and plotted against its REAE
value (Fig.13.5). In addition, the effect of the water and oil UV filter distribution on
the film irregularity, subsequently on the UV performance, was computed. To
describe film irregularity, the calibrated quasi-continuous step film model was used
[10] and is given in following exponential equation:
i c
- B
h ( i ) = A exp n (13.1)
234 M. Sohn
20
15
SPF
10
SPF in vitro
0
0 0.2 0.4 0.6 0.8 1
Fig. 13.5 Variation of the SPF in silico (solid line) considering the effect of oil-water distribution,
SPF invitro (circles, n=96 measurements per formulation) as function of REAE of the UV filter
mixture; dashed line is the SPF simulation without consideration of the oil-water distribution effect
where, h(i) is the height of the film at step i, with i=1, 2,., n, where n is the num-
ber of steps the exponential function is divided into, B and C are parameters deter-
mining the shape of the film, and A is introduced for normalization. The transmission
through the quasi-continuous step film is obtained as the sum of the transmissions
through all steps of height h(i).
Using UV filters in the two phases of the sunscreen formulation, REAE between
0.4 and 0.8 appears to enhance the overall UV performance.
The scattering efficiency highly depends on the particle size range relative to the
light wavelength. Further, for a particle with a given refractive index, the absor-
bance and scattering performance of a given particle run inversely, when varying the
size if scattering increases then absorbance decreases, and there might be an opti-
mum of particle size for a maximized UV attenuation.
Polymer spheres consisting of styrene/acrylates copolymer act as UV booster
material following this principle [36]. In its original product form as well as in the
finished formulation, the sphere is filled with water; the water evaporates during
spreading leaving an air-filled hollow sphere with an external size of approximately
325nm. Supplier of this material recommends using about 45% solids of the
spheres to achieve SPF boosting.
Interestingly, the particulate UV filters such as the inorganic UV filter titanium
dioxide or the organic UV filter bisoctrizole also show scattering properties and are,
therefore, able to amplify the UV performance of the used filtering system through
this additional characteristic. Since these particulate UV filters are basically selected
for their absorbing properties, the additional boosting of efficacy is achieved with-
out the need of a further ingredient that would have merely the scattering activity.
Some authors introduced a method to measure the scattering effect of particulate
UV filters using a spectroscopic setup [37, 38]. In these experiments, the impact of
scattering particles was shown on the absorbance of a dye having its maximum
absorption outside the UV range such as Evans Blue and Patent Blue V.The dye
concentration was maintained constant, and the variation of the dye absorbance was
monitored while varying the amount of scattering particles. Figure13.7b displays
the example of Patent Blue V dye in combination with the organic particulate UV
filter bisoctrizole. The variation of absorbance of a water dispersion of 1.32105 M
Patent Blue V with increasing volume fraction concentration of the increasing par-
ticulate filter bisoctrizole ranging from 0 to 18% was measured. The max of Patent
Blue V dye in water at pH 5 lies around 638nm, outside the absorption range of the
particulate UV filter (Fig.13.7a). The absorbance of the dye could be amplified
reaching a sixfold increase with increased concentration of scattering particles;
achieving a plateau from the incorporation of 5% particles (Fig.13.7b). A similar
boosting is produced using the organic particulate UVB filter with INCI tris-biphenyl
triazine and titanium dioxide particles.
The boosting effect obtained from ingredients with scattering characteristics can
also be shown with SPF invivo measurements. The combination of 5% of the
236 M. Sohn
Absorbance
0.10
increasing concentration of
the organic particulate
broad-spectrum 0.05
bisoctrizole UV filter
0.00
250 350 450 550 650 750 850
Wavelength [nm]
b
8
Amplification factor of absorbance
6
of Patent Blue V dye
4
Patent Blue V dye,
3
max in water, pH 5 = 638 nm
2
1
0
0 5 10 15 20
Volume fraction concentration of bisoctrizole (%)
The intrinsic absorbing properties of the selected UV filters along with their
photocompatibility are primarily responsible for the sunscreen efficacy in terms of
SPF, UVA protection, and photostability [8, 41]. Nevertheless, SPF values appear to
13 UV Booster andPhotoprotection 237
differ between sunscreens containing the same UV filter composition [8, 42].
Homogeneous distribution of the sunscreen product was shown to contribute to the
SPF invivo value [43]. For optimum performance, the sunscreen film should be of
uniform thickness, with an identical thickness over the covered surface area,
similarly to a perfect homogeneous distribution of UV filter solution in an optical
cell. Understandably, this state can never be attained under invivo conditions of
application due to the skin surface topography that preclude the formation of an
even sunscreen film [44]. Furthermore, manual application makes it nearly
impossible to reach film uniformity. The sunscreen film is composed of a multitude
of different thicknesses, and this irregularity of the film thickness may be the reason
for the discrepancy between predictions based on UV transmission of dilute
transparent filter solutions and clinical study results [45]. The objective for optimum
photoprotection is, therefore, to apply the sunscreen product as uniform as possible.
Fig. 13.8Connection
between sunscreen vehicle,
film forming, and delivered
UV protection
Applied
sunscreen film
Sunscreen
Sunscreen film
Skin substrate
Sunscreen Sunscreen
vehicle performance
viscosity sunscreens OW-S and CAS which lacked thickeners exhibited the smallest
average film thickness values that are connected to a greater occurrence of small
film thicknesses. Light transmittance which increases exponentially with decreasing
film thickness is inversely proportional to SPF.Subsequently, low viscosity sun-
screens OW-S and CAS yielded also the lowest SPF values. They may leave larger
areas of ridges virtually uncovered during application while accumulating in the
furrows thus leading to an irregular protective film and a lower SPF value. Therefore,
the presence of viscosity builder in the formulation seems indeed to be a prevailing
prerequisite for UV efficacy.
Further, WO exhibited both the largest average film thickness with a value of
2.9m and the highest SPF.In contrast to the other sunscreens, the UV filters of
WO are distributed in the continuous phase which does not evaporate, thus assum-
ing to form a uniform protecting film with the help of the thickeners.
This study demonstrated that SPF variation observed between sunscreens con-
taining the same filter system arose from the difference in their film thickness distri-
bution that depended on the sunscreen formulation.
Figure 13.8 shows the significant connections between the sunscreen vehicle-
related factor, the film forming, and the measured performance, that is, SPF invitro
of sunscreens.
Finally, Fig.13.9 summarizes the key parameters emphasized in this chapter that
are expected to boost the efficiency of a UV filtering system.
The high expectation of achieving greater UV photoprotection while using
reduced amount of UV filters related to economical, ecological-, sensorial, or
health-related reasons led to a high interest in understanding the factors and their
mechanisms able to influence the efficacy of the UV protection system. Besides the
appropriate selection of the UV filters, including adequate absorption profile,
photostability, and synergy, also the film thickness distribution on the skin is of high
relevance for UV protection. Assessing the effect of individual formulation
excipients on the film formation may offer a novel way to optimize sunscreen
242 M. Sohn
Fig. 13.9Parameters
promoting the boosting of
the performance of a UV
Importance
filtering system
of UVA
protection
Synergy of
Formation water and
of an oil
homogenous dispersed
sunscreen Boosting UV filters
film the UV
protection
Use of
Photocom-
scattering
patibility of
particles
UV filters
photoprotection during the development step. The effect of the distribution of the
UV filters within the applied sunscreen film on the delivered UV protection may be
a further factor to be elucidated.
AcknowledgmentsI would like to thank especially Uli Osterwalder of BASF Personal Care
and Nutrition GmbH for his great support and his confidence in me, Bernd Herzog and
Marcel Schnyder of BASF Grenzach GmbH for their valuable input, and Georgios Imanidis
of the Institute of Pharmaceutical Technology, School of Life Sciences, University of Applied
Sciences Northwestern, Switzerland, for the fruitful discussions.
References
1. Herzog B, Huglin D, Borsos E, Stehlin A, Luther H (2004) New UV absorbers for cosmetic
sunscreens a breakthrough for the photoprotection of human skin. Chimia
58(78):554559
2. de Groot AC, Roberts DW (2014) Contact and photocontact allergy to octocrylene: a review.
Contact Dermatitis 70(4):193204
3. Avenel-Audran M, Dutartre H, Goossens A, Jeanmougin M, Comte C, Bernier C etal (2010)
Octocrylene, an emerging photoallergen. Arch Dermatol 146(7):753757
4. Matsumoto H, Adachi S, Suzuki Y (2005) Estrogenic activity of ultraviolet absorbers and the
related compounds. Yakugaku Zasshi JPharm Soc Jpn 125(8):643652
5. Axelstad M, Boberg J, Hougaard KS, Christiansen S, Jacobsen PR, Mandrup KR etal (2011)
Effects of pre- and postnatal exposure to the UV-filter octyl methoxycinnamate (OMC) on the
reproductive, auditory and neurological development of rat offspring. Toxicol Appl Pharmacol
250(3):278290
6. ISO 24444:2010 Cosmetics sun protection test methods invivo determination of the sun
protection factor (SPF) (2010). http://www.iso.org/iso/catalogue_detail.htm?csnumber=46523
(accessed 2 February 2016)
13 UV Booster andPhotoprotection 243
51. Van Reeth I, Postiaux S, Van Dort H (2006) Silicones bring multifunctional performance to
sun care. Cosmetics & Toiletries magazine 121(10):4154
52. Lademann J, Schanzer S, Richter H, Pelchrzim RV, Zastrow L, Golz K etal (2004) Sunscreen
application at the beach. JCosmet Dermatol 3(2):6268
53. Rhodes LE, Diffey BL (1997) Fluorescence spectroscopy: a rapid, noninvasive method for
measurement of skin surface thickness of topical agents. Br JDermatol 136(1):1217
Chapter 14
Sunscreen Photostability
Key Points
Sunscreens are photochemical systems, and their behavior is best under-
stood through the science of photochemistry.
Deeper understanding of the complex photochemistry of avobenzone has
led to better formulating methods and improved sunscreen performance.
The photostability of sunscreen products is a function of the photostabili-
ties of the individual UV filters and the photochemical and photophysical
interactions between them.
Photostability will retain a leading role in sunscreen product design as
costs and regulatory issues continue to drive sunscreen formulating
worldwide.
Though significant challenges remain, the availability of photostabilizers
and, in many areas, new UV filters has allowed the sunscreen industry to
make great strides in improving photoprotection.
compound was the first and for years remained the only UV filter to be effective
at protecting skin from longer wavelength UVA radiation (320400 nm), widely
believed to be a primary cause of early skin aging and certain skin cancers [1].
Avobenzone degrades rapidly in sunlight [2] and may react chemically with other
organic compounds [3]. This spawned an arms race among both UV filter sup-
pliers and sunscreen manufacturers to discover ways to photostabilize or replace
avobenzone. Scientists in Europe focused on developing photostable UV filters to
compete with avobenzone, while other scientists in the USA and Europe focused
on discovering new photostabilizers. Both groups were successful: the resulting
new UVA filters and photostabilizers are now in widespread use throughout the
world.
Several photostable European UVA filters have been submitted for approval to
the US Food and Drug Administration for inclusion in the monograph for OTC
sunscreen drug products. In 2014, all were deemed by the FDA to have insufficient
data on which to base the requisite generally regarded as safe and effective
(GRASE) determination and were returned to their sponsors for additional informa-
tion [4]. This signals a continuing role for photostabilizers in sunscreens, especially
those to be marketed in the USA but also in other parts of the world where global
acceptability is desired and where cost considerations favor the continued use of
inexpensive avobenzone as the primary UVA filter.
R + hn *R pp R
14 Sunscreen Photostability 249
R + hn *R pc I T P
Transitions are favored between states that look like each other in the sense
that their electronic, vibrational, and spin configurations are similar (Turro et al.
2010, p. 4547, 117). Upon photon absorption, an electron in the singlet ground
state naturally transitions to a singlet excited state and almost never to a triplet
excited state. For many chromophores, the reverse is also true: an electron in the
singlet excited state will tend to relax to the singlet ground state either by dissipat-
ing its excess energy as heat (internal conversion) or by emitting a photon
(fluorescence).
The return to the ground state from the singlet excited state tends to happen
quickly; nanosecond time scales are common. Such a rapid return to the ground
state favors photostability since there is little time for chemical processes to
compete.
Figure 14.2 depicts the electron configurations of the triplet excited state which
a b c
Fig. 14.2 In the triplet state, there are three possible orientations for the electron pair: (a) both
electron spin about up vectors; (b) the electrons spin about opposite vectors while precessing in
phase; (c) both electrons spin about down vectors
energy diagrams, also known as Jablonski diagrams, like the one in Fig. 14.3. Key
parameters for any photophysical process are its energy (E), its quantum yield (),
its rate constant (k), and its lifetime (). (Since 1/k = , it is only necessary to measure
one: either rate constant or lifetime.) Quantum yield is a measure of the efficiency
of a process and is calculated either as the fraction of absorbed photons that produce
a specific sequence or by comparing the rate of a specific pathway to the sum of the
rates of all competing pathways. For example, if 10 out of 100 excited molecules
fluoresce, then the quantum yield of fluorescence is 0.10 (10 %).
Another way for an excited chromophore to return to the ground state is by
transferring its energy to another molecule, known as the quencher, Q. Energy
transfer can be represented schematically by
*R + Q ET R + *Q
where ET is energy transfer (Turro et al. 2010, p. 390). Thus, the excited
chromophore transfers its excited state energy to the ground state quencher which
deactivates the chromophore to the ground state and raises the quencher to the
excited state. The relative efficiency of a quencher to quench the excited state of a
chromophore is characterized by a quenching rate constant, kET, where ET stands for
energy transfer. The actual rate this happens in a solution (or, presumably, in a
sunscreen) is the product of the quenching rate constant and the concentration of the
quencher, [Q], plus the sum of all other deactivation pathways, kD.
kq obs = kD + kET [Q ]
252 C.A. Bonda and D. Lott
Fig. 14.3 A state energy or Jablonski diagram like this is used by photochemists to keep track
of an organic chromophores three most important states: the ground state, S0; the lowest energy
singlet excited state, S1; and the lowest energy triplet state, T1. The upward arrow on the left repre-
sents photon absorption and excitation. The downward and diagonally pointing arrows represent
photophysical processes that drain the chromophores excited state energy. Key parameters are the
energies, quantum yields, and lifetimes of each state and the rates of interstate transitions
where kq obs is the quenching rate observed experimentally (Turro et al. 2010,
p. 390391).
This is the basic mechanistic scheme for most of the photostabilizers to be dis-
cussed later in this chapter. First, we turn to avobenzone as the exemplar of a pho-
tolabile UV filter to find out why photostabilizers are needed in the first place.
Seminal studies published in 1995 by Schwack and Rudolph and in 1997 by Andrae
et al. contributed greatly to the early understanding of this important sunscreen
ingredient.
14 Sunscreen Photostability 253
Fig. 14.4 Graph shows that steady-state irradiation of the enol tautomer (A) generates the keto
tautomer (B) which, when left in the dark, spontaneously converts back to the enol form (C).
(Bonda et al. [2], reprinted with permission)
1
KT represents the lifetime of the keto triplet.
256 C.A. Bonda and D. Lott
1
Es = 73.19 kcal/mol
The enol tautomer Es = 77.20 kcal/mol The keto tautomer
OHO
* O O *
ET = 58.33 kcal/mol
H 3 * H H
OMe
OMe
O
OMe OMe
1, 3-H shift ET = 69.76 kcal/mol
h IC = .25 O O
O O
* O
350-360 nm or h
F O H O H O < 350 nm H H
O H H OMe MeO
H
H E isomer + Photodegadation
Z isomer
3O by Norrish Type I
2 cleavage
OMe 1O
Quenching by 2
OHO .014 O O ground state
oxygen
H H H Cellular
OMe OMe
Enol tautomer damage
Diketo tautomer
Fig. 14.5 A state energy diagram for avobenzone, compiled primarily from studies conducted at
low concentrations in acetonitrile solutions. Photolysis of the enol tautomer drives an increase in
the concentration of the keto tautomer, which, when excited by UVR, undergoes a Norrish type I
cleavage to produce benzoyl and phenacyl radicals. The asterisks signify that the molecule within
the brackets is in an excited state (Bonda et al. [2], reprinted with permission)
Kikuchi et al. determined the triplet excited state lifetime (phos) of the enol form to
be 30 ms and the triplet excited state lifetime of the keto form to be 190 ms.
A compounds fluorescence lifetime puts an upper limit on the lifetime of the
singlet excited state. As reported by Bonda et al. (2009), measurements conducted
at the University of California-Riverside determined the fluorescence lifetime of the
enol form to be 13 ps [15].
From these and other studies, a picture of avobenzones photophysics and
photochemistry has emerged, which is depicted graphically in Fig. 14.5.
Avobenzone is not the only photolabile UV filter used in sunscreens. In fact, there
are no perfectly photostable UV filter, though some are nearly so.
Tarras-Wahlberg et al. irradiated OMC mixed with petrolatum first with 20 MED
of UVB radiation and then with 100 J/cm2 of UVA radiation. They observed slight
loss of peak absorption after the UVB dose and a much larger loss of peak absorp-
tion after the UVA dose. HPLC analysis of the sample following irradiation revealed
formation of a new peak which the researchers attributed to OMCs cis isomer,
indicating that irradiation drove conversion of the normally dominant trans isomer
to its cis counterpart, which absorbs UV with a similar peak but at a significantly
lower molar extinction coefficient [16]. Others have found that when present in high
concentrations, OMC can react with itself as two molecules undergo a [2 + 2] cyclo-
addition reaction [17].
The photostabilities of 18 UVB filters approved for the use in sunscreens in the
EU were studied in vitro by Couteau et al. [18]. Each UV filter was incorporated into
its own standardized oil-in-water emulsion. The researchers applied 30 mg of each
formulation to roughened PMMA plates. The plates were irradiated in a xenon test
14 Sunscreen Photostability 257
chamber filtered to block radiation <290 nm. The SPF was measured at timed inter-
vals with a UV transmittance analyzer. Photodegradation of each formulation was
expressed in three ways: as the number of minutes of irradiation required to cause
the coated plate to lose 50 % of its SPF (t50%); as the number of minutes of irradiation
required to cause the coated plate to lose 10 % of its SPF (t90%); and as the rate con-
stant of photodecay (k) according to the equation SPF/SPF0 = ekt. Table 14.1 pres-
ents the results of the study in rank order from most to least photostable.
Herzog et al. studied the photostabilities of ethylhexyl methoxycinnamate
(OMC), ethylhexyl triazine (EHT), avobenzone, BEMT, and OC [19]. They
incorporated each UV filter into its own oil-in-water emulsion which they applied to
a quartz plate and irradiated in a xenon test chamber. At timed intervals, they used
solvent to extract the residual emulsion containing the UV filter from the quartz
plate and then analyzed the solution by HPLC. After 50 MED, OC and BEMT were
found to be photostable. OMC and avobenzone were strongly degraded (<20 % and
<1 % were recovered, respectively), and EHT was less degraded (approximately
50 % was recovered). The researchers noted degradation of OMC is not observed in
ethanol solutions at low concentrations. A rapid initial loss of absorption is attributed
to a change in the equilibrium between the trans and cis isomers (toward the cis)
which quickly stabilizes and after which no further drop occurs.
258 C.A. Bonda and D. Lott
UV filters are almost never used alone in sunscreen products, which may contain up
to six UV filters. Bimolecular interactions between UV filters of the same or
different species, or between the UV filters and inactive ingredients with which they
are paired, can have a positive, negative, or no effect on the sunscreens photostability,
as illustrated in the studies referenced below.
A major sunscreen manufacturer and marketer in the USA reported studies of the
photostabilities of numerous sunscreen products in their comments to the FDA in
2007 [20] and their follow-up supplement in 2008 [21]. In one study, commercially
available sunscreen products were applied in measured amounts to microscope
slides and exposed to natural sunlight until 7.5 MED was reached as measured by a
radiometer. The UV filters were then assayed by HPLC. Independent labs in Sydney,
Australia, Winston-Salem, North Carolina, and Ormond Beach, Florida, took part.
Some of the products were tested by all three labs, others were tested by two. The
14 products ranged from SPF 30 to SPF 80 and comprised 10 lotions, one lotion
spray, two continuous sprays, and one stick product. Four of the products contained
OMC in combination with avobenzone, and nine combined OC with avobenzone,
two of which also contained OMC. Three contained avobenzone without either OC
or OMC. The results may be found in Table 14.2, which groups the products tested
by the presence or absence of the three UV filters. Clearly, of the products tested,
the most photostable are those that contain OC and avobenzone and no OMC, or do
not contain avobenzone at all. In all 12 of the products containing them, the two
salicylates, octyl salicylate and homosalate, showed significant photolability, declin-
ing on average by about 24 % and 15 %, respectively.
Beasely and Meyer determined the impact of avobenzone photolability on SPF
and UVA-PF [22]. They started with a model SPF 50 sunscreen product which
contained 3 % avobenzone photostabilized with 7 % OC. They then prepared a
Table 14.2 % UV filters remaining after 7.5 MED of natural sunlight by HPLC
OC + Avo, no OC + Avo + Avo + OMC, no OMC, no Avo
UV Filters OMC (N=9) OMC (N=2) OC (N=2) (N=1)
Octocrylene (OC) 100 % 100 % 100 %
Oxybenzone 96.8 % 94.9 % 100 % 97.0 %
Avobenzone (Avo) 91.0 % 59.6 % 25.0 %
OMC 49.4 % 41.0 % 65.1 %
Octyl salicylate 77.8 % 75.8 % 76.3 % 75.1 %
Homosalate 86.4 % 81.4 % 86.8 % 84.2 %
14 commercially available sunscreen products were applied to microscope slides and exposed to
natural 7.5 MED of natural sunlight. The studies were duplicated or triplicated by labs in Australia,
North Carolina, and Florida. After exposure, each sunscreen was extracted from the slide by
solvent and analyzed by HPLC to determine the amount of each UV filter remaining. The two
salicylates showed significant loss in all products. Avobenzone was most photostable when
combined with octocrylene (OC) without OMC
14 Sunscreen Photostability 259
series of four new photostable formulations identical in every way to the original
except that the avobenzone concentration was reduced by 20 %, 33 %, 67 %, and
100 % (no avobenzone), respectively, in order to simulate corresponding degrees of
avobenzone loss due to photodegradation. These products were then tested on
human volunteers and the SPF and UVA-PF determined for each and compared to
the original. As expected, the researchers found that reducing the avobenzone con-
centration had the greatest effect on UVA-PF, though SPF suffered significant losses
as well. Small losses of avobenzone (20 %) had little effect on either SPF or
UVA-PF. However, reductions of avobenzone concentrations of 33 % and 67 %
resulted in the SPF declining from 51 to about 48 and 45, respectively, and the
UVA-PF from about 18 to about 14 and 12, respectively. The formulation contain-
ing no avobenzone, which simulated a complete loss of the UVA filter due to pho-
todegradation, achieved SPF 40 and UVA-PF 8.
To approximate the environment in human skin below the surface, Damiani and
co-workers prepared liposomes containing pairs of UV filters and suspended them
in saline. The suspensions were placed in the wells of cell culture plates and irradi-
ated with UVA delivered by a commercial sun lamp. The total dose was calculated
as equivalent to about 90 min of exposure on the French Riviera on a sunny summer
day. The irradiated samples were collected, diluted with ethyl acetate, and centri-
fuged to recover the UV filters, after which UV absorption measurements were
made and compared to non-irradiated controls. The photostable combinations
paired avobenzone with bis-ethylhexyloxyphenol methoxyphenyl triazine (BEMT),
methylene bis-benzotriazolyl tetramethylbutylphenol (MBBT), and diethylamino
hydroxybenzoyl hexyl benzoate (DHHB). The combination with OC improved avo-
benzones UVA absorption by 35 %, while the combinations with OMC and EHT
showed the least photostability, losing most of their absorption throughout the entire
UVA range. Combinations of OMC with BEMT, MBBT, DHHB, and EHT were
photostable [23].
The oft-used combination of avobenzone and OMC was studied by Herzog and
co-workers (2009). They prepared a sunscreen emulsion containing 3.4 % OMC
and 2.4 % avobenzone and compared the amount of OMC recovered after irradiation
with the amount recovered from the emulsion containing OMC alone. They noted a
significant acceleration of OMC photodegradation when avobenzone was added
and attributed the increase to the availability of a second pathway to a [2 + 2]
cycloaddition (the first being the reaction of OMC with itself) stemming from the
reaction of the enol form of avobenzone with OMC. On the other hand, adding
OMC to avobenzone did not affect the amount of avobenzone recovered, indicating
that the OMC-avobenzone reaction competed successfully with formation of
avobenzones keto form to reduce the pathway to the Norrish type I cleavage.
As of this writing, the FDA does not permit avobenzone to be combined with
either TiO2 or ZnO in sunscreens marketed in the USA [24]. Both combinations are
permitted in many other venues throughout the world however. TiO2 in particular is
widely used in combination with organic UV filters.
Titanium dioxide exists naturally in three crystalline forms: rutile, anatase, and
brookite. The TiO2 grades used in sunscreens are made from rutile or anatase. Both
260 C.A. Bonda and D. Lott
forms are available in a range of particle sizes, from nano to micron. In general, the
larger the particle size, the more whitening is the effect on the skin. Both TiO2 and
ZnO are semiconductors with band gaps in the solar UV range. Absorption of a
photon with energy equal to or greater than the band gap promotes an electron from
the valance band to the conduction band, which creates an electron ()/hole (+) pair.
When this happens, molecules close to or adsorbed to the particle surface can inter-
act with these charge carriers to become reduced (gain an electron) or oxidized (lose
an electron). Because of this, these metal oxides have photocatalytic properties and
can behave as either oxidant or reductant to generate reactive oxygen species (ROS)
such as hydroxyl radicals (OH) and superoxide anion (O2). These ROS in turn can
react with the organic components in sunscreens including UV filters, contributing
to their degradation [25]. Of the TiO2 crystalline forms, anatase is regarded as the
more photocatalytically active [26]. For that reason, there have been recent calls to
limit the TiO2 in sunscreens to grades derived from rutile [27]. Commonly, though
not always, the TiO2 and ZnO grades used in sunscreens are passivated (rendered
less reactive) by treating the surfaces of the particles with chemically inert sub-
stances such as silica, dimethicone, or aluminum hydroxide. Other surface treat-
ments are used to improve the particles oil or water dispersibility [28].
Kockler et al. studied the influence of TiO2 particles size on the photostabilities
of avobenzone and OC by preparing oil-in-water emulsions in which the avobenzone
and OC were dissolved in the oil phase, and various grades of TiO2 were dispersed
in the water phase [29]. TiO2 grades tested included a silica-coated rutile TiO2 with
a mean particle size of 119 nm, an uncoated anatase nano TiO2 with mean particle
size of 25 nm and an uncoated anatase micro TiO2 with a mean particle size of
0.6 m. Measured amounts of the emulsions were applied to glass plates and
irradiated for 14.6 h at 400 W/m2 in a xenon test chamber. After irradiation, solvent
was used to extract residual emulsion from the plates, and the solutions were
analyzed by HPLC. From the emulsions containing avobenzone alone or combined
with coated, micro, and nano TiO2, recovery of avobenzone after irradiation ranged
from 0 to 3.81 %. From the emulsion containing OC alone, or combined with
coated, micro, and nano TiO2, recovery of OC ranged from 88.33 to 99.98 %. From
the emulsions containing avobenzone and OC plus coated, micro, and nano TiO2,
recovery of avobenzone was 16.0 %, 12.6 %, and 0.6 %, respectively, and recovery
of OC was 98.2 %, 95 %, and 92.5 %, respectively. A separate experiment determined
that neither avobenzone nor OC adsorb onto any of the TiO2 particles surfaces. The
authors concluded that uncoated nano-TiO2 is more deleterious to both avobenzone
and OC than either micro or coated TiO2.
Nguyen and Schlossman studied avobenzone photostability in dilute solutions in
ethanol in the presence of various grades of TiO2, coated and uncoated, and one
untreated and four treated ZnO grades [30]. The ethanol solutions contained 0.04 %
avobenzone and 4 % of metal oxide. Each sample was irradiated using a UV lamp
for 1 week. Afterwards, each sample was centrifuged to remove the metal oxide
from the solution, and the solutions UV absorption and transmittance were
measured with a UV/Vis spectrophotometer. Both anatase and rutile forms of TiO2
were tested. Primary particle sizes ranged from 15 nm to 300 nm. Surface treatments
14 Sunscreen Photostability 261
Another UV filter that has considerable overlap with avobenzones absorption spec-
trum is oxybenzone (benzophenone-3). Mendrok-Edinger et al. reported that adding
2 % oxybenzone to 4 % avobenzone increases photostability to 80 % compared to
23 % without [34]. Since it is energetically unlikely that Oxybenzone quenches
a
O
OH O
HO O O
N N
O
N O
O
O HO O
Bis-ethylhexyl hydroxydimethoxy
Bemotrizinol; Bis-ethylhexyloxyphenol benzylmalonate; (HDBM)
methoxyphenyl triazine; (BEMT)
OH O
N OH OH N
N N N
N
Butyloctyl salicylate
O
O O
O HO O O
Diethylhexyl 2,6-naphthalate; (DEHN) O
O
O
Diethylhexyl syringylidene malonate; (DESM)
O
N
O
O
Fig. 14.6 Key chemical compounds discussed in this chapter, identified by their USAN (when
relevant), INCI name, and (abbreviation) as used in this chapter. (a) Photostabilizers including
photostabilizing UV filters, (b) Other UV Filters, (c) Antioxidants
14 Sunscreen Photostability 263
O O O
N
R O R O
O O O R
O n n
O
O O
N
R= or R=
O O
O
O
Polyester-8 Polyester-25
O
CH3 CH3 CH3 CH3 CH3 CH3 CH3
O H3C Si O Si O Si O Si O Si O Si O Si CH3
CH3 CH3 CH2 CH3 CH3 CH3 CH3
(CH2)10
n
O O O
R= N R
O m
O
N O
Octinoxate; Ethylhexyl methoxycinnamate;
Diethylamino hydroxybenzoyl hexyl benzoate; (OMC)
(DHHB)
O
O
NH O
N N O
HN N NH
O O
O H
O
10
Tocopherol; Vitamin E
Coenzyme Q10; Ubiquinone
(also known as Coulombic or Frster energy transfer) in which the electric field
generated by the excited electron of the donor resonates with an electron of the
quencher, essentially transferring the donors energy through space to the quencher
(Turro et al., 2010, p. 399). Thus, the donor returns to the ground state, and the
quencher is raised to the excited state. This mechanism diminishes with the inverse
sixth power of the distance between donor and acceptor (Turro et al., 2010, p. 402).
Energy transfer by the dipole-dipole mechanism is the mechanism most often
responsible for singlet-singlet quenching.
The second mechanism is known as the electron exchange mechanism (also
known as Dexter exchange). In this mechanism, the excited donor (3D*) and
quencher (Q) collide such that the donor exchanges its excited state electron for one
of the quenchers ground state electrons, returning the donor (D) to the ground state
and elevating the quencher (3Q*) to the excited state. Energy transfer by the Dexter
exchange mechanism is easily visualized as follows:
LU
HO + +
3D* Q D 3D*
The Dexter exchange mechanism is the most common one for triplet-triplet
quenching. The majority of photostabilizers on the market today (2014) function as
quenchers of avobenzones triplet excited state.
OC has long been recognized as a triplet quencher for avobenzone. Mendrok-
Edinger et al. (2009) reported that 3.6 % OC added to 4 % avobenzone in a sun-
screen emulsion conferred 90 % photostability. Lhiaubet-Vallet et al. tested
avobenzone alone and in combination with six other UV filters, measuring by HPLC
the amount of avobenzone and UV filter recovered after irradiation for four hours (!)
with a solar simulator. The UV filters tested were OMC, OC, BEMT, diethylamino
hydroxybenzoyl hexyl benzoate (DHHB), EHT, and dioctyl butamido triazone
(DBT). The combination of OC and avobenzone was the clear winner with 84 % of
the avobenzone and 100 % of the OC recovered. Next was BEMT and avobenzone,
with 72 % of the avobenzone and 96 % of the BEMT recovered. With no
photostabilizer, only 41 % of the avobenzone was recovered [36].
Polyester-8 is a low molecular weight (ca. 1900 daltons) organic polymer that is
terminated with cyanodiphenyl propenoic acid, the same chromophore as
OC. According to its manufacturer, it retains OCs ability to photostabilize avoben-
zone by a triplet quenching mechanism though with lower efficiency [37].
Undecylcrylene dimethicone (UCD) is a silicone polymer that also incorporates the
OC chromophore. The manufacturers literature states that it enhances the photo-
stability of the UVA filter avobenzone by quenching its triplet excited state [38].
Ethylhexyl methoxycrylene (EHMC) is a commercially available cosmetic
ingredient that is marketed as a photostabilizer for avobenzone and other photolabile
compounds [39]. Kikuchi and co-workers determined EHMCs excited singlet and
14 Sunscreen Photostability 265
triplet state energies to be 72.3 kcal mol1 and 55.5 kcal mol1, respectively [40].
These excited state energies are below those measured by Kikuchi et al. (2009 and
2010) for avobenzone (73.2 kcal mol1 and 58.3 kcal mol1, respectively) and for
OMC (85.49 kcal mol1 and 55.75 kcal mol1, respectively), making the quenching
of the singlet and triplet excited states of both compounds by EHMC energetically
feasible. Researchers at the University of California-Riverside confirmed the ability
of EHMC to quench avobenzones singlet excited state. The researchers employed
a streak scope (also known as a streak camera) to measure avobenzones fluorescence
lifetime in the absence and presence of varying concentrations of EHMC. At
10 mmol concentration of EHMC, the singlet excited state lifetime of avobenzone
was reduced from 1.3 1011 s to 1.86 1012 s, shorter by about an order of
magnitude [41].
Bonda et al. (2010) compared EHMC and OC to photostabilize the combination
of avobenzone and OMC. The researchers prepared three solutions of 3 %
avobenzone and 7.5 % OMC in ethyl acetate. One solution contained 3 % EHMC,
one contained 3 % OC, and a third control solution contained no photostabilizer.
The solutions were applied to PMMA plates and allowed to dry before they were
irradiated with a solar simulator. After 25 MED, the control with no photostabilizer
retained 44.5 % of its UVA absorbance compared to 53.9 % with 3 % OC and
83.7 % with 3 % EHMC.
4-Methylbenzylidene camphor (4-MBC; USAN Enzacamene) is a UV filter that
functions as an avobenzone photostabilizer, almost certainly by a triplet quenching
mechanism. Though not permitted in the USA, it has been used in Europe for
decades at concentrations up to 4 %. Mendrok-Edinger et al. (2009) prepared a
solution of 4 % 4-MBC and 4 % avobenzone which they applied to a roughened
glass plate and then irradiated with 25 MED. Afterward, the plate was washed with
solvent, and the resulting solution was analyzed by HPLC. Subsequently, 88 % of
the avobenzone was recovered compared to 23 % from the solution containing no
photostabilizer.
Another triplet quencher for avobenzone is diethylhexyl 2,6-naphthalate (DEHN)
[42]. Mendrok-Edinger et al. (2009) found DEHN to be mildly effective. In their
experiment, less than 50 % of avobenzone was recovered after 25 MED. Bonda and
Steinberg reported that matched sunscreens containing 3 % avobenzone and either
0 % or 4 % DEHN were exposed to 10 MED of solar-simulated radiation and then
analyzed on a UV transmittance analyzer. In the sunscreen without DEHN, UVB
and UVA attenuation declined to 77 % and 64 %, respectively, while in the sun-
screen with 4 % DEHN, UVB and UVA attenuation remained at 92 % and 91 %,
respectively [43].
Polyester-25 is a low molecular weight polymer that is marketed as a photostabi-
lizer for avobenzone [44]. Based on examination of its structural components, it
would be expected to function mechanistically in a manner similar to EHMC.
A recent entry to the photostabilizer category is trimethoxybenzylidene
pentanedione (TMBP) [45]. The manufacturer tested ethanol solutions containing
3 % avobenzone, 5 % octisalate, and 15 % homosalate to which was added either
4 % OC, 2 % DESM, or 2 % TMBP, measuring UVA absorption before and after
266 C.A. Bonda and D. Lott
irradiation. After 100 J/cm2, the solution containing TMBP retained about 70 % of
its UVA absorption compared to 60 % for OC and about 30 % for DESM.
Another concept is to use antioxidants to photostabilize avobenzone. Afonso
et al. investigated this strategy by combining ubiquinone (coenzyme Q-10) and
tocopherol (vitamin E) at various ratios with avobenzone in model sunscreen
emulsions [46]. They reported a 62.2 % increase in avobenzone photostability when
avobenzone was combined with ubiquinone at a 2:1 ratio and a 15.3 % improvement
when avobenzone was combined with tocopherol at a 1:2 ratio.
Bis-ethylhexyl hydroxydimethoxy malonate (HDBM) is marketed as an
antioxidant that improves avobenzone photostability. According the manufac-
turer, HDBMs triplet energy is too high to quench avobenzones triplet excited
state. Rudolph et al. tested a solution of 2 % HDBM and 2 % avobenzone in
isopropyl myristate which they spread on PMMA plate. The plate was irradi-
ated in a xenon test chamber with the equivalent of 5 MED, after which the
sample was extracted with solvent and the absorption of the solution measured.
At 355 nm, the avobenzone peak, the sample lost 41 % of its absorbance com-
pared to the control with 2 % avobenzone alone which lost 58 %. A structurally
similar compound, DESM, was also tested. DESM is marketed by its manufac-
turer as both an antioxidant and a triplet quencher for avobenzone. After irra-
diation, the solution of 2 % DESM and 2 % avobenzone lost 29 % of its
absorbance at 355 nm [47].
Butyloctyl salicylate was found by Mendrok-Edinger et al. (2001) to be moder-
ately effective in photostabilizing avobenzone. When butyloctyl salicylate was
added at 5 % to a 4 % avobenzone solution then irradiated with 25 MED, 50 % of
the avobenzone was recovered compared to 23 % without butyloctyl salicylate.
Excited state quenching by butyloctyl salicylate of avobenzone is energetically
unfavorable and is therefore ruled out [48]. As a liquid phenol, butyloctyl salicylate,
like other salicylate esters, is a protic solvent. Recalling that avobenzone is
essentially photostable in protic solvents such as isopropanol, it is likely that the
stabilizing effect on avobenzone is due to butyloctyl salicylates proticity. This
effect was previously reported by Bonda et al. (1997).
Sunscreens that combine avobenzone and OMC present a special challenge for
photostabilization. Under exposure to UVR, avobenzone and OMC engage in a
reaction known as the De Mayo reaction. The De Mayo reaction describes the reac-
tion of an enol with an alkene to produce a [2 + 2] cycloaddition followed by a retro
aldol cleavage [49]. The reaction usually proceeds through the excited enol.
However, in the case of avobenzone and OMC, the reaction probably proceeds
through the excited alkene, OMC. This view is supported by Kikuchi and Yagi who
observed the intermolecular triplet-triplet energy transfer from avobenzone to OMC
through measurements of EPR and time-resolved phosphorescence spectra [50].
First they noted that triplet-triplet energy transfer from avobenzone to OMC is
energetically favorable because avobenzones triplet energy (ET1 enol = 58.3 kcal mol1;
ET1 keto = 69.8 kcal mol1) lies above that of OMC (ET1 = 55.75 kcal mol1), while
avobenzones singlet excited state (ES1 enol = 73.2 kcal mol1) lies below that of OMC
(ES1 = 85.5 kcal mol1), thus ruling out singlet-singlet energy transfer.
14 Sunscreen Photostability 267
R1 R2 R1
R1
R2
these findings, avobenzone efficiently transfers its triplet energy to OMC which
elevates ground state OMC to its triplet excited state. OMC triplets then become the
aggressive species in the previously described De Mayo reaction to photodegrade
both compounds and produce photoproducts. OCs quenching rate constant, at only
about half of OMCs, is not competitive.
Chatelain and Gabard (2001) studied the ability of BEMT to photostabilize the
OMC-avobenzone combination, finding that BEMT exerted a protective effect on
both UV filters. In sunscreens containing 5 % each of avobenzone and OMC, adding
5 % BEMT decreased photodegradation of OMC from about 65 % to about 48 %
and photodegradation of avobenzone from 45 % to about 35 %. Photostabilizing the
combination of OMC and avobenzone remains one of the great challenges in
sunscreen formulating.
There are many ways to measure photostability. In this section we are concerned
only with methods that measure the photostability of fully formulated sunscreen
products as opposed to solvent systems that contain one or two UV filters.
One of the easiest methods to test sunscreen photostability is to monitor the
change in transmission of an otherwise transparent plate (e.g., quartz or PMMA)
that has been coated with the sunscreen being tested while it is being irradiated by
UVR. In this method, the coated plate and suitable controls are placed in the path of
the UV beam. Transmission is monitored by a detector in line with the beam but
placed on the other side of the plate. The change in UV transmission seen by the
detector may be quite rapid for a photolabile product. For example, if the output of
the solar simulator is 150 MED/hour, the solar simulator is emitting approximately
0.042 MED per second or about 1 MED every 24 s. Theoretically, the initial output
through the product covered plate would be 5 MED/hour for an SPF 30 sunscreen.
The MED/hour would rapidly climb for a photolabile product as the sunscreens
ability to absorb UV rapidly declines. The advantage of this method is it is simple
and fast. A second advantage is that it somewhat mimics the SPF test. The sunscreen
product sees the same spectra in the photostability test as it does in the actual SPF
268 C.A. Bonda and D. Lott
The organic UV filters in sunscreens are photochemicals that absorb the energy in
ultraviolet radiation (UVR) by converting it to electronic excitation energy. At a
molecular level, this is understood as the promotion of a single electron in an
outer or valence orbital from its lowest energy state to a previously unoccupied
orbital of higher energy, referred to as the excited state. Subsequently if physical
processes drain the excess energy so that all of the molecules of the compound
return unchanged to the ground state, then the compound is photostable. If, how-
ever, the excess energy fuels chemical processes that change some or all of the
molecules, the compound is photolabile. Photolabile compounds lose effective-
ness as UV absorbers as they are exposed to UVR. So it is with some of the
organic chromophores contained in sunscreens and, therefore, with sunscreens
themselves.
As recognition of the skin-damaging effects of UVA radiation has grown, sun-
screen scientists and photochemists have increasingly turned their attention to
270 C.A. Bonda and D. Lott
understanding avobenzone, still the only effective organic UVA protectant approved
worldwide. Today, after 20 years of study, a comprehensive (though still incom-
plete) picture of avobenzones complex photochemistry has emerged. In a nutshell,
UVR exposure induces fragmentation and radical formation in a dose-related man-
ner. Exactly how this happens is not yet fully understood. What is known is that
avobenzone photodegradation is mitigated or curtailed by combining it with com-
pounds that quench its excited states. When combining avobenzone with TiO2 or
ZnO, coated is better than uncoated, and rutile is better than anatase.
All UV filters have been shown to be photolabile to some degree, though under
conditions of actual use, many can be considered to be photostable. In contrast,
the most widely use UVB filter in the world, OMC, is relatively photostable when
tested at low concentration in ethanol, but quite photolabile when tested at realis-
tic concentrations and in formulated products. When OMC and avobenzone are
combined, UVR catalyzes a photochemical reaction that degrades both com-
pounds, a result that continues to vex sunscreen formulators and for which no
complete cure has yet been found though both BEMT and EHMC have been
reported to help.
A number of photostabilizers have been developed that are more or less effective
at preserving avobenzone from photodegradation. Protic solvents help, as does
increasing optical density. The best photostabilizers quench avobenzones excited
states. Most of these are triplet quenchers; one has been shown to quench avoben-
zones singlet excited state.
Testing sunscreen photostability is straightforward: a measured amount of
product is placed on a substrate and analyzed before and after exposure to UVR and
the results compared. Ideally, the sun would serve as the radiation source. As a
practical matter, solar simulators must suffice for the foreseeable future.
The saga of sunscreen photostability has already produced a lasting dual legacy:
for consumers, the widespread availability in much of the world of photostable sun-
screens, and among sunscreen scientists, a new and deeper understanding of sun-
screen photochemistry. Just as the former promises better health for millions, the
latter portends a future of continual improvement in skin photoprotection.
References
1. Polefka TG, Meyer TA, Agin PP, Bianchini RJ (2012) Effects of solar radiation on the skin.
J Cosmetic Dermatol 11:134143
2. Bonda C, Hu R, Jockusch S (2014) Avobenzone photochemistry: improving photoprotection
by stabilizing avobenzone with ethylhexyl methoxycrylene. HPC Today 9:610
3. Dondi D, Albini A, Serpone N (2006) Interactions between different solar UVB/UVA filters
contained in commercial suncreams and consequent loss of UV protection. Photochem
Photobiol Sci 5:835843
4. http://www.fda.gov/drugs/developmentapprovalprocess/developmentresources/over-the-
counterotcdrugs/statusofotcrulemakings/ucm072134.htm
5. Turro NJ, Ramamurthy V, Scaiano JC (2009) Principles of molecular photochemistry: an
introduction. University Science Books, Sausalito
14 Sunscreen Photostability 271
28. Carlotti ME, Ugazio E, Sapino S, Fenoglio I, Greco G, Fubini B (2009) Role of particle coat-
ing in controlling skin damage photoinduced by titania nanoparticles. Free Radic Res
43:312322
29. Kockler J, Oelgemoller M, Robertson S, Glass BD (2014) Influence of titanium dioxide par-
ticle size on the photostability of the chemical UV-filters butyl methoxy dibenzoylmethane and
octocrylene in a microemulsion. Cosm 1:128139
30. Nguyen U, Schlossman D (2001) Stability study of Avobenzone with inorganic sunscreens.
Poster Presentation at the Annual Scientific Meeting of the Society of Cosmetic Chemists,
New York
31. http://www.justaboutskin.com/sun-protection-guide/how-to-evaluate-uva-protection/sun-
screen-active-ingredients/sunscreen-photostabilizers/www.merck-performance-materials.
com; www.sytheonltd.com; www.personal-care.basf.com
32. Gonzenbach H, Hill TJ, Truscott TG (1992) The triplet energy levels of UVA- and UVB-
sunscreens. J Photochem Photobiol B Biol 16:377379
33. Chatelain EB, Gabard B (2001) Photostabilization of butyl methoxydibenzoylmethane (avo-
benzone) and ethylhexyl methoxycinnamate by bis-ethylhexyloxyphenol methoxyphenyl tri-
azine (tinosorb S), a new broadband filter. Photochem Photobiol 74:401406
34. Mendrok-Edinger C, Smith K, Janssen A, Vollhardt J (2009) The quest for stabilizers and
sunscreen photostability. Cosm Toil 124(2):4754
35. Lin Z-P, Aue WA (1999) Triplet-state energies and substituent effects of excited aroyl com-
pounds in the gas phase. Spectrochimica Acta Part A 56:111117
36. Lhiaubet-Vallet V, Marin M, Jimenez O, Gorchs O, Trullas C, Miranda MA (2010) Filter-filter
interactions. Photostabilization, triplet quenching and reactivity with singlet oxygen.
Photochem Photobiol Sci 9(4):552558
37. HallStar Product Information Sheet PC11030 (2011) http://www.in-cosmeticsasia.com/__nov
adocuments/15139?v=634826419407530000
38. HallStar Product Information Sheet PC1A054 (2011) http://www.in-cosmeticsasia.com/__nov
adocuments/15140?v=634826420506970000
39. HallStar Product Information Sheet PC1H044 http://www.hallstar.com/pis.php?product=1h044
40. Kikuchi A, Hata Y, Kumasaka R, Yagi M (2013) Photoexcited singlet and triplet states of a UV
absorber ethylhexyl methoxycrylene. Photochem Photobiol 89:523528
41. Bonda C, Pavlovic A, Hansen K, Bardeen C (2010) Singlet quenching proves faster is better
for photostability. Cosme Toil 125:4048
42. Bonda CA, Marinelli PJ, Hessefort YZ, Trivedi J, Wentworth G (1999) U.S. Patent No
5,993,789
43. Bonda C, Steinberg D (2000) A new photostabilizer for full spectrum sunscreens. Cosm Toil
115:3745
44. HallStar Product Information Sheet PC11031 (2013) http://www.hallstar.com/pis.
php?product=11031
45. Sytheon brochure for Synoxyl HSS. Available online at http://www.in-cosmetics.com/__nova
documents/46438?v=635285132501130000
46. Afonso S, Horita K, Silva e Sousa JP, Almeida IF, Lobao PA, Costa PC, Miranda MS, da
Esteves Silva JCG, Sousa Lobo JM (2014) Photodegradation of avobenzone: stabilization
effect of antioxidants. J Photochem Photobiol B Biol 140:3640
47. Rudolph T, Pan J, Scheurich R, Pfluecker F, Graf R, Epstein H (2009) Superior two step
approach to completely photoprotect Avobenzone with a designed organic redox pair. SOFW
J 135(9):1419
48. Massaro RD, Blaisten-Barojas E (2011) Theoretical investigation of the photophysics of
methyl salicylate isomers. J Chem Phys 135:164306
49. http://www.rocheresearchgroup.com/wp-content/uploads/2011/06/DeMayo_ReactionSR.pdf
50. Kikuchi AM, Yagi M (2011) Direct observation of the intermolecular triplet-triplet energy
transfer from UV-a absorber 4-tert-butyl-4-methoxydibanzoylmethane to UV-B absorber
octyl methoxycinnamate. Chem Phys Let 513:6366
14 Sunscreen Photostability 273
51. Sayre RM, Dowdy JC, Stanfield JW (2009) In vitro sunscreen transmittance measurement
with concomitant evaluation of photostability: evolution of a method. Photochem Photobiol
85(4):10381040
52. Moyal D, Refregier JL, Chardon A (2002) In vivo measurement of the photostability of sun-
screen products using diffuse reflectance spectroscopy. Photodermatol Photoimmunol
Photomed 18:1422
53. Ou-yang HJ, Stanfield JC, Cole CY, Appa Y (2010) An evaluation of ultraviolet a protection
and photostability of sunscreens marketed in Australia and New Zealand. Photodermatol
Photoimmunol Photomed 26:336337
54. Sayre RM, Dowdy JC (2010) The FDA proposed solar simulator versus sunlight. Photochem
Photobiol Sci 9:535539
55. Sayre RM, Stanfield JW, Bush A, Lott D (2001) Sunscreen standards tested with differently
filtered solar simulators. Photoderm Photoimmunol Photomed 17:278283
56. Uhlmann B, Mann T, Gers-Berlag H, Sauermann A (1996) Consequences for sun protection
factors when solar simulator spectra deviate from the spectrum of the sun. Inter J Cosm Sci
18:1324
57. Young A, Boles J, Herzog B, Osterwalder U, Baschong W (2010) A sunscreens labeled sun
protection factor may overestimate protection at temperate latitudes: a human in vivo study.
J Invest Dermatol 130:24572462
58. Lott D (2013) Testing SPF15-100, indoor vs outdoor. Cosm Toil 9:638647
Chapter 15
Sunscreen Formulation: Optimizing Efficacy
of UVB and UVA Protection
Curtis Cole
Key Points
1. Spectral shape of sunscreen products should be designed to best protect
against the primary causes of sun damage: sunburn, skin cancer, and skin
aging.
2. A spectral absorbance shape with approximately a 3:1 ratio of SPF:UVA-PF
will provide equal protection across the UV range for the three main skin
damages.
3. Vehicle components are chosen to provide the functional and aesthetic
requirements for the use conditions, recreational use in intense sunlight
conditions, or daily moisturization for intermittent or incidental sun
exposure.
4. UV filters can be chemical (organic), physical (inorganic) filters, or a
mixture of both to provide the desired spectral shape and aesthetic
properties.
15.1 Background
C. Cole, PhD
Sun and Skin Consulting, LLC, Ringoes, NJ 08551, USA
e-mail: [email protected]
red veterinary petrolatum product that provided sun protection for use in
tropical regions. During the 1960s, additional UV filters were developed includ-
ing the first filters to block (at least partially) in the UVA region, namely, the
benzophenones and the metal oxides, titanium dioxide and zinc oxide. In 1972,
the US Food and Drug Administration (FDA) initiated the current Monograph
regulatory system that permitted manufacturers to market certain drug products
without pre-market approval from the FDA, as long as the product complied with
the stipulations described in the specific drug product monograph publication.
The first Proposed Rule for the Over-The-Counter Sunscreen Product monograph
was published in 1978 [4] describing the UV-absorbing UV filters that were rec-
ognized as safe and effective for use in these sunscreen products, the allowed
concentrations and combinations permitted, and a test method to evaluate the sun
protection factor (SPF) of the product to be marketed. Only UV filters that were
already in market in 1972 and which had sufficient safety toxicology information
submitted to the FDA were considered to be generally safe and effective
(GRAS/E) and permitted in any new sunscreen products. The benzophenones
(oxybenzone, sulisobenzone, and dioxybenzone) and titanium dioxide were the
only permitted UV filters approved for use in this 1978 Proposed Rule that had
any meaningful protection in the UVA portion of the spectrum. Zinc oxide was
not considered at the time, either by omission or by lack of submitted supporting
safety and efficacy data.
During the 1980s and 1990s, photobiology research focused on the effects of
UVA radiation on skin, assessing its ability to cause skin cancer [58] by itself or
in conjunction with UVB radiation, immune suppression [911], and also to con-
tribute to the photoaging [12, 13] processes. While clearly less efficient on a pho-
ton vs photon basis compared with UVB radiation, UVA radiation is clearly
implicated in virtually all of the same photobiological damage endpoints caused by
UVB radiation, although the photochemical process is typically mediated by oxi-
dative pathways rather than direct UV absorbance and lesion/photoproduct
induction.
Development of commercial sunscreen products progressed throughout this
same period, to provide SPF values beyond the initial envisioned cap of SPF 15,
and was reaching SPF 30+ by the early 1990s. While immediate sunburn protection
was evident via the SPF test for these products, criticism of these high SPF
sunscreen products became more vocal suggesting that while extending the safe
exposure time to acute sunburn, these sunscreens were also allowing for extraordinary
UVA dose exposure as these sunscreens were primarily protecting only against
UVB radiation, with little long-wave UVA protection. The need for broad-spectrum
UVA filters was evident. It was however, not until 1996, when zinc oxide and
avobenzone were approved as Category I monograph filters that formulators could
begin to design truly broad-spectrum protection sunscreens in the USA. But, as
formulators quickly discovered, simply adding these two ingredients into
formulations was no guarantee of functional broad-spectrum performance or high
UVA protection.
15 Sunscreen Formulation: Optimizing Efficacy of UVB and UVA Protection 277
1.0
0.1
Erythema
Relative 'sensitvity'
Elastosis
0.001
0.0001
0.00001
290 310 330 350 370 390
Wavelength (nm)
Fig. 15.1 Ultraviolet action spectra describing the sensitivity of the skin for each endpoint as a
function of the wavelength of the incident UV photons (Y axis is logarithmic scale)
278 C. Cole
1E-06
Winter 60:40 UVB UVA 40 N Lat.
0.9E-06 Solar noon
Measurements
0.8E-06
0.7E-06
0.6E-06
Summer 80:20
0.5E-06
Spring 75:25
0.4E-06 Fall 75:25
0.3E-06 Winter 60:40
0.2E-06
0.1E-06
0.E+00
280 300 320 340 360 380 400
Wavelength (nm)
Fig. 15.2 Plot of erythemal energy from sunlight as a function of the time of year. This demon-
strates the predominance of UVB in causing sunburn compared to UVA The proportions of
UVB:UVA sunburning amounts are shown in the graph legends and range from 80:20 in the sum-
mertime to 60:40 in the winter. Based on noon measurements at 40 N. Lat
contributions of UVB and UVA to each of these damages depend primarily on what
solar spectrum is used, but the ranges are from 87 % UVB: 13 % UVA if using
Australian noontime sun [16] at 19 S latitude to 67 % UVB and 33 % UVA using
an average lower 48 continental United States solar spectrum (ASTM G173-03
[17] Standard tables for reference solar spectral irradiances: Direct Normal and
Hemispherical on 37 Tilted Surface. Book of Standards 14.04 2012). Measurements
taken in New Jersey in summertime in the USA indicate approximately 80:20
UVB:UVA split for sunburn potential energy [18] in the summertime and a roughly
60:40 split in winter (see Fig. 15.2). Solar simulators used for sunscreen testing
purposes are closer in spectral quality to the Australian sun standard, which is a
noontime, low elevation (90 ft. above sea level) observation at the summer sol-
stice, representing a high-level solar exposure situation. This solar spectrum yielded
an average minimal erythema dose (MED) in just over 9 min of exposure time.
These damage action spectra address some but not all of the known UV damages.
Notably missing are action spectra for basal cell skin cancer, malignant melanoma,
and immune suppression. A representative model system for studying basal cell
skin cancer has not been available for developing action spectra; however it is
clearly associated with UV exposures and actinic keratoses [19], as well as squa-
mous cell skin cancer. For many years, the action spectrum for malignant melanoma
has been debated and was proposed to be both UVA and UVB based on a fish model
[20]. More recently, deFabo has utilized a transgenic mouse model for malignant
melanoma and published preliminary action spectrum data indicating that UVB
15 Sunscreen Formulation: Optimizing Efficacy of UVB and UVA Protection 279
radiation is the initiator of solar induced melanoma [21, 22]. Lastly, immune sup-
pression has been demonstrated to have strong UVB sensitivity per photon as well
as sensitivity in the UVA [23], and because of the predominance of UVA in sunlight,
UVA has been suggested to be of particular importance in environmental exposures
[24]. Sunscreens containing both UVB and UVA protection have been shown to
provide protection against immune suppression proportional to the UVA-PF of the
sunscreen [25, 26]. The biological data clearly indicate the need for sunscreens to
provide UVB and UVA protection for both acute and long-term potential damages.
Absorbance Units
1.8 1.8
Critical wavelength = 377 nm Critical wavelength = 377 nm
1.6 1.6
1.4 1.4
1.2 1.2
1.0 1.0
0.8 0.8
0.6 0.6
0.4 0.4
0.2 0.2
0.0 0.0
290 300 310 320 330 340 350 360 370 380 390 400 290 300 310 320 330 340 350 360 370 380 390 400
Wavelength (nm) Wavelength (nm)
Fig. 15.3 Absorbance measurements for two SPF 30 sunscreens having the same critical
wavelength with different absorption distributions and very different UVA protection capabilities
as measured by spectrophotometer and UVA-PF assessments
morphous light eruption (PMLE) [41], providing some validation for use of this test
methods usefulness for predicting biological protection.
We now have three measures for determining optimal efficacy, SPF, UVA-PF,
and critical wavelength to assess both the height and breadth of the UV protection
of sunscreen products. But what should be the proportion of the protection in the
various portions of the ultraviolet wavelength region? Should a sunscreen have a flat
spectral absorbance profile, or more heavily weighted in the UVB, or maybe in the
UVA portion of the spectrum? Do we have enough data to make the determination?
Refregier [42] proposed that the ratio of SPF to UVA protection should be approxi-
mately 3:1 in order to have equivalent damage distributed into the UVB and the
UVA portions of the spectrum, based on fundamental understanding of the relation-
ship between SPF and UVA protection. If a flat sunscreen spectrum is used to
attenuate the suns spectrum, then the same proportion of damage (roughly 80 %
UVB, 20 % UVA) results as with unattenuated solar UV. If, however the spectrum
of the filters used in a sunscreen is weighted in the proportion of SPF: UVA-PF = 3,
then the damage is shifted to the right side of the spectrum and distributing the dam-
age equally into the UVB and the UVA portions of the solar spectrum. Having a
spectrum with SPF:UVA-PF of >3:1 shifts the spectral damage even more deeper
into the UVA range. The choice becomes philosophical at this point as to which
distribution of damage is best, with many regulatory bodies siding with the opin-
ion that a balanced distribution is a better approach and adopting the requirement
for a SPF:UVA-PF ratio of 3.0 in order to make broad spectrum or UVA pro-
tection claims. Coupling this requirement with a critical wavelength measurement
of 370 nm, there is assurance that a product will have significant breadth and
height of UVA protection in addition to the known SPF protection provided by the
product.
With these measures established, the formulator can head to the bench to design
and optimal protection sunscreen product using the tools of the trade.
When starting to formulate a new sunscreen product, the formulator must first
ascertain the intended use of the product (recreational/water resistant or daily-
wear moisturization for incidental UV exposure), the target SPF desired, the
aesthetic or feel characteristics of the product, and the desired delivery system (oil
and water emulsions, liquid, alcohol gel, or spray) format, in order to choose the
appropriate soluble UV filter to be used. The vast majority of products for both
recreational and daily-wear utilize the oil-soluble UV filters due to their superior
ability to absorb UV photons (having a higher extinction coefficient) and deliver
good spreading and dry-down characteristics on the skin contributing to SPF and
UVA protection efficiency on a % weight basis. In the USA, avobenzone is the only
soluble UV filter that can be used to qualify a product for broad-spectrum claims,
282 C. Cole
as it is the only soluble UV filter with absorption past the 370 nm critical
wavelength. Thus, it is the most commonly used UV filter in products today in the
USA. This limited choice of UVA filters dictates much of the formulation options
open to the formulator.
While a highly efficient absorber, avobenzone unfortunately has the tendency to
break down upon UVA photon absorption [43] and requires careful formulation.
Experience has shown that combinations of avobenzone with octinoxate [44] (and
other cinnamate-based filters) or any of the PABA derivative UVB sunscreens leads
to rapid photodegradation of both the UVA and the UVB protection due to the
interaction of UV photons with these filters, resulting in rapidly diminishing
absorption during UV exposure. While capable of delivering the SPF value
determined in clinical testing, constant re-application of product with such a non-
photostable product is needed to maintain meaningful protection during extended
exposures in sunlight. PABA derivative filters have been avoided since the mid-
1980s when concerns regarding allergenicity of Padimate A became evident.
Padimate-O also known as octyldimethyl PABA has strong UVB absorption charac-
teristics, and an excellent safety profile regarding allergenicity, has, nonetheless,
little use in sunscreen products because of its similarity to the Padimate A and
because of this destabilizing effect on avobenzone.
For a photostable broad-spectrum product, the formulator must combine other
UVB filters with the avobenzone to build a broad-spectrum product. The salicylate
filters, homosalate, and octisalate are used to provide UVB protection, despite the
fact that their absorbance extinction coefficient is relatively low compared to other
filters, such that they are typically used at their maximum permitted concentrations
of 15 % and 5 %, respectively. Octocrylene is a good choice to combine with avo-
benzone for several reasons; it has a relatively strong UVB absorbance compared
with the other UVB filters, rapidly increasing SPF values with modest concentra-
tions added, but more importantly, it aids in photostabilization of the avobenzone,
helping to transition the triplet state avobenzone molecule back to ground state in a
timely manner.
The addition of benzophenone filters to this theoretical formulation provides
three additional benefits: it increases the absorbance in the UVB portion of the
spectrum, builds protection in the shortwave UVAII region between 320 and
340 nm, and provides additional photostabilization of avobenzone. This UVAII
region is not strongly served by either the primary UVB filters or by avobenzone
but is still in the biologically sensitive region known to be prone to both direct pho-
ton damage and indirect oxidative damage from reactive oxygen species and free
radicals.
For all of the above reasons, the vast majority of sunscreen products available on
the USA market today consist of combinations of avobenzone, octocrylene,
homosalate, octisalate, and oxybenzone filters. US monograph restrictions currently
prohibit the use of inorganic filters in combination with avobenzone, the most effec-
tive and broadest UVA filter.
15 Sunscreen Formulation: Optimizing Efficacy of UVB and UVA Protection 283
If formulating without avobenzone as the primary UVA filter, the only option for
a broad-spectrum product requires the use of zinc oxide to provide sufficient breadth
of protection to have a critical wavelength of 370 nm. Because it is a photostable
filter, it can be combined with octinoxate, Padimate-O, or other UVB filters as
needed to achieve the desired SPF value.
Water-soluble UV filters become an option when considering daily-wear
moisturizer-type products that do not require water resistance. The best water-
soluble filter for consideration is ensulizole, which has a high and broad UVB
absorbance, but to date, the US monograph currently does not permit marketing of
products containing the combination of ensulizole with avobenzone or zinc oxide
[45, 46], so that there are no options available to formulate a broad-spectrum
product in the US market using only water-soluble filters. The only other water-
soluble filters permitted to be used are sulisobenzone and trolamine salicylate;
however they can be sticky in formulations and are not optimum choice for daily
moisturizing products, particularly those used on the face, that require more elegant
and pleasant tactile properties.
Ex-USA: Formulation options for sunscreens outside of the USA opens many
more options for combinations of soluble UV filters that can provide high SPF and
broad-spectrum characteristics. These include the triazine UVB filters, ethyl hexyl
triazine (Uvinul T-150) and diethylhexyl butamido triazine (Uvasorb -HEB), that
are triple UVB chromophores, with extinction coefficients five to ten times higher
than other UVB filters. Thus with only a few percent of these filters, significant
UVB protection can be provided. Silicone-15 is another novel UVB filter that has a
much more modest extinction coefficient but is reported to boost UVB absorption
and SPFs in a manner disproportionate to its own absorption properties. Its unique
polymeric structure with silicone allows it to provide unique and desirable skin
aesthetics to formulations.
Several other UVA1 filters are also available outside of the US market, namely,
bis-ethylhexyloxyphenol methoxyphenyl triazine (trade name Tinosorb S),
methylene bis-benzotriazolyl tetramethylbutylphenol (trade name Tinosorb M),
and diethyl amino hydroxybenzoyl hexyl benzoate (Uvinul A+). Tinosorb S is an
oil-soluble filter with absorption in the mid-UVA range with a secondary peak in the
UVB range, and while it does not extend its absorbance as far in the long-wave
UVA1 as avobenzone, it has a high extinction coefficient and can provide significant
UVA protection with low percentage quantities in formulations. It is very photostable
and can provide photostabilization to avobenzone in addition to its UVB protection
[47]. Tinosorb M is an insoluble particle (nano-size) that has a broad spectral
absorbance range that extends beyond 380 nm and is the broadest of the UVA
filters in spectral absorption.
Ecamsule (Mexoryl SX) is a mid-range UVA filter (peaking at 340 nm) that is
water soluble, and drometrizole trisiloxane (Mexoryl XL) is an oil-soluble mid-
range UVA filter, with a modest extinction coefficient. These two filters have been
proprietary filters to LOreal. They are typically combined with avobenzone or other
UVA1 absorbers for a broad-spectrum profile.
284 C. Cole
In the mid-1980s, efforts began to improve the effectiveness and cosmetic attributes
of the insoluble inorganic UV filters, titanium dioxide and zinc oxide, by reducing
the particle size of these materials. Making them nano-sized (less than 100 m for
smallest dimension) did two desirable things: it increased the surface area of the
molecules per unit weight and providing higher absorption of the UV photons per
unit weight and making them more transparent in the visible portion of the spectrum
and less visible on the skin. Organic surface coatings added to the molecules
eliminated the potential surface reactivity and made them easier to formulate into
emulsions in either the water phase or the oil phase, depending on the nature of the
surface coating. While never quite achieving ultimate invisibility, significant
progress has been made through careful choices of the suspending excipients and
the emulsifiers used.
While reduction in particle size of titanium dioxide shifts the absorption spec-
trum toward higher UVB protection, and lower UVA protection, and reducing the
particle size of zinc oxide can boost the absorbance in the mid UVA1 region [48].
Anderson et al.[46] have a detailed description regarding the inorganic filter charac-
teristics and their formulation. Nano titanium dioxide and zinc oxide are typically
formulated into lotions and cream emulsion form products, which can be character-
ized as oil-in-water emulsions (oil droplets in a sea of water) or as water-in-
oil emulsion (water droplets in a sea of oil). Each form has its own unique
advantage depending on the intended use of the product and consumer preference.
Different emulsifiers are used for the two forms and with the exterior phase of the
emulsion (the sea portion) typically constituting the larger proportion of the for-
mulation by weight.
Oil-in-water emulsions with the inorganic sunscreen filters will typically have
a more traditional lotion feel and use characteristic, with easier spreading,
more rapid dry-down time, and a less oily/greasy after-feel when the water has
evaporated. This is generally the more consumer preferred form of product. In
contrast, the water-in-oil form of this type of product will have a higher oil
content and thus take more time to dry down with a heavier and perhaps more oily
after-feel. The advantages of this type of emulsion are more moisturization
(especially for very dry skin), higher efficiency of the filters to provide UV
protection per unit UV filter incorporated into the system, and more inherent
water resistance characteristics [49].
As mentioned before, inorganic filters may be combined with all of the soluble
filters in the US except avobenzone. This restriction does not apply outside of the
USA where the combination of titanium dioxide and zinc oxide with the soluble UV
filters is commonly used to augment both the UVB and UVA protection, respectively.
Additionally, ex-US, the insoluble UV filter Tinosorb M can also be added to
provide UVA1 protection beyond the range covered by zinc oxide or avobenzone.
15 Sunscreen Formulation: Optimizing Efficacy of UVB and UVA Protection 285
15.6 Summary
References
11. Iwai I et al (1999) UVA-induced immune suppression through an oxidative pathway. J Invest
Dermatol 112:1924
12. Kligman LH, Sayre RM (1991) An action spectrum for ultraviolet induced elastosis in hairless
mice: quantification of elastosis by image analysis. Photochem Photobiol 53:237242
13. Bissett D et al (1987) An animal model of solar-aged skin: histological, physical, and visible
changes in UV irradiated hairless mouse skin. Photochem Photobiol 46(3):367378
14. McKinley A, Deffey BL (1987) A reference action spectrum for ultraviolet induced Erythema
in human skin. In: Passchier WF, Bosnajakovic BFM (eds) Humen exposure to ultraviolet
radiaton: risks and regulations, International congress series. Elsevier, Amsterdam, pp 8387
15. Setlow R (1974) The wavelengths in sunlight effective in producing skin cancer: a theoretical
analysis. Proc Natl Acad Sci U S A 71(9):33633366
16. Bernhard G, Mayer B, Seckmeyer G (1997) Measurements of spectral solar UV irradiance in
tropical-Australia. J Geophys Res 102(D7):87198730
17. ASTM G173-03(2012) Standard tables for reference solar spectral irradiances: Direct Normal
and Hemispherical on 37 Tilted Surface. Book of Standards 14.04 2012
18. Cole CA (2001) Measurement of sunscreen UVA protectiveness. Review article. Photomedicine
Photoallergy Photodermatology 17(1):210
19. Green A et al (1996) Skin cancer in a subtropical Australian population: incidence and lack of
association with occupation. AM J Epidemiol 144(11):10341040
20. Moan J, Dahlback A, Setlow RB (1999) Epidemiological support for a hypothesis for mela-
noma induction indicating a role for UVA radiation. Photochem Photobiol 70:243247
21. DeFabo E (2006) Initial studies on an in vivo action spectrum for melanoma induction. Prog
Biophys Mol Biol 92(1):97104
22. De Fabo EC, Noonan FP, Fears T, Merlino G (2004) Ultraviolet B but not ultraviolet a radia-
tion initiates melanoma. Cancer Res 64:63726376
23. Ullrich SE, Nghiem DX, Khaskina P (2007) Suppression of an established immune response
by UVA: a critical role for mast cells. Photochem Photobiol 83(5):10951100
24. Damian DL, Matthews YJ, Phan TA, Halliday GM (2011) An action spectrum for ultraviolet
radiation-induced immunosuppression in humans. Br J Dermatol 163(3):657659
25. Moyal D, Fourtanier A (2002) Effects of UVA radiation on an established immune response in
humans and sunscreen efficacy. Exp Dermatol 11(S1):2832
26. Ngheim DX, Kazimi N, Clydesdale G, Ananthaswamy HN, Kripke ML, Ullrich SE (2001)
Ultraviolet a radiation suppresses an established immune response: implications for sunscreen
design. J Invest Dermatol 117:11931199
27. Diffey BL, Tanner PR, Matts PJ, Nash JF (2000) In vitro assessment of the broad-spectrum
ultraviolet protection of sunscreen products. J Am Acad Dermatol 43(6):10241035
28. Cole C (2001) Sunscreen protection in the ultraviolet a region: how to measure effectiveness.
Photoderm Photoimmunol Photomed 17:210
29. Gange R, Soparkar A, Matzinger B, Dromgoole S, Sefton J, DeGryse R (1986) Efficacy of a
sunscreen containing butyl methoxydibenzoylmethane against ultraviolet a radiation in
photosensitized subjects. J Am Acad Dermatol 15:494499
30. Lowe N, Dromgoole S, Sefton JH, Bourget T, Weingarten D (1987) Indoor and outdoor effi-
cacy testing of a broad spectrum sunscreen against ultraviolet a radiation in psoralen sensitized
subjects. J Am Acad Dermatol 17:224230
31. Kollias N, Bykowski J (1999) Immediate pigment darkening thresholds of human skin to
monochromatic (362 nm) ultraviolet a radiation are fluence rate dependent. Photodermatol
Photoimmunol Photomed 15:175178
32. Fourtanier A, Moyal D, Maccario J, Compan D, Wolf P, Quehenberger F, Cooper K, Baron E,
Halliday G, Poon T, Seed P, Walker SL, Young AR (2005) Measurement of sunscreen immune
protection factors in humans: a consensus paper. J Invest Dermatol 125(3):403409
33. Young AR (2004) Methods used to evaluate the immune protection factor of sunscreen: advan-
tages and disadvantages of different in vivo techniques. Cutis 74(5 Suppl):1923
34. Standfield J, Feldt P, Csortan E, Krochmal L (1989) Ultraviolet a sunscreen evaluations in
normal subjects. J Am Acad Dermatol 20:744778
15 Sunscreen Formulation: Optimizing Efficacy of UVB and UVA Protection 287
35. Cole C, Van Fossen R (1992) Measurement of sunscreen UVA protection; an unsensitized
human model. J Am Acad Dermatol 26:178184
36. Cole C (1994) Multicenter evaluation of sunscreen UVA protectiveness with the protection
factor test method. J Am Acad Dermatol 30:729736
37. Chardon A, Moyal D, Hourseau C (1997) Persistent pigment darkening response for evalua-
tion of ultraviolet a protection assays. In: Lowe N, Shaath N, Pathak M (eds) Sunscreens:
development, evaluation and regulatory aspects. Marcel Dekker, New York, pp 582559
38. Japan Cosmetic Industry Association (JCIA) technical bulletin. Measurement standard for
UVA protection efficacy, Issued 21 Nov 1995 as effective as of 1 Jan 1996. (The European
Cosmetic Toiletry and Parfumery Association)
39. COLIPA In Vitro Photo Protection Methods Task Force (2007) Method for the in vitro deter-
mination of UVA protection provided by sunscreen products. Guideline 2007, 2nd edn.
COLIPA, Brussels
40. ISO 244432012 (2012) Test method: cosmetics-sun protection test method determination of
sunscreen UVA photoprotection in vitro
41. Fourtanier A, Moyal D, Selte S (2008) Sunscreens containing the broad-spectrum absorber,
Mexoryl SX, prevent the cutaneous detrimental effects of UV exposure; a review of clinical
study results. Photoderm Photoimmun Photomed 24(4):164174
42. Refregier JL (2004) Relationship between UVA protection and skin response to UV light:
proposal for labeling UVA protection. Int J Cosmet Sci 26:197206
43. Tarras-Wahlberg N, Stenhagen G, Lark O, Rosn A, Wennberg A, Wennerstrm O (1999)
Changes in ultraviolet absorption of sunscreens after ultraviolet irradiation. J Invest Dermatol
113:547553
44. Sayre MR, Dowdy JC, Gerwig AJ, Williams WJ, Lloyd RV (2005) Unexpected photolysis of
the sunscreen octinoxate in the presence of the sunscreen avobenzone. Photochem Photobiol
81(2):452456
45. Federal Register, 21 CFR Parts 201 and 301 (2011) Labeling and effectiveness testing: sun-
screen drug products for over-the-counter human use. 76(117):35623
46. Federal register, 21 CFR Parts 201 and 301 (2007) .Sunscreen products for over-the-counter
human use: propose amendment of final monograph. 72(165):49075
47. Chatelain E, Gabard B (2001) Photostabilization of butyl methoxydibenzoylmethane (avoben-
zone) and ethylhexyl methoxycinnamate by Bis-ethylhexyloxyphenol methoxyphenyl triazine
(tinosorb S), a New UV broadband filter. Photochem Photobiol 74(3):401406
48. Anderson MW, Hewitt JP, Spruce SR (1997) Broad-spectrum physical sunscreens: titanium
dioxide and zinc oxide. In: Lowe N, Shaath N, Pathak M (eds) Sunscreens: development,
evaluation, and regulatory aspects, 2nd edn. Marcel Decker, New York, pp 353396
49. Hewitt J (2013) Formulating with nano technologies in skincare: opportunities and challenges.
In: Nasir A (ed) Nanotechnology in medicine. Springer Science and Business Media LLC,
New York, p 1
50. Osterwalder U, Champ C, Flsser-Mller H, Herzog B (2013) The evoluation of UVA protec-
tion. www.skin-care-forum.basf.com. Sept 2013
51. Diffey BL, Brown MW (2012) The ideal spectral profile of topical sunscreens. Photochem
Photobiol 88(3):744747
Chapter 16
Sunscreen Formulation: Optimising Aesthetic
Elements for Twenty-First-Century
Consumers
Julian P. Hewitt
Key Points
1. Good aesthetic properties (appearance and skin feel) are now seen as an
important aspect of sunscreen products, as they encourage greater con-
sumer compliance and also a means of differentiating products from the
competition.
2. Sensory panel studies indicate that what is usually desired is a product that
spreads easily with a moderately wet feeling during application but feels
smooth and dry afterwards with little or no perceivable residue.
3. With organic UV filters, judicious choice of emollients helps to optimise
both skin feel and efficacy. Improving the efficacy also enables the formu-
lator to improve aesthetic properties as lower concentrations of UV filters
are required to reach the target SPF.
4. With inorganic UV filters, developments in manufacturing and coating
technology have produced materials that are transparent on skin while still
being effective and also deliver elegant skin feel.
16.1 Introduction
Todays sun care formulator must achieve ever more challenging standards for prod-
uct efficacy while also making products cosmetically appealing. Performance and
aesthetics are in fact dependent on one another. Studies have shown that consumers
almost always use less than the recommended amount of sunscreen products [1, 2],
and this means that the effective in-use SPF is considerably less than that tested
J.P. Hewitt
JPH SunCare Technologies Ltd., Durham, UK
e-mail: [email protected]
and displayed on the label [3, 4]. Among the reasons most frequently cited by con-
sumers for underuse (or complete avoidance) of sunscreens are aesthetic issues, for
example, that the products feel too greasy or sticky or they make the skin look shiny
or leave a white residue on skin. It can therefore be expected that products with
improved aesthetics encourage consumers to apply more product and therefore get
closer to the labelled SPF. Conversely, maximising the efficacy of the actives used
enables high SPF products to be created with minimal levels of UV filters, which
allows the formulator greater freedom to optimise skin feel.
Also, in many parts of the world, efficacy claims for sunscreen products are
becoming more regulated and uniform. For example, in Europe, the European
Commission Recommendation on labelling and efficacy of sunscreens [5] provides
a specific list of SPF claims that can be used: 6, 10, 15, 20, 25, 30, 50 and 50+.
Similar restrictions exist in a number of other countries. In terms of UVA claims,
many countries now have a single performance criterion for a UVA or broad-
spectrum claim, and numerical claims to indicate the degree of UVA protection are
either discouraged or explicitly prohibited. This limits the options for manufacturers
and marketers to differentiate their products from the competition based on efficacy
claims. Improving the cosmetic properties of the product has therefore become an
important alternative means of providing differentiating claims. This chapter will
discuss how this can be achieved, depending on the type of vehicle and the active
ingredients used.
Of course, as with any cosmetic or topical product, the optimum aesthetic proper-
ties depend very much on the personal preferences of the individual consumer and
can also be influenced by the environment in which the product is used (e.g. dry or
humid, beach or mountain), level of activity (e.g, sunbathing, walking, sports) and
area of application (face or body). However, there are some general trends that can
be identified.
A study by Vollhardt et al. [6, 2] used descriptive sensory analysis to assess the
sensory properties of over 50 different commercial sunscreen lotions with label SPF
claims of 30 or 50. Since these were products already on the market, it is reasonable
to assume that they give a good representation of the sensory properties that are
accepted/desired by consumers. The sensory parameters assessed were grouped into
three distinct phases of product application: rub-out, immediate after-feel and after-
feel 20 min later. Based on this study, the desirable properties during rub-out can be
summarised as:
Wetness should be neither too high nor too low.
Spreadability should be high.
Thickness should be low.
16 Sunscreen Formulation 291
Whiteness should be low, although the data indicate that consumers expect a
certain degree of whiteness during rub-out.
Oiliness and greasiness should be relatively low but not excessively so,
while waxiness during rub-out was lower in the products tested. This makes
sense, considering the requirement for good spreadability; a product that does
not spread well is perceived as more waxy.
Absorbency should be fast but not too fast; if a product absorbs too quickly
into the skin, it can be difficult to spread over a wide area.
The key attributes in the after-feel, particularly after 20 min, are:
Low gloss
Very low whiteness; note the contrast here with the rub-out phase while
consumers may accept some whitening during rub-out, they want no visible
residue afterwards.
Very low stickiness
High slipperiness in other words consumers are looking for a smooth feeling
on skin after application.
Low residue
Low oiliness and greasiness, with relatively higher waxiness. Again the contrast
with the rub-out phase is instructive here; while a perception of high waxiness
during rub-out would indicate poor spreadability, in the after-feel phase, it indi-
cates a dry after-feel. The prevalence of new sun care products in recent years
with claims of dry skin feel or dry touch shows that this is a desirable
property.
In summary, then, what is usually desired is a product that spreads easily with a
moderately wet feeling during application but feels smooth and dry afterwards with
little or no perceivable (either by touch or sight) residue.
Emulsions remain the predominant form for sun protection products globally, so
this chapter will focus on optimising the aesthetic properties of emulsion-based
sunscreen products.
Generally, oil-in-water (O/W) emulsions have a more preferred skin feel than
water-in-oil (W/O) systems. This can be intuitively understood; with water as the
external phase, there is an immediate sensation of wetness on the skin when an O/W
emulsion is applied. Also, as the water begins to evaporate during application, this
provides a cooling effect to the skin, which can be a pleasant sensory experience
especially for sun care products as they are often applied in hot conditions. This
cooling effect can be enhanced by the addition of alcohol or other volatile
components to the formulation.
W/O emulsions, on the other hand, with oil as the external phase, tend to feel more
oily or greasy upon application and are generally perceived as heavier. In some
niche applications, this can be welcome, for example, in sun care products for winter
sports use, where this more occlusive feel gives a greater sensation of protection in a
cold environment. Another application in which the sensory feel of W/O emulsions
can be preferred is in baby sun care products, where the protective sensation gives
parents a sense of reassurance that they have protected their child. In most beach sun
care products, though, W/O emulsions have traditionally been seen as the less pre-
ferred option in terms of skin feel. However, innovations in W/O emulsifier technol-
ogy [7, 8] mean that it is now possible to formulate W/O emulsions which have a
more elegant skin feel. This allows the sun care formulator to take advantage of the
benefits of W/O systems, such as water resistance and increased efficacy of actives
while still delivering a formulation that is aesthetically pleasing for the consumer.
In any cosmetic emulsion, the spreadability of the oil components exerts a significant
influence on skin feel, in particular during the application of the product. Bruening
et al. [9] described how it is desirable to impart a sensation of smoothness to the skin
16 Sunscreen Formulation 293
There are a number of water-soluble UV filters, but only the following are of
significant commercial importance nowadays:
Benzophenone-4 (UVB/UVA filter)
Disodium phenyl dibenzimidazole tetrasulfonate (UVA filter)
Phenylbenzimidazole sulfonic acid (UVB filter)
Benzylidene camphor sulfonic acid (UVB filter)
Terephthalylidene dicamphor sulfonic acid (UVA filter)
Of these, the last two are proprietary to LOreal, and so for most formulators, the
first three in the above list are the only water-soluble filters available. These all
require neutralisation with a suitable base in order to render them soluble. In terms
of sensory properties, these water-soluble sunscreens tend to have a drying effect on
skin feel; this is often advantageous, but the feel can be excessively dry if a high
concentration of such filters is used.
A relatively new class of sunscreen actives are organic UV filters that are not soluble
in either oil or water and remain in a particulate form in the final formulation. The
first of these was methylene bis-benzotriazolyl tetramethylbutylphenol (MBBT, or
bisoctrizole), which is primarily a UVA filter but has a secondary absorption peak in
the UVB. In 2014, a second material was added to this class of UV filters when tris-
biphenyl triazine (TBPT) was approved in Europe. TBPT gives high absorbance in
the UVB and UVA2 (320-340 nm), the latter being a wavelength region that is not
well covered by most other UV filters. Both of these materials are supplied as
aqueous nanoparticulate dispersions. With regard to sensory properties, both tend to
have a drying effect on skin feel (although less so than most water-dispersed
inorganic sunscreens). Also, being particulates, both can give rise to an undesirable
sensory attribute that is not observed with other organic UV filters, namely,
whitening. Both MBBT and TBPT give a low but significant light extinction in the
visible region of the spectrum; since they are most typically used at relatively low
levels in formulations, this is usually not noticeable, but at higher levels (above 5 %
active), a discernible whitening effect may be observed.
Titanium dioxide (TiO2) and zinc oxide (ZnO) have been used as sun-protective
agents for many years, but it was only with the development of the first fine
particle (a.k.a. microfine, ultrafine) grades of these materials in the late 1980s
16 Sunscreen Formulation 295
Historically, the biggest problem with inorganic sunscreens was whitening on skin.
The original UV-attenuating grades of TiO2 and ZnO that were developed for
personal care use, despite their fine particle size, still often gave a noticeable white
film on skin, especially when incorporated at the concentrations required for SPF
values above 15. In some applications, this is actually perceived as an advantage; for
example, young children tend not to be concerned about the cosmetic appeal of the
sun cream applied to them, but their parents like to be able to see that they have
protected their child from the sun. Those same parents, however, hate to see an
unsightly white film on themselves!
Subsequently, further development of inorganic sunscreens resulted in improve-
ments in transparency. Dransfield et al. [15] discussed how advances in the
296 J.P. Hewitt
technology of titanium dioxide for sunscreens, relating to both the manufacture and
the formulation of fine particle TiO2, resulted in improved transparency. The theory
of light attenuation by titanium dioxide [16] shows that this material becomes pro-
gressively more transparent to visible light as the mean particle size is reduced; with
a mean particle size of 20 nm (0.02 m), TiO2 is essentially completely transparent.
However, with a typical particle size distribution, such a product has very low UV
attenuation, so it has poor efficacy as a sunscreen. This was demonstrated by
Woodruff [17], who compared various grades of titanium dioxide in a standard
frame formulation. Included in this study were two aqueous dispersions of TiO2,
one of which had a mean particle size of 4050 nm and the other a mean particle
size of 1020 nm. While the latter product showed a high degree of transparency, it
gave relatively poor SPF performance, with an SPF of only 7.3 from 5 % TiO2 solids
(compared to 22.6 for the other dispersion at the same solids content).
Dransfield et al showed that by using appropriate manufacturing methods for the
TiO2 and by optimising surface treatments (coatings), solids level, dispersants and
milling processes, it is possible to produce titanium dioxide dispersions which
maintain the optimum mean particle size for UV attenuation but which have a
narrower particle size distribution than previously. Such dispersions therefore have
greater transparency to visible light, but without any loss of UV performance.
A similar approach has also been applied to zinc oxide, providing high
transparency dispersions of this material also. Another approach to making a
transparent zinc oxide is by the use of refractive index matching [18]. In this
technology, the ZnO particles are actually much larger than conventional sunscreen
grades of ZnO (of the order of a micron or more) but have a porous structure that
provides closer matching of refractive index between the particles and the emollient
in which they are dispersed, thus reducing the scattering of visible light and giving
improved transparency.
Even these more transparent materials, however, still need to be formulated
correctly in order to realise the improved transparency. The particle size and size
distribution need to be maintained, as far as possible, in the final formulation; if the
particles agglomerate, they then behave optically as larger particles, and so
whitening is increased. The SPF efficacy of inorganic sunscreens, and also
transparency, can be influenced by emulsifiers, added emollients, rheological
additives and polymers. Each of these can affect the SPF either by influencing the
dispersion degree of the active or by affecting the rheology and spreading properties
of the formulation. For example, the use of waxes to alter rheological properties has
been shown to have a dramatic effect on SPF in W/O emulsions [19]. We can
determine, in at least a qualitative fashion, the relative influences of these two
mechanisms by looking at changes in the UV/visible spectrum as parameters are
altered. If SPF varies solely as a result of changes in rheological/spreading
properties, the shape of the spectral curve does not change, indicating that the
dispersion degree of the TiO2 has not changed. If changing a particular ingredient
does affect the degree of dispersion, this is reflected in a change in the shape of the
UV attenuation curve, as well as a change in SPF. For example, if the dispersion
16 Sunscreen Formulation 297
degree of the TiO2 is improved, the following changes typically occur in the
spectrum:
UVB attenuation increases.
UVA attenuation decreases.
Visible attenuation decreases.
As a result, SPF increases, UVA/UVB ratio decreases, and whitening also
decreases [20]. In other words, ensuring the optimum dispersion of TiO2 promotes
both high SPF and optimum transparency.
The other aesthetic issue to be addressed with inorganic sunscreens is skin feel. The
earliest inorganic sunscreen formulations often had less-than-ideal skin feel, giving
inorganic filter systems a reputation for being dry, draggy, heavy, or sticky.
Fortunately, considerable progress has been made in improving the skin feel of
these systems.
Parameters relating to particulate filters that might be expected to influence skin
feel include particle size, surface treatments, and, in the case of dispersions, the
carrier medium in which the particles are dispersed. A study of formulations
containing oil-dispersed and water-dispersed TiO2 [21] indicated that variations in
particle size at least within the range of sizes typically seen in UV-attenuating
grades of TiO2 have little significant influence on skin feel.
The surface properties of the particles, however, do influence skin feel. All
modern UV-attenuating grades of TiO2 are surface-treated with one or more coating
materials; the main purpose of these coatings is to prevent photocatalytic activity,
but they also aid the dispersibility of the particles and affect sensory properties. The
surface treatments can be either hydrophilic or hydrophobic in nature. Hydrophilic
coatings are typically other inorganic oxides such as silica, alumina, or zirconia.
Hydrophobic surface treatments include organic moieties such as stearate,
organometallics such as isopropyl titanium triisostearate, silicones such as
dimethicone and silanes such as triethoxycaprylylsilane. Not all zinc oxide grades
are coated; ZnO has less photocatalytic activity than TiO2, so there is less of a
requirement for coating in this case. However, many grades do have a surface
treatment to aid dispersion and/or feel. The coating materials used are similar to
those used for TiO2.
Inorganics with a hydrophilic surface tend to impart a dry skin feel, which can
be perceived as draggy when the particles are dispersed in the water phase. This
effect is lessened when the particles are dispersed in the oil phase, particularly if an
effective dispersing agent is included. These dispersing agents are usually surfactants
(often polymeric), which bind to the particle surface and effectively change a
hydrophilic surface to a hydrophobic one.
298 J.P. Hewitt
Of course, with the exception of inorganic-only sun care products (e.g. those making
natural claims or products designed for sensitive skin and/or young children), it is
nowadays very unusual for a sun care product to contain only one active ingredient.
Even low SPF products usually contain a combination of two or more UV filters,
and such combinations are essential for higher SPF products, especially bearing in
mind current requirements for UVA protection (both regulatory and market driven).
It is here that the skill of the sun care formulator in optimising the SPF efficacy of
the formulation also plays a part in optimising the sensory properties.
16 Sunscreen Formulation 299
It is intuitively expected that the higher the SPF, the more oily and/or greasy will
be the skin feel of a sun protection product. However, the study by Vollhardt et al.
[6] indicated that while this may be the case within a single product line, it is not
generally true. The fact that it is not can be attributed at least in part to finding the
right combinations of sunscreen actives to maximise efficacy by taking advantage
of synergies between different UV filters, allowing, for example, SPF 50 to be
achieved with active levels only slightly higher than those needed for SPF 30 [24].
The best synergies are achieved where filters complement each other, for example:
Combine filters that cover different parts of the UV spectrum, to ensure broad-
spectrum protection.
Combine organic filters with inorganic filters, which has been shown to generate
significant synergistic effects [2529].
Combine water-based filters with oil-based filters.
The last of these is of particular interest. The use of water-soluble UV filters, or
aqueous dispersions of inorganic filters, is often avoided in beach products due to
concerns over lack of water resistance. However, the addition of low levels of water-
based filters to a formulation containing an optimised combination of oil-based
filters can give dramatic increases in SPF [24]. Also, the characteristic dry skin
feel of the water-based filters helps to counteract the oily feel associated with, for
example, the liquid organic UV filters.
Of course, the aesthetic properties of sun care formulations are influenced to a large
degree by the other components used as well as by the UV filters themselves. The
effects of emollients have been discussed in the preceding sections; the following is
a brief discussion of how other excipients can affect sensory properties.
16.7.1 Emulsifiers
What is often not appreciated is the effect that emulsifiers have on the sensory
properties of topical skin care products. In fact, it has been demonstrated that during
the rub-out phase, emulsifiers actually exert a greater influence on the skin feel than
the emollients do [30]. Many traditional O/W emulsifiers produce a skin feeling
during rub-out that is more waxy than is ideal for sun care formulations, but
nowadays there are plenty of emulsifier systems that give a more suitable feel. One
example is potassium cetyl phosphate, which facilitates good spreadability upon
application, with a smooth after-feel. It can be combined with co-emulsifiers to
deliver a range of textures and viscosities, from viscous lotions to thin sprayable
milks.
300 J.P. Hewitt
Another class of emulsifiers that are well suited for sun care are those designed
to form liquid crystal networks [3133]. Liquid crystals, in one form or another,
have actually been present in most O/W personal care emulsions for many years, but
it is only within the last 25 years or so that they have been recognised as such, and
formulators have started to deliberately make use of them to achieve specific effects.
Lamellar liquid crystalline phases have been shown to significantly improve
emulsion stability. They also have prolonged hydration properties, due to the fact
that water is bound into the lamellar structure, making it less prone to immediate
evaporation. This helps to give a skin feel that is very well-liked by consumers. For
sun care, the most suitable type of liquid crystal systems is hydrosomes, which
consist of a delocalised network of lamellar liquid crystal structures. In sun care, the
delocalised structure helps to achieve a homogeneous distribution of active
ingredients [34], thus increasing SPF efficacy. In terms of sensory properties, such
systems typically give a light and silky skin feel with excellent skin play.
16.7.2 Thickeners
There are many different types of rheology modifiers that are used in cosmetic O/W
emulsions, including acrylate polymers (e.g. carbomers), natural gums such as
xanthan gum, cellulose derivatives, silicate types such as magnesium aluminium
silicate and starch-based thickeners. Each has its own sensory characteristics, but
the optimum type to use in any given case depends very much on the emulsifier
system being used.
In W/O emulsions, waxes are often used as thickeners and as mentioned earlier
can have a beneficial effect on SPF in sun care formulations [19]. However, care
should be taken to avoid excessive concentrations of wax as this can inhibit the
spreading of the formulation, making it difficult and unpleasant to apply. Fine
particle silica can also be used as a rheological additive in W/O systems, and this
can have a beneficial effect on skin feel, as it counteracts any oiliness or greasiness
from the emollients, delivering a drier feel.
16.7.3 Film-Formers
Film-formers, which are often polymers, are frequently added to sun care
formulations for one or both of two reasons. Firstly, such ingredients can act as SPF
boosters, by giving a more even product film on skin. Secondly, they are used as
water-proofing agents. One of the most common types used are PVP copolymers
[35, 36], for example, VP/eicosene copolymer. However, these polymers can
sometimes give a sticky feel to the formulation, so a number of alternatives have
been developed that confer water resistance while enabling the formulator to
maintain a light, smooth skin feel [37].
16 Sunscreen Formulation 301
16.8 Conclusion
There was a time when sun care products would not be very pleasant to apply and
would not be as cosmetically elegant as, say, daily skin care products, and consum-
ers would either accept this as a necessary evil of protecting themselves from the
sun or would avoid using such products altogether. Nowadays, however, consumers
expect a better sensory experience from their sunscreen products, and manufactur-
ers are increasingly using sensory claims as a way of differentiating their products
from the competition. Sensory analysis by trained panels enables cosmetic scien-
tists to better understand what consumers want in terms of skin feel, and develop-
ments in terms of both active ingredients (UV filters) and formulation excipients are
enabling formulators to develop sun care products that are pleasant to apply, encour-
aging better consumer compliance. This, in turn, makes the products more effective
under real-use conditions than may have been the case in the past.
References
1. Petersen B, Datta P, Philipsen PA, Wulf HC (2013) Sunscreen use and failures on site obser-
vations on a sun-holiday. Photochem Photobiol Sci 12(1):190196
2. Saecker C, Janssen A, Vollhardt J (2013) Are we done by preventing sunburn? In: Proceedings
of the Florida Sunscreen symposium, Orlando, 1821 Sept 2013
3. Ou-Yang H, Stanfield J, Cole C, Appa Y, Rigel D (2012) High-SPF sunscreens (SPF 70) may
provide ultraviolet protection above minimal recommended levels by adequately compensating
for lower sunscreen user application amounts. J Am Acad Dermatol 67(6):12201227
4. Bimczok R, Gers-Barlag H, Mundt C, Klette E, Bielfeldt S, Rudolph T, Pflucker F, Heinrich
U, Tronnier H, Johncock W, Klebon B, Westenfelder H, Flosser-Muller H, Jenni K, Kockott D,
Lademann J, Herzog B, Rohr M (2007) Influence of applied quantity of sunscreen products on
the sun protection factor a multicenter study organized by the DGK Task Force Sun
Protection. Skin Pharmacol Physiol 20(1):5764
5. European Commission (2006) Recommendation of 22 Sep 2006, on the efficacy of sunscreen
products and the claims made relating thereto. Official Journal of the European Union L
265/39L 265/43
6. Vollhardt J, Schoop R, Roos F, Janssen A, Saecker C (2013) The sensory experience of sun-
screens which properties matter? A descriptive sensory analysis of more than 50 sunscreens.
In: Proceedings of the sun protection conference 2013: sun protection for the 21st century,
London, 45 June 2013 (Summit Events Ltd.)
7. Dahms GH (1988) Cosmetic water-in-oil emulsions: application, physical properties and for-
mulations. In: Proceedings of the symposium on The formulation of cosmetic and topical
pharmaceutical products, Bristol, April 1988
8. Tadros ThF et al (1996) Use of a polymeric surfactant (ABA block copolymer) for preparation
of stable water-in-oil (W/O) emulsions and water-in-oil-in-water multiple emulsions. In:
Proceedings of the 19th IFSCC congress, Sydney, October 1996
9. Bruening S, Leonard M, Kawa R, Issberner U, Tomlinson A (2005) Role of emollients and
emulsifiers in sunscreen formulations. In: Shaath NA (ed) Sunscreens: regulations and
commercial development, vol 28, 3rd edn, Cosmetic science and technology series. Taylor &
Francis, Boca Raton, pp 449460
10. Schwarzenbach R, Huber U (2003) Optimization of sunscreen efficacy. In: Verlag fr che-
mische Industrie, H. Ziolkowsky GmbH, Augsburg, pp 131137
302 J.P. Hewitt
11. Wright C (2002) Effects of emollients on efficacy of UV filters. MChem report, York University
12. Hewitt JP (2013) Formulating sun protection products for the 21st century: efficacy, elegance
and globality. In: Proceedings of the Florida Sunscreen Symposium, Orlando, 1821 Sept
2013
13. Nohynek GJ et al (2007) Grey goo on the skin? Nanotechnology, cosmetic and sunscreen
safety. Crit Rev Toxicol 37(3):251277
14. Chen LL, Tooley I, Wang SQ (2013) Nanotechnology in photoprotection. In: Nasir A,
Friedman A, Wang SQ (eds) Nanotechnology in dermatology. Springer, New York, pp 918
15. Dransfield GP, Hewitt JP, Lyth PL (1998) Advances in titanium dioxide technology. In:
Proceedings of the SCS spring conference, London, April 1998
16. Robb JL, Simpson LA, Tunstall DF (1994) Scattering and absorption of UV radiation by sun-
screens containing fine particle and pigmentary titanium dioxide. Drug Cosmet Ind
154(3):3239
17. Woodruff J (1994) Formulating sun care products with micronized oxides. Cosmetics &
Toiletries Manufacture Worldwide, London, Aston Publishing Group, pp 179185
18. McCormick P, Fletcher R, Jones C (2010) Micron-sized zinc oxide dispersion for transparent,
high SPF formulas. Cosmet Toil 125(2):5057
19. Hewitt JP, Dahms GH (1995) The influence of rheology on efficacy of physical sunscreens. In:
Proceedings of the IFSCC conference, Montreux, pp 313323
20. Hewitt JP (1999) Effective use of physical sunscreens: incorporation of physical sunscreens
into water-in-oil emulsions. In: Proceedings of the European UV sunfilters conference, Paris,
November 1999
21. Hewitt JP, Hine H, Kessell LM (2004) Optimising the sensory properties of physical sunscreen
formulations. In: Proceedings of the 23rd IFSCC congress, Orlando, 2427 Oct 2004
22. Kessell LM, Hewitt JP (2008) Water-in-silicone-oil emulsion for use as a sunscreen product.
US patent 2008226727, 18 Sept 2008
23. McNeil K, Sayer R, Miles A (2014) Assessing the sensory performance of novel coating tech-
nology. SOFW J 140(7):27
24. Hewitt JP, Teague J, Sutcliffe P (2014) Efficacy and elegance in 21st century sun care. Cosmet
Toil 129(3):5865
25. Dahms GH (1994) Compositions containing sunscreens. UK patent 2278055, 23 Nov 1994
26. Dahms GH (1994) Compositions containing sunscreens. UK patent 2279007, 21 Dec 1994
27. Nicoll GA, Scott IR (1991) Cosmetic composition. European patent 0456458, 13 Nov 1991
28. Nearn MR, Redshaw SJ, Burgess G (1995) Sunscreen compositions. US patent 5417961, 23
May 1995
29. Deckner GE (1994) Photostable sunscreen compositions. International patent WO9404131, 3
Mar 1994
30. Wiechers JW, Taelman M-C, Wortel VAL, Verboom C, Dederen JC (2002) Emulsifiers and
emollients exert their sensory impact in different phases of the sensory evaluation process but
how does one demonstrate the absence of such an influence? IFSCC Magazine 5(2):99105
31. Dahms GH (1986) Properties of oil-in-water emulsions with anisotropic lamellar phases.
Cosmet Toil 101(11):113115
32. Dahms GH (1990) Optimised formulations for skin care products. SOFW J 1990(10):
388392
33. Loll P (1994) Liquid crystals in cosmetic emulsions. Cosmetics & Toiletries Manufacture
Worldwide, Aston Publishing Group, London, pp 108120
34. Dahms GH (1992) Formulating with a physical sun block. Cosmet Toil 107(10):8792
35. Gupta VK, Zatz JL (1999) In vitro method for modelling water resistance of sunscreen formu-
lations. London, J Cosmet Sci 50:7990
36. Markovic B, Laura D, Rerek M (2001) A laboratory method for measuring the water resistance
of sunscreens. Cosmet Toil 116(9):6168
37. Rigano L (2014) Very water-resistant sunscreens. Cosmet Toil 129(3):6673
Chapter 17
Sunscreen Regulatory Update
Farah K. Ahmed
Key Points
1. Sunscreen active ingredients (or ultraviolet (UV) filters) are regulated
globally under a variety of classifications e.g., over-the-counter drugs,
cosmetics, quasi-drugs and are required to be substantiated for safety and
efficacy.
2. Brief overview of the current state of sunscreen regulation around the
world, with an expanded focus in the United States, will be provided.
3. Worldwide regulation of sunscreens ensures governmental oversight over
the safety and efficacy of sunscreens.
17.1 Introduction
Sunscreen has shown to reduce the risk of developing skin cancer and prevent
UV-induced skin aging when used appropriately and in conjunction with other pro-
tection modalities. In the United States, sunscreens are regulated by the Food and
Drug Administration (FDA) as over-the-counter (OTC) drugs. In many other
countries around the world, there are sets of rigorous specifications governing the
Founded in 1894, the Personal Care Products Council is the leading national trade association
representing the personal care products industry. Our membership includes approximately 300
active member companies that manufacture or distribute personal care products, including OTC
sunscreens. We also represent approximately 300 additional associate members who provide
goods and services to manufacturers and distributors of personal care products.
F.K. Ahmed, BS, JD (*)
Vice President / Chair, Personal Care Products CouncilConsumer Healthcare Products
Association Joint Sunscreen Task Force, Personal Care Products Council,
Washington, DC, USA
e-mail: [email protected]
safety profile of UV filters and product efficacy and labeling guidelines in place to
protect the consumers. To accomplish these tasks, there are regulatory bodies around
the world that create and update their individual rules pertaining to the regulation of
sunscreen products.
This chapter provides a glimpse to the world of regulation associated with sun-
screens. Specifically it will provide (1) a basic background of over-the-counter
(OTC) drug product regulation in the United States, (2) specific information on
sunscreen regulations, (3) an overview of the Sunscreen Innovation Act (SIA), and
(4) a brief summary of global sunscreen ingredient and product regulation.
In the United States, sunscreens primarily fall within the jurisdiction of the US
Food and Drug Administration (FDA or the Agency). FDA regulates all sun-
screens as OTC drugs.1 As such, they must meet standards for safety, efficacy,
good manufacturing practices (GMPs), and labeling. The Agency deems any
topically applied product claiming a sun protection factor (SPF) to be a sun-
screen. Examples of such products include lotions, sprays, daily moisturizers,
foundations, lipsticks, etc.
Two regulatory pathways exist for the legal marketing of OTC drug products: (i)
marketing in compliance with an OTC drug monograph, (ii) marketing under the
authority of an approved product-specific new drug application (NDA), an
abbreviated new drug application (ANDA), or (iii) Rx-to-OTC switch.
The majority of sunscreen products in the United States are marketed under the
OTC sunscreen monograph. An OTC monograph is essentially the recipe for mak-
ing an OTC drug product and based on ingredients FDA preapproved (or permit-
ted) active ingredients that support prescribed labeling claims and, in some
instances, testing (e.g., SPF). Designated OTC monographs represent regulatory
standards for the marketing of nonprescription drug products not covered by new
drug applications. These are OTC drugs related to categories that consumers are
able to self-diagnose, self-treat, and self-manage. Examples of OTC monograph
1
An OTC drug product is a drug product marketed for use by the consumer without the interven-
tion of a health care professional in order to obtain the product.
17 Sunscreen Regulatory Update 305
therapeutic categories include both topical and ingested forms such as sunscreens,
acne, allergy, diaper rash, cough and cold, antiperspirant, dandruff, skin protectant,
external analgesic, psoriasis, etc.
Currently, there are four specific sunscreen formulations approved in the United
States under the new drug application (NDA) process:
Anthelios SX: avobenzone, ecamsule, and octocrylene at 2, 2, and 10 %
Capital Soleil: avobenzone, ecamsule, and octocrylene at 2, 3, and 10 %
Anthelios 20: avobenzone, ecamsule, octocrylene, and titanium dioxide at 2, 2,10,
and 2 %
Anthelios 40: avobenzone, ecamsule, octocrylene, and titanium dioxide at 2, 3, 10,
and 5%
All four NDAs were filed by LOreal.
The term human drug application means an application for approval of a new
drug (the full formulation and labeling).
The following table (Table 17.1) provides a summary of the differences of these
two pathways.
On June 14, 2011, FDA released the following sunscreen-related rulemakings: (1)
final rule on effectiveness testing and labeling for over-the-counter (OTC) sun-
screen products (final rule),2 (2) proposed rule on SPFs above 50, (3) advance
notice of proposed rulemaking (ANPR) on sunscreen dosage forms, (4) draft
guidance on OTC sunscreen drug products, and (5) request for comment on the
final rule.3
The final rule outlines permitted and required claims, testing procedures required to
substantiate those claims, and claims that are not permitted. It is important to note
that these rules amend FDAs drug labeling regulations (i.e., 21 CFR 201) and do
not finalize the sunscreen monograph (i.e., 21 CFR 352) nor lift the stay on the
implementation of the monograph.
In addition, the final rule lifts the delay of the implementation of the 1999 Drug
Facts final rule and requires all sunscreen products to comply with the content and
format requirements of that rule. This includes combination cosmetic sunscreen
products such as lipsticks, foundations, and daily moisturizers that are labeled as
containing an SPF.
Under the Drug Facts rule, if the information listed under Drug Facts requires
more than 60 % of the total available surface area, the Drug Facts labeling can be
reduced as specified in the regulation.4 FDA did not provide for any additional
labeling relief under the Final Rule.
2
The final rule (76 FR 35620), codified in 201.327, establishes labeling and testing requirements
for OTC sunscreen products marketed without approved applications and containing only the
ingredients specified in the stayed 1999 final rule (aminobenzoic acid (PABA), avobenzone, cinox-
ate, dioxybenzone, ensulizole, homosalate, meradimate, octinoxate, octisalate, octocrylene, oxy-
benzone, padimate O, sulisobenzone, titanium dioxide, trolamine salicylate, zinc oxide).
3
All published in the June 17, 2011 Federal Register.
4
21 CFR 201.66(d)(10)
17 Sunscreen Regulatory Update 307
The final rule does not address issues related to sunscreen active ingredients,
including any new active ingredient combinations, or any sunscreen active
ingredients currently under time and extent application (TEA) review.
The final rule effective date was initially set for June 18, 2012, except for products with
annual sales less than $25,000 for which the effective date is June 17, 2013. FDA post-
poned these dates by 6 months to allow companies to comply with the final rule and
ensure no shortage of sunscreens on the market. All products labeled on or after the
effective date must meet all final rule requirements (see below for additional time/
enforcement discretion for SPF testing). Of note, FDA did not require noncompliant
products introduced or delivered for introduction into interstate commerce prior to the
compliance date, June 18, 2012, to be removed from the market; product delivered to
customers, even if in their warehouses, ready to be shipped from manufacturers ware-
houses, or imported prior to June 18, 2012, can continue to be shipped and sold; and
product imported prior to the compliance date would be protected, as would any prod-
uct delivered to customers, even if still in customers warehouses on the effective date.5
The SPF test method was modified to require a smaller number of test subjects to deter-
mine a products SPF (10) compared to the previous methods that required 2025 test
subjects. The reference control formulation was changed from an SPF 4 formulation to
an SPF 15 formulation. The finger cot used for sample application no longer requires
pre-saturation with test product. The minimum size of the test site for product applica-
tion was reduced in area, as was the required minimum area for each individual UV
exposure. The distance between exposure sites in the test area was reduced. Product
application remained at 2 mg/cm2 with test result read at 1624 h postexposure. Solar
simulator specifications were harmonized with those in the International SPF Method.
5
Under the general interpretation of delivered for introduction into interstate commerce, other
warehoused product might also be protected but would have to be evaluated on a case-by-case
basis.
308 F.K. Ahmed
FDA has abandoned the four star rating proposal indicating UVA protection pro-
vided on product labels, in favor of a simple pass/fail in vitro test for broad-
spectrum characteristics known in the industry as the critical wavelength test. The
proposed test methodology differs from previously published methodology to deter-
mine the critical wavelength in several attributes and is different from the ISO in vitro
UVA test method (in development) as well.
A sunscreen must have a critical wavelength of 370 or higher to be able to make
a broad-spectrum claim. A broad-spectrum claim is necessary in order to make
a positive use statement regarding prevention of early skin aging and skin cancer
on products with an SPF of at least 15; otherwise, a warning statement must be used
for the product uses (see below).
The FDA critical wavelength test method prescribes the use of PMMA plates with
a surface roughness from 2 to 7 m, with a sunscreen application density of 0.75 mg/
cm2, and pre-irradiation of the sample with a fixed 4 MED exposure to solar-simulated
radiation. The wavelength at which 90 % of the UV absorbance area under the curve
occurs (when summing from 290 toward 400) is defined as the critical wavelength
and is a measure of the breadth of the protection provided by the product.
The FDA also issued new guidance on labeling of sunscreen products as outlined below.
Uses (indications)
Helps prevent sunburn
If used as directed with other sun protection measures (see Directions),
decreases the risk of skin cancer and early skin aging caused by the sun
[please note: the sunscreen must be broad-spectrum and SPF of at least
15 in order to use this statement]
Warnings
Skin cancer/skin aging alert: Spending time in the sun increases your risk of
skin cancer and early skin aging. This product has been shown only to help
prevent sunburn, not skin cancer or early skin aging [please note: this
statement is required for products that are not labeled as broad-spectrum or
SPF of less than 15]
Directions (for broad-spectrum/SPF 15 and water-resistant)
Apply liberally (or generously and may add and evenly) 15 min before
sun exposure.
Reapply.
After 40 (or 80) minutes of swimming or sweating
17 Sunscreen Regulatory Update 309
6
The agencys reasoning for this allowance is that consumers who are using these products primar-
ily for cosmetic use may be more likely to understand that they might not receive the intended sun
protection if they do not follow the directions in the Drug Facts label.
310 F.K. Ahmed
Fig. 17.1 Illustrates the old vs. the new sunscreen labeling requirements
Water resistance claims on the principal display panel must specify either 40 or
80 min of effectiveness while swimming or sweating, based on testing. Waterproof,
sweatproof, and sun block claims are not permitted. FDA did not explicitly
allow for a sweat resistance claim (Fig. 17.1).
Although FDA acknowledged that SPFs higher than 50 have been substantiated
and results are validated and repeatable, it is proposing to limit SPF to 50+
unless the agency receives data demonstrating additional clinical benefit for
SFPs above 50.
Sunscreens labeled with SPFs above 50 may remain on the market until this
proposed rule becomes final, provided they follow the appropriate SPF test.
Depending on how this proposed rule is finalized, these products may/may not be
able to continue on the market.
17 Sunscreen Regulatory Update 311
Amiloxate, 10 %
Bemotrizinol, 10 %
Bisoctrizole, 10 %
Diethyl butamido triazone, 3 %
Drometrizole trisiloxane, 15 %
Ecamsule, 10 %
Enzacamene, 4 %
Octyl triazone, 5 %
As of the date of this publication, FDA has not approved any of the above
ingredients; rather, the Agency has requested additional data before it can make its
safety and efficacy determination.
On November 26, 2014, President Obama has signed the Sunscreen Innovation Act
(SIA) into law.7 The goal of the SIA is to provide an alternative process for review for
all ingredient TEAs, including prescribed timelines for review, administrative orders
in lieu of rulemaking, and new format for data submissions. The SIA also allows for
advisory committees and requires FDA to regularly update congress and the GAO. Of
note, FDAs safety evaluations and determinations remain with the Agency.
Key aspects of the Act include:
Determining eligibility: FDA TEA eligibility requirements will be maintained an
ingredient must be used safely for at least five years in at least one country.
Eligibility determinations will be made by FDAs Division of Nonprescription
Regulation Development (DNRD). Pending ingredient submissions, already
deemed eligible by FDA, will be considered eligible for the new review and
approval process.
Transparent review: After a finding of eligibility, the ingredient application may be
submitted to the existing FDA Nonprescription Drugs Advisory Committee
(NDAC) for a safety and effectiveness recommendation or may conduct this
review on their own. During the review process, the FDA or the NDAC will
receive data from the public and communicate with the applications sponsor to
seek clarifying or request additional information. FDA will either concur or deny
the NDACs recommendation or come to its own conclusion.
Predictable and reasonable time frame: The SIA sets time frames for the various
stages of the TEA process for both pending and new applications.
Guidance: FDA must issue draft guidance on the implementation of, and compli-
ance with, the requirements with respect to sunscreen TEAs under the Act (e.g.,
format, data requirements).
7
Sunscreen Innovation Act: http://www.gpo.gov/fdsys/pkg/PLAW-113publ195/pdf/PLAW-
113publ195.pdf
17 Sunscreen Regulatory Update 313
On or before May 26, 2018: FDA second report to senate HELP and House E&C
regarding implementation of the SIA.
On or before November 26, 2019: Final sunscreen monograph published.
On or before May 26, 2020:
Subsequent Government Accountability Office (GAO) report.
FDA third report to senate HELP and House E&C regarding implementation
of the SIA.
Figures 17.2 and 17.3 illustrate the new TEA timeline8
8
Images by FDA
17 Sunscreen Regulatory Update 315
Sunscreens are recognized by global health authorities for their ability to pro-
tect consumers from UV exposure and for their role in helping to prevent acute
and chronic damage to the skin, including reducing the incidence of skin can-
cers. These authorities are aligned in that (i) sun protection is a public health
priority, (ii) claims regarding efficacy must be proven, and (iii) sunscreen active
ingredients are reviewed for safety and require premarket approval. While the
above criteria are common among regions, regulatory classifications, available
sunscreen active ingredients and concentration limits, testing requirements,
labeling, approval process and postmarketing requirements differ. Each of these
parameters will be reviewed below for their commonality as well as for their
divergence.
Regulatory classifications for sunscreens vary widely from region to region. At first
glance, one might believe that differences among these classifications are vast; how-
ever, examination of the various definitions reveals common themes. These include
the following: a product placed in contact with the various external parts of the
human body and a product to protect the skin from UV radiation. These similarities
support the common message that sunscreen products, when used as directed, no
matter what the regulatory classification, are designed to provide UV protection to
consumers and sunscreen active ingredients require premarket approval and sup-
porting preclinical/clinical information.
The area where the greatest differences are most evident is in the wide variety of
sunscreen ingredients available to manufacturers and consumers. In the United
States, the monograph allows for 16 current sunscreen ingredients with eight
pending time and extent applications (TEAs). In other countries, up to 38 sun-
screen ingredients are available for manufacturers to formulate sun protection
products. Approvals of sunscreen active ingredients require premarket approval
and supporting preclinical/clinical information. While some approvals appear to
be more stringent, the safety and efficacy of the sunscreen ingredients must be
proven prior to use of the ingredient. This is further discussed in an earlier section
of this briefing document.
316 F.K. Ahmed
Upon examination, ten sunscreen ingredients are permitted for use globally:
1. Octisalate (ethylhexyl salicylate)
2. Homosalate (3,3,5-trimethylcyclohexyl 2-hydroxybenzoate)
3. Octocrylene (2-cyano-3,3-diphenyl acrylic acid, 2-ethylhexyl ester)
4. Octinoate (octyl methoxycinnamate)
5. Zinc oxide*
6. Oxybenzone (benzophenone-3)
7. Ensulizole (phenylbenzimidazole sulfonic acid)
8. Avobenzone (butyl methoxy dibenzoyl methane)
9. Padimate O (ethylhexyl dimethyl PABA)
10. Titanium dioxide*
*Considered to be light-scattering agents in Japan
Globally, all sunscreen active ingredients require premarket approval, and all have
individually been tested in preclinical studies in both short-term and long-term
applications. Within limits determined by individual countries, all have demonstrated
to have acceptable preclinical and/or clinical safety profiles for human use as
directed.
While there is no global agreement as to the SPF limits or the regulatory classification
of specific product types (recreational vs. every day), all sunscreen products no
matter where they are marketed must carry an SPF value. Communication of the
level of UVA protection can vary from a + marking in Japan, the SPF value and a
symbol indicating UVA in Europe, to a broad-spectrum claim in the United
States. All of these statements to the consumer appear on the principal display panel
of the sunscreen product.
All products need to provide information to consumers allowing safe use.
Warning statements and directions for use should be clear and concise to ensure that
even the average consumer can read and understand any risks associated with the
product as well as how to properly apply and use.
As discussed in this section, while the regulatory classification of sunscreens
varies globally, all major markets agree that sun protection is a public health priority
and consider sunscreens a necessary component in preventing both sunburn and
skin cancer. These authorities are aligned in that claims regarding efficacy must be
proven and that sunscreen active ingredients require premarket approval.
17 Sunscreen Regulatory Update 317
17.6 Conclusions
Both the data and regulatory requirements in the United States and globally related
to sunscreens substantiate and ensure the safety and efficacy of these products. In part,
regulations are meant to protect and enhance public health. They also play a major
role in advancing and at time slowing or impeding the technological and scientific
progresses that propel companies and industries to develop novel sunscreen products.
Needless to say, regulations can impact the state of photoprotection offered by
sunscreens.
Chapter 18
Measuring Sunscreen Protection According
totheFDA Final Rule
Key Points
1. The sun protection factor (SPF) is an invivo test that estimates the
protective efficacy of a sunscreen against erythema.
2. The critical wavelength (CW) test is an invitro test that has been accepted
by the US Food and Drug Administration to measure the broad-spectrum
status of sunscreens.
3. Detailed description on the proper steps to conduct SPF and CW tests is
provided.
4. Common pitfalls in conducting SPF and CW tests are also outlined.
18.1 Introduction
Historically, the first known studies establishing the basis for sun protection started
in the 1930s and were published in the 1940s by H.Blum etal. and in the 1950s by
R.Schulze [1, 2]. Professor Franz Greiter invented what is known today as the con-
cept of the sun protection factor (SPF) and introduced sunscreens with SPF labels
in 1962 [3, 4]. The original proposed sunscreen monograph, issued in 1978,
provided the option of an outdoor SPF test using sunlight, as well as a method using
indoor ultraviolet lamps [5].
The advantages of using a natural solar light source were cited as a closer
approximation of the actual use of sunscreens, including the heat and humidity, use
of the full solar spectrum, ability to test several sunscreen products simultaneously,
and the ability to estimate tanning efficacy. Disadvantages of using a natural solar
light source included the uncontrollable variables of weather, changing cloud cover,
changing radiation intensity, changing sun angle, and difficulties of monitoring the
constant changes of sun exposure.
The disadvantages of using the indoor solar simulators, such as xenon arc lamps
included, were the low output in the visible and infrared wavelengths, the difficulty
in measuring the output, and the time-consuming requirement of irradiating only
one test site at a time. This last factor became less significant with the introduction
of multiport systems. On the other hand, the advantages of using the xenon arc lamp
included the constant spectrum and the high UV power output.
It is important to notice that the ratio of UVA (320400nm) and UVB (290320)
radiation in sunlight from 9AM to 3PM is constant and equal to 21:1 [6]. In solar
simulators, the ratio UVA/UVB is around 8:1. This also can be a source of differ-
ence when SPF tests are performed indoors using ultraviolet lamps compared to
tests performed under natural sunlight.
Eventually the consensus of sunscreen manufacturers favored the xenon arc
lamp, especially as SPF values of products began to increase significantly. During
the period of the early 1980s, large solar simulators with 1000 and 2500W were
available and utilized in much of the indoor SPF testing. However, they required
masking and covering of exposed sites, water cooling, and cumbersome power sup-
plies and large space requirements. These large simulators were eventually replaced
by compact xenon arc solar simulators, especially as large-scale SPF testing grew.
The pioneer of the compact xenon arc solar simulator was Daniel S.Berger, from
the Department of Medical Physics at Temple University [7]. The original compact
xenon arc lamp, known as the Berger solar simulator, employed a continuous 150W
xenon arc with optics and filters that produced a uniform beam, approximately one
centimeter in diameter. Its emission spectrum in the UVB region simulated the sun
at an elevation of 70. The goal was to simulate the solar spectrum in the sunburning
UVB region, with minimal long wavelength UVA, visible light, and infrared energy.
This enabled production of erythema on the backs of volunteer subjects, with
relatively short exposure times and minimal discomfort to human volunteer subjects.
The compact solar simulator weighed 5lb and required a 35lb power supply.
The compact xenon arc lamp rapidly became the mainstay of the SPF test. The
output spectrum of the compact xenon arc lamp was considered an acceptable simu-
lation of sunlight, and the FDA and other regulatory agencies worldwide stipulated
spectra for indoor sunscreen testing, designed around the compact xenon arc lamp.
A variation of the original Berger solar simulator employs six liquid light guides
to irradiate six spots, each 8mm in diameter. Use of the multiport solar simulator
was prohibited until the 2011 US FDAs Final Rule on Labeling and Effectiveness
Testing of Sunscreen was issued, because earlier FDA rules required lamp beams at
18 Measuring Sunscreen Protection According totheFDA Final Rule 321
least one centimeter in diameter. Single port and multiport compact xenon arc solar
simulators are now available in 300W versions.
The Berger solar simulator facilitated convenient indoor testing of sunscreen prod-
ucts and made it possible for sunscreen manufacturers to develop products with SPF
ratings higher than 15 and eventually as high as 100. Other than the addition of auto-
matic shutters, solid-state power supplies, and 300W power supplies, the compact
xenon arc lamp solar simulator has not changed significantly in the last three decades.
However, the cutoff filters that are required to diminish the heating of the skin by
the compact xenon arc lamp solar simulator exaggerate the long wavelength UVB
and short UVA and cut away much of the long UVA power [3]. This causes some
overestimation of the SPF measured with compact xenon arc lamp solar simulators,
compared to that of sunlight (Colipa 1994) [9]. See Fig.18.1.
To compensate for the shortcomings of the SPF alone, additional tests of
sunscreens to demonstrate protection against UVA have been developed. Most
notable is the critical wavelength test, devised by Dr. Brian Diffey and published in
1993 [9]. The critical wavelength test will be discussed in Sect.18.4.
The FDA Final Rule stipulates the procedures for measuring both the SPF and the
critical wavelength [10].
Continuous emission spectrum from 290 to 400nm
Emission spectrum measured at least annually and after replacement of lamp
bulb or any change in optical components, using an appropriate spectroradiometer
system that is calibrated to a NIST-traceable source
80 %
60 %
40 %
20 %
0%
290 300 310 320 330 340 350 360 370 380 390 400
Wavelength (nm)
Daily radiation intensity monitored before and after each test, or at least at the
beginning and end of each test day using an erythemally weighted radiometer
with a calibration consistent with the spectroradiometer system
No significant time-related fluctuations in the exposure plane (20%)
Good beam uniformity (20% from centerline reading)
UVAII (320400nm), 20% of total UV irradiance
UVAI (340400nm), 60% of total UV irradiance
Total irradiance from 250 to 1400nm 1500W/m2
The permissible ranges of the percent of erythemal dose contributions are shown
in Table18.1.
According to the FDAs Final Rule on Labeling and Effectiveness Testing, a panel
of ten subjects is required for each test product. Multiple products may be included
on the same subjects, and each subjects test must include the padimate O/
oxybenzone standard. For each test product, a maximum of three subjects may be
rejected due to test failures from the panel and replaced.
Subjects must provide an acceptable written informed consent document and a
medical history, including any instance of skin cancer, dysplastic nevi, current use
18.2.4 Procedures
Test sites are located on the subjects back, between the shoulder blades and the
beltline, and on either side of the midline. Test sites are demarcated using an indel-
ible surgical pen. There are typically four or six horizontally oriented rectangular
sites, with typical dimensions of 5cm by 10cm. The Final Rule requires an area of
at least 30cm2 for each product test site. Within each rectangular site, five sub-sites
are required, for each UV exposure. Above the rectangular sites, a space is reserved
for a horizontal row of two sets of five sub-sites for unprotected minimal erythema
doses. Irradiated sites must be separated by at least 0.8cm. See Fig.18.2. The per-
son who evaluates the results to determine the SPF should be blinded.
On Day 1, after determining that the subject is qualified for participation in the
study, the technician will administer a timed series of five UV doses, increasing in
25% increments.
After determining that the subject has no adverse response, he or she will be
instructed to avoid UV exposure and prohibited medications and given an appoint-
ment to return to the testing laboratory, within 1624h after completion of UV doses.
On Day 2, the subject will return to the testing laboratory within 1624hours after
completion of the unprotected MED doses, for evaluation of responses. The techni-
cian will question the subject nondirectively to assess compliance, to identify
Table 18.2 Fitzpatrick skin 1. Always burns easily; never tans (sensitive)
types
2. Always burns easily; tans minimally (sensitive)
3. Burns moderately; tans gradually (light brown) (normal)
4. Burns minimally; always tans well (moderate brown)
(normal)
5. Rarely burns; tans profusely (dark brown) (insensitive)
6. Never burns; deeply pigmented (insensitive)
Note: Skin type is based on first 3045min of sun exposure after
a winter season of no sun exposure
324 J.W. Stanfield et al.
18.2.7 A
pplication ofTest Products andthePadimate O/
Oxybenzone Standard Sunscreen forSPF
Determination
On Day 2, the technician will apply 2mg per cm2 of each test product and the stan-
dard in its respective designated rectangle. The sunscreens will be applied by spot-
ting the material across the area and gently spreading, using a new finger cot for
each, until a uniform film is applied to the entire area. The finger cots will not be
pre-moistened before the applications.
After at least 15min, the technician will administer a series of five progressively
increasing, timed UV doses to the sites treated with the test products and standard.
The dose series will be determined by the product of the expected SPF of each test
product and the subjects initial unprotected MED.See Table18.3.
18 Measuring Sunscreen Protection According totheFDA Final Rule 325
18.2.9 U
V Doses forRepeat Unprotected Minimal
ErythemaDose
On Day 2, the technician will administer a timed series of five UV doses, increasing
by 25% increments, to an unprotected area of the mid-back. The series of five doses
will include the initial MED in the center as shown in Table18.4.
After determining that the subject has no adverse response, he or she will be
instructed to avoid UV exposure and prohibited medications and given an
appointment to return to the testing laboratory, within 1624h after completion of
UV doses.
On Day 3, the subjects will return to the testing laboratory within 1624h after
completion of the unprotected MED doses, for evaluation of responses. The
technician will question the subject nondirectively to assess compliance, to identify
prohibited concomitant medications and UV exposures, and to identify and record
any adverse events. Then a trained evaluator, who did not participate in product
applications or administration of UV doses, will evaluate all sites that received UV
doses. The technician will determine the repeat unprotected MED as above and
compute the SPF values for the test product SPF and standard sunscreen SPF for
each subject.
The final unprotected MED used for the SPF computation will be the repeat
unprotected MED unless the repeat unprotected MED cannot be determined. In that
case, the initial unprotected MED will be used for the SPF computation.
SPF values for individual subjects will be calculated as:
Table 18.3 Multiple of expected SPF and subject initial MED for each expected SPF
Expected SPF Multiple of expected SPF and subject initial MED
<8 0.64 0.80 1.00 1.25 1.56
815 0.69 0.83 1.00 1.20 1.44
>15 0.76 0.87 1.00 1.15 1.32
Table 18.4 Multiple of expected SPF and subject initial MED for each expected SPF
Multiple of initial MEDu
0.64 0.80 1.00 1.25 1.56
326 J.W. Stanfield et al.
SE = SD / n
where n equals the number of subjects who provided valid test results.
The t value from students t distribution table corresponding to the upper 5%
point with n1 degrees of freedom will be obtained.
The labeled SPF value will be determined as the largest whole number less than
the following calculation:
Water resistance testing will be performed in an indoor fresh water pool, whirlpool,
or hot tub maintained at 2332 Celsius. The pool and air temperature and the
humidity should be recorded.
The labeled SPF will be determined after 40min of water immersion using the fol-
lowing procedure:
. Apply the sunscreen as described in Sect.18.2.7.
1
2. Perform moderate activity in water for 20min.
3. Rest out of water for 15min. Do not towel test site(s).
4. Perform moderate activity in water for 20min.
5. Rest out of water for 15min. Do not towel test site(s).
6. Apply the SPF standard as described above.
7. Expose test sites to UV doses as described above.
The labeled SPF will be determined after 80min of water immersion using the fol-
lowing procedure:
1. Apply the sunscreen as described in Sect.18.2.7.
2. Perform moderate activity in water for 20min.
18 Measuring Sunscreen Protection According totheFDA Final Rule 327
18.4 T
he Broad-Spectrum Protection Test (Critical
Wavelength Test)
18.4.1 Background
The amount of test product applied is 0.75mg/cm2. The test product is applied to the
entire roughened surface of each of at least three PMMA plates, with a roughness
value (Ra) of 27m [11], in a series of small dots. The test product is spread evenly
using a gloved finger, with a very light spreading action for approximately 30s,
followed by spreading with greater pressure for approximately 30s. The plates are
then allowed to equilibrate for 15min in the dark. After equilibration, the plates are
irradiated with a full-spectrum UV dose of four MEDs (800 effective J/m2).
18.4.3 Irradiation
After equilibration, the plates are irradiated with a full-spectrum UV dose of four
MEDs (800 effective J/m2) using a xenon arc solar simulator. The irradiation source
must meet the requirements as described in Sect.18.2.1.
328 J.W. Stanfield et al.
18.4.4 Measurements
After irradiation of the plates, the UV transmission is at wavelengths from 290 to 400nm
at 1nm intervals using a radiometer equipped with an integrating sphere or an ultraviolet
radiation diffuser placed between the sample and the input optics of the spectrometer, to
ensure that the radiation received by the spectrometer is not collimated. The spectrom-
eter input slits must be set to provide a bandwidth that is less than one nanometer. In
addition, the dynamic range of the spectrometer should be sufficient to measure trans-
mittance accurately through a highly absorbing sunscreen product at all terrestrial solar
UV wavelengths (290400nm). Finally the UV dose during one measurement cycle
must not exceed 0.2J/cm2, and a total area of at least 2cm2 is measured on each plate.
The transmission is measured for five locations on the reference plate coated with 15l
of glycerin and five locations on the irradiated plates using a lamp that provides continu-
ous full-spectrum radiation from 290 to 400nm and measuring the transmitted spectral
irradiance. The mean transmittance for each wavelength, T(), is computed as follows:
5
P ( l ) / 5
T (l ) = 1
5
C ( l ) / 5
1
Then
A ( l ) = - log T ( l )
where A ( l ) is the mean absorbance at each wavelength.
The critical wavelength for each plate is then calculated as follows:
lc 400
A ( l )d l = 0.9 A ( l )d l
290 290
where c = critical wavelength
A ( l ) = mean absorbance at each wavelength
d = wavelength interval between measurements
Typical results are shown in Figs.18.3 and 18.4.
18.5.1 Radiometry
1.2
1.0
0.8
Absorbance
0.6
Mean A Pre
Mean A Post
0.4
CW-Pre-Irradiation
CW-Post-Irradiation
0.2
0.0
290 310 330 350 370 390
Wavelength (nm)
Fig. 18.3 Mean absorbance and mean critical wavelength before irradiation and after a full-
spectrum UV dose of four MEDs (800 effective J/m2)
1.2
1.0
0.8
Absorbance
Mean A post
P1-Post
0.6
P2-Post
P3-Post
0.4
0.2
0.0
290 310 330 350 370 390
Wavelength (nm)
Fig. 18.4 Absorbance spectra, the mean absorbance spectrum, and the critical wavelength for
each plate after a full-spectrum UV dose of four MEDs (800 effective J/m2)
Subjects must notify the laboratory of current or recent use of any medication asso-
ciated with sun sensitivity, abnormal responses to sunlight, or phototoxic or photo-
allergic responses. Since prospective subjects often forget or overlook
photosensitizing drugs they may be taking, it is helpful to read a periodically
updated list of potentially photosensitizing drugs to the subject before enrollment in
an SPF test. Lists are available on websites such as Medscape (http://emedicine.
medscape.com/article/1049648-overview).
Subjects that meet the inclusion/exclusion criteria need to be available for testing.
Test subjects must be able to meetall study requirements in the appropriate time
frames. Noncompliance can lead to unreliable data or missed deadlines.
18.6.1 M
easuring theCritical Wavelength intheBroad-
Spectrum Test
According to the FDA Final Rule, the spectroradiometer used to measure the critical
wavelength must provide a bandwidth less than one nanometer (nm). The Labsphere
2000, which is widely used for ISO 24443 and COLIPA tests for critical wavelength
18 Measuring Sunscreen Protection According totheFDA Final Rule 331
As with the invivo test, application can be a factor in unreliable results. The plates
are meant to simulate the surface of human skin. Too much or too little rubbing
during application can lead to differences in film thickness which can cause high
variability.
18.7 Conclusion
The sun protection factor (SPF) is a dimensionless ratio that estimates the protective
efficacy of a sunscreen against erythema. The aim of this chapter was, step by step,
to describe compliance of UV solar simulators, the assessment of the sun protection
factor (SPF) according to the FDA Final Rule, and the determination of the critical
wavelength of sunscreens to determine the degree of broad-spectrum protection.
References
Adnan Nasir
Key Points
Photoprotection is embedded in the DNA of the human species. It has
undergone additional natural and social selection through body hair loss
and skin pigment loss.
Artificial photoprotection predates recorded human history in the form of
grooming habits, clothing, and application of tattoos, muds, and clays.
Some of these may have been comprised of accidental nanomaterials.
Early sunscreens were made of organic compounds with ring structures
which absorbed ultraviolet light and emitted infrared energy. They thus
absorbed UV light, generated heat, and could only be formulated in
lipophilic vehicles.
The modern era of nanoformulation has allowed for these same sunscreens
to be incorporated in a variety of cosmetically elegant vehicles, using
smaller quantities of active ingredient, for better and more stable
photoprotection and enhanced compliance.
Nanoparticles have also been used to develop inorganic and combination
topical sunscreens, as well as entities which combat the effects of
photodamage through a variety of other mechanisms, including physical
blockade, UV absorption, free radical quenching, antioxidant activity, and
delivery of DNA repair enzymes. We are just now witnessing the rosy
fingered dawn of the nano-era of photoprotection.
19.1 Introduction
Less than 1 % of all of the ultraviolet light reaching the surface of the earth is in the
UVC range. About 0.35 % of the total radiation reaching the earth from the sun is
in the UVB range, and 6.5 % is in the UVA range. About 43 % of the light reaching
the earth is visible, and about 49 % is in the infrared range. The effects of these latter
two portions of light spectrum on the skin are not completely understood but are
now increasingly believed to be involved in some form photodamage.
While chronic exposure to UVA and UVB has been shown to contribute to sun-
induced aging of the skin, recent studies have shown that the skin may also be
19 Photoprotection in the Era of Nanotechnology 337
Hair covering the body protects against ultraviolet light, trauma, abrasion, and some
parasites and microorganisms. One Australian study assessed the protective effects
of beards and mostaches under ultraviolet radiation [6]. Facial hair was found to
give the skin an ultraviolet protection factor of 221, depending upon the solar
zenith angle and beard-mostache length. Hair also can insulate and camouflage the
skin. Adaptations of hairlessness include less drag while swimming, less snagging
338 A. Nasir
in dense undergrowth, and reduced risk of overheating. Dark skin may have
developed hand in hand with the loss of body hair. Lack of hair and abundant and
highly active eccrine glands give hominids an advantage in cooling capacity.
Through the process of natural selection, melanin levels have been optimized to
minimize ultraviolet light damage, in order to protect folic acid and DNA from solar
rays while permitting enough light to penetrate the skin to stimulate adequate
vitamin D synthesis and photo conversion. Research has recently shown that sun
protection is probably not the primary driver of skin pigmentation. Common
ancestors shared among humans and chimpanzees have light pigmentation of their
skin covered by dark hair. This lightly pigmented skin is capable of tanning. This
type of response to sun exposure is far more effective at combating skin cancer and
far more adaptive than albinism.
Approximately one million years ago, early humans lost body hair and acquired
pigmented exposed skin in order to adapt to a hot sunny climate. Several lines of
evidence, including analysis of the MC1R gene, suggest that dark pigmentation was
acquired soon after loss of body hair [7]. Tightly curled hair on the head allowed
photoprotection and cooling, allowing air to breeze through, but blocking out
sunlight. Contemporaneously with MC1R gene changes, modifications in the
stratum corneum improved the epidermal barrier against abrasion and microbes.
Stratum corneum keratinocyte response to sun exposure includes immediate dark
pigmentation, which may be due to rearrangement of melanosomes and photo
oxidation of eumelanin.
Individuals with darker Fitzpatrick skin types tend to have a more intense
immediate pigmentary response. The delayed tanning response develops from hours
to days following UV exposure. Because skin cancer, even in sunny climates, tends
to occur after the age of reproduction, selective pressure for dark skin probably was
not designed to reduce the risk of skin cancer. Nonmelanoma skin cancer is typically
not fatal, and its incidence increases with age. Melanoma, while potentially fatal
and tending to occur more frequently in younger individuals, especially young
women, is rare compared to nonmelanoma skin cancer. For millennia, the Inuit have
carved snow goggles from caribou antlers and sinew, creating form-fitting curved
eye masks with narrow slits to limit light exposure to the eyes and prevent
photokeratitis.
Folic acid (vitamin B9) is used for DNA repair in and to prevent neural tube defects.
Low levels of folic acid induced by photolysis can also lead to reduced fertility
because of potentially fatal birth defects [8]. Folic acid is also important in sper-
matogenesis, and reduced levels may also potentially contribute to male infertility.
Ultraviolet light exposure reduces folic acid levels. Diminished folic acid levels are
detrimental to DNA repair and embryogenesis. Selective pressure would promote
tend to promote melanogenesis to preserve optimum folic acid levels. It has been
speculated that hunters and gatherers received significant amounts of vitamin D
19 Photoprotection in the Era of Nanotechnology 339
through their diets of fish and animal livers. With the advent of agriculture, early
Europeans required vitamin D supplementation, and those with less pigment in
their skin were able to synthesize it with suboptimal ultraviolet light exposure.
Vitamin D receptors occur in 36 different tissues, and studies show the importance
of vitamin D in the immune system, the musculoskeletal system, the intestine, the
kidney, and the reproductive system. Vitamin D deficiencies have been associated
with rickets and multiple sclerosis. Because of its effect on reproduction, preserva-
tion of folic acid levels may have exerted more selective evolutionary pressure than
preservation of vitamin D levels.
Evidence suggests that recent changes in the skin, eye, and hair pigmentation have
been due to social and sexual selection. The ancient Egyptians considered light skin
more attractive than dark skin. In the desert environment, it was difficult to maintain
fair skin. Translations of hieroglyphics from Egyptian tombs have revealed
ingredients such as rice bran extract, jasmine, and lupine extract for treating damage
to the skin and reducing the likelihood of a tan or of a sunburn. One of the components
of rice bran extract, oryzanol, has been shown to have UV-absorbing properties [9].
Examples of low-cost photoprotectants using local materials can be found in
Southern Africa. African clays have been used for cosmetic purposes, ceremonial
purposes, ritual coming-of-age ceremonies, local hygiene practices, social signaling,
camouflage, and photoprotection. Aboriginal peoples of South Africa have used
two types of clay for photoprotection: red and white. The Xhosa tribes in the
Amathole Mountain area use red clay for face painting [10]. Clays are composed of
fine grains containing traces of metals such as nanoparticulate aluminum silicates,
organic matter, bound together by water in a mineral structure. In general particle
sizes of clays range from 1000 to 2000 nm. This permits tight aggregation and
packing of clay components. These clays have been studied for their photoprotective
properties [10]. Overall, it has been determined that clays from the Amathole Range
have a low sun protection factor but a broad spectrum of activity.
Before the modern era of photoprotection, sunburn was believed to the caused by
heat rather than ultraviolet light. In 1801, Johann Wilhelm Ritter discovered
ultraviolet rays. He described properties of light with wavelengths shorter than
those in blue and referred to light in this region as infraviolet (which we now term
ultraviolet). While Hippocrates and Aristotle developed early theories of skin color
and climate, it wasnt until 1820 that Everard Home of England dispelled the notion
that heat led to sunburns. He began exploring why inhabitants of temperate climates
have darker skin than inhabitants of northern climates. He found the correlation
surprising given the fact that darker skin tends to absorb more heat and lighter skin
less heat. He would have expected dark skin in colder climates as one means of
capturing as much ambient heat as possible. Home observed this directly when he
covered one of his pale hands with a dark cloth and left the other hand exposed to
sunlight. Even though the hand covered with the dark cloth registered a warmer
340 A. Nasir
temperature, it did not sunburn compared to the exposed hand. He surmised that
skin pigment protected against sunburn and that sunburn was not due to heat. Home
concluded that it was the dark pigment in skin, or melanin, which protected the skin
from ultraviolet light-induced damage.
In 1878, Otto Veiel had shown the benefits of tannin in protecting against
ultraviolet light. The usefulness of tannin was limited by its tendency to stain the
skin. In 1922, it was demonstrated that the wavelengths of light most likely to
induce sunburn were in the 280315 nm wavelength range. By developing filters
which specifically targeted this wavelength, the first sunscreens containing para-
aminobenzoic acid, benzyl salicylate, and benzyl cinnamate were developed.
Sunscreens were initially developed, and their use proliferated during the Second
World War in order to protect soldiers deployed in the Pacific tropics and the African
desert from sunburn causing rays. Early sunscreens blocked rays in the UVB portion
of the solar spectrum. Traditional measurements of sunscreen effectiveness have
focused on this portion of the light spectrum and have not addressed protection
against UVA or UBC or visible or infrared light.
The stigma of dark skin has led to a plethora of skin-lightening products. The impact
of these products has been greatest in countries like South Africa and India. Some of
these products contain mercury and have been the target of FDA regulation and United
States. In India, controversial skin whiteners have also been developed for the vaginal
area. Skin lighteners, when permanently depleting cutaneous melanocytes, can lead to
increased vulnerability to UV light and can mask the appearance of melanoma.
Nanoparticles can be formulated in a broad range of hues and can be manufactured to
make sunscreens which blend in with a variety of skin types. They may prove useful
for individuals who have undergone temporary or permanent lightening procedures.
Inorganic sunscreens typically are clusters of ions such as zinc, iron, or titanium
coupled to oxygen. These clusters are manufactured in particles ranging in size
from 10 to 300 nm. The mechanism of action of inorganic sunscreens differs from
organic sunscreens. Inorganic sunscreen effectively blocks and absorbs UV from
the 200 to 380 nm size range with a steep drop-off after 380 nm. Thus, inorganic
sunscreens have a high utility for UVC, UVB, and UVA light.
Macromolecular clusters of inorganic sunscreens are not generally accepted
by the public because of their opacity. They leave a white residue on the skin and,
if they are used at all, tend to be under used to minimize this effect. The opacity
is due to the light scattering effect of the large clusters of inorganic sunscreens.
Typical clusters of zinc oxide and titanium dioxide tend to be 200 nm or greater.
Clusters of this size are effective at scattering light of a wavelength twice their
diameters. Thus, large clusters of 200 mm or greater tend to scatter light in the
visible range, 400700 nm. Scattering of visible light when reflected to the eyes
appears quite. Organic sunscreens do not scatter visible light because of their
small size. Compared to a 200 nm particle of titanium dioxide capable of scatter-
ing light in the 400700 nm range, a one nanometer particle of methoxycinna-
mate is unable to scatter visible light.
Small particles are small obstacles to longer wavelengths of light. Objects much
smaller than visible light rays (100 nm or smaller) do not scatter visible light
effectively. Therefore, nanoparticulate sunscreens in the 100 nm size range or
smaller appear essentially invisible. Light scatter as a function of wavelength
demonstrates this phenomenon clearly.
19.1.3.3 Nanoformulation
Increased Stability
Some synthetic compounds are also subject to instability and aggregation as well as
potential percutaneous absorption. One example is 4-methylbenzylidine camphor.
When incorporated into microspheres, 4-MBC particles showed the same
photoprotective capacity as the free chemical compound, greater stability, and
significantly slower release kinetics.
Enhanced Compliance
This is discussed in more detail below but occurs as a result of the combination of
benefits and reduced risks of nanoformulation.
Standard methods for evaluating sunscreens include the use of photometers and
animal and human studies to evaluate minimal erythema-inducing doses. Alternatives
to human and animal testing are constantly being developed to assess the protective
19 Photoprotection in the Era of Nanotechnology 343
Topical sun protective agents include metallic physical blockers such as zinc oxide, tita-
nium dioxide; organic chemical filters such as cinnamates, benzophenones, Mexoryl
SX, XL, and Tinosorb M, S; antioxidants such as hydroxycinnamic acids, polyphenols
including flavonoids-genistein, silymarin, equol, quercetin, apigenin, green tea extract,
resveratrol, staxanti, anthocyanins, tannins, pycnogenol, and others (DHA, caffeine,
polygonum multiflorum)-fullerenes, N-(4-pyrodoxylmethylene)-1-serine, creatine, and
idebenone; nonsteroidal anti-inflammatory compounds such as COX-2 inhibitors; and
after-exposure compounds which affect DNA repair, such as the enzymes photolyase,
T4 endonuclease, and DNA oligonucleotides.
19.3.1 Fabrics
Other obvious sources of sun protection include hats, umbrellas, and shade struc-
tures (natural, such as terrain and foliage; as well as artificial, such as awnings and
roofs). The term umbrella derives from the Latin umbra, meaning shade or
shadow. Originally, umbrellas were developed in ancient Egypt and contained
palm fronds for sun protection. Modifications made their way to Europe through
the Greeks. The Chinese developed waterproof umbrellas for rain protection.
Sunglasses and photochromic contact lenses and intraocular lenses provide eye
protection; however, these have proved dangerous in the sudden dark of bridges
and tunnels [22]. As already mentioned, prehistoric circumboreal inhabitants have
carved slits in bones and antlers to develop snow goggles to prevent photokerati-
tis. Photoprotective glass can be found on vehicles and on industrial and residen-
tial windows. Nanomaterials are making inroads in modifying the fabric of
umbrellas and the photoprotective properties of lenses and building materials for
consumer use.
344 A. Nasir
Studies of fabrics have shown that the sun protective factor (UPF) attributable to
clothing is directly proportional to the structure of the fabric and the tightness of the
weave in the fabric [12]. UPF numbers consistently increase after repeated washing
of cotton garments because of shrinkage and reduction in fabric aperture diameter.
For example, for pure cotton T-shirts, UPF increased from a baseline of 1940.6
after weekly washing for 10 weeks. After repeated washing, fabric shrinkage, and
knit hole size reduction, fabric hole area decreased from 8 to 3.9 %. Simple advice
for increasing the sun protective properties of clothing is to wear only after repeated
washing. Darker fabrics may also confer slightly greater photoprotection.
Incorporation of light conducting nanofibers into woven material is the basis for
nanofibers using distributed optics [1418]. Distributed optics can be used for con-
duction and redirection of light of a desired wavelength. Distributed optical fabrics
may be used for biomedical monitoring of the wearer. They may be used to camou-
flage the wearer. They may be used for delivery of phototherapy to the wearers skin.
They may also prove useful for photoprotection using optical interference.
19.3.2 Liposome
19.3.2.1 Definition
These are micro- and nanoparticulate vesicles with lipid bilayers consisting of
charged ionic phospholipids and cholesterol. The amphipathic phospholipids
typically consist of a polar head bound by phosphate and glycerol to two fatty acids
with chains of 1024 carbon atoms and 06 double bonds. Cholesterol is interspersed
in this lipid bilayer. Liposomes can be small (20100 nm) or up to 1000 nm. They
can be unilamellar, oligolamellar, and multilamellar.
19.3.2.2 Background
These are so-called morning-after creams because they are designed to repair
damage to cells after maximal threshold ultraviolet light exposure. One drawback to
these types of agents is thatby giving users an exaggerated sense of confidence
they may promote, rather than prevent, excessive sun exposure.
346 A. Nasir
Photolyase
T4 Endonuclease
19.3.2.5 Disaccharides
Modification of ocular UV protectors with liposomes has shown some benefit for
their use in the skin. Trehalose is a naturally occurring disaccharide which is
typically used as a protein stabilizer to reduce ultraviolet light-induced damage to
the eye when topically applied to cornea. Liposomal formulations of trehalose were
evaluated for their photoprotective effects on human keratinocyte cell lines against
l-carnosine, ergothioneine, l-ascorbic acid, and DL--tocopherol [28]. The
trehalose-laden liposomes showed the greatest protection against the formation of
UV-induced cyclobutane pyrimidine dimers, 8-hydroxy 2-deoxy guanosine, and
protein carbonylation products.
19 Photoprotection in the Era of Nanotechnology 347
19.3.2.8 Resveratrol
19.3.2.9 CDBA
A subset of liposomes are the deformable variety known as elastic liposomes. These
are able to interdigitate between keratinocytes in the stratum corneum and epidermis
to allow for better penetration persistence and distribution upon topical application.
Elastic liposomes loaded with benzophenone-3 in the 100 nm size range at a
concentration of 20.34 % (M/M) showed significant protective effects against
ultraviolet radiation [33].
348 A. Nasir
19.3.4 Niosome
19.3.4.1 Definition
Niosomes are vesicle carriers with an aqueous core surrounded by one or more
layers of phospholipids, typically cholesterol and one or more nonionic surfactants
such as alkyl ethers, alkyl esters, alkyl amides, long chain fatty acids, and amino
acids. Thus, the surfaces of niosomes are nonionic surface active agents, from
which the name is derived. They range in size from 100 to 200 nm. Niosomes tend
to be made from biocompatible and biodegradable agents. They tend to be nontoxic
and non-immunogenic. They are stable and highly resistant to hydrolytic degradation.
They are amphiphilic and can accommodate contents of a wide range of solubilities.
Niosomes are difficult and complex to formulate and may suffer from aggregation,
leaching, or dispersion, which can limit shelf life.
19.3.4.2 Background
Niosomes tend to have greater penetration capability than standard emulsions and
are typically more stable than standard liposomes. Thin film hydration is a common
manufacturing technique and has been used to make niosomes containing minoxidil.
Niosomes and liposomes containing avobenzone and arbutin are under development
to create sunscreens which have added pigment-reduction capacity [34].
19.3.4.3 Polyphenols
Polyphenols from black tea extract have been packaged in multilamellar niosomes
[35]. These were applied topically onto the skin of nude mice and shown to have
enhanced penetration of caffeine and gallic acid than comparable controls dispersed
in aqueous solutions. Black tea extract may be useful in the future as a topical
sunscreen when delivered in a niosome vehicle.
19.3.5 Ethosome
19.3.5.1 Definition
Ethosomes have been developed as transdermal drug delivery systems. They are a
type of soft vesicle composed of phospholipids (such as phosphatidylcholine, phos-
phatidylserine, and phosphatidic acid) in a high concentration of ethanol (2050 %)
and water (from which they derive their name). Ethanol acts as a permeation enhancer,
and its content can be varied to accommodate a broad range of active ingredients.
Ethosome size can be controlled from the nm to sub-mm range. Ethosomes are non-
toxic and can be formulated in creams, lotions, gels, and patches. Ethosomes can be
19 Photoprotection in the Era of Nanotechnology 349
19.3.5.2 Background
19.3.5.4 Apigenin
Apigenin is a bioflavonoid which has been shown to have antioxidant activity and a
number of cellular targets involving GTPase activation, membrane transport, and
mRNA metabolism/alternative splicing [37]. It has been studied as a potential topi-
cal and systemic anti-inflammatory and antitumor agent. In one trial, optimization
studies were conducted to determine ideal formulations for delivery of apigenin to
the skin. Comparisons were made of ethosomes, liposomes, and elastic liposomes.
It was found that increasing phospholipid content in ethosomes (especially Lipoid
S75), propylene glycol content, and ethanol content enhanced skin deposition and
transdermal delivery. Optimized ethosomes showed the greatest reduction of cyclo-
oxygenase-2 levels in mouse skin after exposure to UVB light.
These are colloidal vehicles comprised of solid lipid cores mixed in defined ratios
with water or an aqueous surfactant. The lipids tend to be biocompatible,
biodegradable, and nontoxic, making them ideal for cosmetics and cosmeceutical
350 A. Nasir
preparations. Because of their small size, they pack tightly, have high occlusivity,
promote skin hydration, and limit transepidermal water loss. They are relatively
easy to manufacture, easy to scale up, and easy to sterilize, and dont require special
solvents. Some solid lipid nanoparticles, for example, crystalline cetylpalmitate
nanoparticles (CCP-NP), have inherent photoprotective activity [38]. Native
CCP-NP have about 23-fold greater UV-absorbing properties compared to
traditional emulsions. This effect has been shown to give synergistic and additive
photoprotective effects to sunscreen contents. For example, CCP-NP containing
2-hydroxy-4-methoxybenzophenone (Eusolex 4360) were threefold more
photoprotective compared to reference emulsions.
19.3.6.2 Definition
19.3.6.3 Benzophenone-3
19.3.6.5 Lutein
Lutein has antioxidant and blue light-blocking properties. Because of its poor
solubility, it is an ideal candidate for lipid nanoparticle delivery to the skin. In one
study, lutein was incorporated into nanocarriers such as nanoemulsions (NE), solid
19 Photoprotection in the Era of Nanotechnology 351
lipid nanoparticles (SLN), and nanostructured lipid carriers (NLC) using high-
pressure homogenization [41]. All three were found to be stable. The SLN had the
lowest release (0.4 %) of content after 24 h, compared to NE which had the highest
release (19.5 %). None of the lutein in SLN permeated pig skin after 24 h. Only a
small fraction (0.06 %) of the lutein was degraded after exposure to 10 MED,
compared to 68 % in the NLC, 14 % in the NE, and 50 % for lutein powder
suspended in corn oil.
19.3.6.6 Tocopherol
Synthetic solid lipid nanoparticles were shown in one study to have inherent UV protec-
tive activity. This effect was synergistically augmented with the incorporation of tocoph-
erol acetate. Solid lipid nanoparticles containing tocopherol were twice as effective at
UV blockade than reference emulsions containing identical lipid content [5, 40, 42, 45].
19.3.6.7 Titanium
SLN prepared using conventional and hybrid methodologies were able to enhance
the photoprotectivity of titanium dioxide compared to reference emulsions [43].
They were also able to allow lower concentrations of titanium dioxide for equivalent
photoprotection.
19.3.6.8 Chitin
19.3.7.1 Background
lipids used include bees wax, carnauba wax, Dynasan, precifac, stearic acid, apifil,
and Cutina CP. Typical liquid lipids include Cetiol V, Miglyol, castor oil, oleic acid,
davana oil, palm oil, and olive oil. Sometimes emulsifiers are added to optimize the
blend. These can include Miranol Ultra, PlantaCare, Tween 80, Pluronic F68,
Poloxamer 188, and Phospholipon 90G. In dermatology, nanostructure lipid carrier
preparations have been used for sunscreens and topical drugs (minoxidil, tacrolimus,
miconazole nitrate).
19.3.7.2 Definition
Solid lipid nanoparticles typically have a crystalline matrix with little room for
active ingredients. This often leads to drug expulsion out of the particles over time
and a rapid drop-off in efficacy. Nanostructured lipid carriers are a hybrid of a solid
lipid surrounding a liquid lipid drug carrier space. This leads to enhanced drug
loading and stable storage.
19.3.7.3 Tocopherol
Tocopherol has more potent antioxidant activity than its conjugate tocopherol ace-
tate; however, it is viscous, poorly soluble, and photo-unstable and can cause irritant
dermatitis. In one small study, the high-pressure homogenization technique was
used to create nanostructured lipid carriers and nanoemulsions of tocopherol.
Particle sizes of 67 nm NLC and 586 210 nm NE were formed. About 30 %
tocopherol was released from the NLC within 2 h, while only 4 % was released
from the NE. Both formulations were shown to retain antioxidant activity, to be
non-irritating, and to protect tocopherol from UV degradation.
19.3.7.7 Lycopene
Nanostructured lipid carriers composed of biocompatible lipids from rice oil and
cholesterol were manufactured to be loaded with lycopene. Particle sizes of
nanostructured lipid carriers ranged from 287 to 405 nm. Cholesterol was found to
reduce stability of particles and its exclusion as well as storage at 4 C or room
temperature led to the greatest stability [49, 50].
19.3.8 Microsphere
19.3.9 Gold
Gold metal particles are emerging it has excellent candidates for nanomaterials
with biologic uses. Gold nanoparticles are biocompatible, easy to synthesize,
and easy to conjugate with a number of other compounds. They have been used
for cancer diagnosis and therapy, drug delivery, and as biologic probes.
Phytolatex synthesized gold nanoparticles have been shown to enhance the sun
protection factor of sunscreen when added to native sunscreen at 24 % concen-
trations [52].
19.4 Safety
19.4.1.2 Absorption
19.4.1.4 Instability
Because of their greater efficacy, their high surface-to-volume ratio, their high
occlusivity, and their controlled stability, nanoformulations of sunscreens tend to
require lower total concentrations of active ingredient (organic or inorganic) than
their traditional counterparts. This can lead to lower manufacturing costs and
356 A. Nasir
potentially lower overall exposure to chemical agents. Some studies have shown
that high concentrations of physical blockers can cause perioral dermatitis [61].
19.4.2.2 Detoxification
Concern about hazards associated with metallic nanoparticles stems from in vitro
studies showing the generation of free radicals. Some studies showed that the level
of clastogenicity increased upon UV exposure. However, comparisons of zinc
oxide nanoparticles to known photoclastogens such as 8-MOP [62] showed sub-
stantially minimal effects (24 increase in vitro for zinc oxide, and >15,000
increase for 8-MOP). The Ames test showed no increased mutagenicity for zinc
oxide. Human studies have shown no evidence of phototoxicity. Commercial
nanoparticulate sunscreens have been coated to detoxify them. Coating of nanome-
tallic sunscreens with inert oxides of silica has been shown to eliminate the risk of
reactive oxygen species generation. Furthermore, aggregation of nanoparticles
reduces their surface-to-volume ratio and reactivity. A number of studies have now
shown very little to no dermal penetration of metallic sunscreen nanoparticles fol-
lowing application to the skin. This includes studies of intact, flexed, and stripped
skin. Minimal penetration has been noted on abraded skin. One study of hairless
mice showed slight differences in absorption of nanoparticulate zinc when com-
pared to larger particle size zinc, but no toxicity and no effect on zinc homeostasis
[6365]. Furthermore, permeability comparisons demonstrate that pig and rat skin
are 4- and 911-fold more permeable than human skin. Most of the recent studies
show minimal or no absorption.
Because nanoformulation allows for the precise selection of particle size, shape,
charge, and chemical composition, nanophotoprotective agents can be manufactured
in a wide variety of formats and vehicles to allow for optimal stability, UV protection,
composition, and texture to permit the widest possible range of adoption and
compliance.
19.5 Compliance
References
7. Jablonski NG, Chaplin G (2014) The evolution of skin pigmentation and hair texture in people
of African ancestry. Dermatol Clin 32(2):113121
8. Jablonski NG, Chaplin G (2010) Colloquium paper: human skin pigmentation as an adaptation
to UV radiation. Proc Natl Acad Sci U S A 107(Suppl 2):89628968
9. Shaath NA (2010) Ultraviolet filters. Photochem Photobiol Sci 9(4):464469
10. Dlova NC, Nevondo FT, Mwangi EM, Summers B, Tsoka-Gwegweni J, Martincigh BS,
Mulholland DA (2013) Chemical analysis and in vitro UV-protection characteristics of clays
traditionally used for sun protection in South Africa. Photodermatol Photoimmunol Photomed
29(3):164169
11. Schuch AP, Moraes MC, Yagura T, Menck CF (2014) Highly sensitive biological assay for deter-
mining the photoprotective efficacy of sunscreen. Environ Sci Technol 48(19):1158411590
12. Wang SQ, Kopf AW, Marx J, Bogdan A, Polsky D, Bart RS (2001) Reduction of ultraviolet
transmission through cotton T-shirt fabrics with low ultraviolet protection by various laundering
methods and dyeing: clinical implications. J Am Acad Dermatol 44(5):767774
13. Wu XM, Branford-White CJ, Yu DG, Chatterton NP, Zhu LM (2011) Preparation of core-shell
PAN nanofibers encapsulated -tocopherol acetate and ascorbic acid 2-phosphate for
photoprotection. Colloids Surf B Biointerfaces 82(1):247252
14. DAngelo LT, Weber S, Honda Y, Thiel T, Narbonneau F, Luth TC (2008) System for respira-
tory motion detection using optical fibers embedded into textiles. Conf Proc IEEE Engl Med
Biol Soc 2008:36943697
15. De Jonckheere J, Narbonneau F, Kinet D et al (2008) Optical fibre sensors embedded into
technical textile for a continuous monitoring of patients under magnetic resonance imaging.
Conf Proc IEEE Engl Med Biol Soc 2008:52665269
16. Eden JG, Park S-J, Ostrom NP, Chen K-F (2005) Recent advances in microcavity plasma
devices and arrays: a versatile photonic platform. J Phys D Appl Phys 38:16441648
17. Sorin F et al (2009) Exploiting the collective effects of optoelectronic devices integrated into
a single fiber. Nano Lett 9:26302635
18. Sorin F et al (2007) Multimaterial photodetecting fibers: a geometric and structural study. Adv
Mater 19:38723877
19. Ashley S (2003) X-ray proofing. To save himself, a physician enters the rag trade. Sci Am
288(5):3435
20. Jones MA, Cocker M, Khiani R, Foley P, Qureshi N, Wong KC, Rajappan K, Betts TR (2014)
The benefits of using a bismuth-containing, radiation-absorbing drape in cardiac
resynchronization implant procedures. Pacing Clin Electrophysiol 37(7):828833
21. McCaffrey JP, Tessier F, Shen H (2012) Radiation shielding materials and radiation scatter
effects for interventional radiology (IR) physicians. Med Phys 39(7):45374546
22. Allen MJ (1979) Highway tests of photochromic lenses. J Am Optom Assoc
50(9):10231027
23. Korting HC, Schllmann C (2011) Resistance of liposomal sunscreen formulations against
plain water as well as salt water exposure and perspiration. Skin Pharmacol Physiol
24(1):3643
24. Patsinakidis N, Wenzel J, Landmann A, Koch R, Ger J, Luger TA, Metze D, Surber C, Kuhn
A (2012) Suppression of UV-induced damage by a liposomal sunscreen: a prospective, open-
label study in patients with cutaneous lupus erythematosus and healthy controls. Exp Dermatol
21(12):958961
25. Hofer A, Legat FJ, Gruber-Wackernagel A, Quehenberger F, Wolf P (2011) Topical liposomal
DNA-repair enzymes in polymorphic light eruption. Photochem Photobiol Sci 10(7):11181128
26. Zahid S, Brownell I (2008) Repairing DNA damage in xeroderma pigmentosum: T4N5 lotion
and gene therapy. J Drugs Dermatol 7(4):405408
27. Wolf P, Maier H, Mllegger RR, Chadwick CA, Hofmann-Wellenhof R, Soyer HP, Hofer A,
Smolle J, Horn M, Cerroni L, Yarosh D, Klein J, Bucana C, Dunner K Jr, Potten CS,
Hnigsmann H, Kerl H, Kripke ML (2000) Topical treatment with liposomes containing T4
19 Photoprotection in the Era of Nanotechnology 359
Mary S. Matsui
Key Points
There many reasons for interest in the photoprotective potential of topical
antioxidant botanical extracts. These include consumer demand for non-
sunscreen photoprotective ingredients, the understanding that longwave
(UVA) ultraviolet radiation in particular induces considerable oxidative stress,
and an interest in supplementing sunscreen formulations to increase stability
and protection while decreasing the use of chemical and physical sunscreens.
The botanical product most studied for topical photoprotection is derived
from Camellia sinensis, the tea plant. Topical application of the most potent
constituent, EGCG, has been shown to inhibit UV-induced leukocyte infiltra-
tion, DNA damage, immune suppression, dermal degradation, and erythema.
Other botanicals that have been demonstrated to have photoprotective
properties include genistein, resveratrol, grape seed proanthocyanidins,
Polypodium leucotomos (fern) extract, and certain combinations of ferulic
acid, vitamin C, and vitamin E.
Sunscreens formulated with antioxidants/botanical extracts may have
additive or synergistic photoprotective effects when compared with either
of these agents alone.
Variables that impact the benefits of botanical extracts or other antioxi-
dants include unknown optimal concentrations, possible interactions
between ingredients, and the instability of antioxidants. At present, there is
no labeling information requirement for ingredient concentration, and at
least one study showed that the levels of antioxidants in some off the
shelf sunscreen products were below that required for efficacy.
For several reasons, there is great popular interest in the use of topical antioxidants
for skin photoprotection and antiaging therapies. Many consumers, influenced
particularly by numerous Internet sources giving nonmedical advice, information,
and testimonials, believe that naturally derived ingredients, plant extracts, and
food components are superior or healthier than chemical sunscreens. Other factors
contributing to the popularity of these types of alternative products include
dissatisfaction with the aesthetic properties of current sunscreen/sunblock products,
the perception that the chemicals in OTC products are unhealthy, unsafe, or damage
the environment and the belief that natural ingredients provide additional benefits
not found in sunscreens. Finally, there is interest in supplementing sunscreen
products by manufacturers, as there is some evidence that it is possible to stabilize
sunscreens with antioxidants [2]. This chapter will discuss the topical use of
antioxidants, primarily but not exclusively in the form of botanical extracts and will
review the scientific evidence for their efficacy against the damaging effects of
ultraviolet radiation (UVR) on human skin.
Endogenous antioxidants that scavenge for ROS include superoxide dismutase,
glutathione peroxidase, ascorbate, alpha-lipoic acid, and catalase. Excessive ROS
generated during UV exposure depletes endogenous antioxidants and causes a state
of oxidative stress in cells that can damage cellular proteins, lipids, and DNA,
trigger apoptosis, and contribute to photocarcinogenesis. A review of endogenous
antioxidant strategies and oxidant-induced cellular damage and mechanisms can be
found in Khan et al. [22]. Also, in particular, Table 1 and Fig. 1 contained in an
earlier review [1] are recommended for a concise summary of botanicals and their
mechanisms of action.
Sunscreen/sunblock ingredients protect skin from solar radiation through three
basic mechanisms: reflection, dispersion, and absorption (for review, see Schalka
and Silva dos Reis [34]). In contrast, antioxidants generally have little to no ability
to physically block UVR and act to protect the skin by other mechanisms. The
efficacy of sunscreens to protect skin against UVR is virtually universally measured
and regulated by specific, in vivo (UVB) and in vitro (UVA) assays such as sun
protection factor (SPF) which is based on UVR-induced erythema, immediate
pigment darkening, and critical wavelength. Because erythema is primarily driven
by direct DNA damage and repair and less by oxidative stress, UVR protection
provided by antioxidants is best measured by endpoints other than SPF. In addition,
it should be noted that significant damage, including DNA mutations, immune
suppression, and collagen breakdown can occur in the absence of sunburn.
There is some epidemiological evidence that higher dietary or systemic levels of
antioxidants are associated with a lower risk of nonmelanoma skin cancers and
photoaging signs in humans [3, 7, 15, 33], and there are animal/rodent studies
showing this efficacy for oral supplementation of antioxidants. There is extensive
experimental evidence that exogenous antioxidants are anti-inflammatory and
suppress oxidative stress pathways in in vitro cell culture and that topical antioxidants
are photoprotective in acute and chronic animal models of UVR-induced skin
damage. Reviews of these data can be found in Afaq and Mukhtar [1], Khan et al.
[22], and Passantino et al. [32]. This chapter will focus on the most recent data
20 The Role of Topical Antioxidants in Photoprotection 363
Fig. 20.1 UV radiation initiates production of reactive oxygen species (ROS), which is followed
acutely by secondary messengers and increased expression of proinflammatory cytokines. Long-
term, exposure results in decreased collagen production and increased collagen breakdown,
leading to signs of skin photoaging (Reproduced from Zussman et al. [44])
epidermal hyperproliferation and inflammation [24, 25]. It has also been shown that
ultraviolet B wavelengths (290320 nm), in addition to being directly absorbed by
DNA bases and causing mutagenic lesions such as cyclobutane pyrimidine dimers
(CPDs) and pyrimidine(6-4)pyrimidine photoproducts (6-4PPs), are also capable of
initiating oxidative stress [35]. Increased ROS generation can overwhelm endoge-
nous antioxidant defense mechanisms, resulting in oxidative stress and oxidative
photodamage of proteins and other macromolecules in the skin. These ROS are
believed to be critical mediators of the photoaging and photocarcinogenesis pro-
cesses. Exposure to either UVA or UVB can result in oxidation of amino acid resi-
dues such as lysine, arginine, and proline, which leads to the formation of carbonyl
derivatives that affect the structure and function of proteins. Other protein-related
damage includes tyrosine cross-links, amino acid interconversions, and peptide
bond cleavages. Lipid peroxidation also damages cell membranes, and a major oxi-
dative lesion in DNA is 8-hydroxy-2-deoxyguanosine (8-OHdG). The cascade of
events leading to clinical signs of photoaging is shown in Fig. 20.1. Repeated expo-
sure to ROS leads to an accumulation of cellular damage and visible signs of pho-
toaging. Considering the extent of potential damage, the most important attribute of
antioxidants is their capacity to quench reactive oxygen species (ROS) and prevent
the resulting cascade of protein, lipid, and DNA oxidation, which leads to inflam-
mation, mutation, and structural/functional damage.
Four main categories of research models are available to study the effects of
antioxidants on prevention of photodamage to human skin: in vitro chemical anti-
oxidant properties, in vitro cell-based assays, animal (mouse) in vivo, and human
in vivo models. As to the first category, chemical analytical methods, basic antioxi-
dant properties have been compiled for edible plants and foodstuff in vitro [6, 13,
23]. Screening for active ingredients to include in topical photoprotective products
can be facilitated by assays as the ferric-reducing ability of plasma (FRAP), the
oxygen radical absorbance capacity assay (ORAC) assay, and the DPPH radical
scavenging efficacy. Tissue culture work has provided considerable understanding
of basic mechanisms, and animal studies have shown the benefits of topical antioxi-
dants against UVR-induced carcinogenesis. These have been reviewed elsewhere,
and so in vivo human studies will be emphasized in this chapter.
The botanical product most studied for topical photoprotection is derived from
Camellia sinensis, the tea plant. Tea is one of the most widely consumed beverages
in the world, second only to water, and has been long regarded for its antioxidant,
anti-inflammatory, and anticancer properties. Tea is commercially available mainly
in three forms: green, black, and oolong tea, but white and red can also be found. Of
the total commercial tea consumption worldwide, about 78 % is consumed in the
form of black tea (primarily Europe, Russia, the Middle East, India, and North
America), and about 20 % is consumed in the form of green tea (primarily Asian
countries like, Japan, China, Korea, parts of India, and a few countries in North
Africa and the Middle East). Tea contains variable amounts of three main types of
polyphenols (flavonoids, stilbenes, and lignans). Flavonoids are divided into six
subclasses: flavonols, flavones, isoflavones, flavanones, anthocyanidins, and
flavanols. Of the flavonoids, the majority are monomeric flavanols called catechins.
20 The Role of Topical Antioxidants in Photoprotection 365
Fig. 20.2 Clinical appearance of skin 24 h after having been treated with GTP followed 30 min
later by a 2-MED dose of solar-simulated light. At left is skin treated with vehicle alone. Middle
is skin treated with UVR and vehicle. Right is skin treated with both GTP and UVR. Photo
supplied by SK Katiyar and CA Elmets
A further study sought to examine the protective effects of topical white tea or
green tea against markers of UVR damage that are associated with immune suppres-
sion and carcinogenesis [5]. This was accomplished by performing: (1) immunohis-
tochemical analysis for oxidative DNA damage and for epidermal Langerhans cells
(LCs) from biopsies obtained after in vivo irradiation of human skin in the presence or
absence of the topical tea formulations; (2) assessments of in vivo contact hypersen-
sitivity using the contact sensitizer dinitrochlorobenzene (DNCB); and (3) an analysis
of UVR-induced epidermal LC depletion in vitro, using a skin explant model. In this
study, the SPF for the tea extracts was determined to be 1. Ten volunteers participated
in the study that assessed skin biopsies by immunohistochemical analyses and found
that both green tea and white tea partially prevented UV-induced depletion of CD1a+
cells and ssUVR-induced generation of 8-OHdG, as illustrated in Fig. 20.3a, b. Ninety
subjects were used to assess the ability of tea extracts to prevent ssUVR-induced
immune suppression using 0.75 and 2MED. The results showed a trend (in the face of
large interindividual variability) toward preservation of the ability to sensitize subjects
to DNCB, suggesting that tea-treated subjects had greater preservation of their CHS
response after ssUVR exposure, relative to untreated subjects. In a separate study,
CPDs were found to be significantly reduced after 2MED ssUVR exposure by pre-
treatment with either 0.2 % white tea or 0.5 % green tea extracts. In summary, GTPs in
general and EGCG in particular, have, in human clinical studies, been shown to reduce
UVB-induced erythema, sunburn cell formation, leukocyte infiltration, and protect
against ssUVR-induced Langerhans cell depletion, generation of 8-OHdG, and
immune suppression (CHS). Animal studies support this human evidence that topi-
cal GTP protects from UVR-induced immunosuppression [2021]. In addition, UVR
alteration of IL-10 and IL-12, critical cytokines involved in UVR-induced inflamma-
tion and immune suppression, has been shown to be modified by pretreatment with
GTPs. For example, the reduction in UV-induced DNA damage by GTPs appears to
be mediated via induction of interleukin (IL)-12 [36], previously shown to induce
NER DNA repair. Katiyar [18] has reviewed the evidence for this additional mecha-
nism of action for GTPs in particular that these phytochemicals induce DNA repair
and thereby counteract the effect of UVR exposure on photoaging and carcinogenesis.
20 The Role of Topical Antioxidants in Photoprotection 367
UV + Vehicle
% CD1a + staining
UV + WT
12 UV + WT 20
UV + GT
10 UV + GT
15
8
6 10
4
5
2
0 0
Treatment Groups Treatment Groups
Fig. 20.3 (a) Oxidative damage was measured via levels of 8-hydroxy-2-deoxyguanosine (OhdG)
staining in skin biopsies obtained 72 h after a single 2-MED dose of ssUVR. Untreated and
vehicle-treated skin showed increased 8-OhdG, whereas 8-OhdG levels in white tea (WT)-treated
and green tea (GT)-treated skin were not different from control unirradiated skin. (b) Biopsies
obtained 72 h after a single ssUVR dose of 2 MED showed decreased epidermal CD1a+ Langerhans
cells (LC) in untreated and vehicle-treated skin. White tea (WT) and green tea (GT) application
15 min prior and immediately after irradiation partially prevented SSR-induced LC depletion
Other flavonoids, the isoflavones found in plants such as red clover, soybean,
Psoralea corylifolia, and additional legumes, have been reported to possess signifi-
cant antioxidant, estrogenic, and tyrosine kinase inhibitory activity. Genistein is an
isoflavone and phytoestrogen typically derived from soybeans or red clover and is a
popular nutraceutical. Like GTPs, more work has been done on oral benefits (as
opposed to topical) and on other health issues such as breast and prostate cancers,
postmenopausal syndrome, diabetes, osteoporosis, and cardiovascular diseases.
Some promising work, primarily in animal and cell culture models, has been per-
formed showing genistein is also capable of providing photoprotection [14].
Hairless mice were protected against UVR-induced inflammation, edema, and
immunosuppression by topical applications of genistein, equol, isoequol, or dehy-
droequol [39]. A study published in 2003 demonstrated that topical genistein
potently inhibited UVB-induced photocarcinogenesis, decreased the levels of UVR-
induced CPDs, and blocked signs of photoaging in hairless mice [38]. Another 2003
publication examined the possible molecular signaling mechanisms for the genis-
tein beneficial effect on mediators of photoaging in human subjects [17]. UVR-
induced ROS are critical for MAP kinase activation, which leads to increased
expression of the transcription factor AP-1 (cFos/cJun), which in turn upregulates
MMP gene expression and degradation of the dermal extracellular matrix. This sec-
ond report showed that UVR-induced induction of EGF-R phosphorylation, cJun
protein, JNK MAP kinase, ERK MAP kinases, and MMP-1 was reduced by genis-
tein in human skin in vivo, thus strongly suggesting its value in prevention of
photoaging.
In a further study, genistein ameliorated the detrimental effects of UVB irradia-
tion in a human reconstituted skin model, namely, proliferating cell nuclear antigen
(PCNA) and CPDs [28]. It has been suggested that specific ratios of genistein and
368 M.S. Matsui
another isoflavone, daidzein, when combined and administered at specific ratios and
concentrations, exert a synergistic photoprotective effect that is greater than the
effect obtained with each isoflavone alone [16]. Indeed, the idea that these redox-
active compounds, which cooperate in an integrated manner in plants cells, also
may cooperate in animal cells has been reviewed before [13]. A network of
antioxidants with different chemical structures and properties may be needed for
optimal protection against oxidative damage.
Other botanical extracts that have some human experimental evidence to show
potential for topical photoprotection include resveratrol [9], grape seed [43], and
fern extract [10]. Resveratrol is a chemopreventive phytochemical found in grape
skin and seeds, red wine, peanuts, and fruits. Most works on the benefits of
resveratrol have used it as an oral supplement, but there is an array of animal studies
that support the exploration of topical resveratrol for photoprotection which have
been summarized previously [32]. Topical application of resveratrol in hairless
mice has been shown to reduce signs of oxidative stress and inflammation induced
by UVB exposure. In human subjects, daily topical application of a stabilized
resveratrol derivative, resveratrate, prior to irradiation with solar-simulated UVR
for four consecutive days, provided significant protection against erythema, mela-
nin synthesis, tanning, and sunburn cell formation compared to unprotected skin
[40]. Under the experimental conditions used, a typical antioxidant blend contain-
ing primarily ascorbate and tocopherol was not as effective against these endpoints.
The unique model used in this study, of repetitive irradiation, and further, the use of
solar-simulated UVR rather than UVB alone, has additional relevance and power to
demonstrate the value of this botanical material. Although not specifically on pho-
toprotective capabilities of resveratrol, an interesting report recently suggested that
the combination of topical resveratrol, baicalin, and vitamin E was able to reverse
the signs of skin photoaging by virtue of the blends antioxidant properties and its
ability to upregulate endogenous antioxidant defense systems [9].
The photoprotective properties of grape seed proanthocyanidins has been
demonstrated in human volunteers, also using a repetitive irradiation protocol, with
three UVR exposures (one per day) in which subjects received topical applications
of grape seed extract (GSPE) in solution or vehicle on sites that were then subjected
to 2MED solar-simulated radiation 30 min after treatment [43]. There was a
significant decrease in SBCs and p53+ cells in the GSPE+ UV group compared with
the UV group and the vehicle+ UV group. There was also significant protection
against UV-induced Langerhans cell depletion, illustrated in Fig. 20.4. Virtually all
of the very extensive research on the photoprotective properties of fern extract
(Polypodium leucotomos) have used the oral route of administration; however, there
is at least one report that it is efficacious when applied topically [10].
Ferulic acid, vitamin C, and vitamin E are not usually thought of as botanicals
but of course do exist in plantsferulic acid can be found in wheat, corn, and
legumes, among other sources. Human studies have been conducted with a topical
formulation of 15 % l-ascorbic acid, 1 % -tocopherol, and 0.5 % ferulic acid
(CEFer). In a study using subjects with Fitzpatrick skin type II or III and a protocol
in which CEFer was applied to skin over a 4-day period, skin was exposed to 2, 4,
20 The Role of Topical Antioxidants in Photoprotection 369
200 80
300
Number
Number
Number
150 60
200
100 40
100 *
50 20
*
0 0 0
uv
v
v
uv
V
al
uv
al
al
+u
+u
+u
U
U
rm
rm
rm
E+
E+
E+
vh
vh
vh
no
no
no
SP
SP
SP
G
G
different treatment groups different treatment groups different treatment groups
Fig. 20.4 Characterization of normal skin or skin treated with 2-MED simulated solar radiation
(SSR) only or skin treated with vehicle or GSPE followed 30 min later by 2-MED SSR. Specimens
were taken 24 h after UV exposure. Skin biopsies were analyzed for sunburn cells (SBCs), p53
positive cells, and density of Langerhans cells (Modified from Yuan et al. [43])
6, 8, and 10MED on sites treated with CEFer and evaluated 1 day later [29]. CEFer
was very effective at reducing thymine dimer mutations. It also provided substan-
tial protection against erythema and upregulation of the immune suppressive cyto-
kine IL-10 and significantly decreased sunburn cells, p53 expression, and
proinflammatory cytokine mRNA expression. In another study using Chinese sub-
jects, a single, 5-MED dose of ssUVR substantially induced large amounts of sun-
burn cell formation, thymine dimer formation, overexpression of p53 protein, and
depletion of CD1a+ Langerhans cells [41]. The antioxidant complex containing
vitamins C and E and ferulic acid conferred significant protection against these
endpoints.
Interestingly, the ferulic acid papers lead to another aspect of topical antioxidant
use that has been addressed more than once, following conflicting data on tumor
incidence after long-term antioxidant application and tissue culture experiments
showing a prooxidant effect of (usually high concentrations of) antioxidants. In at
least one study, topical 5 % alpha-tocopherol promoted carcinogenesis when applied
on chronically UVB-irradiated mouse skin [4]. However, a stabilized formulation of
vitamin E combined with vitamin C and ferulic acid decreased tumor number and
tumor burden and prevented the development of malignant skin tumors in female
mice with UVB-irradiated skin.
It has recently become apparent that human skin may be at increased risk of
photoaging from infrared radiation as well as UVA and UVB. The biological
significance and mechanisms of action for visible and IR wavelengths in human
skin is discussed in Chap. 3. The relevance to this chapter is that the mechanism by
which infrared radiation (IR), in particular near-infrared radiation (IRA radiation,
760-1,440 nm), causes damage is through oxidative stress [11]. In a recent study, an
SPF 30 sunscreen was tested versus the same sunscreen supplemented with an anti-
oxidant cocktail containing grape seed extract, vitamin E, ubiquinone, and vitamin
C to evaluate protection against IRA [12]. Exposure to IRA radiation significantly
upregulated MMP-1 expression, and treatment with the SPF30 sunscreen alone did
not provide significant protection, but the MMP-1 response was significantly
reduced if the SPF30 sunscreen plus the antioxidant cocktail was applied prior to
IRA radiation.
370 M.S. Matsui
The IRA study introduces another issue that needs to be addressed when speaking
of topical antioxidants for photoprotection, which is the addition of topical antioxi-
dants to UVA/UVB sunscreens, a scenario already taking place in the consumer land-
scape. Few studies have addressed the effects of this combination, although some
have suggested that because the mechanism of action is different from sunscreens
they would be expected to add to protection provided by sunscreens. Ideally, topical
antioxidants would improve protection against photoaging and carcinogenesis caused
by UV irradiation even in the presence of broad-spectrum UVA+ UVB sunscreens.
Two separate studies using the same formulation of antioxidants and sunscreens
were published that show additional protection is possible even when an SPF 25
broad-spectrum sunscreen is used. A sunscreen containing benzophenone, avoben-
zone, and octyl methoxycinnamate was compared to the same product plus ascorbyl
phosphate, tocopherol acetate, Echinacea pallida extract, chamomile extract, and
caffeine. Because MMP1 is the major enzyme implicated in collagen damage and
photoaging of UV-irradiated human skin, the first study asked whether the addition of
antioxidants to an SPF 25 sunscreen would improve protection against solar-simulated
UVR-induced activation of MMP1 after one exposure [26]. Both sunscreen alone and
sunscreen plus antioxidants reduced the expression of MMP1 relative to unprotected
ssUVR-irradiated control skin (Fig. 20.5). With no protection, the average increase in
MMP1 was 4.75-fold; with sunscreen alone, the increase was 2.4-fold; and in skin
treated with sunscreens plus antioxidants, the increase was only 1.75-fold. The differ-
ence in protection between the sunscreen alone and the sunscreen plus antioxidants
was significant and suggests that additional benefit against sun damage can be gained
by adding antioxidants to sunscreens. In another clinical study with the same sun-
screen and sunscreen plus antioxidants formulas but using a repetitive irradiation
model [42] with exposure to 1.5 MED for 4 consecutive days, additional protection
provided by antioxidants was also shown. Antioxidants alone did not reduce erythema,
10
2 MED + SS vs 2 MED + SS + AOx, P < 0.05
8
(% area)
0
No 2 MED 2 MED +
2 MED
ssUVR + SS SS+Aox
Fig. 20.5 The effect of ssUVR, sunscreens, and sunscreens plus antioxidants on MMP1 expression
in human skin in vivo. The skin of human subjects was treated as indicated: (a) no ssUVR, (b)
2-MED ssUVR, (c) 2 MED plus SSAOx, and (d) 2 MEDSS alone. Biopsies were analyzed by
immunohistochemistry using a monoclonal antibody to MMP1
20 The Role of Topical Antioxidants in Photoprotection 371
a b
160 120
Erythema Index at 24 hours
140
100
120
80
100
80 60
60
40
40
20
20
0 0
Baseline UV Only Vehicle+UV SS+UV AOx+UV SS+AOx SS+AOx Baseline UV Only Vehicle+UV SS+UV AOx+UV SS+AOx SS+AOx
Control Control Control +UV Control Control Control +UV
Treatment Group Treatment Groups
Fig. 20.6 Effect of sunscreens (SS) with or without antioxidants (AOx) on (a) erythema or (b)
melanin formation after repetitive ultraviolet (UV) radiation exposure. Yellow bars, irradiated sites;
blue bars, nonirradiated sites (Modified from [42])
372 M.S. Matsui
exposure levels may not have been sufficiently challenging, or entirely relevant, as
the model was ex vivo pig skin.
Therefore, one of the caveats for phytochemical photoprotection includes the
lack of regulatory standards by which this additional protection can be measured
and then articulated on product labels. In fact, in most of the world, no concentration
needs to be given, and they must be listed as inactive ingredients. More credible
testing of finished product using relevant endpoints on human subjects should be
undertaken, the optimal concentration should be determined for individual
components and mixtures of ingredients, and the stability of ingredients need to be
carefully monitored (as antioxidants are notoriously unstable). In addition, the
bioavailability/delivery of active supplements to the skin needs to be better
understood, as both water- and oil-soluble materials can protect lipids against UVR-
induced peroxidation; however, the bioactive portion must be able to partition into
the lipid bilayers to be protective [27]. There also exists potential synergy or
antagonism between ingredients, which under certain conditions such as high
concentrations, can act as prooxidants. Finally, in part because antioxidants are
frequently supplied as semicharacterized botanical extracts, the risk of adverse reac-
tions such as allergic or irritant sensitization must be considered.
In conclusion, there is ample evidence that certain antioxidants and botanical
extracts have potential to contribute to photoprotection when used topically. While
not to be recommended as alternatives to sun avoidance, broad-spectrum sunscreens,
and protective clothing, they should be considered valuable adjuvants in the preven-
tion of photoaging and skin cancer. Supplemental photoprotection will benefit those
who have heightened personal and professional risk factors such as Fitzpatrick skin
types III; environmental risk factors such as occupational exposure, geographic
location, and elevation; extended outdoor recreational activities; patients with previ-
ous skin cancer, photosensitive dermatological conditions, or patients on medication
that renders them photosensitive; and immunosuppressed patients. However, caution
must be taken when communicating the benefits of topical antioxidants to consumers,
who may already be bombarded with misinformation, such as the statements taken
from Internet sites and which include phrases such as research says antioxidants
work better than sunscreen, our skin is so well designed that when the solar rays hit
it the antioxidants that are in the body actually move up and form a protective shield
and act just like sunscreen, and antioxidants are the exact answerthey act just like
sunscreens. Ideally, any product with a claim to enhance photoprotection with anti-
oxidants would have been clinically tested under relevant and scientifically rigorous
conditions.
Abbreviations
ECG ()-Epicatechin-3-gallate
EGC ()-Epigallocatechin
EGCG ()-Epigallocatechin-3-gallate
GTPs Green tea polyphenols
IR Infrared radiation
LCs Langerhans cells
ROS Reactive oxygen species
SPF Sun protection factor
ssUVR Solar-simulated light
TPs Tea polyphenols
UVR Ultraviolet radiation
References
13. Halvorsen BL, Carlsen MH, Phillips KM, Bhn SK, Holte K, Jacobs DR Jr, Blomhoff R
(2006) Content of redox-active compounds (ie, antioxidants) in foods consumed in the United
States. Am J Clin Nutr 84:95135
14. Huang CC, Hsu BY, Wu NL, Tsui WH, Lin TJ, Su CC, Hung CF (2010) Anti-photoaging
effects of soy isoflavone extract (aglycone and acetylglucoside form) from soybean cake. Int
J Mol Sci 11(12):47824795
15. Ibiebele TI, van der Pols JC, Hughes MC, Marks GC, Williams GM, Green AC (2007) Dietary
pattern in association with squamous cell carcinoma of the skin: a prospective study. Am J Clin
Nutr 85(5):14011408
16. Iovine B, Iannella ML, Gasparri F, Monfrecola G, Bevilacqua MA (2011) Synergic effect of
genistein and daidzein on UVB-induced DNA damage: an effective photoprotective combina-
tion. J Biomed Biotechnol 2011:692846
17. Kang S, Chung JH, Lee JH, Fisher GJ, Wan YS, Duell EA, Voorhees JJ (2003) Topical
N-acetyl cysteine and genistein prevent ultraviolet-light-induced signaling that leads to
photoaging in human skin in vivo. J Invest Dermatol 120(5):835841
18. Katiyar SK (2011) Green tea prevents non-melanoma skin cancer by enhancing DNA repair.
Arch Biochem Biophys 508(2):152158
19. Katiyar SK, Matsui MS, Elmets CA, Mukhtar H (1999) Polyphenolic Antioxidant (-)-epigal-
locatechin-3-gallate from Green Tea Reduces UVB-induced Inflammatory Responses and
Infiltration of Leukocytes in Human Skin. Photochem Photobiol 69:148153
20. Katiyar SK, Challa A, McCormick T, Cooper K, Mukhtar H (1999) Prevention of UVB-
induced immunosuppression in mice by the green tea polyphenol ())-epigallocatechin-3-
gallate may be associated with alterations in IL-10- and IL-12 production. Carcinogenesis
20:21172124
21. Katiyar SK, Elmets CA, Agarwal R, Mukhtar H (1995) Protection against ultraviolet-B radia-
tion-induced local and systemic suppression of contact hypersensitivity and edema responses
in C3H HeN mice by green tea polyphenols. Photochem Photobiol 62:855861
22. Khan N, Afaq F, Mukhtar H (2008) Comprehensive invited review cancer chemoprevention
through dietary antioxidants: progress and promise. Antioxid Redox Signal 10(3) doi:10.1089/
ars.2007.1740, Mary Ann Liebert, Inc
23. Maia Campos PMBG, Gianeti MD, Kanashiro A, Lucisano-Valim YM, Gaspar LR (2006) In
vitro antioxidant and in vivo photoprotective effects of an association of bioflavonoids with
liposoluble vitamins. Photochem Photobiol 82:683688
24. Matsui MS, DeLeo VA (1990) Longwave ultraviolet radiation and promotion of skin cancer.
Cancer Cells 3:812
25. Matsui MS, Scheide S, Laufer L, DeLeo VA (1989) Ultraviolet B radiation inhibits epidermal
growth factor binding to mammalian cells. J Invest Dermatol 92:617622
26. Matsui MS, Hsia A, Miller JD, Hanneman K, Scull H, Cooper KD, Baron E (2009) Non-
sunscreen photoprotection: antioxidants add value to a sunscreen. J Investig Dermatol Symp
Proc 14(1):5659
27. Mishra AK, Mishra A, Chattopadhyay P (2011) Herbal cosmeceuticals for photoprotection
from ultraviolet B radiation: a review. Trop J Pharm Res 10(3):351360
28. Moore JO, Wang Y, Stebbins WG, Gao D, Zhou X, Phelps R, Lebwohl M, Wei H (2006)
Photoprotective effect of isoflavone genistein on ultraviolet B-induced pyrimidine dimer
formation and PCNA expression in human reconstituted skin and its implications in
dermatology and prevention of cutaneous carcinogenesis. Carcinogenesis 27(8):16271635
29. Murray JC, Burch JA, Streilein RD, Iannacchione MA, Hall RP, Pinnell SR (2008) A topical
antioxidant solution containing vitamins C and E stabilized by ferulic acid provides protection
for human skin against damage caused by ultraviolet irradiation. J Am Acad Dermatol
59(3):418425
30. Nichols JA, Katiyar SK (2010) Skin photoprotection by natural polyphenols: anti-inflammatory,
antioxidant and DNA repair mechanisms. Arch Dermatol Res 302(2):7183
20 The Role of Topical Antioxidants in Photoprotection 375
31. OyetakinWhite P, Tribout H, Baron E (2012) Protective mechanisms of green tea polyphenols
in skin. Oxid Med Cell Longev 2012:Article ID 560682
32. Passantino L, Costa M and Matsui MS (2013). Skin Cancer Prevention Strategies.
In: Highlights in Skin Cancer, Dr. Pierre Vereecken (Ed.), ISBN: 978-953-51-1073-6, InTech,
Available from: http://www.intechopen.com/books/highlights-in-skin-cancer/skin-cancer-
prevention-strategies
33. Perner D, Vierktter A, Sugiri D, Matsui M, Ranft U, Esser C, Ethve S, Goralczyk R, Kaneko
N, Yamamoto A, Morita A, Krutmann J, Krmer U (2011) Association between sun-exposure,
smoking behaviour and plasma antioxidant levels with the different manifestation of skin age-
ing signs between Japanese and German women a pilot study. J Dermatol Sci
62(2):138140
34. Schalka S, Manoel Silva dos Reis V (2011) Sun protection factor: meaning and controversies.
An Bras Dermatol 86(3):507515
35. Schneider LA, Bloch W, Kopp K, Hainzl A, Rettberg P, Wlaschek M, Horneck G, Scharffetter-
Kochanek K (2006) 8-Isoprostane is a dose-related biomarker for photo-oxidative ultraviolet
(UV) B damage in vivo: a pilot study with personal UV dosimetry. Br J Dermatol
154(6):11471154
36. Schwarz A, Maeda A, Gan D, Mammone T, Matsui MS, Schwarz T (2008) Green tea phenol
extracts reduce UVB-induced DNA damage in human cells via interleukin-12. Photochem
Photobiol 84(2):350355
37. Wang SQ, Osterwalder U, Jung K (2011) Ex vivo evaluation of radical sun protection factor in
popular sunscreens with antioxidants. J Am Acad Dermatol 65(3):525530
38. Wei H, Saladi R, Lu Y, Wang Y, Palep SR, Moore J, Phelps R, Shyong E, Lebwohl MG (2003)
Isoflavone genistein: photoprotection and clinical implications in dermatology. J Nutr
133:3811S3819S
39. Widyarini S, Spinks N, Husband AJ, Reeve VE (2001) Isoflavonoid compounds from red
clover (Trifolium pratense) protect from inflammation and immune suppression induced by
UV radiation. Photochem Photobiol 74(3):465470
40. Wu Y, Jia LL, Zheng YN, Xu XG, Luo YJ, Wang B, Chen JZ, Gao XH, Chen HD, Matsui M,
Li YH (2013) Resveratrate protects human skin from damage due to repetitive ultraviolet
irradiation. J Eur Acad Dermatol Venereol 27(3):345350
41. Wu Y, Zheng X, Xu XG, Li YH, Wang B, Gao XH, Chen HD, Yatskayer M, Oresajo C (2013)
Protective effects of a topical antioxidant complex containing vitamins C and E and ferulic
acid against ultraviolet irradiation-induced photodamage in Chinese women. J Drugs Dermatol
12(4):464468
42. Wu Y, Matsui MS, Chen JZ, Jin X, Shu CM, Jin GY, Dong GH, Wang YK, Gao XH, Chen HD,
Li YH (2011) Antioxidants add protection to a broad-spectrum sunscreen. Clin Exp Dermatol
36(2):178187
43. Yuan XY, Liu W, Hao JC, Gu WJ, Zhao YS (2012) Topical grape seed proanthocyandin extract
reduces sunburn cells and mutant p53 positive epidermal cell formation, and prevents deple-
tion of Langerhans cells in an acute sunburn model. Photomed Laser Surg 30(1):2025
44. Zussman J, Ahdout J, Kim J (2010) Vitamins and photoaging: do scientific data support their
use? J Am Acad Dermatol 63(3):507525
Chapter 21
The Role of DNA Repair in Photoprotection
Life on earth evolved utilizing solar electromagnetic energy, but at the same time,
this energy has adverse biological effects. The extent of the effects on the skin
depends greatly on the wavelength of light absorbed by its biomolecules. The most
damaging are the shorter wavelengths in the ultraviolet (UV) region because they
are most readily absorbed by the skin.
By convention, UV wavelengths are designated as UVA (320400 nm), UVB
(290320 nm), and UVC (200290 nm). The shorter the wavelength, the greater
absorption of the UV energy by earths atmosphere. UVC, the shortest wavelength
band, is effectively absorbed by atmosphere stratospheric gases and therefore fails
to reach the surface of the earth. The ozone molecules and atmosphere efficiently
filter UVB, so that only a small fraction actually reaches the earth surface (around
5 %). Its local intensity may vary with the solar zenith angle, which differs by the
time of day, the year, the latitude, and the local cloud density. For the long UV
wavelengths, 95 % of UVA energy reaches the earth with its steady presence during
the day, making it the most abundant [35].
Artificial light from incandescent light bulbs and compact fluorescent lamps
present an additional source of UV exposure, mostly UVA. The International
Commission on Illumination recommends maximal UV radiation of 30 J/m2 within
8 h. While the average daily exposure from outdoors is much lower, the cumulative
effects might be significant due to prolonged and continual daily exposures [41].
DNA directly absorbs the energy of UVC and UVB irradiation. The adsorbed energy
causes intranucleotide cross-linking by dimerization of pyrimidines and formation
of cis-syn cyclobutane pyrimidine dimers (CPDs) and pyrimidine (64) pyrimidone
photoproducts (6-4PP) [30, 49]. To a much lesser extent, purine dimers and pyrimi-
dine photohydrates are formed as well. The cyclobutane rings of CPDs are formed
between the 5,6 bonds of two adjacent pyrimidine bases (thymine, cytosine, or
5-methylcytosine). CPD formation is influenced by sequence context [42] and
formed exclusively at dipyrimidines and preferentially at TT sites. The efficiency of
CPD formation at different dipyrimidine sequences is estimated at a ratio of
55:33:11:1 for TT > TC > CT > CC [10]. In addition to the nucleotide sequences, the
chromatin structure and its environment have a significant impact on the distribution
of CPDs and the rate of their repair. Efficient repair in regions of DNA damage
requires nucleosomal rearrangements to allow DNA repair complex initiation.
The formation of (64)PPs arise through a complex electron rearrangement result-
ing in generation of a single covalent bond between position 6 and position 4 of two
adjacent pyrimidine bases [27]. The frequency of (64)PPs formation by UVB is at
the same level as the formation of CPDs but is repaired at much faster rate [49].
For a while it was assumed that UVA could not induce CPDs due to the inability
of DNA to efficiently absorb in the UVA range. However, CPDs were readily
detected upon UVA exposures [2, 33]. After exposure of cultured cells and the skin
to large doses of UVA, higher ratios of oxidized purines to CPDs are found than in
naked DNA [4]. Analysis of the CPDs produced by UVA revealed that the
predominant site for CPD formation is at TT compared to TC and CT sites and that
(64)PPs are almost undetectable.
The exact mechanism of CPD formation upon UVA irradiation is still subject of
debate. Some data suggests involvement of yet undetermined UVA chromophore
that is capable of transferring energy to DNA by photosensitization a triplet energy
transfer mechanism [13]. Other evidence supports direct DNA absorption, with
much lower efficiency than that of UVB. This absorption has a very distinctive
signature exclusive TT dimer formation [15].
Recently, a new pathway for formation of CPDs has been described wherein
fragments of melanin are excited by UV-induced reactive oxygen and nitrogen
species and then transfer the energy to DNA to form CPDs [32] This process is
remarkable in that CPDs continue to form even in the dark. The relative importance
of this photochemical reaction in the overall yield of DNA damage in intact human
skin is an exciting new area of research.
The knowledge of DNA repair pathways has gone from an arcane corner of nucleic
acid biochemistry to the subject of a college textbook [11]. The molecular details of
the reactions that lead to reversal, or removal and resynthesis, of damaged DNA can
be found there. Here we discuss the particular aspects of DNA repair that can
prevent photodamage and their sequelae.
DNA damage induced either directly or indirectly by sunlight is roughly
randomly distributed among the target nucleotides in the genome. However, because
the information content of the nucleotides is not randomly distributed within the
genome, the biological consequences of DNA lesions are not of equal importance.
As a result, repair of a minority of lesions, such as in the exons or on the transcribed
strand, has much greater biological importance than repair of others in the introns
or non-transcribed strands. DNA repair systems, both endogenous and therapeutic,
have indeed focused on repairing some regions, such as transcribed strands, faster
than others, in order to relieve phototoxic effects.
Here we will focus only on the main DNA repair pathways for photodamage.
excision repair recognizes it, and conversely, the more subtle the nucleotide
modification, the longer it takes to find and remove them. The great advantage of
this system is that it is not lesion specific, so that nucleotide excision repair can
remove damage that the organism has never experienced before, including modern
chemical carcinogen adducts that were invented in the last 100 years.
This pathway has many substrates, but it is not fast. It may take only 10 min to
incise UV-induced lesions [18], but following a sunburn it may take 12 h to remove
half the cyclobutane pyrimidine dimers in exposed skin [43].
Base excision repair uses one lead glycosylase enzyme that recognizes a small class
of modified bases and releases them from the phosphodiester backbone to create
vacant (abasic) sites in DNA. These sites are then repaired by a common set of
enzymes that remove the damaged regions on one strand and replace only about 4
nucleotides. The lead enzymes have narrow substrate specificity, but fortunately,
several are custom fit for DNA damage induced by sunlight. Important oxidation
photoproducts, particularly 8-oxo-dGua, are quickly and efficiently repaired by
base excision repair in about 6 h.
One strategy for enhancing DNA repair is to introduce into skin cells glycosylases
specific for cyclobutane pyrimidine dimers. This shifts the repair pathway from
nucleotide to base excision repair. Not only does it speed up repair but it also reduces
the frequency of mutagenic mistakes [46].
21.2.3 Photoreactivation
Human cells harbor a fail-safe mechanism for handling DNA photodamage. They
have polymerase (eta) that, during replication of a photodamaged DNA template,
quickly and efficiently inserts the correct nucleotide opposite a pyrimidine lesion.
21 The Role of DNA Repair in Photoprotection 381
Although this doesnt remove the damage, it preserves the genome integrity until an
excision mechanism can recognize and remove it. A genetic defect in this fail-safe
mechanism produces the cancer-prone xeroderma pigmentosum variant
phenotype.
DNA repair enzymes and pathways are closely coordinated with the rest of the
cells functions. Foremost among these coordinators is the p53 protein. Loss of its
function is a perquisite for many skin cancers. DNA damage triggers release of p53
protein from its inhibitor, which frees it to form a transcription activator for its tar-
get genes. Most of these genes code for cell cycle checkpoints, inhibitors of prolif-
eration and activators of DNA repair. Sustained activation of p53 protein leads to
apoptosis and cell death. In this way, p53 gives the cell a greater opportunity to
repair its DNA and, failing that, a road to suicide to avoid mutations and oncogenic
transformation.
A large number of DNA Damage Response (DDR) proteins, many of them
activated by p53, work together to signal that cell cycling should stop [7]. DNA
repair activity is further tied to the health status of the cell through AMPK
(5-AMP-activated protein kinase), which senses energy levels in cells and whose
activation increases DNA repair [44]. Furthermore, single-stranded breaks in DNA
produced during repair can activate poly(ADP-ribose) polymerase to consume
NAD, which saps the cell of molecules essential to production of ATP and lower
cellular energy.
DNA repair is tied not only to the cell cycle but also to the circadian
rhythm. This should not be surprising since the risk of photodamage to skin
DNA is directly related to the presence of the sun in the sky. The genes and
proteins in human cells that produce a feedback loop to create the circadian
clock (BMal1, Clock, Cryptochrome, and Period) also regulate the cell cycle
and DNA repair [ 34 ]. The peak of DNA repair capacity is late afternoon, just
as the accumulation of daytime sun damage to skin DNA is reaching its
maximum.
The DDR genes, including p53, work through regulation of transcription.
Downstream of transcription, miRNA (micro-RNA) are also modulated following
UV, and they further regulate the DDR genes by increasing or decreasing gene
silencing complexes [31]. Cell survival after UV is dependent on the proper
functioning of the gene silencing apparatus.
Many of the steps of the DDR pathways involve protein modification of the
downstream target. These modifications include classical phosphorylation,
acetylation, and, as we have discussed, poly(ADP-ribosyl)ation, which serve to
activate or inhibit enzyme activity. Another form of modification is ubiquitin and/or
SUMO (small ubiquitin-related modifier) additions to protein, which may coordinate
assembly of protein complexes or designate them for destruction to make way for a
repair response [40].
382 N. Karaman-Jurukovska and D.B. Yarosh
The simplest way to intervene in DNA repair is to accelerate the first step of DNA
repair, the recognition and incision of damaged bases. This has been accomplished
by encapsulating various enzymes in liposomes for delivery into skin cells, including
T4 endonuclease V [47] and M. luteus UV endonuclease [8] for CPDs, OGG1 for
8-oxo-dGua [45], and photolyase for direct reversal of CPDs [39]. These exogenous
but small enzymes are indeed able to enter the nucleus and recognize and then repair
DNA damage in mammalian skin.
The hormone -MSH protects the skin not only by inducing protective pigment
but also by inducing p53 and subsequent reduction in cell cycling and initiation of
DNA repair [14], a property that may be shared with the -MSH analog
afamelanotide, now undergoing clinical testing.
Induction and synchronization of the circadian rhythm by delivery of peptides to
skin cells has been reported to amplify DNA repair [25]. Application of such
peptides at night may therefore accelerate repair of DNA damage accumulated
during the day.
Binding of certain ligands to receptors activates DNA repair even in the absence
of a DNA damage inducing signal. IL-12 binding to its receptor increases repair of
UV-induced cyclobutane pyrimidine dimers [36]. The toll-like receptors TLR-3 and
TLR-4 mediate damage-associated pattern recognition (DAMP). Agonists of these
receptors modulate DNA repair after UV [1, 12]. They may act in part by activating
p53 [26]. Since extracellular DNA is recognized as DAMP and bound by TLRs, this
may explain the observations that dTpT and small oligonucleotides activate DNA
repair through a p53-dependent mechanism [22]. TLRs also distinguish pathogenic
from benign surface bacteria, and this may also explain the long-standing observation
that extracts of probiotic bacteria enhance DNA repair [3].
HMGB1 (high-mobility group protein B1) is a component of histones but also
participates in intercellular communication and recruitment of stem cells to the skin
from bone marrow. It is able to activate DNA repair and increase survival after UV
[21]. This may provide a new use for compounds modulating HMGB1 levels in the
skin.
Antioxidants naturally block oxidation of DNA and are discussed in Chap. 20.
There are recurrent reports of antioxidants inhibiting the formation of cyclobutane
pyrimidine dimers by UV (e.g., [23]). One explanation might be that antioxidant
polyphenols, such as from green tea or polypodium leucotomos, induce IL-12,
which then activates the DNA repair pathways to remove cyclobutane pyrimidine
dimers [17]. Another may be that antioxidants inhibit energy transfer by oxidized
melanin fragments [32].
21 The Role of DNA Repair in Photoprotection 383
References
2. Besaratinia A, Yoon JI, Schroeder C, Bradforth SE, Cockburn M, Pfeifer GP (2011) Wavelength
dependence of ultraviolet radiation-induced DNA damage as determined by laser irradiation
suggests that cyclobutane pyrimidine dimers are the principal DNA lesions produced by
terrestrial sunlight. FASEB J 25(9):30793091
3. Born A, Born W (1987) Replicative and repair DNA synthesis after solar damage. Acta
Dermatol Venerol 134:4042
4. Cadet J, Douki T (2011) Oxidatively generated damage to DNA by UVA radiation in cells and
human skin. J Invest Dermatol 131(5):10051007
5. Cai B-X, Jin S-L, Luo D et al (2009) Ginsenoside Rb1 suppresses ultraviolet radiation-induced
apoptosis by inducing DNA repair. Biol Pharm Bull 32:837841
6. Chen H, Weng Q, Fisher D (2014) UV signaling pathways within the skin. J Invest Dermatol
134:20802085
7. Cotta-Ramusino C, McDonald E, Hurov K et al (2011) A DNA damage response screen identi-
fies RHINO, a 9-1-1 and TopBP1 interacting protein required for ATR signaling. Science
332:13131317
8. DeBoyes T, Kouba D, Ozog D et al (2010) Reduced number of actinic keratoses with topical
application of DNA repair enzyme creams. J Drug Dermatol 9:1151911521
9. Dong K, Damaghi N, Picart S, Markova N, Obayashi K, Okano Y, Masaki H, Grether-Beck S,
Krutmann J (2008) UV-induced DNA damage initiates release of MMP-1 in human skin. Exp
Dermatol 17:10371044
10. Douki T, Cadet J (2001) Individual determination of the yield of the main UV-induced dimeric
pyrimidine photoproducts in DNA suggests a high mutagenicity of CC photolesions.
Biochemistry 40(8):24952501
11. Friedberg E, Walker G, Siede W, Wood R, Schultz R, Ellenberger T (eds) (2006) DNA repair
and mutagenesis. ASM Press, Washington DC
12. Harberts E, Gaspari AA (2013) TLR signaling and DNA repair: are they associated? J Invest
Dermatol 133:296302
13. Ikehata H, Kawai K, Komura J, Sakatsume K, Wang L, Imai M, Higashi S, Nikaido O,
Yamamoto K, Hieda K, Watanabe M, Kasai H, Ono T (2008) UVA1 genotoxicity is mediated
not by oxidative damage but by cyclobutane pyrimidine dimers in normal mouse skin. J Invest
Dermatol 128(9):22892296
14. Kadekaro A, Chen J, Yang J et al (2012) Alpha-melanocyte-stimulating hormone suppresses
oxidative stress through a p53-mediated signaling pathway in human melanocytes. Mol Cancer
Res 10:778786
15. Kappes UP, Luo D, Potter M, Schulmeister K, Runger TM (2006) Short- and long-wave UV light
(UVB and UVA) induce similar mutations in human skin cells. J Invest Dermatol 126(3):667675
16. Katiyar S, Mantena S, Meeran S (2011) Silymarin protects epidermal keratinocytes from ultra-
violet radiation-induced apoptosis and DNA damage by nucleotide excision repair mechanism.
PLoS One 6, e21410
17. Katiyar S, Elmets CA, Katiyar SK (2007) Green tea and skin cancer: photoimmunology,
angiogenesis and DNA repair. J Nutr Biochem 18:287296
18. Kaufmann W, Briley L (1987) Reparative strand incision in saponin-permeabilized human
fibroblasts. Mutat Res 184:237243
19. Kirkland JB (2012) Niacin requirements for genomic stability. Mutat Res 733:1420
20. Koo S-W, Hirakawa S, Fujii S et al (2007) Protection from photodamage by topical application
of caffeine after ultraviolet irradiation. Br J Dermatol 156:957964
21. Lange S, Mitchell D, Vasquez K (2008) High mobility group protein B1 enhances DNA repair
and chromatin modification after DNA damage. Proc Natl Acad Sci U S A 105:1032010325
22. Lee MS, Yaar M, Eller MS et al (2009) Telomeric DNA induces p53-dependent reactive oxy-
gen species and protects against oxidative damage. J Dermatol Sci 56(3):154162
23. Lin JY, Selim MA, Shea CR et al (2003) UV photoprotection by combination topical antioxi-
dants vitamin C and vitamin E. J Am Acad Dermatol 48:866874
24. Lucas D, Ke M, Matsui M, Maes D, Cooper K, Stevens S, Baron E (2008) Immune protective
effect of a moisturizer with DNA repair ingredients. J Cosmet Dermatol 7:132135
21 The Role of DNA Repair in Photoprotection 385
25. Maes D, Pernodet N, Mammone T et al (2010) Skin repair compositions comprising circadian
gene activators and a synergistic combination of sirt 1 gene activators. US Patent Appl
20100028317
26. Menendez D, Shatz M, Azzam K et al (2011) The toll-like receptor gene family is integrated
into human DNA damage and p53 networks. PLoS Genet 7:e1001360. doi:10.1371/
journalpgen.1001360
27. Mitchell DL, Nairn RS (1989) The biology of the (64) photoproduct. Photochem Photobiol
49(6):805819
28. Noonan FP, Zaidi MR, Wolnicka-Glubisz A, Anver MR, Bahn J, Wielgus A, Cadet J, Douki T,
Mouret S, Tucker MA, Popratiloff A, Merlino G, De Fabo EC (2012) Melanoma induction by
ultraviolet A but not ultraviolet B radiation requires melanin pigment. Nat Commun 3:884
29. Pellosi MC, Suzukawa AA, Scalfo AC, Di Mascio P, Martins Pereira CP, de Souza Pinto NC,
de Luna Martins D, Martinez GR (2014) Effects of the melanin precursor 5,6-dihydroxy-
indole-2-carboxylic acid (DHICA) on DNA damage and repair in the presence of reactive
oxygen species. Arch Biochem Biophys 557:5564
30. Pfeifer GP (1997) Formation and processing of UV photoproducts: effects of DNA sequence
and chromatin environment. Photochem Photobiol 65(2):270283
31. Pothof J, Verkaik N, van Ijcken W et al (2009) MicroRNA-mediated gene silencing modulates
the UV-induced DNA-damage response. EMBO J 28:20902099
32. Premi S, Wallisch S, Mano C, Weiner A, Bacchiocchi A, Wakamatsu K, Bechara E, Halaban
R, Douki T, Brash D (2015) Chemiexcitation of melanin derivatives induces DNA
photoproducts long after UV exposure. Science 347:842847
33. Rochette PJ, Therrien JP, Drouin R, Perdiz D, Bastien N, Drobetsky EA, Sage E (2003) UVA-
induced cyclobutane pyrimidine dimers form predominantly at thymine-thymine dipyrimidines
and correlate with the mutation spectrum in rodent cells. Nucleic Acids Res
31(11):27862794
34. Sancar A, Lindsey-Boltz L, Kang T-H et al (2010) Circadian clock control of the cellular
response to DNA damage. FEBS Lett 584:26182625
35. Schuch AP, Yagura T, Makita K, Yamamoto H, Schuch NJ, Agnez-Lima LF, Macmahon RM,
Menck CF (2012) DNA damage profiles induced by sunlight at different latitudes. Environ
Mol Mutagen 53(3):198206
36. Schwarz T, Schwarz A (2009) DNA repair and cytokine responses. J Investig Dermatol Symp
Proc 14:6366
37. Shah P, He Y (2014) Molecular regulation of UV-induced DNA repair. Photochem Photobiol.
doi:10.1111/php.12406
38. Simon JD, Peles D, Wakamatsu K, Ito S (2009) Current challenges in understanding melano-
genesis: bridging chemistry, biological control, morphology, and function. Pigment Cell
Melanoma Res 22(5):563579
39. Stege H, Roza L, Vink A et al (2000) Enzyme plus light therapy to repair DNA damage in
ultraviolet-B-irradiated human skin. Proc Natl Acad Sci U S A 97:17901795
40. Thomson T, Guerra-Rebollo M (2010) Ubiquitin and SUMO signaling in DNA repair.
Biochem Soc Trans 38:116131
41. Tian Y, Liu W, Niu T, Dai C, Li X, Cui C, Zhao X, Yaping E, Lu H (2014) The injury and
cumulative effects on human skin by UV exposure from artificial fluorescence emission.
Photochem Photobiol 90(6):14331438
42. Tornaletti S, Rozek D, Pfeifer GP (1993) The distribution of UV photoproducts along the
human p53 gene and its relation to mutations in skin cancer. Oncogene 8(8):20512057
43. Tung B, McGregor W, Wang Y, Maher V, McCormick J (1996) Comparison of the rate of exci-
sion of major UV photoproducts in the strands of the human HPRT gene of normal and xero-
derma pigmentosum variant cells. Mutat Res 2:6574
44. Wu C, Qiang L, Han W et al (2013) Role of AMPK in UVB-induced DNA damage repair and
growth control. Oncogene 32:26822689
45. Wulff B, Schick J, Thomas-Ahner J et al (2008) Topical treatment with OGG1 enzyme affects
UVB-induced skin carcinogenesis. Photochem Photobiol 84:317321
386 N. Karaman-Jurukovska and D.B. Yarosh
46. Yarosh D (2002) Enhanced DNA repair of cyclobutane pyrimidine dimers changes the biologi-
cal response to UV-B radiation. Mutat Res 509:221226
47. Yarosh D, Alas L, Yee V et al (1992) Pyrimidine dimer removal enhanced by DNA repair
liposomes reduces the incidence of UV skin cancer in mice. Cancer Res 52:42274231
48. Yarosh D, Klein J, OConnor A, Hawk J, Rafal E, Wolf P (2001) Effect of topically applied T4
endonuclease V in liposomes on skin cancer in xeroderma pigmentosum: a randomized study.
Lancet 357:926929
49. Yoon JH, Lee CS, OConnor TR, Yasui A, Pfeifer GP (2000) The DNA damage spectrum
produced by simulated sunlight. J Mol Biol 299(3):681693
50. Yu SH, Bordeaux JS, Baron ED (2014) The immune system and skin cancer. Adv Exp Med
Biol 810:182191
Chapter 22
Oral and Systemic Photoprotection
Key Points
Oral photoprotective agents could potentially be a suitable complement to
topical sunscreen photoprotection.
The effect of oral photoprotective agents is mainly systemic, by reducing
photoinduced oxidation, skin photodamage, and photoaging.
In addition to their photoprotective effect, some of these agents also have
anticancer properties.
Many botanical agents and formulations have antioxidant activity/
properties that provide photoprotection through different mechanisms.
Evaluation of the photoprotective effect of oral agents includes
photoimmunoprotection, antimutagenic, and antioxidant activities.
Photoprotection typically involves applying a thin layer of barrier agents to the skin
prior to sun exposure. There are practical limitations with this approach, e.g., uneven
and/or cumbersome application, short half-life on skin, potential side effect of the
agents, lack of systemic efficiency, etc.
Oral photoprotectives do not protect the skin directly from high-energy pho-
tons; hence, they are not very efficient against solar-induced erythema. However,
they are simple to take (usually as pills), their half-life can be determined phar-
macologically, and they do have systemic effects. These products may contain
one or several active substances that promote different mechanisms of photopro-
tection, particularly related to antioxidant activities [6, 7]. These mechanisms
reload the antioxidant activities of the body after systemic loss of endogenous
antioxidants during UV exposure [8, 9]. In many cases, oral photoprotective
agents also downregulate UV-induced immunosuppression [10]. In the next sec-
tions, we provide an update on the best characteristics of these substances
(Table 22.1).
22.1.1.1 Carotenoids
They are plant pigments present in a wide array of vegetables and fruits, most
notably tomato. Carotenoids are endowed with antioxidant activity. Although one
study did not support their efficacy on skin photoprotection [11], a modest, dose-
dependent photoprotective effect has been reported upon oral administration in
another study [12].
The most represented carotenoid in tomato is lycopene. Lycopene displays
a significant activity as a singlet oxygen scavenger. One study reported that
1012-week treatment with oral lycopene (16 mg daily) renders subjects less
prone to develop erythema in response to UV [13]. In addition, oral admin-
istration of a mixture of 2.5 mg lycopene, 4.7 mg beta-carotene, and 5 108
Lactobacillus johnsonii protected the development of UVA-induced polymor-
phous light eruption [14].
Other carotenoids, globally named xanthophylls, include astaxanthin,
lutein, and zeaxanthin. These have been shown to prevent sun-induced ery-
thema when administered orally, particularly in combination with topical
administration of the same compounds [15]. Combined oral and topical admin-
istration of lutein and zeaxanthin provided higher degree of antioxidant protec-
tion that either one alone. Astaxanthin has been shown to inhibit the production
of lipid peroxides and decrease the accumulation of polyamines induced by
UVA photons [16].
22 Oral and Systemic Photoprotection 389
22.1.1.2 Nicotinamide
It is the amide version of vitamin B3. It has proven useful for the management of
acne, photoaging, and photoimmunosuppression. The underlying mechanism
involves modulation of inflammatory cytokine expression and other enzymatic
mechanisms related to DNA repair [17]. Oral administration of nicotinamide or
390 S. Gonzlez et al.
These two vitamins with potent antioxidant properties are not photoprotective when
used separately [2022]. However, they enhance the photoprotective effects of each
other [23, 24]. Furthermore, T opical application of combinations of both vitamins
with melatonin also enhanced the photoprotective response against UV-induced
erythema [25]. The mechanism of this synergy is unclear, but it may depend on the
ability of ascorbate to reduce tocopherol, transferring the free radicals captured by
the latter to the medium, where they are quenched by other antioxidant systems
present in the skin [26].
22.1.2.3 Seresis
This general term includes antioxidant and anti-inflammatory flavonoids and other
phenolics found in vegetable foodstuffs. Flavonoids are the most important natural
antioxidants due to their chemical nature, which contains phenolic rings that can
absorb free radicals to form phenoxy radicals [29]. There are different subfamilies
22 Oral and Systemic Photoprotection 391
Extracts made from leaves of the fern Polypodium leucotomos are rich in
polyphenols. These antioxidant compounds are the molecular basis for the common
use of Polypodium leucotomos extracts to treat inflammatory skin conditions in
indigenous Central American cultures. In addition, these extracts can modulate
immune phenomena in response to inflammatory insult, e.g., UV-induced sunburn.
Fernblock is active topically and orally. At a molecular level, it scavenges free
radicals, e.g., singlet oxygen, hydroxyl, and superoxide, and prevents lipid peroxi-
dation [41, 42]. Fernblock enhances the function of endogenous antioxidant sys-
tems such as glutathione S-transferase (GST) [43], and it inhibits the isomerization
392 S. Gonzlez et al.
and inactivation of trans-urocanic acid [44]; it also prevents oxidant DNA damage
(8-hydroxyguanine) and accelerates the repair of damaged DNA, particularly T-T
dimers [45], which underlies its ability to prevent immunosuppression [46]. Finally,
Fernblock prevents inflammation by inhibiting UV-induced expression of TNF-
alpha and inducible nitric oxide synthase [47] and COX-2 [45].
The role of Fernblock has been assessed in psoralen-UVA (PUVA) and UVB-
induced changes [4850]. In both cases, oral Fernblock administration elevated the
threshold of UV-induced tanning (melanogenic dose) and the minimum UV dose to
cause erythema (erythematogenic dose) [28] . Beyond the new threshold, Fernblock
decreased the degree of erythema (i.e., the erythema vs. time slope was less steep),
reduced the number of sunburn cells, and ameliorated skin immune cell depletion
[51, 52]. A recent study described that oral administration of Fernblock in a hair-
less mouse model delayed the onset of skin tumors and increases p53 expression in
UV-irradiated skin [53].
Topical treatment with Fernblock had comparable effects in skin sensitivity to
UV radiation and immune depletion, with similar mechanisms involved, i.e.,
inhibition of oxidation and inflammation and immune protection. In addition,
topical application assays have indicated anti-photoaging capability and long-term
immune protection, including reduced elastosis and development of skin tumors in
response to chronic exposure to UVB [54]. It has also been shown to decrease the
development of polymorphous light eruption and solar urticaria [50].
22.1.3.3 Isoflavones
the bioavailability of other drugs [65], which may compromise its usefulness in
oral photoprotection schemes.
Quercetin: It is a flavonoid mainly found in oak bark and many other vegetables and
seeds. It is a very potent antioxidant with topical photoprotective effect [66], but
it has not been assessed as an oral photoprotection agent. Similar to silymarin, it
may affect the bioavailability of other drugs, e.g., paclitaxel [67], and it has an
effect on DNA cleavage [68]. Hence, its potential as an oral photoprotector is
controversial.
Apigenin: It is a flavonoid endowed with antitumor capability [69]. Topically, it
decreases tumor emergence upon exposure to UV photons in a rodent model. This
effect seems to be caused, at least in part, by inhibition of COX2 expression [70].
The flesh, peel, and seeds of pomegranate contain high amounts of polyphenols,
including catechins, anthocyanidins (e.g., delphinidin, cyanidin, and pelargonidin),
and tannins. Pomegranate has a strong reputation as a natural antioxidant [71]. As
an oral photoprotector, the Mukhtar group has described its efficacy in preventing
photocarcinogenesis in a UVB-irradiated mouse model [72, 73].
22.1.3.7 Resveratrol
It is a polyphenolic phytoalexin found in the peels and seeds of grapes, nuts and
fruits, and red wine. Its topical photoprotective effects are well documented [77].
Oral administration of resveratrol in a p53-sensitive mouse tumor model decreases
the onset of UV-mediated tumorigenesis [78], and this effect is related to its ability
to modulate TGF-beta [78] and NF-kB [79]. In addition, resveratrol boosts the
response to radiation therapy in hyperproliferative, precancerous, and neoplastic
conditions [80].
22.1.3.9 Pycnogenol
Pycnogenol is an extract of the bark of Pinus pinaster Ait. It is endowed with potent
antioxidant, anti-inflammatory, and anticarcinogenic properties, but it has only been
used topically to confer photoprotection [84].
22.1.3.10 Sulforaphane
22.1.3.11 Forskolin
Cats claw is a climbing plant indigenous from the Andes region. Water-soluble
extracts from its leaves have displayed high efficacy in topical photoprotection
assays, with a remarkable ability to enhance cyclobutyl pyrimidine dimer repair
[95]. Orally, cat claw extracts have not been assayed for photoprotection, but it is
well tolerated and decreases experimental endometriosis in a rodent model [96].
It is an extract from the root of Polygonum multiflorum (PM), with a long history in
Traditional Chinese Medicine. It displays antibacterial, antifungal, and antiaging
properties and has topical photoprotective effect [97]. However, its efficacy as an
oral photoprotector remains to be determined.
22.1.4.2 Probiotics
22.1.4.3 Idebenone
Idebenone is a more lipophilic analog of coenzyme Q10, which has higher skin pen-
etrance. Idebenone alleviates the onset of UV-induced photoaging [101], although
this is controversial [102]. Its efficacy as an oral photoprotector has not been
addressed, but its oral administration increases nerve growth factor (NGF) production
[103], and it is beneficial in patients with Lebers hereditary optic neuropathy [104].
human use, but it can be obtained illegally on the Internet and other sources [110] for
tanning and cosmetic and recreational purposes [111]. Very recent studies report anec-
dotal coincidence of melanotan II use with the emergence of melanoma [112, 113].
Non-topical photoprotection is a rapidly expanding field that still lacks gold standards
and is vulnerable to counterfeit and fraud (e.g., the current situation with the distribu-
tion and effect of melanotan II). But the premise of non-topical, especially oral, pho-
toprotection holds undeniable promise. Of course, oral photoprotective agents are not
meant to completely substitute topical photoprotection. Although some substances,
e.g., forskolin, increase epidermal thickening, UV irradiation of the skin will always
damage unprotected cells. Oral supplementation is aimed at countering the long-term
effects of sun exposure, which are more related to immunosuppression, chronic
inflammation, and photocarcinogenesis. Our current view is that the field strongly
needs standardization for the assessment of the effectiveness of oral photoprotection,
398 S. Gonzlez et al.
References
18. Gensler HL, Williams T, Huang AC, Jacobson EL (1999) Oral niacin prevents photocarcino-
genesis and photoimmunosuppression in mice. Nutr Cancer 34(1):3641
19. Surjana D, Halliday GM, Martin AJ, Moloney FJ, Damian DL (2012) Oral nicotinamide
reduces actinic keratoses in phase II double-blinded randomized controlled trials. J Invest
Dermatol 132(5):14971500
20. Fuchs J, Kern H (1998) Modulation of UV-light-induced skin inflammation by D-alpha-
tocopherol and L-ascorbic acid: a clinical study using solar simulated radiation. Free Radic
Biol Med 25(9):10061012
21. McArdle F, Rhodes LE, Parslew R, Jack CI, Friedmann PS, Jackson MJ (2002) UVR-induced
oxidative stress in human skin in vivo: effects of oral vitamin C supplementation. Free Radic
Biol Med 33(10):13551362
22. McArdle F, Rhodes LE, Parslew RA, Close GL, Jack CI, Friedmann PS et al (2004) Effects
of oral vitamin E and beta-carotene supplementation on ultraviolet radiation-induced
oxidative stress in human skin. Am J Clin Nutr 80(5):12701275
23. Eberlein-Konig B, Ring J (2005) Relevance of vitamins C and E in cutaneous photoprotec-
tion. J Cosmet Dermatol 4(1):49
24. Zussman J, Ahdout J, Kim J (2010) Vitamins and photoaging: do scientific data support their
use? J Am Acad Dermatol 63(3):507525
25. Dreher F, Gabard B, Schwindt DA, Maibach HI (1998) Topical melatonin in combination
with vitamins E and C protects skin from ultraviolet-induced erythema: a human study
in vivo. Br J Dermatol 139(2):332339
26. Wefers H, Sies H (1988) The protection by ascorbate and glutathione against microsomal
lipid peroxidation is dependent on vitamin E. Eur J Biochem 174(2):353357
27. Cesarini JP, Michel L, Maurette JM, Adhoute H, Bejot M (2003) Immediate effects of UV
radiation on the skin: modification by an antioxidant complex containing carotenoids.
Photodermatol Photoimmunol Photomed 19(4):182189
28. Greul AK, Grundmann JU, Heinrich F, Pfitzner I, Bernhardt J, Ambach A et al (2002)
Photoprotection of UV-irradiated human skin: an antioxidative combination of vitamins E and C,
carotenoids, selenium and proanthocyanidins. Skin Pharmacol Appl Skin Physiol 15(5):307315
29. Graf E (1992) Antioxidant potential of ferulic acid. Free Radic Biol Med 13(4):435448
30. Agati G, Brunetti C, Di Ferdinando M, Ferrini F, Pollastri S, Tattini M (2013) Functional
roles of flavonoids in photoprotection: new evidence, lessons from the past. Plant Physiol
Biochem 72:3545
31. Katiyar S, Elmets CA, Katiyar SK (2007) Green tea and skin cancer: photoimmunology,
angiogenesis and DNA repair. J Nutr Biochem 18(5):287296
32. Elmets CA, Singh D, Tubesing K, Matsui M, Katiyar S, Mukhtar H (2001) Cutaneous photo-
protection from ultraviolet injury by green tea polyphenols. J Am Acad Dermatol
44(3):425432
33. Lee SR, Im KJ, Suh SI, Jung JG (2003) Protective effect of green tea polyphenol ()-epigal-
locatechin gallate and other antioxidants on lipid peroxidation in gerbil brain homogenates.
Phytother Res 17(3):206209
34. Rutter K, Sell DR, Fraser N, Obrenovich M, Zito M, Starke-Reed P et al (2003) Green tea
extract suppresses the age-related increase in collagen crosslinking and fluorescent products
in C57BL/6 mice. Int J Vitam Nutr Res 73(6):453460
35. Vayalil PK, Mittal A, Hara Y, Elmets CA, Katiyar SK (2004) Green tea polyphenols prevent
ultraviolet light-induced oxidative damage and matrix metalloproteinases expression in
mouse skin. J Invest Dermatol 122(6):14801487
36. Kim S, Lee MJ, Hong J, Li C, Smith TJ, Yang GY et al (2000) Plasma and tissue levels of tea
catechins in rats and mice during chronic consumption of green tea polyphenols. Nutr Cancer
37(1):4148
37. Farris P (2007) Idebenone, green tea, and Coffeeberry extract: new and innovative antioxi-
dants. Dermatol Ther 20(5):322329
38. Dvorakova K, Dorr RT, Valcic S, Timmermann B, Alberts DS (1999) Pharmacokinetics of
the green tea derivative, EGCG, by the topical route of administration in mouse and human
skin. Cancer Chemother Pharmacol 43(4):331335
400 S. Gonzlez et al.
39. Nwuha V, Nakajima M, Tong J, Ichikawa S (1999) Solubility study of green tea extracts in
pure solvents and edible oils. J Food Eng 40(3):161165
40. Hsu S (2005) Green tea and the skin. J Am Acad Dermatol 52(6):10491059
41. Gonzalez S, Pathak MA (1996) Inhibition of ultraviolet-induced formation of reactive oxy-
gen species, lipid peroxidation, erythema and skin photosensitization by Polypodium leucoto-
mos. Photodermatol Photoimmunol Photomed 12:4556
42. Gomes AJ, Lunardi CN, Gonzalez S, Tedesco AC (2001) The antioxidant action of
Polypodium leucotomos extract and kojic acid: reactions with reactive oxygen species. Braz
J Med Biol Res 34(11):14871494
43. Mulero M, Rodriguez-Yanes E, Nogues MR, Giralt M, Romeu M, Gonzalez S et al (2008)
Polypodium leucotomos extract inhibits glutathione oxidation and prevents Langerhans cell
depletion induced by UVB/UVA radiation in a hairless rat model. Exp Dermatol
17:653658
44. Capote R, Alonso-Lebrero JL, Garcia F, Brieva A, Pivel JP, Gonzalez S (2006) Polypodium
leucotomos extract inhibits trans-urocanic acid photoisomerization and photodecomposition.
J Photochem Photobiol B 82(3):173179
45. Zattra E, Coleman C, Arad S, Helms E, Levine D, Bord E et al (2009) Oral polypodium leu-
cotomos decreases UV-induced Cox-2 expression, inflammation, and enhances DNA repair
in Xpc +/ mice. Am J Pathol 175:19521961
46. Siscovick JR, Zapolanski T, Magro C, Carrington K, Prograis S, Nussbaum M et al (2008)
Polypodium leucotomos inhibits ultraviolet B radiation-induced immunosuppression.
Photodermatol Photoimmunol Photomed 24(3):134141
47. Janczyk A, Garcia-Lopez MA, Fernandez-Penas P, Alonso-Lebrero JL, Benedicto I, Lopez-
Cabrera M et al (2007) A Polypodium leucotomos extract inhibits solar-simulated radiation-
induced TNF-alpha and iNOS expression, transcriptional activation and apoptosis. Exp
Dermatol 16(10):823829
48. Nestor M, Bucay V, Callender V, Cohen JL, Sadick N, Waldorf H (2014) Polypodium leu-
cotomos as an adjunct treatment of pigmentary disorders. J Clin Aesthet Dermatol
7(3):1317
49. Gonzalez S, Gilaberte Y, Philips N (2010) Mechanistic insights in the use of a Polypodium
leucotomos extract as an oral and topical photoprotective agent. Photochem Photobiol Sci
9(4):559563
50. Choudhry S, Bhatia N, Ceilley R, Hougeir F, Lieberman R, Hamzavi I, Lim HW (2014) Role
of oral polypodium leucotomos extract in dermatologic diseases: a review of the literature.
J Drugs Dermatol 13(2):148153
51. Middelkamp-Hup MA, Pathak MA, Parrado C, Garcia-Caballero T, Rius-Diaz F, Fitzpatrick
TB et al (2004) Orally administered Polypodium leucotomos extract decreases psoralen-
UVA-induced phototoxicity, pigmentation, and damage of human skin. J Am Acad Dermatol
50(1):4149
52. Middelkamp-Hup MA, Pathak MA, Parrado C, Goukassian D, Rius-Diaz F, Mihm MC et al
(2004) Oral Polypodium leucotomos extract decreases ultraviolet-induced damage of human
skin. J Am Acad Dermatol 51(6):910918
53. Rodriguez-Yanes E, Cuevas J, Gonzalez S, Mallol J (2014) Oral administration of Polypodium
leucotomos delays skin tumor development and increases epidermal p53 expression and the
anti-oxidant status of UV-irradiated hairless mice. Exp Dermatol 23(7):526528
54. Alcaraz MV, Pathak MA, Rius F, Kollias N, Gonzlez S (1999) An extract of Polypodium
leucotomos appears to minimize certain photoaging changes in a hairless albino mouse
animal model. Photodermatol Photoimmunol Photomed 15:120126
55. Messina MJ, Persky V, Setchell KD, Barnes S (1994) Soy intake and cancer risk: a review of
the in vitro and in vivo data. Nutr Cancer 21(2):113131
56. Zhang T, Wang F, Xu HX, Yi L, Qin Y, Chang H et al (2013) Activation of nuclear factor
erythroid 2-related factor 2 and PPARgamma plays a role in the genistein-mediated
attenuation of oxidative stress-induced endothelial cell injury. Br J Nutr 109(2):223235
57. Zhai X, Lin M, Zhang F, Hu Y, Xu X, Li Y et al (2013) Dietary flavonoid genistein induces
Nrf2 and phase II detoxification gene expression via ERKs and PKC pathways and protects
against oxidative stress in Caco-2 cells. Mol Nutr Food Res 57(2):249259
22 Oral and Systemic Photoprotection 401
58. Wei H, Saladi R, Lu Y, Wang Y, Palep SR, Moore J et al (2003) Isoflavone genistein: photo-
protection and clinical implications in dermatology. J Nutr 133(11 Suppl 1):3811S3819S
59. Widyarini S, Spinks N, Husband AJ, Reeve VE (2001) Isoflavonoid compounds from red
clover (Trifolium pratense) protect from inflammation and immune suppression induced by
UV radiation. Photochem Photobiol 74(3):465470
60. Widyarini S, Husband AJ, Reeve VE (2005) Protective effect of the isoflavonoid equol
against hairless mouse skin carcinogenesis induced by UV radiation alone or with a chemical
cocarcinogen. Photochem Photobiol 81(1):3237
61. Reeve VE, Widyarini S, Domanski D, Chew E, Barnes K (2005) Protection against photoag-
ing in the hairless mouse by the isoflavone equol. Photochem Photobiol 81(6):15481553
62. Schwen RJ, Nguyen L, Plomley JB, Jackson RL (2012) Toxicokinetics and lack of utero-
tropic effect of orally administered S-equol. Food Chem Toxicol 50(5):17411748
63. Pinnell SR (2003) Cutaneous photodamage, oxidative stress, and topical antioxidant protec-
tion. J Am Acad Dermatol 48(1):119; quiz 2022
64. Crocenzi FA, Basiglio CL, Perez LM, Portesio MS, Pozzi EJ, Roma MG (2005) Silibinin
prevents cholestasis-associated retrieval of the bile salt export pump, Bsep, in isolated rat
hepatocyte couplets: possible involvement of cAMP. Biochem Pharmacol 69(7):11131120
65. Park JH, Park JH, Hur HJ, Woo JS, Lee HJ (2012) Effects of silymarin and formulation on
the oral bioavailability of paclitaxel in rats. Eur J Pharm Sci 45(3):296301
66. Casagrande R, Georgetti SR, Verri WA Jr, Dorta DJ, dos Santos AC, Fonseca MJ (2006)
Protective effect of topical formulations containing quercetin against UVB-induced oxidative
stress in hairless mice. J Photochem Photobiol B 84(1):2127
67. Choi JS, Jo BW, Kim YC (2004) Enhanced paclitaxel bioavailability after oral administration
of paclitaxel or prodrug to rats pretreated with quercetin. Eur J Pharm Biopharm
57(2):313318
68. Robinson MJ, Corbett AH, Osheroff N (1993) Effects of topoisomerase II-targeted drugs on
enzyme-mediated DNA cleavage and ATP hydrolysis: evidence for distinct drug interaction
domains on topoisomerase II. Biochemistry 32(14):36383643
69. Torkin R, Lavoie JF, Kaplan DR, Yeger H (2005) Induction of caspase-dependent, p53-
mediated apoptosis by apigenin in human neuroblastoma. Mol Cancer Ther 4(1):111
70. Tong X, Van Dross RT, Abu-Yousif A, Morrison AR, Pelling JC (2007) Apigenin prevents
UVB-induced cyclooxygenase 2 expression: coupled mRNA stabilization and translational
inhibition. Mol Cell Biol 27(1):283296
71. Afaq F, Mukhtar H (2006) Botanical antioxidants in the prevention of photocarcinogenesis
and photoaging. Exp Dermatol 15(9):678684
72. Afaq F, Saleem M, Krueger CG, Reed JD, Mukhtar H (2005) Anthocyanin- and hydrolyzable
tannin-rich pomegranate fruit extract modulates MAPK and NF-kappaB pathways and
inhibits skin tumorigenesis in CD-1 mice. Int J Cancer 113(3):423433
73. Afaq F, Khan N, Syed DN, Mukhtar H (2010) Oral feeding of pomegranate fruit extract
inhibits early biomarkers of UVB radiation-induced carcinogenesis in SKH-1 hairless mouse
epidermis. Photochem Photobiol 86(6):13181326
74. Perez-Sanchez A, Barrajon-Catalan E, Caturla N, Castillo J, Benavente-Garcia O, Alcaraz M
et al (2014) Protective effects of citrus and rosemary extracts on UV-induced damage in skin
cell model and human volunteers. J Photochem Photobiol B 136:1218
75. Scapagnini G, Davinelli S, Di Renzo L, De Lorenzo A, Olarte HH, Micali G et al (2014)
Cocoa bioactive compounds: significance and potential for the maintenance of skin health.
Nutrients 6(8):32023213
76. Heinrich U, Neukam K, Tronnier H, Sies H, Stahl W (2006) Long-term ingestion of high
flavanol cocoa provides photoprotection against UV-induced erythema and improves skin
condition in women. J Nutr 136(6):15651569
77. Flamini R, Mattivi F, De Rosso M, Arapitsas P, Bavaresco L (2013) Advanced knowledge of
three important classes of grape phenolics: anthocyanins, stilbenes and flavonols. Int J Mol
Sci 14(10):1965119669
78. Kim KH, Back JH, Zhu Y, Arbesman J, Athar M, Kopelovich L et al (2011) Resveratrol tar-
gets transforming growth factor-beta2 signaling to block UV-induced tumor progression.
J Invest Dermatol 131(1):195202
402 S. Gonzlez et al.
79. Adhami VM, Afaq F, Ahmad N (2003) Suppression of ultraviolet B exposure-mediated acti-
vation of NF-kappaB in normal human keratinocytes by resveratrol. Neoplasia 5(1):7482
80. Reagan-Shaw S, Mukhtar H, Ahmad N (2008) Resveratrol imparts photoprotection of normal
cells and enhances the efficacy of radiation therapy in cancer cells. Photochem Photobiol
84(2):415421
81. Saija A, Tomaino A, Trombetta D, De Pasquale A, Uccella N, Barbuzzi T et al (2000) In vitro
and in vivo evaluation of caffeic and ferulic acids as topical photoprotective agents. Int
J Pharm 199(1):3947
82. Lin FH, Lin JY, Gupta RD, Tournas JA, Burch JA, Selim MA et al (2005) Ferulic acid stabi-
lizes a solution of vitamins C and E and doubles its photoprotection of skin. J Invest Dermatol
125(4):826832
83. Nair SC, Panikkar B, Akamanchi KG, Panikkar KR (1991) Inhibitory effects of Ixora javan-
ica extract on skin chemical carcinogenesis in mice and its antitumour activity. Cancer Lett
60(3):253258
84. Sime S, Reeve VE (2004) Protection from inflammation, immunosuppression and carcino-
genesis induced by UV radiation in mice by topical Pycnogenol. Photochem Photobiol
79(2):193198
85. Cheung KL, Kong AN (2010) Molecular targets of dietary phenethyl isothiocyanate and
sulforaphane for cancer chemoprevention. AAPS J 12(1):8797
86. Dinkova-Kostova AT, Jenkins SN, Fahey JW, Ye L, Wehage SL, Liby KT et al (2006)
Protection against UV-light-induced skin carcinogenesis in SKH-1 high-risk mice by
sulforaphane-containing broccoli sprout extracts. Cancer Lett 240(2):243252
87. Dickinson SE, Melton TF, Olson ER, Zhang J, Saboda K, Bowden GT (2009) Inhibition of
activator protein-1 by sulforaphane involves interaction with cysteine in the cFos DNA-
binding domain: implications for chemoprevention of UVB-induced skin cancer. Cancer Res
69(17):71037110
88. Dinkova-Kostova AT (2008) Phytochemicals as protectors against ultraviolet radiation: ver-
satility of effects and mechanisms. Planta Med 74(13):15481559
89. DOrazio JA, Nobuhisa T, Cui R, Arya M, Spry M, Wakamatsu K et al (2006) Topical drug
rescue strategy and skin protection based on the role of Mc1r in UV-induced tanning. Nature
443(7109):340344
90. Spry ML, Vanover JC, Scott T, Abona-Ama O, Wakamatsu K, Ito S et al (2009) Prolonged
treatment of fair-skinned mice with topical forskolin causes persistent tanning and UV
protection. Pigment Cell Melanoma Res 22(2):219229
91. Scott TL, Christian PA, Kesler MV, Donohue KM, Shelton B, Wakamatsu K et al (2012)
Pigment-independent cAMP-mediated epidermal thickening protects against cutaneous UV
injury by keratinocyte proliferation. Exp Dermatol 21(10):771777
92. Passeron T, Namiki T, Passeron HJ, Le Pape E, Hearing VJ (2009) Forskolin protects kerati-
nocytes from UVB-induced apoptosis and increases DNA repair independent of its effects on
melanogenesis. J Invest Dermatol 129(1):162166
93. Gonzalez-Sanchez R, Trujillo X, Trujillo-Hernandez B, Vasquez C, Huerta M, Elizalde A
(2006) Forskolin versus sodium cromoglycate for prevention of asthma attacks: a single-
blinded clinical trial. J Int Med Res 34(2):200207
94. Bristow MR, Ginsburg R, Strosberg A, Montgomery W, Minobe W (1984) Pharmacology
and inotropic potential of forskolin in the human heart. J Clin Invest 74(1):212223
95. Emanuel P, Scheinfeld N (2007) A review of DNA repair and possible DNA-repair adjuvants
and selected natural anti-oxidants. Dermatol Online J 13(3):10
96. Nogueira Neto J, Coelho TM, Aguiar GC, Carvalho LR, de Araujo AG, Girao MJ et al (2011)
Experimental endometriosis reduction in rats treated with Uncaria tomentosa (cats claw)
extract. Eur J Obstet Gynecol Reprod Biol 154(2):205208
97. Hwang IK, Yoo KY, Kim DW, Jeong SJ, Won CK, Moon WK et al (2006) An extract of
Polygonum multiflorum protects against free radical damage induced by ultraviolet B
irradiation of the skin. Braz J Med Biol Res 39(9):11811188
22 Oral and Systemic Photoprotection 403
98. Rhodes LE, OFarrell S, Jackson MJ, Friedmann PS (1994) Dietary fish-oil supplementation
in humans reduces UVB-erythemal sensitivity but increases epidermal lipid peroxidation.
J Invest Dermatol 103(2):151154
99. Bouilly-Gauthier D, Jeannes C, Maubert Y, Duteil L, Queille-Roussel C, Piccardi N et al
(2010) Clinical evidence of benefits of a dietary supplement containing probiotic and
carotenoids on ultraviolet-induced skin damage. Br J Dermatol 163(3):536543
100. Weill FS, Cela EM, Paz ML, Ferrari A, Leoni J, Gonzalez Maglio DH (2013) Lipoteichoic
acid from Lactobacillus rhamnosus GG as an oral photoprotective agent against UV-induced
carcinogenesis. Br J Nutr 109(3):457466
101. McDaniel D, Neudecker B, Dinardo J, Lewis J 2nd, Maibach H (2005) Clinical efficacy
assessment in photodamaged skin of 0.5% and 1.0% idebenone. J Cosmet Dermatol
4(3):167173
102. Tournas JA, Lin FH, Burch JA, Selim MA, Monteiro-Riviere NA, Zielinski JE et al (2006)
Ubiquinone, idebenone, and kinetin provide ineffective photoprotection to skin when
compared to a topical antioxidant combination of vitamins C and E with ferulic acid. J Invest
Dermatol 126(5):11851187
103. Takeuchi R, Murase K, Furukawa Y, Furukawa S, Hayashi K (1990) Stimulation of nerve
growth factor synthesis/secretion by 1,4-benzoquinone and its derivatives in cultured mouse
astroglial cells. FEBS Lett 261(1):6366
104. Klopstock T, Yu-Wai-Man P, Dimitriadis K, Rouleau J, Heck S, Bailie M et al (2011) A ran-
domized placebo-controlled trial of idebenone in Lebers hereditary optic neuropathy. Brain
134(Pt 9):26772686
105. Kitazawa M, Ishitsuka Y, Kobayashi M, Nakano T, Iwasaki K, Sakamoto K et al (2005)
Protective effects of an antioxidant derived from serine and vitamin B6 on skin photoaging in
hairless mice. Photochem Photobiol 81(4):970974
106. Fischer SM, Lo HH, Gordon GB, Seibert K, Kelloff G, Lubet RA et al (1999) Chemopreventive
activity of celecoxib, a specific cyclooxygenase-2 inhibitor, and indomethacin against
ultraviolet light-induced skin carcinogenesis. Mol Carcinog 25(4):231240
107. Dorr RT, Lines R, Levine N, Brooks C, Xiang L, Hruby VJ et al (1996) Evaluation of mela-
notan-II, a superpotent cyclic melanotropic peptide in a pilot phase-I clinical study. Life Sci
58(20):17771784
108. Fabrikant J, Touloei K, Brown SM (2013) A review and update on melanocyte stimulating
hormone therapy: afamelanotide. J Drugs Dermatol 12(7):775779
109. Biolcati G, Marchesini E, Sorge F, Barbieri L, Schneider-Yin X, Minder EI (2014) Long-term
observational study of afamelanotide in 115 patients with erythropoietic protoporphyria. Br
J Dermatol 172(6):16011612
110. Langan EA, Nie Z, Rhodes LE (2010) Melanotropic peptides: more than just Barbie drugs
and sun-tan jabs? Br J Dermatol 163(3):451455
111. Wessells H, Fuciarelli K, Hansen J, Hadley ME, Hruby VJ, Dorr R et al (1998) Synthetic
melanotropic peptide initiates erections in men with psychogenic erectile dysfunction:
double-blind, placebo controlled crossover study. J Urol 160(2):389393
112. Hjuler KF, Lorentzen HF (2014) Melanoma associated with the use of melanotan-
II. Dermatology 228(1):3436
113. Ong S, Bowling J (2012) Melanotan-associated melanoma in situ. Australas J Dermatol
53(4):301302
114. Fourtanier A, Moyal D, Maccario J, Compan D, Wolf P, Quehenberger F et al (2005)
Measurement of sunscreen immune protection factors in humans: a consensus paper. J Invest
Dermatol 125(3):403409
115. Maier T, Korting HC (2005) Sunscreens which and what for? Skin Pharmacol Physiol
18(6):253262
Chapter 23
Photoprotection from Sunless Tanning
Products and Colored Cosmetics
Z.D. Draelos, MD
Department of Dermatology, Duke University School of Medicine,
2444 North Main Street, Durham, NC 27262, USA
e-mail: [email protected]
CH2OH
DHA is the active agent in all presently marketed sunless tanning products and is a
3-carbon sugar appearing as a white, crystalline hygroscopic powder. DHA is
formed when glycerol is fermented by Gluconobacter oxydans. It interacts with
amino acids, peptides, and proteins to form chromophores known as melanoidins
[2]. Melanoidins structurally have some similarities to skin melanin, but are not able
to function as electron donors to reactive oxygen species [3]. Tanning occurs when
melanin becomes oxidized, a reaction that cannot occur with DHA.
DHA interacts with the stratum corneum to form the melanoidins as the entire
brown color can be removed by tape stripping the skin. No DHA is found in the
viable epidermis or dermis after topical application accounting for its systemic
safety profile. Thus, the thicker the stratum corneum, the more deeply the skin will
pigment. For this reason, the brown is less intense on the face where the stratum
corneum is thin and more intense on the elbows and knees where the stratum
corneum is thicker. It also produces a much darker stain on the palms and the soles,
areas that do not normally tan.
In addition to the thickness of the stratum corneum, the color produced by DHA
is controlled by skin pH and the pH of the sunless tanning product. If the skin or the
formulation is alkaline, the DHA color will be more orange. Conversely, if the skin
or the formulation is acidic, the DHA color will be pinkish yielding a more natural
appearance. For this reason, manufacturers typically formulate their products at a
pH of 56 to yield the best color development.
The amount of water in the formulation can also affect the sunless tanning
product color. If too much water is present, the DHA color development will be less
as the water inhibits the melanoidin formation. For this reason, DHA products are
not formulated with glycerin, which is a humectant capable of attracting water.
Instead the DHA is placed in a propylene glycol and sorbitol vehicle to increase
melanoidin formation and the intensity of the stain produced.
23 Photoprotection from Sunless Tanning Products and Colored Cosmetics 407
The reaction that occurs on the skin surface creating the melanoidins, which yield
the simulated tan color, is known as the Maillard reaction (Fig. 23.2). The Maillard
reaction occurs when a protein binds to a sugar. Thus, the keratin protein of the skin
reacts with the sugar DHA to create the browning reaction [4]. DHA is technically
categorized as a colorant or colorless dye. It reacts with amines, peptides, and free
amino acids in the stratum corneum. The first step is the conversion of DHA to
pyruvaldehyde with the elimination of water. Then the ketone or aldehyde interacts
with skin keratin to form an imine [5]. The remaining specifics of the reaction are
still unknown, but the resulting products are cyclic and linear polymers that have a
yellow or brown color.
The chemical reaction is usually visible within 1 h after DHA application, but
maximal darkening may take 824 h [6]. Many sunless tanning products contain a
temporary dye to allow the user to note the sites of application and to promote even
application, but this immediate color should not be confused with the Maillard
reaction.
Fig. 23.2 The Maillard reaction. The Maillard reaction involves the interaction of the DHA sugar
with the stratum corneum keratin protein to produce a pigmented substance known as a melanoidin
408 Z.D. Draelos
DHA is a nontoxic ingredient both for ingestion and topical application. It has a
proven safety record with only a few reported cases of allergic contact dermatitis [9].
In the 1920s, it was determined that large quantities of oral DHA did not produce
toxicity, and the LD50 in rats is over 16 g/kg. It is interesting to note that the phos-
phate of DHA is one of the intermediates in the Krebs cycle known as dihydroxyac-
etone monophosphate. Topically applied DHA reacts immediately upon contact
with the stratum corneum amines and is not absorbed for this reason. DHA has not
been detected in the urine or serum of volunteers following topical application [7].
Patients who are allergic to one sunless tanning product may be allergic to all
sunless tanning products as DHA is a common ingredient. While the reported
instances of allergic contact dermatitis to DHA are few, practical experience
indicates a much higher incidence. The author personally sees two patients per
month with allergic contact dermatitis to sunless tanning products. To confirm the
allergy, it is best to patch test the patient to the product the patient purchased under
an occlusive bandage for 48 h followed by removal and evaluation 24 and 48 h later.
23 Photoprotection from Sunless Tanning Products and Colored Cosmetics 409
The final question remains the ability of DHA to provide photoprotection. DHA
was listed on the sunscreen monograph at one time but has since been replaced
by better agents with superior UV-absorbing qualities; therefore, it is no longer
considered a sunscreen ingredient. DHA absorbs long wavelength UV in the
300380 nm range [10]. It has an SPF of 34 [11].
What does this mean? It means that no SPF rating can be assigned to DHA, as it
now has become simply a cosmetic and not an over-the-counter drug. Nevertheless,
there is some photoprotective value to DHA. It was originally used in a 3 % concen-
tration in combination with 0.25 % lawsone. The advantage to DHA is that an irre-
versible reaction occurs when the DHA sugar binds to the keratin protein that cannot
be removed by rubbing. This is not the case with modern sunscreen formulations.
While it is important to remind patients that the simulated tanned appearance cre-
ated by sunless tanning creams requires additional sunscreen use, a sunless tanning
preparation applied first several days before an extended outdoor outing with addi-
tional sunscreen applied on top 30 min before sun exposure might be helpful.
In addition, there are some formulations of sunless tanning products that contain
monographed sunscreen ingredients. DHA can be combined with organic sunscreens
that do not contain amino groups, such as octyl methoxycinnamate, homosalate,
octocrylene, and benzophenone. It also can be combined with inorganic sunscreens
(zinc oxide and titanium dioxide). The challenge with inorganic sunscreen
combinations is that the zinc oxide and titanium dioxide can discolor brown in the
bottle if 5 % DHA is combined with 5 % inorganic sunscreen after only a few days.
Nevertheless, the use of sunless tanning creams containing sunscreens may
encourage application compliance.
after it has completely dried; it can also increase the SPF of a sunscreen by provid-
ing an additional layer of organic sunscreen ingredients (Fig. 23.3). In addition,
there are some pigmented powder sunscreen formulations that have become very
popular for patients with multiple allergies and sensitive skin, since the powder
formulations have ewer ingredients than cream or spray sunscreens with similar
SPF values.
Facial powders are composed of talc, also known as hydrated magnesium silicate,
combined with pigments that camouflage the underlying skin. The pigments used in
face powder listed in order of increasing opaqueness are: titanium dioxide, kaolin,
magnesium carbonate, magnesium stearate, zinc stearate, prepared chalk, zinc
oxide, rice starch, precipitated chalk, and talc. It is generally accepted that the
optimum opacity is achieved with a particle size of 0.25 m. This is important
because the opacity of a facial powder directly correlates with its ability to shield
the skin from UV radiation. Opaque facial powders that are used as stand-alone
sunscreens rely on higher concentrations of titanium dioxide, a monographed
inorganic sunscreen ingredient, to achieve their SPF rating. Adding various
concentrations of iron oxides, to match the various brown tones representative of the
human population, minimizes the cosmetically unattractive whiteness of the powder.
In addition to iron oxides as the main pigment, other inorganic pigments such as
ultramarines, chrome oxide, and chrome hydrate may be used. All of the additional
pigments may increase the product SPF, even though they are not monographed
sunscreen ingredients.
Facial powders are available in two formulations: opaque and transparent. The
opaque powders, previously discussed, mitigate the penetration of UV radiation and
can be applied on top of a spray, lotion, or cream sunscreen to augment the ability
of the sunscreen to shield the skin or used alone as an SPF-rated powder sunscreen.
It also prevents the transmission of visible light. Transparent powders are more
natural appearing due their ability to allow some light to reach the skin surface,
Fig. 23.3 Facial powder. Titanium dioxide and talc that form the basis for facial powder are also
used as inorganic sunscreens
23 Photoprotection from Sunless Tanning Products and Colored Cosmetics 411
but their sun-protective ability is diminished. Transparent powders have the same
formulation as full coverage powders except they contain less talc, titanium dioxide,
or zinc oxide, since coverage is not a priority. Transparent facial powders com-
monly have a light reflective shine, produced by nacreous pigments, such as bis-
muth oxychloride, mica, titanium dioxide-coated mica, or crystalline calcium
carbonate. They are not used as stand-alone sunscreens but can be dusted over a
sunscreen for added protection.
While facial powders contain ingredients that function as sunscreens, powder
can also increase the ability of the sunscreen film to remain in place on the skin
surface. One of the most common causes of sunscreen failure is removal of the
product due to rubbing, wiping, and water contact. The sunscreen film is also
degraded as it mixes with sweat and sebum. Magnesium carbonate can be added to
facial powders to absorb sebum, thus minimizing the ability of sebum to destroy the
sunscreen emulsion. Kaolin, also known as hydrated aluminum silicate, may also
function to absorb oil and perspiration [12]. Other specialty additives in more
expensive boutique powders include partially hydrolyzed ground raw silk, corn silk,
treated starch, and synthetic resins for increased oil and perspiration absorption.
Powder can be applied to the face over sunscreen with a brush, pad, or fingers.
Brushing the powder is least effective as the particles sit on top of the sunscreen, but
represents the easiest application method. Pressing the powder into the sunscreen
film with a pad or fingers is more effective as the powder becomes embedded in the
sunscreen, increasing the longevity of both products on the face. In general, a
transparent powder can increase the SPF of a sunscreen by 2 SPF points, for
example, from an SPF of 7 to an SPF of 9, while an opaque powder can increase the
sunscreen SPF by five numerical points. It is important to remember that the powder
does not improve the water resistance of the sunscreen film to abundant sweat,
precipitation, or submersion in water. Nevertheless, facial powders are a valuable
sunscreen adjuvant for some patients.
Facial foundations are another important category of cosmetic that can supplement
facial photoprotection. If you take the facial powder formulation discussed
previously and add a moisturizer, you end up with a facial foundation. Older
nomenclature labeled facial foundation a liquid powder. Facial foundations are
available in a variety of formulations to include liquid, mousse, water-containing
cream, souffl, anhydrous cream, stick, cake, and shake lotion [13]. Each of these
formulations offers a different degree of facial protection, partly due to the
ingredients in the facial foundation and to the addition of monographed sunscreen
ingredients. Thus, a more modern name for facial foundations might be a pigmented
sunscreen. The role of facial foundations in photoprotection will be explored.
The most popular facial foundations are liquid formulations containing water,
oils, titanium dioxide, and iron oxides. The liquid formulations can be further
412 Z.D. Draelos
characteristics. The cream can be applied to the face as dipped from a jar, wiped
from a compact, or stroked from a rod packaged in a roll-up tube [14]. For patients
with exquisite sun sensitivity, superior photoprotection is achieved by first applying
a traditional sunscreen followed by application of an anhydrous waterproof cream
foundation. For patients with the inability to use traditional sunscreens, the
anhydrous waterproof cream foundation can be used alone.
In summary, the ability of a facial foundation to provide sun protection is directly
proportional to its ability to conceal or cover the underlying skin, a quality known
as coverage. Further, the coverage of a foundation is directly related to the amount
of titanium dioxide, zinc oxide, talc, and kaolin in the formulation. Sheer coverage
foundations with minimal titanium dioxide are almost transparent and have an SPF
around 2, moderate coverage foundations are translucent and have an approximate
SPF of 45, anhydrous high-coverage foundations with large amounts of titanium
dioxide may be opaque, acting as a total physical sunblock which protects against
UV and visible light. Thus, facial foundations can be important tools for
photoprotection.
23.2.3 Lipsticks
The final colored cosmetic that has important photoprotective qualities is lipstick.
Lipsticks contain pigments that can function as sun-protective ingredients but can
also contain monographed sunscreen ingredients allowing them to possess an SPF
rating. The lips are a common site of actinic cheilitis and may also be afflicted with
squamous cell carcinoma. Lipsticks are an excellent cosmetic for preventing lip
photodamage.
Lipsticks are mixtures of waxes, oils, and pigments in varying concentration to
yield the characteristics of the final product (Fig. 23.4). Several different lipstick
formulations are currently marketed. Lipsticks labeled as long wearing are
excellent for photoprotection and are designed to remain on the lips for a prolonged
period of time. They are composed of high wax, low oil, and high pigment
concentrations, which accounts for their intrinsic SPF of 45 even though they do
not contain monographed sunscreen ingredients [15]. The waxes incorporated into
lipstick formulations are white beeswax, candelilla wax, carnauba wax, ozokerite
wax, lanolin wax, ceresin wax, and other synthetic waxes. Lipsticks combine these
waxes to achieve a desired melting point that controls the hardness of the lipstick
and the ability of the lipstick to coat the lips when applied. Oils are then selected,
such as castor oil, white mineral oil, lanolin oil, hydrogenated vegetable oils, or
oleyl alcohol, to form a film suitable for application to the lips. The thickness of the
film over the lips determines the degree of photoprotection provided and the ability
of the film to remain in place on the lips, but the photoprotection is due to the
pigments dispersed in the oil and the suspended in the waxes.
A variety of coloring agents are used in lipsticks to achieve the wide variety of
shades available in the marketplace. Since lipsticks are removed by eating, speaking,
414 Z.D. Draelos
Fig. 23.4 Lipstick. Opaque lipstick confers excellent photoprotection to the lips
and lip licking, they commonly are ingested. Thus, the US Food and Drug
Administration controls the coloring agents that can be used in lipsticks, which also
provide photoprotection. The Food and Drug Administration divides certified colors
into three groups: Food, Drug, and Cosmetic (FD&C) colors, Drug & Cosmetic
(D&C) colors, and External Drug & Cosmetic colors. Only the first two groups can
be used in lipsticks [16]. While these pigments can provide some photoprotection,
they are not monographed sunscreen ingredients. Additional monographed
ingredients can be added, converting the lipstick into an OTC drug. Most lipsticks
do not have an SPF over 30 because the addition of higher concentrations of
monographed sunscreen ingredients will give the product a bitter unpalatable taste.
The best lipsticks are those with a high titanium dioxide and pigment load that are
completely opaque when applied to the lips.
One unique type of lipstick that provides long-lasting protection contains a lip
stain [17]. These lip stains contain indelible coloring agents known as bromo acids,
consisting of fluoresceins, halogenated fluoresceins, and related water-insoluble
dyes [18]. These lipsticks are colored red and stain the lips a reddish color. The stain
produces some minimal photoprotection but is best combined with a sunscreen-
containing lip balm. The lip stain is applied first followed by the lip balm slightly
boosting the lip balm SPF.
23.3 Summary
Dermatologists typically think of sunscreen sprays, lotions, and creams when body
and facial photoprotection is required. This chapter expands the number of products
that should be considered. While sunless tanning creams offer minimal
photoprotection with an SPF of 34, they can be used as a safer tanning alternative
23 Photoprotection from Sunless Tanning Products and Colored Cosmetics 415
References
Key Points
Clothing provides simple and effective broad-spectrum photoprotection.
Photoprotection by clothing is affected by several factors, including the
material, thickness, and color.
Ultraviolet protection factor (UPF) is the in vitro assessment of
photoprotection of fabric. Similar to SPF, it is weighted toward the
assessment of protection against erythema, the predominant effect of UVB.
Clothing with UPF of at least 40 is preferred.
24.1 Background
Skin cancer is the most common type of cancer in the United States. In 2006, more
than one million people were diagnosed with basal cell carcinoma (BCC) or squa-
mous cell carcinoma (SCC). Malignant melanoma (MM), the third and most often
fatal type of skin cancer, is expected to be diagnosed in approximately 60,000 people
and hold into account for over 8000 deaths in 2007. Between 1975 and 2004, the
annual age-adjusted incidence rate for MM (new cases diagnosed per 100,000 people)
nearly tripled, from about 7 to 19 cases per 100,000 [13]. Solar UV radiation is ubiq-
uitous during daylight hours. Ambient ground-level UV is comprised mainly of UVA
(320400 nm) plus a small proportion (<10 %, variable by time of day, season, and
location) of UVB (290320 nm). Within-person and between-person UV doses vary
greatly, depending on location, time of day and season, clothing habits, and skin pig-
mentation [4]. Exposure to UV radiation on the skin results in demonstrable
mutagenic effects. The p53 suppressor gene, which is frequently mutated in skin can-
cers, is believed to be an early target of UV radiation inducing neoplasms such as SCC
[5]. Fair-skinned individuals, who are more sensitive to the effects of exposure at these
wavelengths, are at higher risk of developing skin cancer. The amount of average
annual UV radiation correlates with the incidence of skin cancer. There is a direct
relationship between the incidence of nonmelanoma skin cancer and latitude. The
closer an individual is to the equator, the greater the UV energy to which they are
exposed [6, 7]. MM mortality in the United States and Canada has also been shown to
directly correlate with ambient UV exposure. The correlation of MM incidence to UV
radiation exposure is greater when ambient UVA radiation is also included [69].
Apart from sun avoidance, the most frequently used form of UV protection is the
application of sunscreens. The use of textiles as a means of sun protection has been
underrated in previous education campaigns, even though suitable clothing potentially
offers usually simple and effective broad-spectrum protection against sunlight [1014].
In Australia, Cancer Council education campaigns have long urged the use of clothing
in conjunction with hats, sunglasses, and sunscreens as UV protection [15]. However,
a number of studies have recently shown that, contrary to popular opinion, some tex-
tiles provide insufficient UV protection [16]. In addition to skin cancer formation,
photoaging and photosensitive disorders (e.g., polymorphous light eruption, lupus ery-
thematosus, porphyrias, solar urticaria, and phototoxic/photoallergic reactions) may
also be prevented by UV protective clothing. Consequently, the use of suitable textiles,
which block UVB as well as UVA radiation, has been recommended for photosensitive
patients [1721]. Most of the photosensitive diseases are provoked by wavelengths in
the UVA range [19]. In some of these disorders (e.g., solar urticaria, chronic actinic
dermatitis), even very small UV doses can lead to exacerbation. The data of several
studies indicate that some aspects of sun protection are being practiced consistently,
while others, such as the use of UV protective clothing, are not [10, 22, 23].
Direct and diffuse UV transmittance through fabric is the crucial factor determining
the grade/amount of UV protection of textiles. Spectroradiometers and
spectrophotometers are suitable for the assessment of the spectral irradiance. These
measurements are usually performed in the wavelength range of 290400 nm and
operated in five or fewer nm steps. They are generally made under worst-case
conditions, with collimated radiation beams at a right angle to the fabric [10, 24]. To
determine the in vitro ultraviolet protection factor (UPF), the spectral irradiance
(both source * and transmitted # spectrum) is weighted against the erythemal action
spectrum [25]. The UPF is calculated as follows:
UPF = El S l d l / El S l Tl d l
24 Photoprotection by Clothing and Fabric 419
were tested on skin. The inconsistency of the data in previous studies is certainly
due to different methodologies (e.g., different test protocols, UV sources, and textile
materials).
For undyed fabrics, there are differences in the UV-absorbing properties of the fiber.
Summer clothing is usually made of cotton, viscose, rayon, linen, and polyester or
combinations thereof. Other materials such as nylon or elastane are also found in
bathing suits and nylon stockings (Table 24.1). Usually, consumers consider
lightweight non-synthetic fabrics, e.g., cotton, viscose, and linen, the most
comfortable for summer textiles [10, 42]. Comparing different types of material in
relation to the UPF is difficult and is only possible in a limited number of cases. In
the case of synthetic fibers (e.g., polyester, polyamide), the analysis is even more
difficult because the UV protection of these materials depends on the type and
amount of additives to the fiber (e.g., antioxidants or UV stabilizers). In particular,
polyester usually has good UV-blocking properties, since this fabric provides
relatively low UVB transmission. This is most likely due to a large conjugated
system in the polymer chains [43, 44]. Polyester or polyester blends may be the
most suitable type of fabric for UV protective garments. However, its permeability
for wavelength in the UVA range is frequently higher compared to other types of
fiber [18]; this is of crucial significance for many patients suffering from
photosensitive disorders. Bleached cotton and viscose rayon provide relatively low
UV protection. This was confirmed by a study of Crews et al. who reported that
bleached cotton print clothing had a UV transmission of 23.7 %, whereas unbleached
cotton print cloth showed a UV transmission of 14.4 %.
The influence of bleaching was also evident among silk fabrics in their study.
Compared to bleached textiles, unbleached fabrics such as cotton and silk have
better UV protective properties due to natural pigments absorbing UV radiation and
Table 24.1 Summary of parameters significantly influencing the UPF of apparel textiles
Fabric material UPF of cotton, viscose, rayon < linen, nylon, wool,
silk < polyestera
Fabric porosity, weight, and UPF increases with small yarn-to-yarn spaces, fabric weight,
thickness and thickness
Fabric color UPF increases with darker colors
UV absorbers UPF is improved by UV absorbers
Stretch UPF decreases under stretch
Wetness UPF decreases for wet cotton
Wash UPF increases for cotton fabrics
a
When other parameters are kept constant
24 Photoprotection by Clothing and Fabric 421
other impurities. Very few studies have considered the fiber-fabric construction
processing history of fabrics to fully elucidate the UV protection abilities of fab-
rics. Sarkar [45] recently reported the effect of fabric processing treatments, both
chemical and biochemical, on the transmission of UV radiation through selected
white and undyed fabrics. He reported that chemical processing methods such as
desizing and bleaching have a deleterious effect on UV transmission through fabric
[45]. Biochemical processing such as the use of enzymes is comparatively benign
and does not adversely impact the UV protective ability of cotton fabric. Grifoni
et al. [46] studied the UV protection properties of two fabrics made of natural fibers
(flax and hemp) which were dyed with some of the most common natural dyes. UV
transmittance of fabrics was measured by a spectrophotometer, and outdoor
measurements were taken by a spectroradiometer. Experimental results revealed
that natural dyes could confer good UV protection, depending mainly on their
different UV-absorbing properties, provided that the fabric construction already
guaranteed good cover. The authors also confirmed that UPFs calculated by in vitro
measurements were generally lower than those based on outdoor data, indicating an
underestimation of the actual level of protection of tested fabrics assessed by the
in vitro test [46].
Sarkar [47] investigated the UV properties of natural fabrics dyed with natural
colorants. Three cotton fabrics were dyed with three natural colorants. Fabrics were
characterized with respect to fabric construction, weight, thickness, and thread
count. A positive correlation between the weight of the fabric and their UPF values
was observed [47]. Similarly, thicker fabrics offered more protection from UV rays.
Thread count appears to negatively correlate with UPF. Dyeing with natural
colorants dramatically increased the protective abilities of all three fabric
constructions.
The fabric construction is a primary determinant of fabric porosity followed
by fabric weight and thickness of the textile [43]. An increased density concern-
ing the weave or knitting technique (smaller yarn-to-yarn spaces) leads to a
decreased fabrics porosity and consequently less UV radiation is transmitted.
Spaces between the yarns are frequently larger in a knit than in a woven textile.
Besides, plain-woven textiles have a lower porosity than textiles using other
weaves [48].
Thickness is a useful parameter for understanding differences in UV protec-
tion between fabrics. Crews et al. [43] reported that thicker, denser fabrics
transmitted less UV radiation. Therefore, they concluded that thickness is most
useful in explaining differences in UV transmission when differences in per-
centage cover are also accounted for [49]. By contrast, Kan and Au [50] recently
found that fabric weight is the most important factor to affect the UPF while
thickness and stitch density were not the main parameters determining UV
protection.
The color of the fabric may also influence the UPF since some dyes have an
absorption spectrum extending into the UV spectrum. Enhanced UV protection of
dyed textiles depends on the position and intensity of the absorption bands of the
dyes within the UV wavelength and the concentration of the dye in the textile.
422 T. Gambichler et al.
The absorbance of UV radiation can influence many substrate attributes, e.g., fluo-
rescence, photodegradation, and UV protection. Generally, dark colors provide bet-
ter UV protection due to increased UV absorption. This holds only true for the same
UV absorbent dye provided that other characteristics of the textile, e.g., fabric type
and construction, are the same [10]. However, dyes within particular hue types can
vary considerably in the degree of UV protectiveness due to their individual trans-
mission/absorption characteristics [51].
In order to improve UV protection, UV absorbers have been added recently with
different techniques [50]. UV absorbers are colorless compounds that absorb in the
wavelength range from 280 to 400 nm. Hilfiker et al. [52] found the cover factor to
be useful in predicting the maximum UPF that could be achieved by treating the
yarns with UV absorbers. Thus, fabrics could be made opaque to UV radiation with
a sufficient level of UV absorber impregnation. The corresponding UPFs approached
the theoretically predicted levels based on the cover factor. Osterwalder and Rohwer
[53] demonstrated that a UV absorber can be brought into contact with a fabric
during the wash or rinse cycle of a laundry operation. The high UV transmittance of
30 % of a thin, bleached cotton swatch in the dry state (UPF 3) can be reduced
tenfold to about 3 % (UPF >30) in ten washing cycles.
Titanium dioxide is frequently used as a UV-blocking substance in fabrics.
However, the absorptive and scattering properties of titanium dioxide particles in
the UVA wavelength range are different and depend mainly on the particle size and
geometry. Nevertheless, UV absorbers are suitable for significantly increasing UPF,
especially that of nondyed lightweight summer fabrics such as cotton and viscose
fabrics [10, 52, 5456]. Recently, Wang et al. [57] presented a facile process to
prepare uniform dumbbell-shaped ZnO crystallites. They discovered a unique
morphological effect on the UV-blocking property. The as-prepared ZnO crystallites
were characterized by different criteria including UV blocking and Raman scattering
spectra. The as-prepared structural material demonstrated a significant advance in
protective functional treatment and provided a potential commercialization [58].
Furthermore, Behler et al. [58] showed that the use of electro-spun nanofibers with
a high load of nanodiamond can provide UV protection.
Moon and Pailthorpe [59] showed that stretching elastane-based garments about
10 %, in both the machine and the cross-machine directions, causes a dramatic
decrease in the measured UPF of a textile. Their consumer survey also showed that,
on average, about 15 % stretch is achieved when these textiles are worn. However,
the 15 % stretch refers to power stretch, which is only a small segment of the
clothing market. Elastane-based textiles for tight fitting should not be considered
as defined UV protective clothing. Kimlin et al. [60] reported that the UPF of 50
denier stockings decreased 868 % when stretched 30 % of their original size.
24 Photoprotection by Clothing and Fabric 423
Notably, the most popular type of stockings (15 denier) provides a UPF less than 2
[61]. The maximum stretch point on the body for tight-fitting garments is the upper
back, where textiles can be stretched up to 15 %. However, realistically, the effect
of stretch on the UPF of a textile may be of significance only for garments with a
non-stretched UPF of less than 30, particularly leggings, womens stockings, and
swimsuits [10].
When textiles become wet, by humidity in the air, perspiration, or water, UV
transmission through the fabric can significantly change [62]. A marked reduction
of the UPF was observed for textiles made from cotton and cotton blends. However,
Wong et al. [63] recently reported that knitted fabrics with miss stitches retained
good UV protection even when the fabrics were stretched by 20 % of its original
dimensions. In a field-based study, it was shown that significant UV exposures may
occur underneath garments, particularly white cotton fabrics in a wet state. Similar
results were also observed in in vivo measurements of cotton and polyester blends
[37, 64, 58]. In case of fabrics made of viscose or silk, or in fabrics that have been
treated with broadband UV absorbers, the UPF frequently increases when the textile
becomes wet. This was also observed in a recent study of modal fabrics treated with
titan dioxide [37, 54]. Thus, UV protection of wet garments is not always poor.
Most of the fabrics will undergo a combination of relaxation shrinkage and
consolidation shrinkage when washed [65]. Therefore, the spaces between the yarns
will decrease and UV protection increases. The effect of laundering on the UPF puts
into perspective other fabric parameters and factors which decrease the UPF [10].
Stanford et al. [66] showed that UPFs of cotton T-shirts increased after the first
washing and did not change significantly with subsequent washing. Wang et al. [67]
observed only a moderate UPF increase of cotton fabrics after laundering. Adding
UV-absorbing agents during laundering was found to substantially enhance UPF
[67, 68]. Recently, Zhou and Crews [69] reported that UPF of cotton or cotton/
polyester blended fabrics can be significantly enhanced by repeated laundering of
the garment in a detergent containing optical brightening agent. This was not the
case for fabrics comprised entirely of polyester or nylon [69]. Prolonged wear and
tear beyond the standard lifetime of a garment may eventually cause thinning of
the individual fibers and consequently alter the UPF. Photostability of a textile and
its UV protectiveness is an important requirement for sun protective clothing [62].
Unfortunately, there are only limited data on the stability of the UV protectiveness
of a textile against UV radiation or infrared [10].
The first standard for sun protective clothing was published by the Australian
Standardisation Institute in 1996. This standard, referred to as AS/NZS 4399, has
set requirements for determining and labeling the UPF of sun protective fabrics and
other items that are worn in close proximity to the skin [26]. Based on the standard,
spectrophotometrically assessed UPF is for a specific type of fabric and does not
424 T. Gambichler et al.
address the degree of protection that is afforded by the design of a garment. The
effects of stretch, wetness, wear, and use are not included in the AS/NZS 4399.
According to the Australian/New Zealand standard, UPFs are classified in three
categories: UPFs of 1524 (ratings 15 and 20) offer good protection; UPFs of 2539
(ratings 25, 30, and 35), very good protection; and UPFs of 40 and higher (ratings
40, 45, 50, and 50+), excellent protection. Fabrics with a UPF of less than 15 are not
labeled. Three standard documents that pertain to the testing and labeling of UV
protective textile products were also published by the American Society for Testing
and Materials [70] and the American Association of Textile Chemists and Colorists
[71]. More recently, the European Committee for Standardization (CEN) has
developed a standard on requirements for test methods and labeling of sun protective
garments. The first part of the standard (EN 13758-1 [72],) includes all details of
test methods (e.g., spectrophotometric measurements) for textile materials, and part
2 (EN 13758-2 [73],) covers the classification and marking of apparel textiles [10].
UV protective clothing must fulfill all stringent instructions of testing, classification,
and marking including a UPF larger than 40 (UPF 40+), average UVA transmission
lower than 5 %, and design requirements as specified in part 2 of the standard to
claim the European standard as described above. A pictogram, which is marked
with the number of the standard EN 13758-2 and the UPF of 40+, shall be attached
to the garment if it is in compliance with the standard [74]. Moreover, British,
Canadian, South African, and multinational groups, including Commission on
Illumination (CIE) and also the International Organization for Standardization
(ISO), have been engaged in writing UV protective fabric standard documents [10].
24.6 Conclusions
Defined UV-blocking fabrics are important element not only in campaigns against
skin cancer but also in prevention of photosensitive disorders and photoaging. The
UPF of a garment depends on a variety of parameters, including fabric construction,
type, color, weight, thickness, finishing processes, and presence of additives such as
UV-absorbing substances (e.g., titan dioxide, brightening agents) (Table 24.2).
Moreover, UV protection of a garment during use depends on wash and wear,
including stretch and hydration [10]. Optimally, apparel textiles assigned for UV
protective clothing should be therefore measured and labeled in accordance with a
standard document (e.g., AATCC 183:1998; AS/NZS 4399:1996; EN 13758-
1:2002). Sun protective clothing needs to be designed with special types of complex
weaves allowing the passage of air to promote wearer comfort but to block the pas-
sage of sunlight through the textile. Fabrics may include UV absorbers of various
types to increase UV protection [74]. It will of course be essential to select sub-
stances that have a low potential for irritation and sensitization. Moreover, stringent
requirements for the design should be complied with garments assigned for sun
protective clothing (EN 13758-2:2203). A recent German study indicated that more
counseling on UV protective clothing is needed for young, male, and lower educated
24 Photoprotection by Clothing and Fabric 425
individuals [42, 75]. The textile industry should be aware of the increasing demand
for labeled sun protective clothing, in particular clothing segments such as baby
wear, children wear, and leisure and outdoor worker wear [76, 77]. Light-weighted,
breathable, natural fabrics made of cotton and linen are preferred textiles. The
textile industry may consider such fabrics for the production of labeled sun protective
clothing. Nevertheless, peoples compliance of buying and wearing sun protective
clothing may be impaired by several factors such as price, lack of knowledge, and
desire to tan [10].
References
1. Lucas RM, McMichael AJ, Armstrong BK, Smith WT (2008) Estimating the global disease
burden due to ultraviolet radiation exposure. Int J Epidemiol 37:654667
2. Tucker MA (2009) Melanoma epidemiology. Hematol Oncol Clin North Am 23:383395
3. Cho YR, Chiang MP (2010) Epidemiology, staging (new system), and prognosis of cutaneous
melanoma. Clin Plast Surg 37:4753
4. Holick MF, Jung EG (1999) Biologic effects of light 1998, proceedings of a symposium,
Basel, Switzerland, November 1-3, 1998. Kluwer Academic Publishers, Norwell
5. Soehnge H, Ouhtit A, Ananthaswamy ON (1997) Mechanisms of induction of skin cancer by
UV radiation. Front Biosci 2:D538D551
6. Setlow RB, Grist E, Thompson K, Woodhead AD (1993) Wavelengths effective in induction
of malignant melanoma. Proc Natl Acad Sci U S A 90:66666670
7. Boniol M, De Vries E, Coebergh JW, Dore JF (2005) Seasonal variation in the occurrence of
cutaneous melanoma in Europe: influence of latitude. An analysis using the EUROCARE
group of registries. Eur J Cancer 41:126132
8. Moan J, Dahlback A, Setlow AB (1999) Epidemiological support for a hypothesis for
melanoma induction indicating a role for UVA radiation. Photochem Photobiol 70:243247
9. Rigel DS, Rigel EG, Rigel AC (1999) Effects of altitude and latitude on ambient UVB radia-
tion. J Am Acad Dermatol 40:1141111416
426 T. Gambichler et al.
10. Gambichler T (2011) Ultraviolet protection of clothing. In: Functional textiles for improved
performance, protection and health. Woodhead Publishing, Philadelphia, pp 4563
11. Gies P (2007) Photoprotection by clothing. Photodermatol Photoimmunol Photomed
23:264274
12. Hoffmann K, Lappere J, Avermaete A, Altmeyer P, Gambichler T (2001) Defined UV
protection by apparel textiles. Arch Dermatol 137:10891094
13. Glanz K, Yaroch AL, Dancel M, Saraiya M, Crane LA, Buller DB, Manne S, ORiordan DL,
Heckman CJ, Hay J, Robinson JK (2008) Measures of sun exposure and sun protection
practices for behavioral and epidemiologic research. Arch Dermatol 144:217222
14. Hatch KL, Osterwalder U (2006) Garments as solar ultraviolet radiation screening materials.
Dermatol Clin 24:85100
15. Dobbinson S, Wakefield M, Hill D, Girgis A, Aitken JF, Beckmann K, Reeder AI, Herd N,
Fairthorne A, Bowles KA (2008) Prevalence and determinants of Australian adolescents and
adults weekend sun protection and sunburn, summer 2003-2004. J Am Acad Dermatol
59:602614
16. Gambichler T, Rotterdam S, Altmeyer P, Hoffmann K (2001) Protection against ultraviolet
radiation by commercial summer clothing: need for standardised testing and labelling. BMC
Dermatol 1:6
17. Aguilera J, de Glvez MV, Snchez-Roldn C, Herrera-Ceballos E (2014) New advances in
protection against solar ultraviolet radiation in textiles for summer clothing. Photochem
Photobiol 90:11991206
18. Gambichler T, Laperre J, Altmeyer P, Hoffmann K (2002) UVA and UVB transmission of
fabrics: critical wavelength based on absorbance and effective dose. Exog Dermatol
1:290295
19. Gambichler T, Al-Muhammadi R, Boms S (2009) Immunologically mediated photodermato-
ses: diagnosis and treatment. Am J Clin Dermatol 10:169180
20. OQuinn RP, Wagner RF Jr (1998) Unusual patterns of chronic photodamage through clothing.
Cutis 61:269271
21. Roelandts R (2000) The diagnosis of photosensitivity. Arch Dermatol 136:11521157
22. Barankin B, Liu K, Howard J, Guenther L (2001) Effect of a sun protection program targeting
elementary school children and their parents. J Cutan Med Surg 5:27
23. Robinson JK, Rigel DS, Amonette RA (2000) Summertime sun protection used by adults for
their children. J Am Acad Dermatol 42:746753
24. Laperre J, Gambichler T (2003) Sun protection offered by fabrics: on the relation between effec-
tive doses based on different action spectra. Photodermatol Photoimmunol Photomed 19:1116
25. Diffey BL (1998) The CIE ultraviolet action spectrum for erythema. In: Mathes R, Sliney D
(eds) Measurements of optical radiation hazards. Mrkl-Druck, Mnchen, pp 6367
26. Australian/New Zealand Standards AS/NZS (1996) Sun protective clothing evaluation and
classification. Homebush, NSW, Australia/Wellington, New Zealand, AS/NZS 4399
27. Gies HP, Roy CR, Elliott G, Zongli W (1994) Ultraviolet radiation protection factors for cloth-
ing. Health Phys 67:131139
28. Hoffmann K, Kesners P, Bader A, Avermaete A, Altmeyer P, Gambichler T (2001) Repeatability
of in vitro measurements of the ultraviolet protection factor (UPF) by spectrophotometry with
automatic sampling. Skin Res Technol 7:223226
29. Laperre J, Gambichler T, Bhringer B, Driscoll C, Varieras S, Gassan U (2001) Determination
of the ultraviolet protection factor of textile materials: measurement reliability. Photodermatol
Photoimmunol Photomed 17:223229
30. Holman CDJ, Gibson IM, Stephanson M, Armstrong BK (1983) Ultraviolet irradiation of
human body sites in relation to occupation and outdoor activity: field studies using personal
UVR dosimeters. Clin Exp Dermatol 8:269277
31. Moehrle M, Garbe C (2000) Solar UV-protective properties of textiles. Dermatology 201:82
32. Parisi AV, Kimlin MG, Mulheran L, Meldrum LR, Randall C (2000) Field-based measure-
ments of personal erythemal ultraviolet exposure through a common summer garment.
Photodermatol Photoimmunol Photomed 16:134138
24 Photoprotection by Clothing and Fabric 427
33. Ravishankar J, Diffey BL (1997) Laboratory testing of UV transmission through fabrics may
underestimate protection. Photodermatol Photoimmunol Photomed 13:202203
34. Quintern LE, Furusawa Y, Fukutsu K, Holtschmidt H (1997) Characterization and application
of UV detector spore films: the sensitivity curve of a new detector system provides good
similarity to the action spectrum for UV-induced erythema in human skin. J Photochem
Photobiol B Biol 37:158166
35. Gambichler T, Hatch KL, Avermaete A, Bader A, Herde M, Altmeyer P, Hoffmann K (2002)
Ultraviolet protection factor of fabrics: comparison of laboratory and field-based measurements.
Photodermatol Photoimmunol Photomed 18:135140
36. Gambichler T, Avermaete A, Bader A, Altmeyer P, Hoffmann K (2001) Ultraviolet protection
by summer textiles. Ultraviolet transmission measurements verified by determination of the
minimal erythema dose with solar-simulated radiation. Br J Dermatol 144:484489
37. Gambichler T, Hatch KL, Avermaete A, Altmeyer P, Hoffmann K (2002) The influence of
wetness on the ultraviolet protection factor (UPF) of textiles: in vitro and in vivo measurements.
Photodermatol Photoimmunol Photomed 18:2935
38. Gies HP, Roy CR, Holmes G (2000) Ultraviolet radiation protection by clothing: comparison
of in vivo and in vitro measurements. Radiat Prot Dosimetry 91:247250
39. Greenoak GE, Pailthorpe M (1996) Skin protection by clothing from the damaging effects of
sunlight. Australas Text 16:61
40. Lowe NJ, Bourget TD, Hughes SN, Sayre RM (1995) UV protection offered by clothing: an
in vitro and in vivo assessment of clothing fabrics. Skin Cancer 10:8996
41. Menzies SW, Lukins PB, Greenoak GE, Walker PJ, Pailthorpe M, Martin JM (1991) A compara-
tive study of fabric protection against ultraviolet-induced erythema determined by spectrophoto-
metric and human skin measurements. Photodermatol Photoimmunol Photomed 8:157163
42. Gambichler T, Dissel M, Altmeyer P, Rotterdam S (2009) Evaluation of sun awareness with an
emphasis on ultraviolet protection by clothing: a survey of adults in Western Germany. J Eur
Acad Dermatol Venereol 24:155162
43. Crews PC, Kachmann S, Beyer AG (1999) Influences on UVR transmission of undyed woven
fabrics. Text Chemist Colorist 31:1726
44. Davis S, Capjack L, Kerr N, Fedosejevs R (1997) Clothing as protection from ultraviolet radia-
tion: which fabric is most effective? Int J Dermatol 36:374379
45. Sarkar AK (2007) On the relationship between fabric processing and ultraviolet radiation
transmission. Photodermatol Photoimmunol Photomed 23:191196
46. Grifoni D, Bacci L, Zipoli G, Carreras G, Baronti S, Sabatini F (2009) Laboratory and outdoor
assessment of UV protection offered by flax and hemp fabrics dyed with natural dyes.
Photochem Photobiol 85:313320
47. Sarkar AK (2004) An evaluation of UV protection imparted by cotton fabrics dyed with natu-
ral colorants. BMC Dermatol 4:15
48. Capjack L, Kerr N, Fedosejevs R, Hatch KL, Markee NL (1994) Protection of humans from
ultraviolet radiation through the use of textiles: a review. Fam Consum Sci Res J 23:198218
49. Pailthorpe M (1994) Textile and sun protection: the current situation. Australas Text
14:5466
50. Kan CW, Au CH (2014) Effect of biopolishing and UV absorber treatment on the UV protec-
tion properties of cotton knitted fabrics. Carbohydr Polym 101:451456
51. Srinivasan M, Gatewood BM (2000) Relationship of dye characteristics to UV protection pro-
vided by cotton fabric. Text Chemist Colorist 32:3643
52. Hilfiker R, Kaufmann W, Reinert G, Schmidt E (1996) Improving sun protection factors of
fabrics by applying UV-absorbers. Text Res J 66:6170
53. Osterwalder U, Rohwer H (2002) Improving UV protection by clothing recent develop-
ments. Recent results. Cancer Res 160:6269
54. Hoffmann K, Hoffmann A, Hanke D, Bhringer B, Schindling G, Schn U (1998) Sun pro-
tected from optimally designed fabrics. Hautarzt 49:1016
55. Hoffmann K (1998) UV protective clothing in Europe: recommendation of European working
party. J Eur Acad Dermatol Venereol 11:198199
428 T. Gambichler et al.
56. Eckhardt C, Rohwer H (2000) UV protector for cotton fabrics. Text Chemist Colorist
32:2123
57. Wang RH, Xin JH, Tao XM (2005) UV-blocking property of dumbbell-shaped ZnO crystallites
on cotton fabrics. Inorg Chem 44:39263930
58. Behler KD, Stravato A, Mochalin V, Korneva G, Yushin G, Gogotsi Y (2009) Nanodiamond-
polymer composite fibers and coatings. ACS Nano 3:363369
59. Moon R, Pailthorpe M (1995) Effect of stretch and wetting on the UPF of elastane fabrics.
Australas Text 15:3942
60. Kimlin MG, Parisi AV, Meldrum LR (1999) Effect of stretch on the ultraviolet spectral trans-
mission of one type of commonly used clothing. Photodermatol Photoimmunol Photomed
15:171174
61. Sinclair SA, Diffey BL (1997) Sun protection provided by ladies stockings. Br J Dermatol
136:239241
62. Khazova M, OHagan JB, Grainger KJ (2007) Radiation and chemical degradation of UVR
protection characteristics of fabrics. Radiat Prot Dosimetry 123:369377
63. Wong WY, Lam JK, Kan CW, Postle R (2014) In vitro assessment of ultraviolet protection of
coloured cotton knitted fabrics with different structures under stretched and wet conditions.
Radiat Prot Dosimetry ncu276 164(3):325334
64. Jevtic AP (1990) The sun protective effect of clothing, including beachwear. Australas
J Dermatol 31:57
65. Kaskel P, Rohwer H, Osterwalder U, Peter RU (2001) Improving textile sun protection by
regular washing of clothing. Fortschr Med Orig 119(Suppl 2):9194
66. Standford DG, Georgouras KE, Pailthorpe MT (1995) Sun protection by a summer-weight
garment: the effect of washing and wearing. Med J Aust 162:422425
67. Wang SQ, Kopf AW, Marx J, Bogdan A, Polsky D, Bart RS (2001) Reduction of ultraviolet
transmission through cotton T-shirt fabrics with low ultraviolet protection by various laundering
methods and dyeing: clinical implications. J Am Acad Dermatol 44:767774
68. Osterwalder U, Schlenker W, Rohwer H, Martin E, Schuh S (2000) Facts and fiction on UV
protection by clothing. Radiat Prot Dosimetry 91:255260
69. Zhou Y, Crews PC (1998) Effect of OBAs and repeated launderings on UVR transmission
through fabrics. Text Chemist Colorist 30:1924
70. American Society for Testing and Materials. ASTM (2000) ASTM D 6544: standard practice
for the preparation of textile prior to UV transmittance testing. ASTM, Conshohocken
71. American Association of Textile Chemists and Colorist. AATCC (2000) AATCC TM 183-
1998 transmittance or blocking of erythemally weighted ultraviolet radiation through fabrics.
AATC Tech Manual. AATCC, Research Park
72. EN 13758-1 (2002) Fabrics solar UV protective properties Method of test for apparel fab-
rics. Brussels, European Standard, CEN
73. EN 13758 2 (2003) Fabrics solar UV protective properties classification and marking of
apparel, Brussels, European Standard, CEN
74. Gambichler T, Laperre J, Hoffmann K (2006) The European standard for sun-protective cloth-
ing: EN 13758. J Eur Acad Dermatol Venereol 20:125130
75. Eichhorn C, Seibold C, Loss J, Steinmann A, Nagel E (2008) Knowledge about UV-radiation
and sun protection: survey of adolescents and young adults in Bavaria. Hautarzt 59:821827
76. Turner D, Harrison SL (2014) Sun protection provided by regulation school uniforms in
Australian Schools: an opportunity to improve personal sun protection during childhood.
Photochem Photobiol 90(6):14391445
77. Wolska A, Owczarek G, Bartkowiak G (2010) UV protective textile clothing for workers
exposed to natural and artificial UV radiation. Conf Proc IEEE Eng Med Biol Soc
2010:62606263
Chapter 25
Photoprotection by Glass
Key Points
Glass has the ability to block all ultraviolet-B (290320 nm) and a variable
amount of ultraviolet-A (320400 nm). Factors affecting ultraviolet
radiation transmission include glass type, thickness, color, and film coating.
Glass, window films, and sunglasses play an important yet possibly under
recognized role in our effort to decrease UVR damage.
Sunglasses should meet one of the national lens safety standards, be of
adequate circumference, wrap around the eye, and be as close to the
forehead as possible.
25.1 Introduction
Glass has chemical properties similar to that of a liquid with a melting point of
around 1700 C but at room temperature behaves as a solid [4]. Glass is mainly
made of silica from sand, soda ash, and limestone which are melted together and
mixed with various other chemical to change its properties and color. The float
process is the classical method of creating smooth sheet glass. Melted glass is
poured over a bath of molten tin leaving a perfectly smooth surface as it spreads and
cools [5].
Annealed glass is the most basic type of glass produced from the float process. It is
usually the starting type of glass that can later be modified via lamination,
toughening, etc. It is often used in double glazed windows; when broken, it results
in large sharp pieces [6] (Table 25.1).
Tempered or toughened glass is created by gradual heating and sudden cooling
of the glass. It breaks into small pieces that are less likely to cause injury and is four
times stronger than annealed glass. It is commonly used in car side windows, glass
sliding doors, and shower enclosures [6].
Laminated glass is created when two laminae are fused to a middle plastic PVB
(polyvinyl butyral) layer. When this composite is broken, the pieces of glass adhere
to the plastic preventing injury and maintaining the glass integrity. This is the main
glass type used in front windshields of cars to prevent the passengers from being
ejected from the vehicle; it is also increasingly being used for the side windows to
increase passenger safety [7].
25 Photoprotection by Glass 431
Coated glass allows the glass to be modified, such as being scratch resistant,
increased reflectivity or transmissibility, and corrosion resistance. It is created by
allowing the coating vapor to bind to the surface of the glass often during the float
process.
Patterned glass is created by making a pattern on the surface of the glass. This is
usually done by passing the heated glass between rollers with an imprint on the
rollers. The glass can take on any pattern and it is often used to allow in light but to
prevent transparency [6].
Glass in general has the ability to block all UVB (290320 nm) and a variable
amount of UVA (320400 nm) depending on the type of glass [8]. The various
factors that affect UVR transmission include glass type, thickness, color, and film
coating.
In the glass film industry, UVA transmission is often measured up to 380 nm; in
general, there is a sharp increase in the UVA transmission between 380 and 400 nm.
According to a study performed using 40 different films on museum glass, the
protection ranged from 86 to 99 %, only two products actually blocked 99 % of the
UVR up to 400 nm [9].
A study by Duarte et al. measured the UVA and UVB penetration through
different types of glass of varying colors at different distances from the UVA source
[7]. They found that laminated glass blocked all the UVA regardless of distance
from the UV source. At 0 cm from the UVA source, the greatest UVA transmission
was through annealed glass (74 %), followed by tempered glass (71 %) and patterned
glass (44 %).
The color of the glass also had an effect on the transmission of UVA. At 0 cm
from the light source, the amount of transmission of UVA through green glass was
0 %, followed by yellow glass (1.3 %), wine glass (31.1 %), colorless glass (36.5 %),
and blue glass (56.8 %).
432 M. Shaban and F. Almutawa
The transmission of radiation was decreased with thicker glass but not significantly,
showing that the color of the glass was a more important variable than glass thickness.
Glass thickness of 0.2 cm allowed 75.7 % of UVA transmission, and the thickest glass
of 1 cm allowed 51.4 % of UVA transmission at 0 cm from the UV source [7].
A study by Ding et al. showed that the average driving time for Australians of 45
years of age or older in New South Wales was 84 mins/day [10]. This was similar to
the result of a US study that shows that the average time in a car was 12 h per day
in 169 individuals [11]. The clinical relevance of these findings was shown by the
observation that patients with very severe polymorphic light eruption may be
triggered by UVA doses of 5 J/cm2 which can be achieved from 30 to 60 min of UV
exposure through tempered glass [8].
In a study of UV exposure in cars, for a left-sided driver in a nonconvertible car
with the windows rolled up, the maximal exposure was on the left arm (34 % of the
ambient radiation) followed by left lateral head. With the windows rolled down, UV
exposure was 25 % of the ambient UVR, and in a convertible car, this reached 61 % of
the ambient UVR [12]. The size of the car also plays a role as a study by Kimlin et al.
found that the average daily UVA exposure was 1.3 times higher in a large family
sedan when compared to a small hatchback [13]. The annual UV exposure in people
who drive as their primary occupation has been estimated to be around 35 MED which
is approximately equivalent to a 1 week of skiing without UV protection [14].
In the USA, two retrospective studies showed that there was a slightly greater
increase in basal cell carcinomas, squamous cell carcinomas, Merkel cell carcinomas,
and melanomas on the left side corresponding to the drivers side [15, 16]. In
Australia, where the drivers seat is on the right, two studies confirmed an increase
in actinic keratosis and lentigo maligna on the right side [17, 18].
A recent meta-analysis reported higher risks for melanoma among pilots and cabin
crew [19]. Another study showed an increase in mortality related to melanoma in
pilots [20]. While these observations could be accounted for by the higher probability
of intermittent, high-intensity UV exposure among pilots and cabin crew, the contribu-
tory factor of prolonged, intense UV exposure in the cockpit needs to be considered.
It has now been made compulsory to have laminated glass in the front windshield as the
plastic layer prevents the passenger from being ejected. In contrast, safety tempered
glass is usually used in the side windows which shatter into small pieces. The factors
that affect UV penetration include glass type, color, protective films, and thickness.
25 Photoprotection by Glass 433
The use of window films was started in the 1960s and was boosted during the 1970s with
the energy crisis as a means to reduce heat loss to the external environment; films were
also found to reflect infrared radiation back into the interior space [21]. In the USA,
windshields have to maintain an American standard rating of 1(AS-1) which is the high-
est optical clarity allowing more than 70 % transmission of visible light [22]. The allow-
able side and back window tinting is highly variable according to state regulations [23].
Most films consist of several layers:
1. Protective release layer: This is polyester layer that is removed to expose the
adhesive layer.
2. Adhesive layer: This is made of a transparent high-quality adhesive which does
not distort and fixes the film to the glass.
3. A multilayered polyester film.
4. Metals, alloys, dyes, and UV inhibitors.
5. Scratch-resistant coating made from acrylic.
The metals, alloys, dyes, and filters work by either reflecting or absorbing the
UVR. The most common method used is an individual film layer of UV-blocking
material [24]. Bernstein et al. used fibroblast death from UVA exposure as an
endpoint to correlate the protection of a UV film on a tempered side vehicle glass
[25]. They found that before the film, the glass blocked 21 % of the UVR versus
99.6 % after the film application. Another study using a G50 sunlight control film
and tempered 3 mm vehicle glass showed that in the presence of the film, 100 % of
UVA transmission was blocked versus 82.4 % without the film [7].
434 M. Shaban and F. Almutawa
There are currently three major national guidelines on sunglasses: (1) the Australian/
New Zealand standards AS/NZS 1067:2003, (2) the American standard ANSI
Z80.3 updated in 2010, and (3) the European standard EN 1836:2005, which will be
replaced by the EN ISO 123121:2013 by March 2015. The new European standard
will include transmittance and refractive changes, resistance to sweat and damage,
temporal protection with highly tinted lenses, and increased coverage of the eye
[2931]. The Australian and European standards share the lens category definition
but differ on the allowed UVB transmission (Table 25.2) [32, 33]. The American
standard categorizes the lenses according to purpose, i.e., cosmetic versus
professional use (Table 25.3).
A study performed in 2003 showed that 17 % of 646 sunglasses tested under the
European standard failed to meet this standard, showing that self-regulation was
insufficient. The Australian and EU standards are now mandatory for eyeglass pro-
ducers, with the Australian requiring a third party for testing the lenses [34].
There is currently a proposal for the development of an eye-sun protection factor
(E-SPF). It integrates the UV reflectance and transmission of the lens to act as an aid
similar to skin SPF protection [26] (Table 25.4).
25 Photoprotection by Glass 435
25.10 Summary
Glass, window films, and sunglasses play an important yet possibly under recognized
role in our effort to decrease UVR damage. The most important factors in choosing
glass with the highest UVA protection would be lamination, color, and possibly thick-
ness. Sunglasses should meet one of the national lens safety standards, be of adequate
circumference, wrap around the eye, and be as close to the forehead as possible.
References
14. Sasaki H, Sakamoto Y, Schnider C, Fujita N, Hatsusaka N, Sliney DH et al (2011) UV-B expo-
sure to the eye depending on solar altitude. Eye Contact Lens 37(4):191195, PubMed
15. Butler ST, Fosko SW (2010) Increased prevalence of left-sided skin cancers. J Am Acad
Dermatol 63(6):10061010, PubMed
16. Paulson KG, Iyer JG, Nghiem P (2011) Asymmetric lateral distribution of melanoma and
Merkel cell carcinoma in the United States. J Am Acad Dermatol 65(1):3539, PubMed
Pubmed Central PMCID: 3117975
17. Foley P, Lanzer D, Marks R (1986) Are solar keratoses more common on the drivers side? Br
Med J 293(6538):18, PubMed Pubmed Central PMCID: 1340770
18. Foley PA, Marks R, Dorevitch AP (1993) Lentigo maligna is more common on the drivers
side. Arch Dermatol 129(9):12111212, PubMed
19. Sanlorenzo M, Wehner MR, Linos E, Kornak J, Kainz W, Posch C et al (2015) The risk of
melanoma in airline pilots and cabin crew: a meta-analysis. J Am Med Assoc 151(1):5158
20. Yong LC, Pinkerton LE, Yiin JH, Anderson JL, Deddens JA (2014) Mortality among a cohort
of U.S. commercial airline cockpit crew. Am J Ind Med 57(8):906914
21. European Window Film Association (2014) Window film manufacturing process. Available
from: http://www.ewfa.org/. Accessed 3 Nov 2014
22. Car windshields (2014) Car windshield markings. Available from: http://www.carwindshields.
info/windshieldmarkings. Accessed 2 Dec 2014
23. International Window Film Association (2014) State window tinting rules & laws [updated
January 2013]. Available from: http://www.iwfa.com/. Accessed 20 Oct 2014
24. Almutawa F, Vandal R, Wang SQ, Lim HW (2013) Current status of photoprotection by win-
dow glass, automobile glass, window films, and sunglasses. Photodermatol Photoimmunol
Photomed 29(2):6572, PubMed
25. Bernstein EF, Schwartz M, Viehmeyer R, Arocena MS, Sambuco CP, Ksenzenko SM (2006)
Measurement of protection afforded by ultraviolet-absorbing window film using an in vitro
model of photodamage. Lasers Surg Med 38:337342
26. Behar-Cohen F, Baillet G, de Ayguavives T, Garcia PO, Krutmann J, Pena-Garcia P et al
(2014) Ultraviolet damage to the eye revisited: eye-sun protection factor (E-SPF(R)), a new
ultraviolet protection label for eyewear. Clinical Ophthalmol 8:87104, PubMed Pubmed
Central PMCID: 3872277
27. Taylor HR, West S, Munoz B, Rosenthal FS, Bressler SB, Bressler NM (1992) The long-term
effects of visible light on the eye. Arch Ophthalmol 110(1):99104, PubMed
28. Sui GY, Liu GC, Liu GY, Gao YY, Deng Y, Wang WY et al (2013) Is sunlight exposure a risk
factor for age-related macular degeneration? A systematic review and meta-analysis. Br
J Ophthalmol 97(4):389394, PubMed
29. Australian Competition & Consumer Commission (2003) Sunglasses & fashion spectacles.
Available from: http://www.productsafety.gov.au. Accessed 3 Dec 2014
30. American National Standards Institute (2010) Nonprescription sunglass and fashion eyewear
requirements. Available from: http://www.ansi.org. Accessed 3 Dec 2014
31. Bureau Veritas Consumer Product Services (2013) Withdrawal of the EN 1836 sunglasses
standard and its replacements by two new EN ISO standards. Available from: http://www.
bureauveritas.com. Accessed 7 Dec 2014
32. Dain SJ (2003) Sunglasses and sunglass standards. Clin Exp Optom 86(2):7790, PubMed
33. Wang SQ, Balagula Y, Osterwalder U (2010) Photoprotection: a review of the current and
future technologies. Dermatol Ther 23(1):3147, PubMed
34. Dain SJ, Ngo TP, Cheng BB, Hu A, Teh AG, Tseng J et al (2010) Sunglasses, the European
directive and the European standard. Ophthalmic Physiol Opt 30(3):253256, PubMed
35. Rosenthal FS, Bakalian AE, Lou CQ, Taylor HR (1988) The effect of sunglasses on ocular
exposure to ultraviolet radiation. Am J Public Health 78(1):7274, PubMed Pubmed Central
PMCID: 1349214
36. Krutmann J, Behar-Cohen F, Baillet G, de Ayguavives T, Ortega Garcia P, Pena-Garcia P et al
(2014) Towards standardization of UV eye protection: what can be learned from
photodermatology? Photodermatol Photoimmunol Photomed 30(23):128136, PubMed
Chapter 26
Augmenting Skin Photoprotection Beyond
Sunscreens
Key Points
Sun exposure generates an abundance of reactive oxygen species (ROS)
within skin, which overwhelm skins natural defenses (leading to oxidative
stress) and which over the course of our lives exact a toll on skins health
and appearance (especially photoaging).
Recent research establishes that generation of sun-induced ROS within
skin occurs from exposure not only to the ultraviolet (UV) but also to the
visible and infrared spectral regions; these results prompt thinking of new
strategies for photoprotection that go beyond the UV attenuation capacities
of sunscreen filters.
In addition to sunscreen filters, antioxidants (AOX) and quenchers of
photoexcited states (QPES) represent promising, complimentary
intervention strategies for topical products that can suppress or scavenge
ROS and thereby optimize skins protection against the harmful effects of
sun-induced ROS formation.
Selection of AOX and QPES for use in sunscreens needs to be conducted
judiciously, since they have potential to function as pro-oxidants (i.e.,
photosensitizers) when applied to skin and exposed to the sun, which
would exacerbate the burden of excess ROS formation within skin.
Addition of appropriate AOX to sunscreens can significantly improve
protection against ROS formation within skin over a broad range of low to
high SPFs.
26.1 Introduction
Reactive oxygen species (ROS) are a class of oxygen derivatives which have the
potential to initiate radical reactions, and nowhere is this potential more evident
than in a cell, where ROS can exhibit a duality in behavior. They occur naturally to
maintain healthy cell function, most clearly represented by their integral role in
mitochondrial respiration and participation in cellular signaling, and are typically
inhibited from undergoing uncontrolled radical reactions through an endogenous
network of both enzymatic and small-molecule antioxidants [17, 18]. However,
when the AOX network fails, oxidative stress ensues from ROS, initiating chain
radical reactions on cellular targets such as polyunsaturated fatty lipids, proteins,
and both mitochondrial and nuclear DNA [1922]. Failure of the AOX network in
the skin can result from a depletion in AOX as well as an increase in the concentration
of pro-oxidant ROS that overwhelms the skins intrinsic AOX network [1, 22, 23].
The end result is the same an increase in oxidative stress in the skin through ROS-
initiated radical reactions [1, 2].
ROS include both free radical and non-radical species (Table 26.1). In general
free radicals, like hydroxyl radical (OH), superoxide anion (O2), and peroxide
radical (ROO), have an unpaired electron centered on the oxygen, in contrast to
non-radical species like hydrogen peroxide (H2O2), which have full electron shells
on all atoms. Oxygen is a particularly unique ROS because of its diradical nature
and ground-state triplet character, making it a highly reactive molecule, and of
particular importance in biological systems. Its reactivity can be more fully
understood through a diagram of the distribution of its valence electrons. Recalling
that electrons in molecules are contained in molecular orbitals (MO), where each
MO can hold two electrons with opposite spins denoted by an up or down arrow, we
can see that most molecules fill their MOs with paired spins, such that no net spin
exists, and the molecule would be considered to be in a singlet state. The exception
to this rule is molecular oxygen, which, because of Hunds rule, has two electrons
with the same spin in two different molecular orbitals of equal energy (Fig. 26.1)
yielding a net spin such that the molecule exists in a triplet state (3O2). This is
an important characteristic of oxygen because, as a triplet, it can participate in trip-
let-state reactions (Fig. 26.1), which can lead to the formation of the highly reactive
singlet oxygen (1O2) as well as the superoxide anion (O2). Both are highly destruc-
tive ROS, but 1O2 warrants a particular focus because in its short lifetime (2 s in
H2O), it has the ability to act in signal transduction as well as in more destructive
442 T. Meyer et al.
reactions with saturated bonds of cellular molecules like lipids and proteins forming
derivatives of hydroperoxides, endoperoxides, and cycloaddition products. Its
highly reactive nature can be more fully understood, again, by looking at its valence
electrons (Fig. 26.1), which show that upon energy transfer by another triplet-state
molecule (i.e., triplet sensitization), one electron flips spin and joins its pair in one
of two available MOs such that there is no net spin and the molecule is considered
a singlet state. Unlike most singlet states, 1O2 is highly unstable compared to its
3
O2 ground state and thus is highly reactive.
In order to interact with skin photochemically to induce ROS formation, solar
radiation must be absorbed by chromophores resident in skin cells and extracellular
matrix. Table 26.2 lists common chromophores in the skin and the ROS that they
have been found to sensitize. Each reaction that leads to ROS generation is unique
to the energetics and kinetics of the chromophore involved; however, in general,
understanding how these molecules can generate ROS can be gained through a
Jablonski diagram (Fig. 26.2), which shows that following absorption of a photon,
b c
* *
1O O2.
2
3O
2
Fig. 26.1 Abbreviated molecular orbitals for ground state oxygen (a), the lowest excited state of
oxygen (b), and superoxide anion (c). In its ground state, because of Hunds rule, the outermost
electrons of the molecule have parallel spins in the two p* orbitals, indicating a triplet state. When
excited, the one of these electrons flips a spin and can pair with the other electron in one p orbital,
thus forming an unstable and thus highly reactive singlet state. Superoxide anion forms (c) when
an extra electron is donated to 3O2 from another molecule
26 Augmenting Skin Photoprotection Beyond Sunscreens 443
Radical Reactions:
Electron Transfer, H+Abstraction,
Photofragmentation
S1 Intersystem
Crossing Excited State Rxn
1
M* M* O2
Internal Conversion
Fluorescence or
T1 S1 T1
hv Absorption
hv Absorption
S0
M
S0
Fig. 26.2 (a) A Jablonski diagram showing the possible energy levels and reaction pathways of a
skin chromophore M, where S0 is the ground state, S1 is the first excited singlet state, and T1 is the
first triplet state. (b) A more detailed description of the potential excited state reactions that may
occur from S1 or T1. ROS can be formed through energy transfer through the triplet manifold to
form singlet oxygen, or other radical reactions can occur with proteins, lipids, DNA, sugars. Non-
radical reactions can lead to the formation of photoproducts
a molecule (M) is excited from the ground (S0) to, most often, the excited singlet
state (S1) (the exception being molecular oxygen as discussed above). From S1 a
molecule can dissipate the excess energy via innocuous mechanisms of internal
conversion (heat) or fluorescence (light). However, as described in Fig. 26.2b, for
444 T. Meyer et al.
ROS sensitization we become concerned when two pathways are favored: (1)
intersystem crossing to the excited state triplet manifold (T1) or (2) excited state
reactions from S1 or T1. If the molecule forms a triplet, then sensitization of singlet
oxygen (1O2) can occur through the triplet manifold with ground-state molecular
oxygen 3O2. Additionally, if the energetics and kinetics are favorable, the excited
molecule (M*) in S1 or T1 may undergo both non-radical reactions (isomerization,
dimerization) and radical reactions with proteins, lipids, nucleic acids, and sugars
(R) to form a radicalized R. Multiple radical reactions are possible, including
peroxidation. Figure 26.2 is a simplified drawing to illustrate some of the potential
pathways by which a chromophore may sensitize ROS. It is important to emphasize
that any chromophore, either endogenous in the skin or exogenously applied to the
skin if it is energetically and kinetically allowed, may sensitize ROS formation.
The information in Table 26.2 also raises the general recognition that skin
contains many different types of chromophores that may serve to sensitize ROS
formation over a broad range of wavelengths, including UV (290400 nm), visible
(400770 nm), and IRA (7701400 nm) radiation. The different wavelength regions
comprise vastly different energies and have different capacities to penetrate skin.
Owing to these factors, Grether-Beck et al. emphasized that the three different
wavelength bands likely interact with different chromophores in different cellular
compartments to exert their biological effects [15]. A good example is ROS
overproduction by IRA radiation where the main chromophore has been identified
as the copper complex of intramitochondrial cytochrome-C complex IV of dermal
fibroblasts. The resulting increase in intracellular ROS correlates with the
upregulation of the matrix metalloproteinase-1 (MMP-1) enzyme, which degrades
collagen in the extracellular matrix in a process that is associated with many of the
hallmark signs of photoaging, including coarse wrinkles and skin laxity. While
the specific ROS generated by IRA radiation have not been elucidated to date as the
research is so new, this example among others listed in Table 26.2 stresses the need
for additional skin photoprotective strategies that go beyond the traditional
protection afforded by UV filters in sunscreen products.
K Endogenous Chromophores
I
Photoexcited States (including1o2)
N
QPES (suppress ROS formation)
ROS
AOX (neutralize ROS if formed)
Decreased ROS to Interact with Skin
ROS within skin. Sunscreens filter suns UV radiation at skins surface to attenuate
levels of UVR that can reach and interact with endogenous chromophores in the
underlying skin, whereas QPES and AOX each work within skin below the protec-
tive sunscreen film. QPES function upstream of ROS formation by relaxing photo-
excited states via electron transfer or energy transfer pathways before they can
sensitize the formation of ROS. Antioxidants, on the other hand, scavenge ROS
once formed before they can initiate damaging radical interactions with skin.
Sunscreens represent the first line of defense against ROS formation from UVR
when skin is exposed to the sun. While exposure to UVB and UVA both induce
ROS formation, research has firmly established that filtration of UVA rays plays a
more important role in reducing ROS formation within skin, in agreement with the
free radical effectiveness spectrum [12, 4345]. Indeed, sunscreens containing
combinations of UVB, UVA, and broadband UVA/UVB sunscreen actives can be
highly effective at reducing ROS generation within skin. For example, Flober-
Muller et al. reported a radical skin/sun protection factor (RSF) as high as 51 was
achieved for a lotion formulation containing a combination of the UVA sunscreen
diethylamino hydroxybenzoyl hexyl benzoate (5 %) with the UVB sunscreen
actives octocrylene (5 %) and octinoxate (5 %) [45]. As RSF represents the ratio of
the number of free radicals generated in unprotected skin to the number of free radi-
cals generated in protected skin, a value of 51 means that the broad-spectrum sun-
screen lotion reduced free radical generation caused by UVR exposure within skin
by about 98 %.
While their ability to protect against UVR damage associated with both acute
and chronic skin damage is undisputed, sunscreens lack ability to neutralize ROS,
and they cannot prevent ROS formation stimulated by wavelengths outside their UV
attenuation capacities (290400 nm). This latter point is important since it is now
446 T. Meyer et al.
appreciated (a) that as much as 50 % of ROS formed in human skin exposed to solar
radiation may be caused by visible radiation (400700 nm) and (b) that near-infrared
radiation (IRA, 7701400 nm) can also generate ROS in dermal fibroblasts through
mitochondrial interactions that appear to have clinical relevance for photoaging of
human skin [12, 15].
As depicted in Fig. 26.3, the protective action of sunscreens against sun-induced
ROS formation can be augmented by the inclusion of QPES or AOX along with
sunscreens. QPES is a term coined by Wondrak et al. to describe agents that can
assist excited states of skins endogenous chromophores (including singlet oxygen)
by dissipating their excess energies acquired from absorption through alternative
pathways (energy or charge-transfer reactions) that are harmless to skin [46]. QPES
neither absorb radiation directly nor become consumed during the process, so they
can continue to catalyze relaxation of skins endogenous excited states as long as
skin is exposed to the sun. In separate publications, Wondrak et al. outlined the vari-
ous mechanisms by which QPES can inactivate photoexcited states and described a
battery of test methods to identify QPES, including use of reconstructed human skin
exposed to solar-simulated UVR [46, 47]. Wondrak lists several effective QPES
agents from his and other research groups, including molecules that incorporate
secondary cyclic amines (L-proline methyl ester, ectoine, mycosporine amino acids)
or plant-derived polyphenols (genticaulein) [48]. Most recently, Jockusch et al.
identified that cyanoacrylates with fused aromatic rings effectively quenched
excited states of porphyrins to suppress formation of singlet oxygen [49]. Porphyrins
cause photosensitivity skin disorders called porphyrias, which are caused by an
abnormality in the heme metabolic pathway leading to an accumulation of porphy-
rins in the skin and other body tissues. Exposure to visible radiation (400410 nm)
triggers the disease, which manifests clinically with vesicles, bullae, and hyper- or
hypopigmentation [50].
AOX, on the other hand, help neutralize ROS once formed in skin before they
can oxidize biomolecules or influence signal transduction pathways. Classic AOX
typically function by one-electron or hydrogen atom donation to neutralize free
radicals and help terminate chain reactions. As indicated earlier, while skin has a
full complement of enzymatic (superoxide dismutase, catalase, peroxidases) and
nonenzymatic (vitamin E, ascorbic acid, glutathione) antioxidants to cope with
excess ROS formation, exposure to sun produces such an abundance of ROS that
skins own defenses become easily overwhelmed [1, 2]. Supplementation of topical
products with AOX can bolster skins natural antioxidant defenses and help prevent
UV-induced oxidative damages [18, 51, 52]. However, as will be shown below,
selection of AOX for use in sunscreens needs to be conducted judiciously. For
example, care must be taken to ensure that AOX themselves do not become strong
pro-oxidants (i.e., photosensitizers) when applied to skin and subsequently exposed
to UVR.
Thus, sunscreens with high UVA protection factors combined with QPES or
AOX represent promising complimentary intervention strategies to optimize
protection of skin against the harmful effects of sun-induced ROS formation. It
must be emphasized, however, that both QPES and AOX must be present at the right
26 Augmenting Skin Photoprotection Beyond Sunscreens 447
levels and must be within close physical proximity to their intended targets within
skin in order to perform their functions successfully. Formulations must be designed
to release AOX and QPES so they become bioavailable. Many in vivo human stud-
ies now document the ability of various combinations of AOX from topical applica-
tions to exert protective effects within epidermis and dermis from ROS induced by
UVR, visible, or IRA radiation [12, 14, 50]. Especially significant is the finding that
topical AOX can reduce expression of MMP-1 within skin. MMP-1 is the main
matrix metalloproteinase enzyme responsible for degradation of collagen from
exposure to solar radiation and is now accepted as a major biomarker of photoaging
in human skin [53].
There are many in vitro assays based on transfer of a single electron or hydrogen
atom that have been used to assess the relative performance of AOX to quench free
radicals in solution [54]. While these methods are useful to measure antioxidant
capacities in various biological matrices (plasma, saliva, food extracts) or even track
AOX integrity and stability in finished product formulations, they have limited utility
to predict AOX efficacy in skin exposed to solar radiation. These assays neither pro-
vide any indication of AOX bioavailability within different cellular compartments of
skin nor take into account possible photochemical reactions of AOX when they are
applied to skin and exposed to solar radiation. The importance of using methods that
include exposures to solar radiation to qualify AOX for use in sunscreen products is
critical, since AOX can become powerful photosensitizers when exposed to solar
radiation. Under these circumstances, AOX can significantly increase rather than
decrease ROS formation within skin, which is exactly opposite of the intended effect.
As reported below, this is especially true for some botanical AOX.
The ability of AOX to neutralize ROS within skin is typically assessed by employ-
ing either spectroscopic techniques to measure changes in ROS levels or biological
assays to track various biomarkers that result from ROS damage. A summary of the
experimental methods appears in Table 26.3. All of these methods involve exposure
to UVR and interestingly comprise a mixture of in vitro and in vivo methods plus
invasive and noninvasive in vivo techniques. While it is beyond the scope of this
chapter to review all these different techniques, in the remaining sections below, we
provide more complete descriptions of the methods we have used to screen AOX
and to confirm AOX compatibility for use in sunscreen products.
448 T. Meyer et al.
Pre-UV Post-UV
a b
c d
e f
Min Max
Fig. 26.4 Two-photon fluorescence images of skin tissue incubated with the ROS probe DHR
both before (a, c, e) and after (b, d, f) solar-simulated UV irradiation (22 mJ cm2 UVB, 660 mJ
cm2 UVA) at ca. 5 m (a, b), 20 m, (c, d) and 70 m (e, f)
250
150
100
Antioxidant
50
0
)
5% )
1%
)
%
)
)
)
t.E %)
)
t.E 0.1%
5% )
SC %)
%
%
5%
(0 .25%
)
(0 0.1
%
1
.1
5% )
.5
.1
5
0.
C )
)
BP 1.0
)
(0 .0%
0.
TH .1%
.0
.2
5
)
(0
(0
(0
T(
(
.2
%
(
it. ET
(
)
(0
(0
.1
(0
(
C
G
)
a
.2
(0
(1
.1
.1
G
m a (0
O
TH
ic
.2
t.E
T(
(0
(0
Vi
.1
AC
.2
bl
us
a
)/G
Vi
Vi
os
(0
E
Em
)/E lic
a
% erm
N
ic
ic
R
H
b
5%
E
bl
bl
)V
5% m
ic
)/T
it.
Th
m
bl
E
%
.2
)/V
5%
)/E
(0 %)/
.1
(0
)/E
(0
%
t.E
.2
1
.0
ET
.0
05
(0
%
Vi
(1
.2
(0
t.E
.5
.
(0
us
(0
E
Vi
E
E
t.
m
t.E
Vi
t.
t.
er
Vi
Vi
Vi
Th
Fig. 26.5 Examples of antioxidants (AOX) showing their ability to inhibit or enhance UVR-
induced formation of lipid hydroperoxides (LOOH) in an ex vivo study using tape strips to collect
lipids from human skin in the absence or presence of different types and levels of AOX. The results
highlight the need to qualify AOX for use in sunscreen products. NAC n-Acetyl cysteine, Emblica
Phyllanthus emblica fruit extract, Vit.E vitamin E, GT green tea extract, THC tetrahydrocurcuminoids,
ROO Rosmarinus officinalis oleoresin, Rose G Rose gallica extract, BPSC bioactive photosynthetic
complex from green tea, Thermus Thermus thermophilus ferment, ET ergothiotaine
452 T. Meyer et al.
The experimental design provides additional advantages in that (a) each subject
serves as their own control; (b) the tape strips fix the lipids, AOX, and squames to
its surface in a similar spatial arrangement as existed on the skin; and (c) the method
eliminates the need to irradiate skin of human subjects.
As reported above, during the assessment of AOX to protect lipids removed from
human skin against UVR- induced peroxidation, we observed that many botanical
AOX behaved as strong pro-oxidants to increase LOOH levels significantly above
the maximum levels formed in unprotected skin in the absence of AOX (Fig. 26.5).
This result was also confirmed for Rosa G extract using two-photon fluorescence
imaging of the nucleated epidermis, which shows that application of Rosa G extract
(1 %) increases the ROS probe fluorescence significantly compared to the control
(Fig. 26.6). Similar results using TPM were also obtained for other botanical AOX,
including chardonnay grape extract, vitamin C complex, and fennel seed extract
(Fig. 26.6).
Botanical AOX typically comprise complex mixtures of polyphenols. Many plant
polyphenols have been shown to exert strong photoprotective effects in skin, includ-
ing catechins from green tea, proanthocyanidins from grape seeds, and anthocyani-
dins from berries, among others [75, 76]. Indeed, we also observed that green tea,
tetrahydrocurcumoids, and Phyllanthus emblica fruit extract conferred significant
protection against UVR-induced lipid peroxidation (Fig. 26.5). However, plant poly-
phenols can also be potent sensitizers of ROS formation when exposed to UVR, as
recently reported for verbascoside, isoverbascoside, and tyrosol or silibinin [77, 78].
Fig. 26.6 Two-photon fluorescence images (z = 3050 mm) of skin applied with or without
botanical extracts and obtained post-solar-simulated UVB/UVA irradiation. The images show that
each AOX becomes pro-oxidative in nucleated epidermis under UV radiation. These data are
confirmed by the LOOH test for Rosa G (Fig. 26.5) and dramatically show how under UVR some
botanical AOX become pro-oxidants
26 Augmenting Skin Photoprotection Beyond Sunscreens 453
Interestingly, of the seven natural extracts tested by us in Fig. 26.5, four were found
to be strong pro-oxidants.
These data illustrate the point made in the Jablonski diagram of Fig. 26.2
depicting how a molecule including an AOX can undergo radical reactions once
it reaches its excited state following absorption of a photon and indicate that not
all AOX may be equally effective at quenching light-induced ROS. These results
also underscore the need to qualify AOX selected for use in sunscreen products
using appropriate methods to ensure that when applied to skin, they help reduce
rather than exacerbate the burden of excess ROS formation induced by sun
exposure.
Use of AOX in finished sunscreen products necessitates that AOX remain physi-
cally and chemically stable from their point of manufacture until the product has
been used up for all practical purposes by consumers. By virtue of their ability to
scavenge ROS, AOX themselves are reactive molecules. Hence, during formula-
tion development, it is important to ensure that AOX are chemically compatible
with all the ingredients comprising a formulation and that once incorporated into a
formulation, the AOX remain stable and can continue to scavenge ROS
effectively.
An easy way to monitor AOX activity in finished formulations is by using one of
the routine in vitro techniques that measures the capacity of an AOX to quench a
free radical in solution. One such test is based on use of ,-diphenyl--
picrylhydrazyl (DPPH), which is a stable organic free radical with an intense purple
coloration (max = 515 nm). When dissolved in methanol (or other appropriate
solvent) and exposed to AOX, the purple color fades as DPPH is reduced. The
extent to which the color fades can be readily measured using a spectrophotometer,
and the color change can be used to construct a scale of relative effectiveness to rank
AOX or to track AOX stability within a given formulation. Either hydrophilic or
hydrophobic AOX can be assessed using DPPH provided the AOX are soluble in the
solvent selected to conduct the assay.
An example to illustrate the usefulness of DPPH to monitor the stability of AOX
in a finished sunscreen formulation appears in Fig. 26.7. The extent to which an
aliquot of the lotion caused the purple color of DPPH in methanol to fade (referred
to as antioxidant reducing units) was measured at regular intervals using a defined
protocol after the lotion was stored either at room temperature or 50 C for 30 days.
In this case, the results show that AOX in the formulation was stable and maintained
its activity over the entire period of the stability test. Advantages of the method are
that few formulation ingredients, including sunscreens, interfere with the absor-
bance readings at 515 nm, it is simple and fast to perform, and it provides a mea-
surement on whether the AOX capacity of the formulation remains intact or has
degraded.
454 T. Meyer et al.
10
6
5 Room Temperature
50 deg.C
4
3
1
0
0 5 10 15 20 25 30 35
Time (days)
Fig. 26.7 A plot illustrating the usefulness of DPPH to monitor the stability of AOX in a finished
sunscreen product stored at room temperature or 50 C for 30 days
TPM has proven highly effective in probing how sunscreens and AOX affect
UV-induced ROS density in the epidermis and in redefining what constitutes effica-
cious photoprotection. Sunscreens at any SPF yield some protection against the
generation of UV-induced ROS in the lower stratum corneum, from their inherent
ability to absorb UV photons at the skin surface before they penetrate deeper into
the skin; however, they afford incomplete photoprotection against ROS. For exam-
ple, Fig. 26.8 shows a series of TPM images of ex vivo skin applied with a broad-
spectrum SPF 30 sunscreen that have been irradiated with UVB-UVA radiation
from a solar simulator fitted with WG 320 and UG 11 filters (4 MED, 88 mJ cm2
UVB; 2.6 kJ cm2 UVA (Solar Light Company); note that the output from this lamp
contained negligible visible light). The control image (Fig. 26.8a) of unprotected
skin shows the maximum fluorescence detected, and by comparison, we see that a
broad-spectrum SPF 30 sunscreen affords some protection against UV-induced
ROS with a 39 % decrease (fROSSPF30 = 0.61) compared to unprotected skin
(Fig. 26.8b). In terms of photoprotection against ROS, there is room for improve-
ment, which we see with the addition of two antioxidants to the SPF 30 formulation
0.5 % vitamin E and 0.1 % Phyllanthus emblica fruit extract (Fig. 26.8c). These
data show that skin applied with the SPF30 + AOX formulation yields a dramatic
decrease in fluorescence intensity of the ROS probe, corresponding to a 73 %
decrease (fROSSPF30+AOX = 0.27) in UV-induced ROS compared to unprotected skin
and a twofold increase in ROS photoprotection compared to the SPF 30-AOX for-
mulation itself .
To gain a better understanding of the effect that SPF with and without AOX has
on ROS generation in the skin, we performed TPM experiments on a series of
formulations in a common placebo base with increasing SPF and either with or
without AOX. A comparison of the sunscreen actives and AOX appears in Table 26.4.
26 Augmenting Skin Photoprotection Beyond Sunscreens 455
Min Max
Fig. 26.8 Two-photon fluorescence images of skin (z = 5 m) incubated with DHR and a base
formula (a), the based + a broad-spectrum SPF 30 sunscreen (b), or the base + sunscreen + 0.5 %
vitamin E and 0.1 % Emblica fruit extract (c). All images are post-solar-simulated UV irradiation
(88 mJ cm2, 2.6 kJ cm2 UVA). fROS of the SPF 30 only is 0.61 (39 % reduction in ROS), and
fROS of the SPF 30 + AOX is 0.27 (71 % reduction in ROS)
Table 26.4 Sunscreen actives, antioxidant combinations, and broad-spectrum indications for the
formulas created to investigate the role of AOX to attenuate UV-induced levels of ROS in
sunscreens with increasing levels of SPF
SPF SPF SPF
Placebo 4 15 50 SPF 70
Oxybenzone 3.0 5.0 6.0
Octinoxate 2.0
Homosalate 5.0 10.0 15.0
Octisalate 5.0 5.0 5.0
Octocrylene 3.0 2.0 10.0 10.0
Avobenzone 2.0 3.0 3.0
Tocopherol (vitamin E) 0.5 0.5 0.5 0.5
Diethylhexyl syringlidene malonate (DESM) 0.9 0.9 0.9 0.9
Broad spectrum (c 370 nm) ? N/A No Yes Yes Yes
Just as increasing SPF correlates with greater erythemal protection, so does increas-
ing the SPF correlate with greater UV-induced ROS protection because of the
increasing optical density provided by the UV filters at the skin surface. This effect
can be seen in Fig. 26.9 where the fraction of UV-induced ROS detected in the lower
stratum corneum decreases with increasing SPF. The data also show that the same
formulations can yield significantly improved ROS photoprotection through the
456 T. Meyer et al.
0.7
Expected if ROS protection = SPF
without AOX
0.6 with AOX
Fraction ROS Detected
0.5
0.4 *
0.3
0.2
*
*
0.1
0.0
SPF 4 SPF 15 SPF 50 SPF 70
Fig. 26.9 The fraction of ROS detected in the lower stratum corneum based upon two-photon
images and at different SPF values. The addition of AOX improves ROS protection for each SPF,
although much less than what is predicted if ROS protection was equivalent to the SPF. Each SPF
formula without AOX was compared to the same SPF formula with AOX using an unpaired t-test
(*denotes p < 0.5)
26.6 Conclusions
Exposure to solar radiation induces abundant levels of ROS within skin. Biologically
relevant levels of ROS originate not only from the UV but also from the visible and
IRA regions of the solar spectrum. ROS stimulated by the different portions of solar
spectrum likely provoke biological responses within different epidermal and dermal
compartments and are strongly associated with photoaging of skin. Sunscreens con-
taining high levels of UVA filters combined with QPES and AOX represent promis-
ing intervention strategies to optimize protection of skin against the harmful effects
26 Augmenting Skin Photoprotection Beyond Sunscreens 457
References
1. Rhie G, Shin MH, Seo JY, Choi WW, Cho KH, Kim KH, Park KC, Eun HC, CHung JH (2001)
Aging-and photoaging-dependent changes of enzymic and non-enzymic antioxidants in the
epidermis and dermis of human skin in vivo. J Invest Dermatol 117:12121217
2. Thiele JJ, Traber MG, Packer L (1998) Depletion of human stratum corneum vitamin E: an
early and sensitive in vivo marker of UV induced photo-oxidation. J Invest Dermatol
110:756791
3. Hellemans L, Corstjens H, Neven A, Declrq L, Maes D (2003) Antioxidant enzyme activity in
human stratum corneum shows seasonal variation with an age-dependent recovery. J Invest
Dermatol 120:434439
4. Xu F, Fisher GJ (2002) Ultraviolet (UV) light irradiation induced signal transduction in skin
photoaging. Arch Dermatol 138:14621470
5. Gutteridge JMC (1987) Thiobarbituric acid-reactivity following iron-dependent free-radical
damage to amino acids and carbohydrates. FEBS Lett 128:343346
6. Bickers DR, Athar M (2006) Oxidative stress in the pathogenesis of skin disease. J Invest
Dermatol 126:25652575
7. Biniek K, Levi K, Dauskardt RH (2012) Solar UV radiation reduces the barrier function of
human skin. Proc Natl Acad Sci U S A 109:16
8. Fisher JF (2002) Mechanisms of photoaging and chronological skin aging. Arch Dermatol Res
138:14621470
9. Pathak MA, Stratton K (1968) Free radicals in human skin before and after exposure to light.
Arch Biochem Biophys 123:468476
10. Jurkiewicz BA, Buettner GR (1996) EPR detection of free radicals in UV-irradiated skin:
mouse versus human. Photochem Photobiol 64:918922
11. Hanson KM, Clegg RM (2002) Observation and quantification of UV-induced ROS generation
in ex vivo human skin. Photochem Photobiol 76:5763
12. Zastrow L, Groth N, Klein F, Kockott D, Lademann J, Renneberg R, Ferrero L (2009) The
missing link-light induced (280 Nm 1600 Nm) free radical formation in human skin. Ski
Pharmacol Physiol 22:3144
13. Liebel F, Kaur S, Ruvolo E, Kollias N, Southall MD (2012) Irradiation of skin with visible
light induces reactive oxygen species and matrix-degrading enzymes. J Invest Dermatol
132:19011907
14. Grether-Beck S, Marini A, Jaenicke T, Krutmann J (2015) Effective photoprotection of human
skin against infrared a radiation by topically applied antioxidants: results from a vehicle con-
trolled, randomized study. Photochem Photobiol 91(1):248250
15. Grether-Beck S, Marini A, Jaenicke T, Krutmann J (2014) Photoprotection of human skin
beyond ultraviolet radiation. Photodermatol Photoimmunol Photomed 30:167174
16. Darvin ME, Haag S, Meinke M, Zastrow L, Sterry W, Lademann J (2001) Radical production
by infrared a irradiation in human tissue. Skin Pharmacol Physiol 23:4046
17. Droge W (2002) Free radicals in the physiological control of cell function. Physiol Rev
82:4795
18. Chen L, Hu JY, Wang SQ (2012) The role of antioxidants in photoprotection: a critical review.
J Am Acad Dermatol 67:10131024
458 T. Meyer et al.
19. Pryor WA (1986) Oxy-radicals and related species: their formation, lifetimes, and reactions.
Annu Rev Physiol 48:657667
20. Rubbo H, Radi R, Trujillo M, Telleri R, Kalyanaraman B, Barnes S, Kirk M, Freeman BA
(1994) Nitric oxide regulation of superoxide and peroxynitrite-dependent lipid peroxidation.
Formation of novel nitrogen-containing oxidized lipid derivatives. J Biol Chem
269:2606626075
21. Sanders CS, Chang H, Salzmann S, Muller SL, Ekanayake-Mudiyanselage S, Elsner P, Thiele
JJ (2002) Photoaging is associated with protein oxidation in human skin in vivo. J Invest
Dermatol 118:618625
22. Richter C, Park JW, Ames BN (1988) Normal oxidative damage to mitochondrial and nuclear
DNA is extensive. Proc Natl Acad Sci U S A 85:64656467
23. Thiele JJ, Schroeter C, Hsieh SN, Podda M, Packer L (2001) The antioxidant network of the
stratum corneum. In: Thiele J, Elsner P (eds) Current problems in dermatology: oxidants and
antioxidants in cutaneous biology. Karger, Basel, pp 2642
24. Rosenstein BS, Ducore JM, Cummings SW (1983) The mechanism of bilirubin-photosensi-
tized D N a strand breakage in human cells exposed to phototherapy light. Mutat Res
112:397406
25. Christensen T, Roll EB, Jaworska A, Kinn G (2000) Bilirubin- and light induced cell death in
a murine lymphoma cell line. J Photochem Photobiol B 58:170174
26. Wondrak GT, Jacobson EL, Jacobson MK (2002) Photosensitization of DNA damage by gly-
cated proteins. Photochem Photobiol Sci 1:355363
27. Masaki H (1999) Generation of active oxygen species from advanced glycation End-products
(AGEs) during ultraviolet light a (UVA) irradiation and a possible mechanism for cell
damaging. Biochim Biophys Acta 1428:4556
28. Wondrak GT, Roberts MJ, Jacobson MK, Jacobson EL (2002) Photosensitized growth inhibi-
tion of cultured human skin cells: mechanism and suppression of oxidative stress from solar
irradiation of glycated proteins. J Invest Dermatol 119:489498
29. Wondrak GT, Roberts MJ, Cervantes-laurean D, Jacobson MK, Jacobson EL (2003) Proteins
of the extracellular matrix are sensitizers of photo-oxidative stress in human skin cells. J Invest
Dermatol 121:578586
30. Karu TI (2008) Mitochondrial signaling in mammalian cells activated by red and near-IR
radiation. Photochem Photobiol 84:10911099
31. Simon JD (2000) Spectroscopic and dynamic studies of the epidermal chromophores trans
-urocanic acid and eumelanin. Acc Chem Res 33:307313
32. Sarna T, Sealy RC (1984) Free radicals from eumelanins: quantum yields and wavelength
dependence. Arch Biochem Biophys 232:574578
33. Cunningham ML, Johnson JS, Giovanazzi SM, Peak MJ (1985) Photosensitized production of
superoxide anion by monochromatic (290405 Nm) ultraviolet irradiation of NADH and
NADPH coenzymes. Photochem Photobiol 42:125128
34. Cunningham ML, Krinsky N, Giovanazzi SM (1985) Superoxide anion is generated from cel-
lular metabolites by solar radiation and its components. J Free Radic Biol Med 1:381385
35. Peak JG, Peak MJ, MacCoss M (1984) DNA breakage caused by 334 Nm ultraviolet light is
enhanced by naturally occurring nucleic acid components and nucleotide coenzymes.
Photochem Photobiol 39:713716
36. Menon IA, Becker MAC, Persad SD, Haberman HF (1989) Quantitation of hydrogen peroxide
formed during UV-visible irradiation of protoporphyrin, coproporphyrin and uroporphyrin.
Clin Chim Acta 186:375381
37. Moan J, Berge K (1991) The photodegradation of porphyrins in cells can be used to estimate
the lifetime of singlet oxygen. Photochem Photobiol 53:549553
38. Reddi E, Jori G, Rodgers MAJ, Spikes JD (1983) Flash photolysis studies of hemato- and
copro-porphyrins in homogeneous and microheterogeneous aqueous dispersions. Photochem
Photobiol 38:639645
39. Speck WT, Rosenkranz S, Rosenkranz HS (1976) Further observations on the photooxidation
of DNA in the presence of riboflavin. Biochim Biophys Acta 435:3944
26 Augmenting Skin Photoprotection Beyond Sunscreens 459
40. Smith GJ (1992) Singlet oxygen produced by UV irradiation of wool keratin. J Photochem
Photobiol B 2:173178
41. Craggs J, Kirk SH, Ahmad SI (1994) Synergistic action of near-UV and phenylalanine, tyro-
sine or tryptophan on the inactivation of phage T7 : role of superoxide radicals and hydrogen
peroxide. J Photochem Photobiol B 24:123128
42. Hanson KM, Simon JD (1998) Epidermal trans-UA and the UVA-induced photoaging of the
skin. Proc Natl Acad Sci U S A 95:1057610578
43. Herrling T, Jung K, Chatelain E, Langenquer M (2006) Radical skin/sun protection factor
RSF-protection against UV-induced free radicals. SOFW J 132:2429
44. Osterwalder U, Jung K, Seifert M, Herrling T (2009) Importance of UVA sun protection: a
comparative analysis of different quality control methods. SOFW J 135:213
45. Flober-Muller H, Champ S, Kandzia C, Jung K, Siefert M, Herrling T (2008) Strategy for
efficient prevention of photo-ageing. SOFW J 134:2329
46. Wondrak GT, Jacobson MK, Jacobson EL (2005) Identification of quenchers of photoexcited
states as novel agents for skin photoprotection. J Pharmacol Exp Ther 312:482491
47. Wondrak GT, Jacobson MK, Jacobson EL (2006) Endogenous UVA-photosensitizers: media-
tors of skin photodamage and novel targets for skin photoprotection. Photochem Photobiol Sci
5:215237
48. Wondrak GT (2007) Let the sun shine in: mechanisms and potential for therapeutics in photo-
damage. Curr Opin Investing Drugs 8:390400
49. Jockusch S, Bonda C, Hu S (2014) Photostabilization of endogenous porphyrins: excited state
quenching by fused ring cyanoacrylates. Photochem Photobiol Sci 13:11801184
50. Lowe NJ (2006) An overview of ultraviolet radiation, sunscreens, and photo-induced derma-
toses. In: Thiers BH, Draelos ZD (eds) Dermatologic clinics. Saunders, Philadelphia, pp 917
51. Oresajo C, Yatskayer M, Galdi A, Foltis P, Pillai S (2010) Complementary effects of antioxi-
dants and sunscreens in reducing UV-induced skin damage as demonstrated by biomarker
expression. J Cosmet Laser Ther 12:157162
52. Matsui MS, Hsia A, Miller JD, Hanneman K, Scull H, Cooper KD, Baron E (2009) Non-
sunscreen photoprotection: antioxidants add value to a sunscreen. J Invest Dermatol
14:5659
53. Brenna M, Bhatti H, Nerusu KC, Bhagavathula N, Kang S, Fisher GJ (2003) Matrix metallo-
proteinase-1 is the major collagenolytic enzyme responsible for collagen damage in
UV-irradiated human skin. Photochem Photobiol 78:4348
54. Serafini M (2008) Role of the antioxidant network in the prevention of age-related disease. In:
Miwa S, Beckman KB, Muller FL (eds) Aging medicine: oxidative stress in aging: from model
systems to human diseases. Humana Press, Totowa, pp 269289
55. Baier J, Maisch T, Maier M, Landthaler M, Baumler W (2007) Direct detection of singlet
oxygen generated by UVA irradiation in human cells and skin. J Invest Dermatol
127:14981506
56. Lauer A, Groth N, Haag SF, Darvin ME, Lademann J, Meinke MC (2013) Radical scavenging
capacity in human skin before and after vitamin C uptake: an in vivo feasibility study using
EPR spectroscopy. J Invest Dermatol 133:11021104
57. Hanson KM, Beasley DG, Meyer TA, Bardeen CJ (2001) Antioxidants in sunscreen for
improved ROS photoprotection. Cosm Toil 126:712716
58. Hanson KM, Bardeen CJ (2009) Applications of nonlinear optical microscopy for imaging
skin. Photochem Photobiol 85:3344
59. Hanson KM, Gratton E, Bardeen CJ (2006) Sunscreen enhancement of UV-induced reactive
oxygen species in the skin. Free Radic Biol Med 41:12051212
60. Mudiyanselage SE, Hamburger M, Elsner P, Thiele JJ (2003) Ultraviolet a induces generation
of squalene monohydroperoxide isomers in human sebum and skin surface lipids in vitro and
in vivo. J Invest Dermatol 120:915922
61. Jeanmaire C, Danoux L, Pauly G (2001) Glycation during human dermal intrinsic and actinic
aging: an in vivo and in vitro model study. Br J Dermatol 145:1018
460 T. Meyer et al.
62. Halliday GM (2010) Common links among pathways leading to UV-induced immunosuppres-
sion. J Invest Dermatol 130:12091212
63. Crow JP, Beckman JS, McCord JM (1995) Sensitivity of the essential zinc-thiolate moiety of
yeast alcohol dehydrogenase to hypochlorite and peroxynitrite. Biochemistry 34:35443552
64. Kooy NW, Royall JA, Ischiropoulos H, Bekman JS (1994) Peroxynitrite-mediated oxidation of
dihydrorhodamine 123. Free Radic Biol Med 16:149156
65. Royall JA, Ischiropoulos H (1993) Evaluation of 2,7-dichlorofluorescin and dihydrorhoda-
mine 123 as fluorescent probes for intracellular H2O2 in cultured endothelial cells. Arch
Biochem Biophys 302:348355
66. Henderson LM, Chappell JB (1993) Dihydrorhodamine 123: a fluorescent probe for superox-
ide generation? Eur J Biochem 217:973
67. Master BR, So PTC (1999) Multiphoton excitation microscopy and confocal imaging of
in vivo human skin: a comparison. Mircosc Microanal 5:282289
68. Ragan TR, Huang H, So PTC (2003) In vivo and ex vivo tissue applications of two-photon
microscopy. Methods Enzymol 361:481506
69. Master BR, So PTC, Gratton E (1997) Multiphoton excitation fluorescence microscopy and
spectroscopy of in vivo human skin. Biophys J 72:24052412
70. Yuen KS, Halliday GM (1997) -tocopherol, an inhibitor of epidermal lipid peroxidation,
prevents ultraviolet radiation from suppressing the skin immune system. Photochem Photobiol
65:587592
71. Tanaka N, Tajima S, Ishibashi A, Uchida K, Shigemastsu T (2001) Immunohistochemical
detection of lipid peroxidation products, protein-bound acrolein and 4-hydroxynonenal protein
adducts in actinic elastosis of photodamaged skin. Arch Dermatol Res 293:363367
72. Miyamoto S, Matinez GR, Medeiros MHG, Mascio PD (2003) Singlet molecular oxygen gen-
erated from lipid hydroperoxides by the russell mechanism: studies using 18O-labelled lin-
oleic acid hydroperoxide and monomol light emission. J Am Chem Soc 125:61726179
73. Chiba K, Kawakami K, Sone T, Onoue M (2003) characteristics of skin wrinkling and dermal
changes induced by repeated application of squalene monohydroperoxide to hairless mouse
skinitle. Skin Pharmacol Appl Skin Physiol 16:242251
74. Meyer TA, Hanson KM, Beasley DG (2008) Judicious Use of antioxidants in broad spectrum
sunscreens enhances protection beyond that afforded by sunscreens alone. J Am Acad
Dermatol Suppl 58:112
75. Vayalil P, Mittal A, Hara Y, Elmets CA, Katiyar SK (2004) Green tea polyphenols prevent
ultraviolet light-induced oxidative damage and matrix metalloproteinases expression in mouse
skin. J Invest Dermatol 122:14801487
76. Nichols JA, Katiyar SK (2010) Skin photoprotection by natural polyphenols: anti-inflamma-
tory, antioxidant and DNA repair mechanisms. Arch Dermatol Res 302:7183
77. Narayanapillai S, Agarwal C, Tilley C, Agarwal R (2012) Silibinin is a potent sensitizer of
UVA radiation-induced oxidative stress and apoptosis in human keratinocyte HaCaT cells.
Photochem Photobiol 88:11351140
78. Lecci RM, Logrieco A, Leone A (2014) Pro-oxidative action of polyphenols as action mecha-
nism for their pro-apoptotic activity. Anticancer Agents Med Chem 14:113
79. Hanson K, Behne M, Barry N, Mauro T, Gratton E, Clegg R (2002) Two-photon fluorescence
lifetime imaging of the skin stratum corneum pH gradient. Biophys J 83:16821690
80. Behne M, Meyer J, Hanson K, Barry N, Murata s, Crumrine D, Clegg R, Gratton E, Holleran
W, Elia P, Mauro T (2002) NHE1 regulates the stratum corneum permeability barrier homeo-
stasis J Biol Chem 277: 4739947406
81. Plasencia I, Norlen L, Bagatolli L (2007) Direct visulation of lipid domains in human skin
stratum corneum lipid membranes: effect of pH and temperature 93: 31423155
Part IV
Chapter 27
Education, Motivation, and Compliance
Key Points
Nearly five million people are treated annually for skin cancer in the United
States, with an estimated cost of over $8 billion.
While knowledge that photoprotection from ultraviolet radiation can
reduce the incidence of skin cancer which is high, meaningful behavioral
changes have not yet been achieved.
A multifaceted approach to improve compliance is required, combining
ongoing public education as primary prevention, stricter indoor tanning
legislation, a change in social norms regarding tanned skin, and community-
level interventions at schools and the workplace.
Increased research, surveillance, and monitoring can measure the effects of
our prevention efforts and assist in designing future efforts to motivate
behavioral change and reduce the incidence of skin cancer.
27.1 Introduction
Skin cancer is the most common form of cancer in the United States and is a major
public health concern [13]. Nearly five million people in the United States are
treated for skin cancer every year, and the incidence and associated healthcare
expenditures of skin cancer continue to rise, currently with an estimated annual cost
of over $8 billion [46]. The vast majority of skin cancers are nonmelanoma skin
cancer (NMSC), which can be treated with topical medications, radiation, or sur-
gery with good prognosis. Although the risk of metastasis is low, NMSC is locally
destructive and can impair quality of life. While cutaneous melanoma makes up
only about 5 % of the total number of skin cancers, it is more deadly [3, 5]. In the
United States in 2014, it is estimated that there would be over 76,000 new cases of
invasive melanoma and 9710 melanoma-related deaths [7].
It is well established that ultraviolet (UV) radiation plays a major role in the
development of skin cancer [1, 8, 9]. Comprehensive photoprotective strategies
have been shown to reduce the incidence of skin cancer [10]. While the general
knowledge regarding the harmful effects of UV radiation and safe sun behaviors
in Western countries is high [11, 12], this knowledge has not always translated
into meaningful change of behavior. Studies continue to show significant levels
of sun exposure in children and adults with inadequate protection from UV radia-
tion [1319]. Only 3 in 10 adults routinely practice sun protection behaviors in
the United States [20]. Surveys found the majority (83 %) of teenagers, and
nearly half (37 %) of all adults experienced at least one sunburn in the previous
year [21, 22]. Furthermore, approximately 30 million people in the United States
use tanning beds each year, including 2.3 million adolescents [23]. Among ado-
lescents aged 1319, 11 % of males and 37 % of females report ever using indoor
tanning [24]. These numbers increase to 38 % of males and over 73 % of females
by age 40 [23].
As highlighted by these statistics, there is a dramatic need for further public
education and new motivational strategies to effectively change sun-protective
behaviors. This chapter will discuss the major participants in the public health
campaign, the current disconnect between knowledge and behaviors among the
general population, and provide suggestions for improving motivation and
compliance when it comes to skin cancer prevention.
The medical community plays a key role in educating the public regarding proper
photoprotection. When it comes to skin cancer prevention and education,
dermatologists are the most experienced and have led the way. As skin specialists,
they are able to appeal to both health- and appearance-based motivations to change
behaviors. For example, for patients with a history of skin cancer, the annual skin
exam is the perfect time to reinforce the importance of proper photoprotection to
prevent further malignancies. Similarly, clinic visits for patients presenting for
cosmetic procedures can also be an ideal time to remind the patient that
photoprotection slows the signs of aging. In addition, the American Academy of
Dermatology (AAD), along with other organizations, has launched health cam-
paigns to educate the public about the need for photoprotection.
27 Education, Motivation, and Compliance 465
Aside from dermatologists, primary care physicians are in the unique position to
provide counseling to patients. In 2011, a systematic review for the US Preventive
Services Task Force (USPSTF) found that counseling in the primary care setting
can increase photoprotective behaviors and decrease intentional indoor and outdoor
tanning [25]. As a result, the USPSTF recommends physicians counsel fair-skinned
patients aged 1024 years to minimize their UV exposure and effectively reduce
their risk of skin cancer [26]. By incorporating together a total skin examination and
photoprotection counseling, primary care providers can efficiently combine both
primary and secondary prevention strategies into their visits.
Physicians from other specialties, such as pediatricians, obstetricians, and
gynecologists, can also deliver education guidelines. For pediatricians in particular,
evidence has shown that education to adolescents can be especially effective in
reducing UV exposure [26, 27]. Additionally, pediatric checkups are generally
oriented toward anticipatory guidance and serve as an ideal platform to integrate
cancer prevention education into the visit for both the child and parent [28]. As
individuals age, annual physical exams are a good time to plant the seed and keep
encouraging proper skin care with a consistent and clear message.
While nearly all physicians agree that education regarding photoprotection is
important, only a small percentage actually counsels their patients in practice. In a
review of over 18 billion total patient visits between 1989 and 2010, sunscreen was
mentioned at only 0.9 % of patient visits associated with a diagnosis of skin disease
[29]. Commonly cited reasons include not remembering and lack of knowledge.
Other major barriers include lack of time and lack of monetary incentive, as proce-
dures, diagnostics, and other interventions are favored over preventive care [30, 31].
The US government has played a role in educating the public about skin cancer
prevention. For most areas of the United States, the National Weather Service
provides UV index measurements that the EPA publishes with suggested UV
466 B.P. Hibler and S.Q. Wang
The traditional media, such as magazines, newspaper, radio, and television, have
played an important role in disseminating information related to the skin cancer
prevention message. Although members of the medical community are most trusted,
the media channels are the primary source where large segments of public learn
about UV risks and photoprotection [41]. In the past decades, the Internet and social
media have overtaken traditional media. In addition to distributing information,
social media can also play a role in influencing behavioral change by opening a
forum for interaction with peers and family who also engage in certain behaviors
[42]. As technologies advances, novel media approaches have been introduced to
reach larger and targeted demographics. A recent example is the development of a
short messaging service (SMS)-based sun safety intervention which improved skin
cancer prevention behaviors and knowledge among adolescents [43].
In summary, members of the medical community, educational organizations,
government agencies, and media and social marketing groups all play a collective
role in educating the public about photoprotection. Through their combined efforts,
they are able to reach a significant proportion of the public in a myriad of ways to
increase skin cancer prevention awareness.
risk of skin cancers is too distant [44, 45]. In view of the continual interest in
cosmetic procedures and antiaging therapies, sunscreen marketed as a cosme-
ceutical with focus on its antiaging properties may increase use and compli-
ance in the younger population and ultimately have an effect on future skin
cancer rates.
less [50]. Moreover, to achieve the desired protection, sunscreens must be applied
30 min prior to exposure and reapplied every 2 h when outdoor. Lastly, SPF is mea-
sured based on protection against erythema or burning as an indicator of photodam-
age, which is mainly produced by UVB. Thus, the SPF rating does not indicate the
level of protection from UVA, which is known to cause reactive oxygen species-
mediated damage to cells [51, 52]. As a result, individuals must be cognizant to
choose a sunscreen that also contains UVA active ingredients affording
broad-spectrum protection. In the United States and many other countries, these are
specific testing guidelines mandated by regulatory agencies for sunscreen product
to be labeled as broad-spectrum [53].
27 Education, Motivation, and Compliance 469
As stated above, despite the evidence showing most people understand the harmful
effects of UV radiation, this knowledge has not translated into meaningful behavioral
change [54]. A number of barriers to making these changes have been identified. First,
in the Western societies, tanned skin is viewed as attractive, healthy, and affluent. As
a result, many individuals, especially young women, are influenced by their peers and
trends in the media to purposefully receive high doses of UV exposure to achieve a
darker and tanned appearance [12]. Second, there are health benefits associated with
UV and sunlight exposure. UV radiation, specifically UVB, is needed to synthesize
vitamin D, which is important for bone health [55]. Sunlight regulates the circadian
rhythm and natural sunlight is effective to ameliorate seasonal affective disorder [56].
Being outdoors and living an active lifestyle is important to combat the obesity epi-
demic and other public health concerns, including diabetes and heart disease [49].
Third, changing behavior for preventive health actions is intrinsically challenging as
seen in weight loss and smoking cessation programs. It is difficult to persuade indi-
viduals to make these behavioral changes when there is a lack of immediate rewards.
Furthermore, for young individuals, the future risk of skin cancer is too distant to
motivate and compel individuals to change. Lastly, maintaining these improved health
habits requires sustained commitment. Far too often, the change on the part of indi-
viduals may be disruptive and inconvenient. For example, wearing protective clothing
and applying sunscreens may be considered as hassles in ones busy daily routine.
The need to bridge the gap between knowledge and behavior is clear. Attempts at
further increasing public awareness regarding the association between sun expo-
sure and skin cancer risk may have diminished effects on influencing behaviors,
as most people are already aware of this connection [57]. As a result, it is neces-
sary to understand the key concepts behind behavior modification in order to
develop an effective marketing campaign that targets specific demographics and
to take a collaborative approach to improve compliance among the population.
cancer. However, our endeavors must continue to build upon increasing knowledge
and move toward focusing on behavior-based interventions to encourage change.
Behavior change requires lifelong education and intervention; however, many inter-
ventions are complex, expensive, and difficult to implement. While education alone
is not an effective way to change behavior, it is the first and foremost approach to
change behaviors, especially during the early stages of behavior modification.
These stages include the pre-contemplation and contemplation stages, in which
individuals have either a favorable attitude toward sun-seeking behaviors or consid-
erable ambivalence, as is common in the case of photoprotection. These individuals
place more weight on the benefits of UV exposure, either its physiological effects
or the cosmetic appeal of having tanned skin, rather than the harmful effects of
sunbathing. For example, healthy teenagers are not concerned with the far-off
thought of skin cancer, which inhibits their progression to making sustainable
behavior change.
Education also needs to be taken into account for individuals in the other stages
of behavior modification. Those who are in the later stages of behavior modification
understand that the benefits of photoprotection outweigh the risks of unprotected
UV exposure. In those cases, education needs to be focused more on the interventional
side, with an emphasis placed on learning habits and methods to maintain healthy
lifestyle adjustments. Furthermore, photoprotection messages should offer a
comprehensive approach, including seeking shade; wearing protective clothing, hat,
and sunglasses; and applying sunscreen appropriately.
A substantial portion of US adults do not perceive cancer as preventable and are less
likely to engage in sun protection behaviors. Avoiding sunbathing and indoor
tanning is one key component of education that must be emphasized. Indoor tanners
may incorrectly believe that tanning indoors is safer and has health benefits
compared to outdoor tanning. In actuality, indoor tanning is responsible for an
estimated 450,000 NMSC and more than 10,000 melanoma cases in Europe,
Australia, and the United States each year [58]. In order to reduce the harms from
indoor tanning, it is necessary to further evaluate the attitudes and behaviors of
indoor tanners. Targeted messages should be developed that resonate with the
different groups who participate in indoor tanning and sunbathing. Additionally, the
medical professionals need to assist the Federal Communications Commission in
identifying and correcting the deceptive and misleading advertisements.
27 Education, Motivation, and Compliance 471
27.5 Conclusion
Despite ongoing public health campaigns to raise awareness of the skin cancer epi-
demic and need for improved photoprotection, there exists a disconnect between
knowledge and behavior. The development of successful interventions relies heavily
on a thorough understanding of the attitudes and beliefs that influence specific behav-
iors. The best strategy is to align efforts in a variety of settings that target different
demographics with a coordinated approach. Enhanced sun protection knowledge and
awareness is crucial for initiating behavioral change, and the public needs informa-
tion required to make informed choices. We should continue to support and reinforce
these efforts through community-level interventions, along with increased research,
surveillance, and monitoring to determine the utility of these approaches and measure
the effect of our prevention efforts. With this comprehensive approach, we can poten-
tially motivate behavioral change and reverse the trend of increasing skin cancer.
References
1. Armstrong BK, Kricker A (2001) The epidemiology of UV induced skin cancer. J Photochem
Photobiol B 63(13):818
2. Rogers HW et al (2010) Incidence estimate of nonmelanoma skin cancer in the United States,
2006. Arch Dermatol 146(3):283287
3. Wang SQ, Halpern AC (2008) Public education in photoprotection. In: Clinical guide to sun-
screens and photoprotection. New York: Informa Healthcare, pp 281291
27 Education, Motivation, and Compliance 473
4. U. S. Department of Health and Human Services (2014) Reports of the surgeon general. In:
The surgeon generals call to action to prevent skin cancer. Office of the Surgeon General (US),
Washington (DC)
5. Jemal A et al (2011) Recent trends in cutaneous melanoma incidence and death rates in the
United States, 19922006. J Am Acad Dermatol 65(5 Suppl 1):S1725.e13
6. Lazovich D, Choi K, Vogel RI (2012) Time to get serious about skin cancer prevention. Cancer
Epidemiol Biomarkers Prev 21(11):18931901
7. Siegel R et al (2014) Cancer statistics, 2014. CA Cancer J Clin 64(1):929
8. Saraiya M et al (2003) Preventing skin cancer: findings of the task force on community preven-
tive services on reducing exposure to ultraviolet light. MMWR Recomm Rep 52(Rr-15):112
9. Berwick M, Pestak C, Thomas N (2014) Solar ultraviolet exposure and mortality from skin
tumors. Adv Exp Med Biol 810:342358
10. Simard EP et al (2012) Cancers with increasing incidence trends in the United States: 1999
through 2008. CA Cancer J Clin 62(2):118128
11. MacKie RM (2004) Awareness, knowledge and attitudes to basal cell carcinoma and actinic
keratoses among the general public within Europe. J Eur Acad Dermatol Venereol
18(5):552555
12. Spradlin K et al (2010) Skin cancer: knowledge, behaviors, and attitudes of college students.
South Med J 103(10):9991003
13. Lovato CY et al (1998) Canadian national survey on sun exposure & protective behaviours:
youth at leisure. Cancer Prev Control 2(3):117122
14. Shoveller JA et al (1998) Canadian national survey on sun exposure & protective behaviours:
adults at leisure. Cancer Prev Control 2(3):111116
15. Hall HI et al (1997) Sun protection behaviors of the U.S. white population. Prev Med
26(4):401407
16. Robinson JK et al (1997) Summer sun exposure: knowledge, attitudes, and behaviors of
Midwest adolescents. Prev Med 26(3):364372
17. Jones SE et al (2012) Trends in sunscreen use among U.S. high school students: 19992009.
J Adolesc Health 50(3):304307
18. Basch CH et al (2012) Improving understanding about tanning behaviors in college students:
a pilot study. J Am Coll Health 60(3):250256
19. Ellis RM et al (2012) Sunscreen use in student athletes: a survey study. J Am Acad Dermatol
67(1):159160
20. Buller DB et al (2011) Prevalence of sunburn, sun protection, and indoor tanning behaviors
among Americans: review from national surveys and case studies of 3 states. J Am Acad
Dermatol 65(5 Suppl 1):S114S123
21. Holman DM et al (2014) The association between demographic and behavioral characteristics
and sunburn among U.S. adults National Health Interview Survey, 2010. Prev Med 63:612
22. Geller AC et al (2002) Use of sunscreen, sunburning rates, and tanning bed use among more
than 10 000 US children and adolescents. Pediatrics 109(6):10091014
23. Levine JA et al (2005) The indoor UV tanning industry: a review of skin cancer risk, health
benefit claims, and regulation. J Am Acad Dermatol 53(6):10381044
24. Demko CA et al (2003) Use of indoor tanning facilities by white adolescents in the United
States. Arch Pediatr Adolesc Med 157(9):854860
25. Lin JS, Eder M, Weinmann S (2011) Behavioral counseling to prevent skin cancer: a system-
atic review for the U.S. Preventive Services Task Force. Ann Intern Med 154(3):190201
26. Moyer VA (2012) Behavioral counseling to prevent skin cancer: U.S. Preventive Services Task
Force recommendation statement. Ann Intern Med 157(1):5965
27. Balk SJ (2011) Ultraviolet radiation: a hazard to children and adolescents. Pediatrics
127(3):e791e817
28. Crane LA et al (2006) A randomized intervention study of sun protection promotion in well-
child care. Prev Med 42(3):162170
474 B.P. Hibler and S.Q. Wang
54. Kristjansson S, Ullen H, Helgason AR (2004) The importance of assessing the readiness to
change sun-protection behaviours: a population-based study. Eur J Cancer 40(18):27732780
55. OLeary RE, Diehl J, Levins PC (2014) Update on tanning: more risks, fewer benefits. J Am
Acad Dermatol 70(3):562568
56. Wirz-Justice A et al (1996) Natural light treatment of seasonal affective disorder. J Affect
Disord 37(2-3):109120
57. Peacey V et al (2006) Ten-year changes in sun protection behaviors and beliefs of young adults
in 13 European countries. Prev Med 43(6):460465
58. Wehner MR et al (2014) International prevalence of indoor tanning: a systematic review and
meta-analysis. JAMA Dermatol 150(4):390400
59. US Food and Drug Administration (2014) FDA to require warnings on sunlamp products. FDA
News Release. 2014 May 29. Available from: http://www.fda.gov/NewsEvents/Newsroom/
PressAnnouncements/ucm399222.htm
60. Ernst A, Grimm A, Lim HW (2015) Tanning lamps: health effects and reclassification by the
Food and Drug Administration. J Am Acad Dermatol 72(1):175180
61. Chen LL, Wang SQ (2012) Post-California tanning ban: a brief update on current youth access
laws. Arch Dermatol 148(9):10711072
62. Grewal SK et al (2013) Compliance by California tanning facilities with the nations first
statewide ban on use before the age of 18 years. J Am Acad Dermatol 69(6):883889.e4
63. Guy GP Jr et al (2014) State indoor tanning laws and adolescent indoor tanning. Am J Public
Health 104(4):e69e74
64. Simmons RG, Smith K, Balough M (2014) Decrease in self-reported tanning frequency among
Utah teens following the passage of Utah senate bill 41: an analysis of the effects of youth-
access restriction laws on tanning behaviors. J Skin Cancer 2014:839601. http://www.ncbi.
nlm.nih.gov/pubmed/25215240
65. Hester EJ et al (2005) Compliance with youth access regulations for indoor UV tanning. Arch
Dermatol 141(8):959962
66. Mayer JE et al (2015) The impact of indoor tanning legislation: newspaper coverage of the
risks of indoor tanning before and after the California indoor tanning Ban for minors. J Cancer
Educ 30(1):124129. http://www.ncbi.nlm.nih.gov/pubmed/24882438
67. Geller AC et al (2002) The environmental protection Agencys national SunWise school pro-
gram: sun protection education in US schools (19992000). J Am Acad Dermatol
46(5):683689
68. Glanz K, Halpern AC, Saraiya M (2006) Behavioral and community interventions to prevent
skin cancer: what works? Arch Dermatol 142(3):356360
69. Horsham C et al (2014) Interventions to decrease skin cancer risk in outdoor workers: update
to a 2007 systematic review. BMC Res Notes 7:10
Index
A Alpha-melanocyte-stimulating hormone
AAD. See American Academy of (-MSH), 396
Dermatology (AAD) American Academy of Dermatology (AAD),
Ablative lasers, 15 115, 116, 464
Acne vulgaris treatment American Association of Textile Chemists
IR effects, 13 and Colorists, 424
visible light effects, 10 American Medical Association, 106
Actinic keratoses (AKs) prevention, 3031 American Society for Testing and Materials, 424
Actinic prurigo, 45 American standard ANSI Z80.3:2010, 434, 435
Active outdoor lifestyles, 126 Annealed glass, 430, 431
Acute cutaneous affects, 430 Antioxidants (AOX)
Aesthetic properties enzymatic and nonenzymatic, 446
in after-feel, 291 fluorescence probe, 448
area of application, 290 LOOH, 450452
combine filters, 299 oxidize biomolecules/signal transduction
combine organic filters, 299 pathways, 446
combine water-based filters, 299 polyphenols, 452
consumer personal preferences, 290 spectroscopic and biological assays, 447, 448
emulsifiers, 299300 SPF, 454456
film-formers, 300 sun-induced ROS formation, 445, 446
inorganic UV filters sunscreen formulations, 453, 454
skin feel, 297298 TPM, 448450
TiO2, 295 Apigenin, 349, 393
transparency, 295297 Apple peel ethanolic extracts (APETE), 342
ZnO, 295 Atopic eczema, 8687
insoluble particulate organic filters, 294 Australian/New Zealand standards (AS/NZS
level of activity, 290 1067:2003), 434, 435
liquid UV filters, 292293 Avobenzone (AVO), 352
oil-soluble solid UV filters, 293 BEMT, 257, 259, 261, 264
O/W emulsions, 292 benzoyl radical, 253
during rub-out, 290291 bimolecular quenching rate constant, 267
thickeners, 300 butyloctyl salicylate, 266
water-soluble solid UV filters, 294 in commercially available sunscreen
W/O emulsions, 292 products, 254
,-diphenyl--picrylhydrazyl (DPPH), 364, in concentrated solutions, 254
371, 453, 454 DEHN, 265
B
BAD. See British Association of D
Dermatologists (BAD) Daily facial moisturizer, 128
Basal cell carcinoma (BCC), 2731, 108, 109 Daily UV radiation (DUVR), 207209,
Bemotrizinol, 231, 237 216, 218
BEMT. See Bis-ethylhexyloxyphenol DEHN. See Diethylhexyl 2,6-naphthalate
methoxyphenyl triazine (BEMT) (DEHN)
Berger solar simulator, 320321 Delayed tanning (DT), 68
Bis-ethylhexyl hydroxydimethoxy malonate Dermatoheliosis. See Photoaging
(HDBM), 266 Dermatology, 336
Bis-ethylhexyloxyphenol methoxyphenyl Dermatomyositis, 52
triazine (BEMT), 257, 259, 261, 264 Dexter exchange mechanism, 264
British Association of Dermatologists (BAD), 117 Diethylamino hydroxybenzoyl hexyl benzoate
(DHHB), 259, 352
Diethylhexyl 2,6-naphthalate (DEHN), 265
C Dihydrorhodamine (DHR), 448450
CE. See Cosmetic Europe (CE) Dihydroxyacetone (DHA)
Chronic actinic dermatitis (CAD), 9, 4445, 87 chemical structure of, 405, 406
Chronic effects, 430 chemistry of, 406
Clear lipo-alcoholic spray (CAS), 240241 formulations, 408
Clothing Maillard reaction, 407
broad-spectrum protection, 418 photoprotection, 409
UPF (see Ultraviolet protection factor safety record, 408
(UPF)) Discoid lupus erythematosus (DLE), 79
Index 479
H
HDBM. See Bis-ethylhexyl hydroxydimethoxy L
malonate (HDBM) Laminated glass, 430, 431
Hunds rule, 441 Lasers
Hydroa vacciniforme, 4344 ablative, 15
Hyperbilirubinemia, 10 hair removal, 15
Hypermelanosis, 65 keloids and hypertrophic scars, 15
Hyperparathyroidism, 101 pigmented lesion removal, 14
Hyperpigmentation, 200 types, 14
uses, 14
for vascular lesions, 1314
I wavelength peaks, 14
Immediate pigment darkening (IPD), 68 Lipid hydroperoxides (LOOH), 450452
Immunologically mediated photodermatoses Liposome
(IMPs) daylong actinica, 345
actinic prurigo, 45 definition, 345
CAD, 4445 disaccharides, 346
hydroa vacciniforme, 4344 DNA repair enzymes, 345346
PMLE, 4142 elastic, 347
solar urticaria, 4243 octyl methoxycinnamate, 347
Index 481