You are on page 1of 19

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/41669565

Imaging skin: Past, present and future perspectives

Article  in  Giornale Italiano di Dermatologia e Venereologia · February 2010


Source: PubMed

CITATIONS READS

10 13,183

1 author:

Marty O. Visscher
Cincinnati Children's Hospital Medical Center
160 PUBLICATIONS   4,621 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Tyrosinase Inhibitors, Synthesis and Biological Activity View project

Ontoegny of Skin Barrier Maturation in Premature Infants; Effect of Emollient Therapy on Skin Barrier Integrity in Rural Nepal View project

All content following this page was uploaded by Marty O. Visscher on 16 March 2016.

The user has requested enhancement of the downloaded file.


IMAGING OF THE SKIN
REVIEWS
G ITAL DERMATOL VENEREOL 2010;145:11-28

CA Imaging skin: past, present and future perspectives

DI M. O. VISSCHER

E
M ®
Skin imaging modalities relevant to the range of skin conditions
encountered in clinical settings are described with respect to the
information provided, advantages and limitations, current sta-
The Skin Sciences Institute
Division of Neonatology and Pulmonary Biology
Cincinnati Children’s Hospital Medical Center

A
tus and indications for further development. The methods use the and Department of Pediatrics

V T
interaction of energy with the skin, penetrating to various depths
in the stratum corneum, epidermis, dermis and subcutaneous lay-

H
ers. They include a detection system such as the retina, film or a
digital array, and a processing system to deconstruct, analyze and
University of Cincinnati College of Medicine
Cincinnati, OH, USA

R
interpret the information. Similarly, the areas of interest, or tar-

G
gets, have common features. The skin conditions deviate from the

E I
ideal or normal state with respect to skin integrity and function.
The deviations include evidence of barrier disruption, inflam-
mation, dispigmentation, and vascular change. The user of skin
tered in the clinical setting in comparison to normal,
healthy skin and, thereby, to address readers from the
health care, clinical, research and industrial settings.

IN YR
imaging is often interested in the extent and severity of disease. Skin imaging modalities will be described with respect
Part of the task in skin imaging is to establish the criteria for the to the goals, methodologies, current status, advantages
normal condition. The review encompasses the past, present and
future of visual assessment, photographic image collection, spec-
and limitations, i.e., opportunities for further devel-
opment. Practical considerations for implementing

M P
trophotometric techniques, noninvasive histology, and three
dimensional scanning. The analytical techniques for processing skin imaging techniques in clinical and research set-
and extracting specific parameters that inform about the under- tings will be presented. A further objective is to show

O
lying biological status are presented. the commonality among imaging modalities, i.e., to
Key words: Skin diseases - Imaging, three-dimensional - Der- inform the user about the biological response to a stres-
sor under specified conditions and without artifacts

C
moscopy.
from information processing. It is, however, beyond the
scope to review the plethora of methods in detail.
Skin imaging has been comprehensively reviewed,
T his paper will review imaging of the skin. Con-
ceptually, imaging will be broadly defined as “the
creation of visual representations of objects for the
particularly with respect to the advantages it may offer
for clinical dermatology. Dr. Perednia discussed pho-
purpose of data collection or medical diagnosis using tography and image analysis, ultrasound, magnetic
any of a variety of usually computerized techniques”.1 resonance imaging (MRI) methods and three-dimen-
The goal is to present imaging in a way that is relevant sional reconstruction for non-visible light imaging,
to the range of skin “conditions” typically encoun- cataloging of patient histories, education, communi-
cation, evaluating changes over time in comparison
to standard visual methods, and diagnosis.2 He dis-
Corresponding author: M. O. Visscher, PhD, The Skin Sciences Insti-
tute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave- cussed the advantages of these quantitative objective
nue, Cincinnati, OH 45229 USA. E-mail: Marty.Visscher@cchmc.org measures in terms of increased clinician efficiency for

Vol. 145 - No. 1 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA 11


VISSCHER IMAGING SKIN

ment, determine the stage of disease progression, estab-


lish the efficacy of a treatment, select an appropriate
treatment, generate evidence for clinical practice, iden-
tify the biological changes that define the disease state,
determine the physical and chemical composition, and

A
relate physiological changes to biomarkers (or labora-
tory measures) to assess the mechanism of action. Use
of skin imaging provides the opportunity to relate clin-

IC ical judgment to patient/client response and to estab-


lish the relationship between patient expectations and
treatment efficacy as judged by clinicians.

E D Skin: energy interactions

Methods of imaging skin use energy of various

M ®
Figure 1.—The electromagnetic spectrum. wavelengths in the electromagnetic spectrum to extract
information and determine specific features. Energies
ranging from the low frequency (longer wavelengths)

A T
overcoming the limitations posed by recall and objec-
tivity. In 2004, Rallan and Harland reviewed the top-

V
ic with a provocative title “Skin imaging: is it clinically
of 7 MHz to the high frequency (shorter wavelengths)
of x-radiation (~1013 MHz) have been used to evalu-
ate and collect information from the skin (Figure 1). For

R H
useful?”.3 The detailed review of imaging methods
emphasizes surface textural assessment and subsur-

G
face techniques (e.g., dermoscopy) to characterize
example, MRI uses radio frequency energy coupled
with a magnetic field to evaluate subsurface features
of skin, such as subcutaneous fat. Ultrasound methods

E I
lesions and classify moles. In vivo microscopic meth- use the interaction of high frequency sound waves to
ods were discussed in terms of future potential as sub- characterize skin. Devices range from 7 MHz (wave-
stitutes for the invasive skin biopsy. They cautioned length ~200 micrometers) for deep tissues to 20 MHz

IN YR
about the limited predictive value and the potential frequencies (wavelength ~70 micrometers) for evalu-
for misinterpretation. The specialty of plastic surgery ating dermal thickness and to characterize the three
has relied on photography to document patient char- dimensional structure. The stratum corneum (SC) and
acteristics, for treatment planning, and to evaluate sur- upper epidermis can be examined with 50 MHz (~30

M P
gical outcomes for over fifty years.4 Galdino et al.
compared the newer high resolution cameras and found
micrometers) probes. Dual energy absorptiometry
(DEXA scan) uses low exposure to two different X-ray

O
variability in quality and contrast among specific energy levels to quantify fat and muscle beneath the
brands. To overcome this potential limitation and to skin surface. Optical coherence tomography obtains 3D
facilitate effective comparison of images taken at dif- images by using wavelength in the near infrared region

C
ferent times, the lighting, collection and image pro-
cessing procedures must be standardized.4
A search of “skin imaging” in PubMed today pro-
duces about 9800 citations, more than 900 of which
appeared in the past twelve months. The topic is pre-
and relies on optical scattering in the tissue to gener-
ate 3D images. Resolution is at the micrometer level.
In vivo confocal Raman spectroscopy is a relatively
new technique that uses inelastic light scattering and
a specific wavelength of laser light to determine the
sented across the range of medical specialties, e.g., nurs- concentration and depth of molecular species in the
ing, radiology, surgery, perinatal medicine, dermatology, upper epidermis.5 The wavelengths of the scattered
emergency medicine and biomedical engineering. The light are slightly longer than visible light. The wave-
specific methods include “traditional” imaging such as length and shape provide a molecular “fingerprint”
photography as well as “internal” methods including that can be assigned to a specific compound. Many
magnetic resonance imaging, interventional radiology skin imaging methods use the “optical” spectrum,
and in vivo confocal microscopy. Generally, the pur- defined as wavelengths from 250 nm (ultraviolet) to
poses of “imaging skin” are to: quantify clinical judg- 3 000 nm (infrared), to discern features of interest.6

12 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA February 2010


IMAGING SKIN VISSCHER

Incident energy
Reflection
Fluorescence
Stratum corneum
(10 mm)

A
Epidermis
(100 mm)

C
Dermis Scattering Absorption
(3 mm)

DI
Figure 2.—Human color perception.
Dermal capillaries
Melanocytes
Melanosomes,
Melanin transfer

E
Human color perception and visual skin imaging
Figure 3.—Energy interactions with skin.

M ®
One may assert that the most important skin imag-
ing method is human color vision. Visual methods are
color specified by dominant wavelength) of red, green,

A
and blue additive primaries (Figure 2). It is used in

T
the foundation of clinical care and the “gold standard” digital cameras, monitors, televisions, image pro-
for skin color assessment.7 Clinical judgments depend

V
cessing programs, etc.14

H
upon perception of the skin surface.8 Human beings use Perceived skin color arises when visible light inter-
skin coloration to infer physiological health status.9

R
acts with components within the skin including the
Subjects viewed high resolution images of human

G
constitutive pigments melanins (yellow to brown),
faces and were instructed to adjust the red color to

E I
oxygenated hemoglobin (red), deoxyhemoglobin (blue-
achieve a “healthy appearance”. All subjects increased purple), bilirubin (yellow) and carotene (yellow).15
the red color, regardless of the inherent pigmentation, The observed skin color arises from the interplay of

IN YR
but subjects with dark skin themselves increased the red
light with components of the stratum corneum, epi-
component of dark skin photos more than for lighter
dermis and dermis. It is determined by diffuse reflec-
skin.9 Visual responses to facial image sets, standard-
tion and scattering and absorption of light inside the
ized for shape and surface features, were recorded

M P
with eye-tracking methods. Images with more uni- skin.15, 16 Of the total incident light contacting the
form skin coloration drew more attention and were skin.6 The remainder is transmitted, absorbed or scat-
perceived to be younger than those with greater color tered by structural and chemical elements within the

O
variability.10 The distribution of skin color on the face skin corneum, epidermis, dermis and subcutaneous
has been associated with the perception of overall tissues, as shown in Figure 3. The stratum corneum

C
health.11 transmits light, the epidermis and dermis absorb light
Humans perceive color when three types of cone due to the presence of melanin and hemoglobin and
cells on the retina receive and mediate light. Long (L) scatter some light, and the subcutaneous fat scatters
cones have peak absorption at 564-580 nm (range 500- light back.17 Melanin is synthesized by the melanocytes
700), medium (M) cones at 534-545 nm (450-630), (dendritic cell) in the basal layer of the epidermis and
and short (S) cones at 420-440 nm (400-500), corre- transferred to the keratinocytes throughout the epi-
sponding roughly to the red, green and blue regions of dermis.18 Oxygenated blood in the dermal capillaries
the color spectrum (Figure 1).12, 13 A color of 600 nm and vascular plexis and deoxygenated blood (blue-
would produce a response in both the L and M cones. purple) in the dermal venules contribute to skin color.19
Processing of incoming light occurs in the optic nerves The yellow pigment carotene is in the epidermis. Biliru-
and the visual cortex of the occipital lobe. The red, bin (yellow) is in the epidermis due to result of pre-
green, blue (RGB) tristimulus color space is an addi- cipitation in phospholipid membranes and leakage as
tive model based on the chromaticities (quantity of a complex with albumin into extravascular regions.20

Vol. 145 - No. 1 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA 13


VISSCHER IMAGING SKIN

TABLE I.—Eczema Area and Severity Index (ESAI). blanchable erythema of stage I pressure ulcers is a
Body Site Region Score recognized limitation of the current staging system.29
The visually based pressure ulcer scale for healing
Head/Neck (E + I + Ex + L) × Area × 0.1 (PUSH) has been developed for the determination of
Upper limbs (E + I + Ex + L) × Area × 0.2
Trunk (E + I + Ex + L) × Area × 0.3 healing status and is the method recommended by the
(E + I + Ex + L) × Area × 0.4

A
Lower limbs National Pressure Ulcer Advisory Panel.30 However,
Total EASI Score Sum of Regions
only a 43% agreement between the PUSH results and
typical nursing assessments of healing (i.e., deterio-

IC
E = erythema, I = induration/palpulation, Ex = excoriation, L = lichenification,
are each evaluated on a severity scale of 0 – 3, where 0 = none, 1 = slight, 2 = mode-
rate, and 3 = severe.
rating, unchanged, improved) was observed, indicat-
ing that the development/implementation of an accu-
rate system is essential to improved clinical out-

E D
Visual skin imaging methods

Visual skin methods commonly use descriptors


comes.31

Atopic dermatitis and psoriasis


The need for an evaluation system that would effec-

M ®
such as slight, mild, moderate, severe to indicate tively measure response to treatment over a wide range
severity of damage, e.g., inflammation and erythema, of presentations and that would be consistent, valid,
and area (percent) of involvement relative to nearby reliable, simple to use in a clinical setting motivated the

A
areas of normal.21, 22 One advantage is that they are development of the eczema area and severity index

V HT
clinically relevant and rely on experience with devi-
ations from the “normal” skin features. Examples of
the clinical conditions and the evaluation methods
are as follows.
(EASI).32 It is based on two components of skin dis-
ease, total area of involvement and disease intensity
measured as erythema (E), induration/papulation (I),

R
excoriation (Ex), and lichenification (L) The common

G
eczema features of scaling, xerosis, oozing, crusting

E I
Inflammation and pruritus were not included in the schema because
they are not essential markers of the disease and/or
Skin inflammation is a common feature of many are subjective. Each attribute is scored from 0 to 3

IN YR
conditions, including irritant contact dermatitis, aller- (0=none, 1=slight, 2=moderate, 3=severe, with half
gic contact dermatitis, acne, psoriasis, eczema, pres- grade increments as needed). The area of involvement
sure ulcers and wounds. Minor abrasions, scratches is estimated and assigned a number from 0-6, where
and fissures are characterized by erythema (redness), 0≤10%, 2≤10-29%, 3≤30-49%, 4≤50-69%, 5≤70-89%,

M P
edema, exudate, and minor bleeding, usually sur-
rounded by areas of normal, non-damaged skin. The
extent of injury is typically determined with visual
and 6≥90-100%. The total EASI score is derived from
the severity and area values over the entire body (Table

O
I). The EASI is based on the scale for judging psoria-
scoring methods based on severity (e.g., slight, mild, sis.33
moderate, severe) and area (percent) of tissue damage. The European Task Force on Atopic Dermatitis for-

C
Erythema (abnormal redness from capillary dilation
and increased blood flow) is the visible indicator of
inflammation, infection or irritation.23 The degree of
erythema is typically assessed with somewhat sub-
jective methods.24-26 Inherent skin pigmentation (i.e.,
mulated a similar method for younger patients.34 Area
(A) is estimated using the rule of nines and severity (B)
of erythema, edema/population, oozing/crust, exco-
riation, and lichenification are each rated from 0 to 3
(0 = none, 1=slight, 2=moderate, 3=severe). Pruritus
brown, yellow, red colors due to melanin) influences and sleep loss (C) are reported by caregivers on 0-10
the interpretation of visual erythema. Changes in scales. The score (SCORAD) is calculated as follows:
blood, e.g., pooling in the tissues, affects the appear- A/5+7B/2+C. There was good agreement between
ance of skin pigmentation.27 Ben-Gashir reported that scores by expert and non-expert evaluators indicating
skin pigmentation, particularly in dark skinned applicability for use by a variety of personnel.34 The
patients, influenced the assessment of erythema in SCORAD items of intensity, area and symptoms (pru-
atopic dermatitis and cautioned that the severity could ritus, sleep) were positively correlated to disease mark-
be underestimated. 28 The evaluation of the non- ers serum eosinophil cationic protein and urinary

14 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA February 2010


IMAGING SKIN VISSCHER

CA
DI
E
M ®
VA T
R G H
E I
IN YR
M P
Figure 4.—Diaper Dermatitis Grading Scale.

O
eosinophil protein X among infants and children.35
Area of involvement has also been determined by

C
shading involved regions on body diagrams and ana-
lyzing area with software (Scion Image beta 4.02,
National Institutes of Health: www.scioncorp.com/
Infant skin
Methods have been devised for evaluating extent
and severity of irritant diaper dermatitis. To address the
skin type heterogeneity, each region (perineal, geni-
frames/fr_scion_products.htm) to create the Objec- tal, buttocks, and intertriginous) is assessed for ery-
tive Severity Assessment of Atopic Dermatitis Score thema and rash using standardized scales (7-point 0-
(OSAADS).36 It incorporates values of skin barrier 3, 0.5 grade increments) with scores based on sur-
function, i.e., transepidermal water loss and skin hydra- face area of coverage and intensity of the compro-
tion, measured as differences from normal values, by mise.38 A visually based version has been developed
the affected area at each severity level and is an effec- for use in clinical and research settings to monitor
tive scheme for assessment of adult atopic patients.37 treatment effects by noting the area of involvement (%
In the adults, OSAADS scores were generally corre- area covered) and severity of damage (faint erythema,
lated (r=0.56, P<0.05) for interleukin-16 as a marker definite redness, intense redness, rash, bleeding, etc)
of atopic disease intensity. (Figure 4).39

Vol. 145 - No. 1 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA 15


VISSCHER IMAGING SKIN

TABLE II.—Global Severity Assessment Scale for Acne. Scars


Grade Classification Attributes Deep tissue injuries such as burns, wounds and sur-
0 None No inflammatory lesions gical incisions can result in the formation of hyper-
trophic scars that are then treated to improve function
1 Only an occasional small inflammatory lesion
and aesthetic appearance of the effected area. The Van-

A
2 Mild Few scattered lesions, mild erythema on less couver Scar Scale (VSS) is used to evaluate four char-
than half of the face
acteristics of burn scars following injury or recon-

C
3 Moderate number of inflammatory lesions,
increasing erythema, over wide area of face structive surgery by assigning numerical values to the

I
four parameters of vascularity, pigmentation, height and
4 Moderate Moderate number of inflammatory lesions,
some large, increasing erythema, over wide pliability (Table IV).45 Vascularity is inferred from

D
area of face color descriptors, pigmentation from color due to
5 Papules and pustules with larger inflamed melanin, height relative to the surrounding tissue, and

E
lesions, pronounced erythema, covers much of pliability by palpation. Pliability reflects a mechanical
face tissue property. Since the assessment requires tactile
6 Severe Large papules and pustules, pronounced interaction with the skin, it does not fit the traditional

M ®
erythema, covers most of face criteria for visual assessment.

A
Acne Imaging skin: instrumental methods

V T
A visual global severity assessment scale has been
developed for acne and later refined (Table II).40 This

H
method incorporates the extent of disease by incor-
Visual skin assessment based on relevant features of
the underlying biology is a mainstay for effective clin-
ical care. However, the methods often are limited.

ER
porating the number of lesions, their size and the extent

IG
of inflammation. Representative photographs are often
used during evaluation to provide reminders of the
grades and severity. In one study, a grade change of ≥1
There are frequently no “universal standards” for nor-
mal skin integrity and color making them difficult to
implement uniformly across institutions and geogra-
phies. Drawbacks also include low reproducibility,

IN YR
was considered to be clinically meaningful.41 As with variation among skilled, experienced observers and
other visual scales, training and inter-rater calibration low reliability.24, 25 The accuracy of visual methods is
is necessary for comparison of results across multi- significantly impacted by the inherent skin pigmenta-
ple clinical sites. tion, varying from very light (Caucasian) to very dark

Wounds
M P (African). Erythema is difficult to assess in the dark-
er skin due to masking by the red, yellow and brown
pigments. This tissue damage may be underdeter-

O
Wounds are evaluated for severity, depth, healing,
etc., by a wide range of health care specialties (e.g.,

C
certified wound ostomy and continence nurses, sur-
geons, dermatologists, general physicians, and nurs-
es) because wounds transcend underlying disease
mined. Of particular importance is the determination
of skin compromise in dark skinned patients who risk
more severe injury and for whom skin pigmentation
interferes with detection of erythema.46, 47 In another
example, visual evaluation of neonatal jaundice under-
processes. The Pressure Ulcer Scale for Healing estimated serum bilirubin in 17-40% and over esti-
(PUSH) is a comprehensive visual assessment scheme mated levels in 5-36% in a sample of 517 infants.48
for pressure ulcers that includes sub-scores of wound In examining the need and utility of objective quan-
size, exdudate amount and stage of healing by tissue titative measurements of skin condition, disease process
type (epithelial, granulation, slough, necrotic) (Figure and response to treatment, consider the report of Ben-
5).42 Dermatitis secondary to radiation therapy has deck and Jacobe who provide the criteria for a good
been evaluated by several visual methods to encom- outcome measure.49 They assert that measurements
pass skin damage (erythema, erosion) and responses that need to 1) determine whether a real change has
indicate healing (desquamation, hyperpigmentation).43, occurred as a result of an intervention; 2) accurately
44 One example is shown in Table III. show that a condition is different from the normal; 3)

16 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA February 2010


IMAGING SKIN VISSCHER

CA
DI
E
M ®
VA T
R G H
E I
IN YR
M P
Figure 5.—The Pressure Ulcer Scale for Healing (PUSH).

CO
be valid as defined by correlating to the gold standard
of visual clinical assessment, standard; 4) be repro-
ducible; and 5) be feasible.49 The following skin imag-
ining methods are reviewed with these criteria in mind.
mentation, abrasion).17, 51, 52 Under standardized con-
ditions (lighting, focal length, white balance and col-
or correction), photographs serve as a permanent clin-
ical records and can be evaluated by multiple exam-
iners. Standardization of image collection (controlled
lighting, white balance, color correction) facilitates
Photography and digital imaging comparison of images taken at different times. Pre
versus post-treatment comparisons can occur in real
Recent estimates suggest that 85% of dermatolo- time. They have been used to objectively assess pho-
gists use photography at least some of the time and toaging, blanching, skin atrophy, wounds, burns,
20% take photographs as a standard procedure for all lesions, and disease (e.g., psoriasis).25, 26, 52-55
patients.50 Photographic and imaging techniques have For image capture (photography) and analysis to be
been developed to improve the objectivity and quan- relevant, practical, feasible and useful, the informa-
tify the particular skin features (e.g., erythema, dispig- tion must at some point be related back to the live clin-

Vol. 145 - No. 1 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA 17


VISSCHER IMAGING SKIN

TABLE III.—Radiation Dermatitis Severity Assessment. TABLE IV.—Vancouver Scar Scale.


Characteristic Score Characteristic Score

Induration 0 normal Vascularity 0 normal


1 slight 1 pink
2 moderate 2 red

A
3 severe 3 purple
Desquamation 0 normal Pigmentation 0 normal
1 slight 1 hypopigmentation

C
2 moderate 2 mixed pigmentation

I
3 severe 3 hyperpigmentation
Erythema 0 normal Height 0 flat
1 slight 1 less than 2 mm

D
2 moderate 2 2 to 5 mm
3 severe 3 more than 5 mm

E
Hyperpigmentation 0 normal Pliability 0 normal
1 slight 1 supple
2 moderate 2 yielding
3 severe 3 firm

M ®
4 ropes
Excoriation 0 normal 5 contracture
1 slight
2 moderate
3 severe Total Sum of individual scales

A
Maximum = 14
Total

V HT Maximum = 15

for grades of slight and moderate erythema, but not

R
ical examination. The perception of color depends for intense erythematic lesions when visual erythema

G
upon the features of the illuminating light (energy was high. They suggested that other symptoms accom-

E I
used to collect information), the traits of the observer panying the erythema (i.e. edema, infiltration, vesi-
(eye physiology, interpretation), and the interaction cles, etc) masked the actual red color52. Oduncu, et

IN YR
of light with the object.56 Given this, colors repre- al., used the hue, saturation and intensity (HIS) color
sented in the RGB model described above cannot be model measurements for analysis of chronic wounds.55
easily related to the way the human eye perceives col- The amount of slough was quantified and compared
or. To address this, the values in RGB were used to with expert visual inspection scores. The overall cor-

M P
generate the CIE 1976 L* a* b* (CIELAB) color val- relation between the methods was excellent for lower
ues. As a result of the conversion, colors are in a uni- grades but moderate overall.55
form three-dimensional space where L* is the light- We evaluated repetitive hand hygiene procedures

O
ness-darkness (L* of 100=white and L* of 0=black),
a* is red-green (positive to negative, respectively) and

C
b* is yellow-blue (positive to negative) (Figure 6).13,
57 Values of CIE L*, a*, and b* were determined from

reflectance spectroscopy measurements of adult fore-


in health care workers found the hand skin to be appre-
ciably compromised (dryness and erythema) regardless
of season.58 Erythema was particularly difficulty to
assess, partly due to the extent of epidermal barrier
compromise and the regional heterogeneity across the
arm skin over 400-700 nm and color differences were hands. We determine the effects of insults and treat-
observed across the population. As a result, the L*a*b* ments. We acquired high resolution images (RBG for-
system was proposed for the objective clinical assess- mat) under controlled lighting conditions and ana-
ment of human skin.56 lyzed them to objectively quantify skin erythema.
Setaro et al. used color markers on the skin to cor- White balance was corrected and tonal value (bright-
rect for variations in lighting and analysis of RGB ness) enhanced to improve visual image. White point
images to quantify erythema in patients (multicenter, was selected and remapped and tonal information
N=348) after esthetic treatments (e.g., chemical peel, images were compressed (4:1) in the lossless (TIFF)
laser resurfacing).52 They found a good correlation format to retain all detail of the original image. To
between digital image analysis (DIA) and visual scores amplify subtle color differences and remove the con-

18 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA February 2010


IMAGING SKIN VISSCHER

CA
DI
E
M ®
VA T
R G H
Figure 6.—RGB Color Model and CIE L*a*b* Color space.

E I
founding effects of dependent color components, the
images were transformed into luminance and chromi-
A significant correlation between color changes by
DIA and laser Doppler values suggested an association

IN YR
nance representation by conversion to L*a*b* color between erythema and blood flow.25 Nystrom et al.
and separating them into three distinct images (L, a*, used near-infrared (NIR) spectroscopy, laser Doppler
and b*).55, 60, 61 The enhanced a* component image perfusion imaging (LDPI), and digital photography
was saved as a grayscale image and used for analysis to measure erythema of breast cancer patients (N=28)

M P
of redness. The a* component was selected since it is
correlated with the visual evaluation of skin erythema.62
Means (µ) and standard deviations (σ) from the a*
receiving different doses of radiation.26 From image
analysis, the authors proposed a skin redness index
that increased with higher radiation. All methods cor-

O
channel histogram data were calculated with ImageJ
(National Institutes of Health, Washington, USA) to

C
generate µ+σ. Pixels with redness higher than µ+σ, to
188, were considered to represent excess erythema.63
This method has been used to examine treatment effects
related to varying degrees.26
Skin conditions such as wounds, burns and pressure
ulcers are very heterogeneous over the affected area
with respect to severity, depth and stages of compromise
or healing. Image analysis methods that use thresh-
for irritant hand dermatitis.64 olds, to determine percent area of involvement over
Digital image analysis (DIA) has been compared the mean or compared to a normal skin site are useful.
with other skin color methods.25, 26 Mattsson et al. Segmentation algorithms have been developed for ana-
obtained clinical photographs from subjects (N=12) lyzing complex wounds such as pressure ulcers.65 Fig-
exposed to a mild experimental thermal injury and ure 7 shows a pressure ulcer with varying stages of
used normalized RGB and for image analysis.25 Ery- injury before and after computer segmentation. The
thema by both methods was most pronounced during relevant features are extracted and classified by human
the first hour postburn then gradually decreased while experts. Image processing routines (e.g., edge detection)
skin perfusion (measured with laser Doppler meth- are applied and the resultant segmented images used to
ods) peaked at 30 minutes and continuously decreased. “train” the computer for routine analysis.65

Vol. 145 - No. 1 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA 19


VISSCHER IMAGING SKIN

CA
I
Figure 7.—Wound (pressure ulcer) with varying stages of severity befo-
re (left) and after image segmentation (right). From Kosmopoulos DI,
Tzevelekou FL. Automated pressure ulcer lesion diagnosis for telemedi-

D
cine systems.65

E
Visual perception system method
Skin features can be evaluated from pairs of high res-

M ®
olution images, provided they were taken under con-
trolled lighting conditions, at a fixed distance from
the subject, with standardization for subject position-
Figure 8.—Skin absorption spectra.

A T
ing and color corrected as describe above. The method
depends upon accurate repositioning so the judges

V
cannot use subtle differences to infer time or treat-
method, “judges” first view single images to obtain a
frame of reference for the range of severity of a given

H
ment effects. Image pairs are presented at random

R
using an algorithm and viewed on a high resolution

G
monitor (e.g., flat screen, 12×16 inches). Judges are
attribute, e.g., pigmentation, erythema, scaling. Pairs
of images are then presented to allow observers to
detect small differences in images.68 In the two-alter-

E I
blinded with respect to both order and pair. Images native forced-choice (TAFC) method, judges score
can be compared to specific attributes (e.g., erythe- each image within a pair. The method assumes that
the image with the higher score has the more severe

IN YR
ma, lightness, edema, dryness). The high resolution
allows “magnification” photograph after resolution to condition.68 Randomized pairs of varying differences
examine specific regions and/or detail not easily in severity are evaluated to determine the threshold of
observed with the naked eye. Judges use a fixed line on incremental discrimination (i.e. grade 1 vs. 7, 6-incre-
ment difference). Pairs of identical images serve as

M P
the screen to indicate the magnitude of difference (e.g.,
0=no difference, 100=largest possible difference, controls.
etc.).63 Alternatively, they can select descriptors (e.g.,

O
a) I think there is a difference; b) I know there is a dif-
ference; c) there is a moderate difference; d) there is a Effect of skin color and pigmentation

C
very large difference).66 The VPS method was used
to compare images of facial skin with hyperpigment- Photographic methods typically use the entire range
ed spots before and after 8 weeks of treatment.67 This of wavelengths from white light (e.g., flash) for inter-
allows comparison of temporal effects and does not rely actions with the skin. Melanin, water, hemoglobin and
on written documentation or the clinician’s memory to other chromophores absorb the incident light to vary-
evaluate ontogeny and treatment progression. ing extents, depending upon the wavelength (Figure 8).
The inherent skin coloration and the response to ultra-
violet radiation are used to assign a skin type from I-
Naïve judge/patient assessment
VI, known as the Fitzpatrick Skin Type system (Table
In a similar fashion, images that represent a range of V).69 The anthropologist Fredrick Von Luschan devel-
severity for the condition of interest can be used to oped a color chart to classify and describe the col-
determine the magnitude of differences that naïve oration in the late 1900s (Figure 9).70 The influence of
judges, patients or clients can detect and/or to relate chromophores on incident light varies considerably
their perception to expert clinical opinion. In this across clinical populations. Skin coloration for a giv-

20 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA February 2010


IMAGING SKIN VISSCHER

TABLE V.—Skin Classification by Pigmentation (see Figure 9 for Luschan’s color).


Von Luschan’s Von Luschan’s
Type Effect of Light Exposure Description Description Color

I Always burns, never tans Pale, fair, freckles Very light 1-5
II Usually burns, sometimes tans Fair Light 6-10

A
III May burn, usually tans Light brown Intermediate 11-15
IV Rarely burns, always tans Olive brown Mediterranean 16-21
V Moderate constitutional pigmentation Brown Dark or brown 22-28

C
VI Marked constitutional pigmentation Black Very dark or black 29-36

DI
en individual varies with season due to differences in
the amount of light exposure.71
The variation in light absorption needs to be taken

E
into account with either image processing and/or wave-
length of image collection. One approach has been to

M ®
use reflectance spectroscopy. Reflectance spectrome-
ters use diodes to supply light of specific wavelengths,
i.e., 568 nm (green), 660 nm (red) and 880 nm

A
(infrared), to measure 1) erythema as hemoglobin from

T
absorbed and reflected light (red, green); and 2)

V
melanin from absorbed and reflected red or combina-

H
tions of red and infrared.72 Melanin (M) content is
approximated from red reflectance using this equa-

ER
tion: M=log10 (1/% red reflectance) and erythema (E)

IG
from: E=log10 (1/% green reflectance) - log10 (1/% red
reflectance).73 Shriver et al. used narrow band
reflectance spectroscopy and CIE L*a*b* tristimulus
Figure 9.—Von Luschan skin color chart.

IN YR
color (three specific wavelengths) to measure erythe- methods for separating erythema and pigmentation
ma, melanin, L* and a* in people of varying ethnici- have been a “necessary outgrowth” of research on
ty.73 The results (Table VI) show differences in skin understanding the effects of environmental exposure
lightness and pigmentation across racial groups for to ultraviolet radiation (UVB) on human skin and the

M P
healthy, nondamaged skin E and M are relatively inde-
pendent for low pigmentation and can be used to esti-
mate erythema and melanin content. However, for
influence of inherent skin pigmentation, dose, previ-
ous exposure, etc. In the L*a*b* color system, the

O
Individual Typology Angle (ITA) has been used to
dark skinned subjects, they are correlated and cannot determine pigmentation as follows:15
be infer redness or melanin content. This finding

C
demonstrates the difficulty in the evaluation of ery-
thema in dark-skinned patients and indicates that oth-
er methods must be used.
For skin conditions, e.g., irritant dermatitis, acne,
eczema, the extent of inflammation is of interest and
ITA=[arctan((L* - 50)/b*)] × 180/π

However, changes in color cause by changes in blood


flow can influence this attribute.16 Wagner et al., made
refinements to the measurement of erythema and col-
apparent color is influenced by post inflammatory or by adjusting the melanin (M) and erythema (E)
responses (hypo and hyperpigmentation). Successful indices by the slope of the absorbance from 650 to 700
evaluation of treatment efficacy depends upon ade- nm, to generate AM and EM, respectively.21 They con-
quately separation of the responses due to increased cluded that L*, M and AM were valid methods for
blood flow (e.g., irritant phase of irritant dermatitis) determining inherent color, but only the change in AM
from changes in barrier properties (i.e., skin dryness could be used to assess skin responses (in this case to
due to hyperproliferation and altered desquamation) or UV treatment). The erythema response could be deter-
changes in pigmentation. The development of new mined from changes in both a* and AE.

Vol. 145 - No. 1 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA 21


VISSCHER IMAGING SKIN

Quantitation of specific areas of increased or Contrast agents


decreased skin pigmentation over a large area of is of
The extent of epidermal tissue injuries such as abra-
interest for solar lentigines, acne, etc. Miyamoto et sions, fissures, and cracks can be difficult to assess
al. developed a controlled digital image capture and accurately, particularly in darker skinned patients.
analysis system to quantify the total area constituted Contrast agents, e.g., toluidine blue, fluorescein, cou-

A
by multiple hyperpigmented spots on the face (cheek pled with digital fluorescence image capture were
and periorbital regions).74 Following extraction of the used to quantify the extent of epidermal and oral

C
blue channel of the RGB image, the spots were select- mucosal injury as a function of skin pigmentation.78
ed, extracted and compared to the surrounding skin

I
Images were taken with blue light where fluorescein
using CIE L*a*b* values. The method is of sufficient emits an intense green light and analyzed for area and
sensitivity to determine the effectiveness of treatment intensity in groups with dark (inherent pigmentation,

E D
modalities relative to a vehicle control.75

Diffuse reflectance spectroscopy: recent developments


More recently, diffuse reflectance spectrometers
Type V & VI, with L from 35-48) and light (type I &
II, L from 63-72) skin. The extent of injury could be
quantified without interference from inherent pig-
mentation and may be applicable to other situations of

M ®
epidermal compromise.
(DRS) have added components (e.g., gratings) to
increase the number of discrete wavelengths and obtain
information at increments of 5-20 nm within the region

A
Imaging skin lesions

T
of 400-700 nm.27 In this mode, DRS can be used to sep-
arate the pigmentation (melanin), oxyhemoglobin, and Dermoscopy

V
deoxyhemoglobin. Reference spectra (white, dark)

H
are collected, the region of interest is measured, the

R
contribution due to surface reflection (specular
Dermoscopy is a simple imaging technique that
uses low magnification (10×), cross-polarization to

G
reflectance) is subtracted, and the apparent absorp- remove surface reflection and application of a liq-

E I
tion (A) is computed for each wavelength.76 The uid to increase light transmission into the epidermis
absorbance from melanin (Am) is derived from the and upper dermis.79 A meta-analysis of published
studies indicated that the use of a dermatoscope

IN YR
individual values in the region from 630-700 nm. After
increases the sensitivity for detection of melanoma
Am is subtracted from A, the absorbances for oxy and
relative to the naked eye alone.80 The capabilities of
deoxyhemoglobin are calculated from absorbtion val-
these hand held devices have been extended by the
ues at 580 nm (oxy-HB maximum) and 560 (deoxy-Hb
development and implementation of spectroscopic

M P
maximum). Values of the three chromophores are inde- methods. The goal is to obtain more detailed infor-
pendent when this method is used.76 mation, particularly about melanoma where diag-
The diffuse reflectance spectrometers described thus

O
nostic accuracy is difficult. Dwyer et al used DRS
far are designed to measure local areas of ~10-50 mm to collect spectral images from 400-700 nm at 20 nm
in diameter with a probe in contact with the skin sur- intervals and compared them with image analysis of

C
face. Additional information about the skin status with
respect to water, e.g., edema, can be obtained by adding
the near infrared region and collecting multiple absorp-
tion spectra from 400 to 1 000 nm, referred to as
“hyperspectral” imaging.77 As described for DRS to
3 mm excised tissue biopsies stained for melanin
(Masson Fontana). They determined melanin from
the difference in L* for 400 nm versus 420 nm images
and found an excellent correlation with convention-
al histology (r=0.68).81
separate the components of melanin and hemoglobin, The method of spectrophotometric intracutaneous
images are collected at 10-20 nm intervals. When large analysis (SIA) has moved from the research to the
areas are sampled with hyperspectral “cameras”, the clinical setting.82 SIA employs diffuse reflectance
output data is a stack of images at each wavelength. spectroscopy over the range of 400-1 000 nm of ener-
Image analysis methods extract the specific informa- gy and extracts features that can be related to the mor-
tion, e.g., color, hemoglobin, water. Erythema data phology of the lesion for a sample area of 12-24 mm
collected this way was highly correlated to the expert in diameter. The output was validated with conven-
clinical assessment (grades 0-4).77 tional histology of tissue biopsies obtained after image

22 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA February 2010


IMAGING SKIN VISSCHER

Poor Moderate Excellent Almost perfect

Dermal melanin
Blood
displacement
Dermal melanin
globules

CA Blood
displacement
with blush

I
Asymmetry

Biaxial symmetry

D
Collagen holes
Erythematous

E
blush

Melanin globules

Blood globules

M ®
Figure 10.—Representative images from spectrophotometric intercuta-
neous analysis (SIA).
0 0.2 0.4 0.6
Bars indicate 95% confidence levels
0.8 0

A
Adapted from Moncrieff, British Journal of Dermatology, 2002, 246, 448-457

V T
collection. The results showed good specificity (80.1%)
and sensitivity (82.7%).82 The image processing pro-

H
cedures use the near infrared scans to map the quanti-
Figure 11.—Agreement between skin features from the SIA method and
conventional histology.

R
ty of collagen in the papillary dermis. Next, the total Other modalities: laser Doppler perfusion,

G
melanin, oxy-Hb and deoxy-Hb are extracted, sepa- ultrasound, and thermal imaging

E I
rated and quantified. Once they have been removed, the
amount of melanin that is physically located below While instruments like the Vancouver General Hos-
the dermal-epidermal junction can be isolated and

IN YR
pital Scar Scale and the Lund and Browder chart esti-
analyzed. Representative images are shown in Figure mation are used for burn area and severity, they are
10.82 Features typically identified from standard his- not effective for determining the extent of healing or
tology, including as dermal melanin, blood displace- treatment effectiveness.85 Scanning laser Doppler per-
ment with blush, collagen holes, melanin globules,

M P
fusion imaging was effective for determining the rate
were compared for the SIA images and biopsies. Fig-
of re-epithelialization, need for skin grafting and treat-
ure 11 indicates the reliability and supports the use of
ment planning for scars.85 Scanning laser Doppler per-

O
the SIA method for most of the features. SIA has also
been applied to the quantitation of the effects of long fusion imaging was used to evaluate blood flow among
term ultraviolet exposure on facial skin, e.g., hyper- subjects wearing transparent burn masks. The results

C
pigmented spots and to evaluate the perception of age
and health status.83 The applicability of SIA for the
evaluation of common skin conditions has been demon-
strated by Kollias et al.84 Changes in erythema could
be used to quantify the irritant response to sodium lau-
showed significant differences in perfusion with and
without pressure and as a function to specific mask
treatment.86 The healing status of tissue injuries such
as wounds and burns is difficult to assess by live visu-
al and optically based imaging systems (e.g., photog-
ryl sulfate, inflammation due to histamine release, raphy) perfusion cannot be easily determined in the
changes in blood flow (by application of pressure), presence of other chromophores. Knowledge of per-
inflammation induced by ultraviolet exposure and for fusion status is essential for treatment planning and
conditions of reactive hyperemia. Their findings sug- determination of progress. Laser Doppler imaging was
gest applicability of this technique for the evaluation used to evaluate the status of burns among 48 pedi-
of common skin conditions including irritant contact atric patients with 85 burns over a period of 186 hours.
dermatitis, allergic dermatitis, and pressure induced Perfusion data was related to re-epithelialisation and
injury. predictive of grafting and scar management outcomes87.

Vol. 145 - No. 1 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA 23


VISSCHER IMAGING SKIN

Three instruments, the TiVi imaging system (Wheels- focal microscopy (RCM) and fluorescence excitation
Bridge AB), the laser Doppler scanner (Moor Instru- spectroscopy, to determine the effect of skin pig-
ments) and the laser speckle perfusion imager, were mentation (African American versus Caucasian) on the
compared in detail to assess their relative clinical util- irritant response to surfactant.92 Both groups showed
ity.88 They were evaluated in conditions of known SC disruption and epidermal spongiosis but damage

A
changes in blood flow, i.e., a topical analgesic (men- was more severe in lighter skin. Although a greater per-
thol), vasodilatation (methyl nicotinate), blanching centage of the fluorescence was absorbed in dark skin
(steroid), and post occlusive reactive hyperemia. The (due to increased melanin concentrations), the relative

IC
laser speckle and TiVi systems were able to detect
changes in the superficial papillary plexus that could
not be detected by the laser Doppler instrument. The
changes in epidermal proliferation versus baseline
were the not different. RCM holds promise as a bet-
ter and more accurate method for diagnosing malig-

D
laser Doppler was effective at measuring changes in the
dermis. The effects of occlusion could be measured

E
with both laser instruments but not readily with the
TiVi. O’Doherty urged consideration of the features of
each prior to use for a specific clinical condition and
nant melanoma and may allow more rapid interven-
tion.93 Multiphoton microscopy (MM) uses lasers
with pulses of femtoseconds and simultaneous absorp-
tion of two photons in the near infrared region to
achieve resolution at the sub-cellar level.94 Although

M ®
noted the need for further investigation.88
When combined information from laser Doppler per-
fusion imaging, thermal imaging, and nailfold capil-
work is in relatively early stages, investigators antic-
ipate its use for evaluating processes in transdermal
drug delivery and modifications at the level of the

A T
laroscopy, ultrasound imaging could differentiate patients
with systemic sclerosis from those with Raynaud’s dis-

V
ease and from healthy controls.89 Low echo (10 MHz)
stratum corneum.

H
ultrasound and thermal imaging detected inflammation

R
on the feet of diabetic patients that could not be found

G
with visual inspection, most likely due to interference
3D skin surface imaging

Many of the skin imaging methods are inherently

E I
from the thickened stratum corneum of the callus.90 two-dimensional in nature or rely on sequential 2D
scans to create a three dimensional image. Natural
curvatures of any body site can constrain the area of

IN YR
Confocal microscopy interest to relatively “flat” regions. The skin is high-
ly textured, as easily seen by inspection. Evaluation
In an effort to obtain highly detailed “histological” of textural features such as wrinkles and fine lines is
information within the stratum corneum, epidermis frequently accomplished by inspection of high qual-

M P
and dermis, noninvasive in vivo microscopic tech-
niques have been developed and evaluated in research
ity 2D images taken with careful repositioning. Skin
surface texture can be measured from surface replicas

O
and clinical settings. The goal is to replace the invasive made with a silicone polymer. Stereomicroscopy, pro-
tissue biopsy with a technique that is not limited by the filometry and 3D camera systems with fringe projec-
artifacts of tissue preparation and sectioning prior to tion optics (e.g., PRIMOS) have been used to quantify

C
microscopy. The techniques for imaging skin include
optical coherence tomography (OCT), reflectance con-
focal microscopy, multiphoton microspectroscopy,
and confocal Raman spectroscopy. In OCT, light from
two paths achieves coherence and can penetrate up to
texture (roughness) from replicas or skin in vivo (up
to ~ 25 cm2).95 The skin surface features of larger
areas and/or for irregularities in actual clinical situa-
tions, e.g., wounds, surgical scars, malformations,
burns, radiation dermatitis (breast tissue) or trauma can
2 mm at with high resolution (10 µm).91 It has been be difficult to evaluate with these more localized meth-
used to quantitatively characterize nonmelanoma skin ods. 3D skin surface data can be obtained from scan-
cancer because it can distinguish tumor borders from ning systems (e.g., mounted on a platform and con-
normal skin. Epidermal thickening and edema have trolled by data acquisition software.96 Laser-based
been demonstrated for irritant dermatitis and psoriasis scanners (e.g., Cyberware Rapid 3D Digitizer, Cyber-
and shows promise for characterizing sub-epidermal ware, Inc., Monterrey, CA) operate on the principle of
features in blistering disease.91 triangulation. As a helium-neon laser light source
Astner et al. used a combination of reflectance con- passes through two cylindrical lenses, the resulting

24 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA February 2010


IMAGING SKIN VISSCHER

vertical plane of light projects onto the surface of the related to, and usually correlated with clinical judg-
object. The highlighted profile is reflected from the ment. Perhaps the clinician is relying more on imag-
image mirrors to a video sensor and digitized in a ing methods than previously with the benefit of
raster fashion to determine the 2D coordinates of 256 increased efficiency. With “future vision”, the
points along the profile surface. The scanner moves approaches will be multimodal. One method is unlike-

A
along a linear trajectory performing 512 individual ly to tell the story. The era of “globalization” demands
surface contour scans in equal increments. Trigono- that skin imaging is done successfully and complete-
metric calculations of the 2D coordinates to 3D space ly across an extraordinarily diverse population of

IC
are performed. Resulting images are processed to
remove extraneous features, e.g., LS 3rd order poly-
nomial equation to remove the thigh curvature and
humans. The need for methods of clinical utility and
rapid data reduction is driven by an increased under-
standing of the complex processes in the skin and a

D
filtering utilities to remove noise from movement or
hair. Quantitative surface roughness parameters are

E
then calculated from the scan data with appropriate
software (e.g., TrueMap Software, TrueGage, N. Hunt-
ingdon, PA).97
desire to deliver cost-effective care that meets
patient/consumer expectations. Prevention and early
detection of tissue injury is dependent on objective,
quantitative skin imaging methods. The generation
of a coherent framework with quantitative endpoints

M ®
As an example 3D surface scanning was used to
determine differences in the severity of gynoid lipody-
strophy (cellulite). The effect of weight loss on sever-
may be facilitated by the reapplication of validated,
published strategies and approaches from one condi-
tion to another. Based on the current research, the

A T
ity was determined by comparing 3D surface rough-
ness, thigh fat levels and dermal tissue density (ultra-

V
sound) over six months. Significant differences were
future goal of better, less invasive diagnostic proce-
dures for skin disorders is likely to be reached. In

H
1966, renowned dermatologist, Dr. Albert M. Kligman
found for cellulite severity: Sa (average roughness, delivered a lecture entitled “Blind Man Dermatol-

R
average of the absolute distances of the surface profile ogy” in which he stressed the importance of quanti-

G
from the reference plane), Sq (square root of Sa, width tative skin measurements.100 He stated, “I hope to be

E I
or variance of the amplitude distribution function), St alive when the first blind medical graduate applies for
(vertical distance from the highest peak to the lowest residency training in dermatology; if he is as confident
valley), Spm (mean height of the highest peaks over the

IN YR
as I now am that sightlessness will be an inconve-
sample length), and Sz (mean of the vertical distance nience rather than an insuperable disability, the gold-
from the highest peaks to the lowest valleys over the en age will have begun.” It is possible to make the
sample length).98 The percent thigh fat and subregion
case that the golden age is here.
fat decreased and lean tissue increased. The dermal

M P
tissue density (ultrasound) decreased with weight loss,
but dermal thickness and the dermal-subcutaneous Riassunto

O
surface area were not different. Recently, Horton et
al., have demonstrated the use of computed tomogra- Metodiche di imaging cutaneo: passato, presente e pro-
phy with multidectector hardware for 3D volume ren-

C
spettive future
dering of skin.99 They propose exciting uses of this
Le modalità di imaging cutaneo importanti nella valuta-
modality for skin surface characterization in condi- zione della varietà di patologie cutanee riscontrate in ambi-
tions such as cellulite. to clinico vengono descritte con riferimento alle informa-
zioni fornite, ai vantaggi e alle loro limitazioni, allo stato
attuale e alle indicazioni per un ulteriore sviluppo. Le diver-
Future perspective se metodologie utilizzano l’interazione di energia con la
cute, che penetra a diverse profondità nello strato corneo,
In keeping with the title, imaging skin today con- nell’epidermide, nel derma e nello stato sottocutaneo, un
jures “images” of the past and present. Based on the sistema di rilevamento come la retina, pellicola oppure un
array digitale, e un sistema di processazione per frammen-
rich past history of skin evaluation, it seems clear that tare, analizzare e interpretare l’informazione. Allo stesso
the live visual clinical evaluation is still the gold stan- modo, le aree di interesse, o obiettivi, hanno caratteristiche
dard. Presently, the data suggests that the output of comuni. Le patologie cutanee deviano dallo stato ideale o
energy based (optical) imaging methods is always normale per quanto riguarda l’integrità e la funzione della

Vol. 145 - No. 1 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA 25


VISSCHER IMAGING SKIN

cute. Una parte del compito nell’ambito delle metodiche Freinkel, Woodley, D.T. editor. The biology of the skin. New York:
per immagine cutanee è di stabilire i criteri della condizio- Parthenon Publishing Group; 2001. p. 113-31.
ne normale. Le deviazioni includono l’evidenza di rottura 19. Morgan JE, Gilchrest B , Goldwyn RM. Skin pigmentation. Current
concepts and relevance to plastic surgery. Plast Reconstr Surg
della barriera, di infiammazione, di alterazione della pig- 1975;56:617-28.
mentazione e di modifiche vascolari. Chi utilizza l’ima- 20. Knudsen A , Brodersen R. Skin colour and bilirubin in neonates. Arch
ging cutaneo è spesso interessato dalla valutazione dell’e- Dis Child 1989;64:605-9.

A
stensione e della gravità della malattia. Questa revisione 21. Wagner JK, Jovel C, Norton HL, Parra EJ , Shriver MD. Comparing
include il passato, il presente e il futuro della valutazione quantitative measures of erythema, pigmentation and skin response
visiva, della raccolta di immagini fotografiche, delle tecni- using reflectometry. Pigment Cell Res 2002;15:379-84.

C
22. Jordan WE, Lawson KD, Berg RW, Franxman JJ, Marrer AM. Diaper
che spettrofotometriche, dell’istologia non invasive, e del- dermatitis: frequency and severity among a general infant popula-

I
la scansione tridimensionale. Inoltre, gli autori presentano tion. Pediatr Dermatol 1986;3:198-207.
le tecniche analitiche per la processazione e l’estrazione di 23. Elsner P. Skin color. In: Berardesca E, Elsner P, Wilhelm KP, Maibach
specifici parametri che forniscono informazioni circa il sot- HI, editors.0 Bioengineering of the skin: methods and instrumentation.

E
nale - Dermoscopia.D
tostante stato biologico.
Parole chiave: Malattie della pelle - Immagine tridimensio-
24.

25.
Boca Raton: CRC Press; 1995. p. 29-40.
Kawai K, Kawai J, Nakagawa M, Kawai K. Effects of detergents. In:
Wilhelm K, Elsner P, Berardesca E, Maibach HI, editors. Bioengi-
neering of the skin: skin surface imaging and analysis. Boca Raton:
CRC Press; 1997. p. 303-14.
Mattsson U, Jonsson A, Jontell M , Cassuto J. Digital image analysis

M ®
(DIA) of colour changes in human skin exposed to standardized ther-
References mal injury and comparison with laser Doppler measurements. Com-
put Methods Programs Biomed 1996;50:31-42.
1. Imaging. The American Heritage Science Dictionary. Orlando, FL 26. Nystrom J, Geladi P, Lindholm-Sethson B, Rattfelt J, Svensk AC ,

A
USA: Houghton Mifflin Company; 2005. Franzen L. Objective measurements of radiotherapy-induced erythe-

T
2. Perednia DA. What dermatologists should know about digital imag- ma. Skin Res Technol 2004;10:242-50.
ing. J Am Acad Dermatol 1991;25:89-108. 27. Stamatas GN, Kollias N. Blood stasis contributions to the perception

V
3. Rallan D, Harland CC. Skin imaging: is it clinically useful? Clin Exp of skin pigmentation. J Biomed Opt 2004;9:315-22.

H
Dermatol 2004;29:453-9. 28. Ben-Gashir MA , Hay RJ. Reliance on erythema scores may mask
4. Galdino GM, Vogel JE , Vander Kolk CA. Standardizing digital pho- severe atopic dermatitis in black children compared with their white

R
tography: it’s not all in the eye of the beholder. Plast Reconstr Surg counterparts. Br J Dermatol 2002;147:920-5.

G
2001;108:1334-44. 29. Black J, Baharestani M, Cuddigan J, Dorner B, Edsberg L, Langemo
5. Caspers PJ, Lucassen GW, Carter EA, Bruining HA, Puppels GJ. In D et al. National Pressure Ulcer Advisory Panel’s updated pressure

E I
vivo confocal Raman microspectroscopy of the skin: noninvasive ulcer staging system. Urol Nurs 2007;27:144-50, 56.
determination of molecular concentration profiles. J Invest Dermatol 30. Thomas DR, Rodeheaver GT, Bartolucci AA, Franz RA, Sussman
2001;116:434-42. C, Ferrell BA et al. Pressure ulcer scale for healing: derivation and val-

IN YR
6. Anderson RR , Parrish JA. The optics of human skin. J Invest Dermatol idation of the PUSH tool. The PUSH Task Force. Adv Wound Care
1981;77:13-9. 1997;10:96-101.
7. Taylor S, Westerhof W, Im S, Lim J. Noninvasive techniques for the 31. George-Saintilus E, Tommasulo B, Cal CE, Hussain R, Mathew N,
evaluation of skin color. J Am Acad Dermatol 2006;54:S282-90. Dlugacz Y et al. Pressure ulcer PUSH score and traditional nursing
8. Serup J. Skin irritation: objective characterization in a clinical per- assessment in nursing home residents: do they correlate? J Am Med

M P
spective. In: Wilhelm KP, Elsner P, Berardesca E, Maibach I, editors. Dir Assoc 2009;10:141-4.
r6Bioengineering of the Skin: Skin Surface Imaging and Analysis. 32. Hanifin JM, Thurston M, Omoto M, Cherill R, Tofte SJ , Graeber M.
Boca Raton: CRC Press; 1997. p. 261-73. The eczema area and severity index (EASI): assessment of reliabili-
9. Stephen ID, Coetzee V, Law Smith M , Perrett DI. Skin blood perfu- ty in atopic dermatitis. EASI Evaluator Group. Exp Dermatol

O
sion and oxygenation colour affect perceived human health. PLoS 2001;10:11-8.
One 2009;4:e5083. 33. Fredriksson T , Pettersson U. Severe psoriasis—oral therapy with a new
10. Fink B, Matts PJ, Klingenberg H, Kuntze S, Weege B, Grammer K. retinoid. Dermatologica 1978;157:238-44.

C
Visual attention to variation in female facial skin color distribution. J 34. Oranje AP, Stalder JF, Taieb A, Tasset C, de Longueville M. Scoring
Cosmet Dermatol 2008;7:155-61. of atopic dermatitis by SCORAD using a training atlas by investiga-
11. Fink B , Matts PJ. The effects of skin colour distribution and topog- tors from different disciplines. ETAC Study Group. Early Treatment
raphy cues on the perception of female facial age and health. J Eur Acad of the Atopic Child. Pediatr Allergy Immunol 1997;8:28-34.
Dermatol Venereol 2008;22:493-8. 35. Pucci N, Novembre E, Cammarata MG, Bernardini R, Monaco MG,
12. Hunt RWG. The Reproduction of Color Chichester: Wiley; 2004. Calogero C et al. Scoring atopic dermatitis in infants and young children:
13. Wyszecki G, Stiles, W.S. Color Science: Concepts and methods, quan- distinctive features of the SCORAD index. Allergy 2005;60:113-6.
titative data and formulae. New York: Wiley; 1982. 36. Sugarman JL, Fluhr JW, Fowler AJ, Bruckner T, Diepgen TL, Williams
14. Susstrunk S, Buckley R, Swen S. Standard RBG color spaces. ML. The objective severity assessment of atopic dermatitis score: an
IS&T/SID 7th Color Imaging Conference1999. p. 127-34. objective measure using permeability barrier function and stratum
15. Chardon A, Cretois I, Hourseau C. Skin colour typology and suntan- corneum hydration with computer-assisted estimates for extent of
ning pathways. Int J Cosmet Sci 1991;13:191-208. disease. Arch Dermatol 2003;139:1417-22.
16. Stamatas GN, Zmudzka BZ, Kollias N, Beer JZ. Non-invasive mea- 37. Angelova-Fischer I, Bauer A, Hipler UC, Petrov I, Kazandjieva J,
surements of skin pigmentation in situ. Pigment Cell Res 2004;17:618- Bruckner T et al. The objective severity assessment of atopic der-
26. matitis (OSAAD) score: validity, reliability and sensitivity in adult
17. Takiwaki H. Measurement of skin color: practical application and patients with atopic dermatitis. Br J Dermatol 2005;153:767-73.
theoretical considerations. J Med Invest 1998;44:121-6. 38. Odio MR, O’Connor RJ, Sarbaugh F , Baldwin S. Continuous topi-
18 Nordlund JJ, Boissy RE. The biology of melanocytes. In: R. K. cal administration of a petrolatum formulation by a novel disposable

26 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA February 2010


IMAGING SKIN VISSCHER

diaper. 2. Effect on skin condition. Dermatology 2000;200:238-43. dards: Algorithms and Architectures. Norwell: Kluwer Academic
39. Visscher MO. Update on the use of topical agents in neonates. New- Publishers; 1997.
born & Infant Nursing Reviews 2009;9:31-47. 61. Mark R, Billo, E. Application of digital image enhancement in rock
40. Horfelt C, Funk J, Frohm-Nilsson M, Wiegleb Edstrom D, Wennberg art recording. Fifth International Rock Art Symposium. Tarija2000.
AM. Topical methyl aminolaevulinate photodynamic therapy for 62. Westerhof W, van Hasselt BA , Kammeijer A. Quantification of UV-
treatment of facial acne vulgaris: results of a randomized, controlled induced erythema with a portable computer controlled chromame-
study. Br J Dermatol 2006;155:608-13. ter. Photodermatol 1986;3:310-4.

A
41. Leyden JJ, Shalita A, Thiboutot D, Washenik K, Webster G. Topical 63. Canning J, Barford B, Sullivan D, Wickett R , Visscher M. Use of
retinoids in inflammatory acne: a retrospective, investigator-blind- digital photography and image analysis techniques to quantify erythema
ed, vehicle-controlled, photographic assessment. Clin Ther in health care workers. Skin Res Technol 2009;15:24-34.

C
2005;27:216-24. 64. Visscher M, Davis J, Wickett R. Effect of topical treatments on irri-
42. Pressure Ulcer Scale for Healing (PUSH). PUSH Tool 3.0: National tant hand dermatitis in health care workers. Am J Infect Control

DI
Pressure Ulcer Advisory Panel; 1998. p. [Internet]. Available at
www.npuap.org [cited 2010, Feb 2].
43. Liguori V, Guillemin C, Pesce GF, Mirimanoff RO, Bernier J. Dou-
ble-blind, randomized clinical study comparing hyaluronic acid cream
to placebo in patients treated with radiotherapy. Radiother Oncol
65.

66.
2009;37:842 e1- e11.
Kosmopoulos DI, Tzevelekou FL. Automated pressure ulcer lesion
diagnosis for telemedicine systems. IEEE Eng Med Biol Mag
2007;26:18-22.
Visscher M, Chatterjee R EJ, LaRuffa AA, Hoath SB. Biomedical

E
1997;42:155-61. assessment and instrumental evaluation of healthy infant skin. Pedi-
44. Schmuth M, Wimmer MA, Hofer S, Sztankay A, Weinlich G, Linder atr Dermatol 2002;19.
DM et al. Topical corticosteroid therapy for acute radiation dermati- 67. Hakozaki T, Minwalla L, Zhuang J, Chhoa M, Matsubara A, Miyamo-
tis: a prospective, randomized, double-blind study. Br J Dermatol to K et al. The effect of niacinamide on reducing cutaneous pigmen-

M ®
2002;146:983-91. tation and suppression of melanosome transfer. Br J Dermatol
45. Sullivan T, Smith J, Kermode J, McIver E , Courtemanche DJ. Rating 2002;147:20-31.
the burn scar. J Burn Care Rehabil 1990;11:256-60. 68. Denecker K, De Neve P, Van Assche S, Van de Walle R, Lemahieu I,
46. Barczak CA, Barnett RI, Childs EJ, Bosley LM. Fourth national pres- Philips W. Psychovisual Evaluation of Lossy CMYK Image Com-

A
sure ulcer prevalence survey. Adv Wound Care 1997;10:18-26. pression for Printing Applications. Computer Graphics Forum

T
47. Henderson CT, Ayello EA, Sussman C, Leiby DM, Bennett MA, 2002;21:5-17.
Dungog EF et al. Draft definition of stage I pressure ulcers: inclusion 69. Weller R, Hunter J, Savin J, Dahl M. Cinical dermatology. 4th edition.

V
of persons with darkly pigmented skin. NPUAP Task Force on Stage Malden, Massachusetts, USA: Blackwell Publishing; 2008.

H
I Definition and Darkly Pigmented Skin. Adv Wound Care 1997;10:16- 70. von Luschan F. Beitrage zur Volkerkunde der Deutschen Schutzge-
9. bieten. Berlin: Deutsche Buchgemeinschaft; 1897.

R
48. De Luca D, Zecca E, Zuppa AA , Romagnoli C. The joint use of 71. Lock-Andersen J , Wulf HC. Seasonal variation of skin pigmenta-

G
human and electronic eye: visual assessment of jaundice and tran- tion. Acta Derm Venereol 1997;77:219-21.
scutaneous bilirubinometry. Turk J Pediatr 2008;50:456-61. 72. Clarys P, Alewaeters K, Lambrecht R, Barel AO. Skin color mea-

E I
49. Bendeck SE , Jacobe HT. Ultrasound as an outcome measure to assess surements: comparison between three instruments: the Chromame-
disease activity in disorders of skin thickening: an example of the ter(R), the DermaSpectrometer(R) and the Mexameter(R). Skin Res
use of radiologic techniques to assess skin disease. Dermatol Ther Technol 2000;6:230-8.

IN YR
2007;20:86-92. 73. Shriver MD, Parra EJ. Comparison of narrow-band reflectance spec-
50. Rubegni P, Nami N, Poggiali S, Tataranno D, Fimiani M. Digital troscopy and tristimulus colorimetry for measurements of skin and hair
image management project for dermatological health care environ- color in persons of different biological ancestry. Am J Phys Anthro-
ments: a new dedicated software and review of the literature. Skin pol 2000;112:17-27.
Res Technol 2009;15:148-56. 74. Miyamoto K, Takiwaki H, Hillebrand GG , Arase S. Development

M P
51. Coelho SG, Miller SA, Zmudzka BZ , Beer JZ. Quantification of UV- of a digital imaging system for objective measurement of hyperpig-
induced erythema and pigmentation using computer-assisted digital mented spots on the face. Skin Res Technol 2002;8:227-35.
image evaluation. Photochem Photobiol 2006;82:651-5. 75. Bissett DL, Robinson LR, Raleigh PS, Miyamoto K, Hakozaki T, Li
52. Setaro M , Sparavigna A. Quantification of erythema using digital J et al. Reduction in the appearance of facial hyperpigmentation by top-

O
camera and computer-based colour image analysis: a multicentre ical N-undecyl-10-enoyl-L-phenylalanine and its combination with
study. Skin Res Technol 2002;8:84-8. niacinamide. J Cosmet Dermatol 2009;8:260-6.
53. Aspres N, Egerton IB, Lim AC , Shumack SP. Imaging the skin. Aus- 76. Stamatas GN, Zmudzka BZ, Kollias N, Beer JZ. In vivo measure-

C
tralas J Dermatol 2003;44:19-27. ment of skin erythema and pigmentation: new means of implementation
54. Fogelberg A, Ioffreda M , Helm KF. The utility of digital clinical of diffuse reflectance spectroscopy with a commercial instrument.
photographs in dermatopathology. J Cutan Med Surg 2004;8:116- Br J Dermatol 2008;159:683-90.
21. 77. Stamatas GN , Kollias N. In vivo documentation of cutaneous inflam-
55. Oduncu H, Hoppe A, Clark M, Williams RJ , Harding KG. Analysis mation using spectral imaging. J Biomed Opt 2007;12:051603.
of skin wound images using digital color image processing: a pre- 78. Visscher MO, Sullivan D, Sullivan S, Barford B, Dock M , Sommers
liminary communication. Int J Low Extrem Wounds 2004;3:151-6. MS. Quantitation of epidermal and mucosal tissue injury using con-
56. Weatherall IL , Coombs BD. Skin color measurements in terms of trast agents and imaging techniques. Skin Res Technol 2009;15:180-
CIELAB color space values. J Invest Dermatol 1992;99:468-73. 6.
57. Colorimetry. 2nd edition. CIE publications No. 15.2. Colorimetric 79. Malvehy J, Puig S, Argenziano G, Marghoob AA, Soyer HP. Der-
Illuminants (CIE S001); Colorimetric Observers (CIE S002). Vienna: moscopy report: proposal for standardization. Results of a consensus
Central Bureau of the CIE; 1986. meeting of the International Dermoscopy Society. J Am Acad Dermatol
58. Visscher M, Canning J, Said D, Wickett R, Bondurant P. Effect of 2007;57:84-95.
hand hygiene regimens on skin condition in health care workers. Am 80. Vestergaard ME, Macaskill P, Holt PE , Menzies SW. Dermoscopy
J Infect Control 2006;34:S98-110. compared with naked eye examination for the diagnosis of primary
59. Hinkel B. The focal easy guide to photoshop CS2. Burlington: Focal melanoma: a meta-analysis of studies performed in a clinical setting.
Press; 2006. Br J Dermatol 2008;159:669-76.
60. Bahaskaran V, Konstantinides K. Image and Video Compression Stan- 81. Dwyer T, Muller HK, Blizzard L, Ashbolt R, Phillips G. The use of

Vol. 145 - No. 1 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA 27


VISSCHER IMAGING SKIN

spectrophotometry to estimate melanin density in Caucasians. Can- 91. Mogensen M, Thrane L, Joergensen TM, Andersen PE, Jemec GB.
cer Epidemiol Biomarkers Prev 1998;7:203-6. Optical coherence tomography for imaging of skin and skin dis-
82. Moncrieff M, Cotton S, Claridge E, Hall P. Spectrophotometric intra- eases. Semin Cutan Med Surg 2009;28:196-202.
cutaneous analysis: a new technique for imaging pigmented skin 92. Astner S, Burnett N, Rius-Diaz F, Doukas AG, Gonzalez S , Gonzalez
lesions. Br J Dermatol 2002;146:448-57. E. Irritant contact dermatitis induced by a common household irri-
83. Matts PJ. New insights into skin appearance and measurement. J tant: a noninvasive evaluation of ethnic variability in skin response.
Investig Dermatol Symp Proc 2008;13:6-9. J Am Acad Dermatol 2006;54:458-65.

A
84. Kollias N, Seo I, Bargo PR. Interpreting diffuse reflectance for in 93. Hofmann-Wellenhof R, Wurm EM, Ahlgrimm-Siess V, Richtig E,
vivo skin reactions in terms of chromophores. J Biophotonics Koller S, Smolle J et al. Reflectance confocal microscopy—state-of-
2010;3:15-24. art and research overview. Semin Cutan Med Surg 2009;28:172-9.

C
85. Cho JK, Moon DJ, Kim SG, Lee HG, Chung SP, Yoon CJ. Relation- 94. Tsai TH, Jee SH, Dong CY, Lin SJ. Multiphoton microscopy in der-
ship between healing time and mean perfusion units of laser Doppler matological imaging. J Dermatol Sci 2009;56:1-8.

86.

I
imaging (LDI) in pediatric burns. Burns 2009;35:818-23.
Allely RR, Van-Buendia LB, Jeng JC, White P, Wu J, Niszczak J et al.
Laser Doppler imaging of cutaneous blood flow through transparent

D
face masks: a necessary preamble to computer-controlled rapid pro-
totyping fabrication with submillimeter precision. J Burn Care Res
95. Jacobi U, Chen M, Frankowski G, Sinkgraven R, Hund M, Rzany B
et al. In vivo determination of skin surface topography using an opti-
cal 3D device. Skin Res Technol 2004;10:207-14.
96. Smalls LK, Lee CY, Whitestone J, Kitzmiller WJ, Wickett RR , Viss-
cher MO. Quantitative model of cellulite: three-dimensional skin

E
2008;29:42-8. surface topography, biophysical characterization, and relationship to
87. Mill J, Cuttle L, Harkin DG, Kravchuk O, Kimble RM. Laser Doppler human perception. J Cosmet Sci 2005;56:105-20.
imaging in a paediatric burns population. Burns 2009;35:824-31. 97. Precision Devices, Inc. Surface Metrology Guide2003. [Internet].
88. O’Doherty J, McNamara P, Clancy NT, Enfield JG, Leahy MJ. Com- Available from http://www.predev.com [cited 2010, Feb 2].

M ®
parison of instruments for investigation of microcirculatory blood 98. Smalls LK, Hicks M, Passeretti D, Gersin K, Kitzmiller WJ, Bakhsh
flow and red blood cell concentration. J Biomed Opt 2009;14:034025. A et al. Effect of weight loss on cellulite: gynoid lypodystrophy.
89. Murray AK, Moore TL, Manning JB, Taylor C, Griffiths CE, Herrick Plast Reconstr Surg 2006;118:510-6.
AL. Noninvasive imaging techniques in the assessment of scleroder- 99. Horton KM, Johnson PT, Heath DG, Ney DR, Fishman EK. Three-

A
ma spectrum disorders. Arthritis Rheum 2009;61:1103-11. dimensional volume rendering and display of skin and subcutaneous

T
90. Nishide K, Nagase T, Oba M, Oe M, Ohashi Y, Iizaka S et al. Ultra- tissues. Applied Radiology 2009;6-13.
sonographic and thermographic screening for latent inflammation in 100. Kligman AM. Blind man dermatology. Journal of the Society of

V
diabetic foot callus. Diabetes Res Clin Pract 2009;85:304-9. Cosmetic Chemists 1966:506-9.

R G H
E I
IN YR
M P
CO

28 GIORNALE ITALIANO DI DERMATOLOGIA E VENEREOLOGIA February 2010

View publication stats

You might also like