Shinjita Das M.D., Rachel Reynolds M.D. (Auth.), Joshua Zeichner (Eds.) - Acneiform Eruptions in Dermatology - A Differential Diagnosis-Springer-Verlag New York (2014) PDF
Shinjita Das M.D., Rachel Reynolds M.D. (Auth.), Joshua Zeichner (Eds.) - Acneiform Eruptions in Dermatology - A Differential Diagnosis-Springer-Verlag New York (2014) PDF
Zeichner Editor
Acneiform Eruptions
in Dermatology
A Differential Diagnosis
Acneiform Eruptions in Dermatology
Joshua A. Zeichner
Editor
Acneiform Eruptions
in Dermatology
A Differential Diagnosis
Editor
Joshua A. Zeichner, M.D.
Assistant Professor
Director of Cosmetic and Clinical Research
Dermatology Department
Mount Sinai Medical Center
New York, NY, USA
An estimated 40–50 million people in the United States suffer from acne, and up to
85 % of people experience acne at some point in their lives. Dermatologists, pri-
mary care doctors, and pediatricians see these patients every day in practice. It is
important to treat the skin effectively not only to reduce the risk of physical scarring
but also to address the negative psychosocial impact this disease carries. Improving
the skin can improve self-confidence, interpersonal relationships, and performance
in school or at work.
While the majority of acne patients seen in practice truly have run-of-the-mill
acne, not all “acne” is really acne vulgaris. It may be exception rather than the rule
for a patient to have a condition that mimics acne, but it is our responsibility as health
care providers to be up to date and educated on acne’s broad differential diagnosis. If
patients do not have any comedonal lesions, if they have an atypical medical history,
or if they are not responding to traditional therapies, then alternative diagnoses
should be considered. Proper diagnosis will allow us to prescribe proper treatments
and ultimately improve clinical outcomes and patient satisfaction.
This book will help to provide a broad overview of acne vulgaris itself as well as
conditions that manifest with acneiform eruptions in the skin.
vii
Acknowledgment
We are all influenced by the people with whom we surround ourselves. I have had
the privilege of learning from the greatest minds in Dermatology, who have
shaped the person I am today. While I have had countless mentors touch my career,
I must take this opportunity to acknowledge one person in particular. Dr. Mark
Lebwohl is truly a mentor of mentors. He has taught me not only how to be a
good dermatologist, but also, more importantly, how to be a great doctor, colleague,
and teacher. This book would not have been possible without his support, advice,
and encouragement.
ix
Contents
xi
xii Contents
43 Rosacea..................................................................................................... 309
Joseph Bikowski
44 Rosacea Fulminans ................................................................................. 317
Cristina Caridi and Joshua A. Zeichner
45 Sarcoidosis ............................................................................................... 321
Laura Thornsberry and Joseph English III
46 Seborrheic Dermatitis............................................................................. 329
Elizabeth Farhat and Linda Stein Gold
47 Steatocystoma Multiplex ........................................................................ 343
Alejandra Vivas and Jonette Keri
48 Xanthomas ............................................................................................... 349
Libby Rhee and Mark Kaufmann
xv
xvi Contributors
1.1 Introduction
Acne vulgaris is the result of multifactorial processes in and around the pilosebaceous
unit. Currently, the major pathogenic factors are thought to be:
1. Androgens [1–3]
2. Sebum [4]
3. Abnormal keratinization [5]
4. Propionibacterium acnes (P. acnes) [6]
5. Innate immune system-mediated inflammation [6, 7]
The traditional notion that these factors contribute independently to acne
development has been replaced by a more nuanced understanding of their complex
interplay. Though the multifactorial nature of acne pathogenesis makes effective
treatment challenging, research has shed light on both the mechanisms of action of
current treatments and discovered new targets for acne therapy.
S. Das, M.D.
Department of Dermatology, Harvard Combined Dermatology
Residency Program, Boston, MA, USA
e-mail: [email protected]
R. Reynolds, M.D. (*)
Department of Dermatology, Beth Israel Deaconess Medical Center,
330 Brookline Avenue, Brookline, MA 02215, USA
e-mail: [email protected]
The immune system consists of both innate (rapid response but no memory to
pathogens) and adaptive (delayed, antigen-specific response forming memory)
mechanisms against pathogens [8]. The skin is a first-line agent of the innate
immune system. As an anatomic barrier, skin physically prevents invasion of exter-
nal toxins and maintains an acidic stratum corneum (from free fatty acids generated
by P. acnes) that limits bacterial colonization [9–11]. It can also generate soluble
immune factors (antimicrobial peptides, complement factors, chemokines, and
cytokines) and express pattern recognition receptors (PRRs) to mediate responses
against pathogen-associated molecular patterns (PAMPs) via effector cells, such
as monocytes/macrophages, natural killer (NK) cells, neutrophils, and dendritic
cells (DCs) [12–15].
1.3.1 Androgens
1.3.2 Sebum
Fig. 1.1 P. acnes promotes acne pathogenesis through multiple pathways involving the innate
immune system
1.4 Conclusions
References
1. Thiboutot D. Hormones and acne: pathophysiology, clinical evaluation, and therapies. Semin
Cutan Med Surg. 2001;20(3):144–53.
2. Thiboutot D, Jabara S, McAllister JM, Sivarajah A, Gilliland K, Cong Z, Clawson G. Human
skin is a steroidogenic tissue: steroidogenic enzymes and cofactors are expressed in epidermis,
normal sebocytes, and an immortalized sebocyte cell line (SEB-1). J Invest Dermatol.
2003;120(6):905–14.
3. Thiboutot D. Acne: hormonal concepts and therapy. Clin Dermatol. 2004;22(5):419–28. Review.
4. Thiboutot D. Regulation of human sebaceous glands. J Invest Dermatol. 2004;123(1):1–12.
Review.
5. Plewig G, Fulton JE, Kligman AM. Cellular dynamics of comedo formation in acne vulgaris.
Arch Dermatol Forsch. 1971;242(1):12–29.
6. Vowels BR, Yang S, Leyden JJ. Induction of proinflammatory cytokines by a soluble factor of
Propionibacterium acnes: implications for chronic inflammatory acne. Infect Immun. 1995;
63(8):3158–65.
10 S. Das and R. Reynolds
7. Guy R, Green MR, Kealey T. Modeling acne in vitro. J Invest Dermatol. 1996;106(1):176–82.
PubMed PMID: 8592071.
8. Parkin J, Cohen B. An overview of the immune system. Lancet. 2001;357:1777–89.
9. Fluhr JW, Kao J, Jain M, Ahn SK, Feingold KR, Elias PM. Generation of free fatty acids from
phospholipids regulates stratum corneum acidification and integrity. J Invest Dermatol.
2001;117(1):44–51.
10. Elias PM. Stratum corneum defensive functions: an integrated view. J Invest Dermatol.
2005;125(2):183–200. Review.
11. Elias PM. The skin barrier as an innate immune element. Semin Immunopathol. 2007;29(1):
3–14. Review.
12. Braff MH, Bardan A, Nizet V, Gallo RL. Cutaneous defense mechanisms by antimicrobial
peptides. J Invest Dermatol. 2005;125(1):9–13. Review.
13. Schauber J, Gallo RL. Antimicrobial peptides and the skin immune defense system. J Allergy
Clin Immunol. 2008;122(2):261–6.
14. Walport MJ. Complement. First of two parts. N Engl J Med. 2001;344(14):1058–66. Review.
15. Modlin RL, Miller LS, Bangert C, Stingl G. Innate and Adaptive Immunity in the Skin. In:
Goldsmith LA, Katz SI, et al., editors. Fitzpatrick’s dermatology in general medicine. 8th ed.
New York, NY: McGraw-Hill; 2012.
16. Jugeau S, Tenaud I, Knol AC, Jarrousse V, Quereux G, Khammari A, Dreno B. Induction of
toll-like receptors by Propionibacterium acnes. Br J Dermatol. 2005;153(6):1105–13.
17. Kim J. Review of the innate immune response in acne vulgaris: activation of Toll-like receptor
2 in acne triggers inflammatory cytokine responses. Dermatology. 2005;211(3):193–8. Review.
18. McInturff JE, Kim J. The role of toll-like receptors in the pathophysiology of acne. Semin
Cutan Med Surg. 2005;24(2):73–8. Review.
19. McInturff JE, Modlin RL, Kim J. The role of toll-like receptors in the pathogenesis and treat-
ment of dermatological disease. J Invest Dermatol. 2005;125(1):1–8. Review.
20. Perisho K, Wertz PW, Madison KC, Stewart ME, Downing DT. Fatty acids of acylceramides
from comedones and from the skin surface of acne patients and control subjects. J Invest
Dermatol. 1988;90(3):350–3.
21. Elias PM, Brown BE, Ziboh VA. The permeability barrier in essential fatty acid deficiency:
evidence for a direct role for linoleic acid in barrier function. J Invest Dermatol. 1980;
74(4):230–3.
22. Jeremy AH, Holland DB, Roberts SG, Thomson KF, Cunliffe WJ. Inflammatory events are
involved in acne lesion initiation. J Invest Dermatol. 2003;121(1):20–7.
23. Freedberg IM, Tomic-Canic M, Komine M, Blumenberg M. Keratins and the keratinocyte
activation cycle. J Invest Dermatol. 2001;116(5):633–40. Review.
24. Guy R, Kealey T. Modelling the infundibulum in acne. Dermatology. 1998;196(1):32–7.
25. Guy R, Kealey T. The effects of inflammatory cytokines on the isolated human sebaceous
infundibulum. J Invest Dermatol. 1998;110(4):410–5.
26. Deplewski D, Rosenfield RL. Role of hormones in pilosebaceous unit development. Endocr
Rev. 2000;21(4):363–92. Review.
27. Gilliver SC, Ashworth JJ, Ashcroft GS. The hormonal regulation of cutaneous wound healing.
Clin Dermatol. 2007;25(1):56–62. Review.
28. Thiboutot D, Harris G, Iles V, Cimis G, Gilliland K, Hagari S. Activity of the type 1 5 alpha-
reductase exhibits regional differences in isolated sebaceous glands and whole skin. J Invest
Dermatol. 1995;105(2):209–14.
29. Merke DP, Bornstein SR. Congenital adrenal hyperplasia. Lancet. 2005;365(9477):2125–36.
Review.
30. Lolis MS, Bowe WP, Shalita AR. Acne and systemic disease. Med Clin North Am. 2009;
93(6):1161–81.
31. Maluki AH. The frequency of polycystic ovary syndrome in females with resistant acne
vulgaris. J Cosmet Dermatol. 2010;9(2):142–8.
32. Imperato-McGinley J, Gautier T, Cai LQ, Yee B, Epstein J, Pochi P. The androgen control of
sebum production. Studies of subjects with dihydrotestosterone deficiency and complete
androgen insensitivity. J Clin Endocrinol Metab. 1993;76(2):524–8.
1 Acne Pathophysiology 11
peripheral blood mononuclear cells and normal human keratinocytes. J Dermatol Sci.
2005;38(1):47–55.
55. Nagy I, Pivarcsi A, Koreck A, Széll M, Urbán E, Kemény L. Distinct strains of Propionibacterium
acnes induce selective human beta-defensin-2 and interleukin-8 expression in human keratino-
cytes through toll-like receptors. J Invest Dermatol. 2005;124(5):931–8.
56. Nagy I, Pivarcsi A, Kis K, Koreck A, Bodai L, McDowell A, Seltmann H, Patrick S, Zouboulis
CC, Kemény L. Propionibacterium acnes and lipopolysaccharide induce the expression of
antimicrobial peptides and proinflammatory cytokines/chemokines in human sebocytes.
Microbes Infect. 2006;8(8):2195–205.
57. Dessinioti C, Katsambas AD. The role of Propionibacterium acnes in acne pathogenesis: facts
and controversies. Clin Dermatol. 2010;28(1):2–7.
58. Jalian HR, Liu PT, Kanchanapoomi M, Phan JN, Legaspi AJ, Kim J. All-trans retinoic acid
shifts Propionibacterium acnes-induced matrix degradation expression profile toward matrix
preservation in human monocytes. J Invest Dermatol. 2008;128(12):2777–82.
59. Choi JY, Piao MS, Lee JB, Oh JS, Kim IG, Lee SC. Propionibacterium acnes stimulates
pro-matrix metalloproteinase-2 expression through tumor necrosis factor-alpha in human der-
mal fibroblasts. J Invest Dermatol. 2008;128(4):846–54.
60. Chronnell CM, Ghali LR, Ali RS, Quinn AG, Holland DB, Bull JJ, Cunliffe WJ, McKay IA,
Philpott MP, Müller-Röver S. Human beta defensin-1 and -2 expression in human piloseba-
ceous units: upregulation in acne vulgaris lesions. J Invest Dermatol. 2001;117(5):1120–5.
61. Lu PT, Phan J, Tang D, Kanchanapoomi M, Hall B, Krutzik SR, Kim J. CD209(+) macro-
phages mediate host defense against Propionibacterium acnes. J Immunol. 2008;180(7):
4919–23.
62. Jarrousse V, Castex-Rizzi N, Khammari A, Charveron M, Dréno B. Modulation of integrins
and filaggrin expression by Propionibacterium acnes extracts on keratinocytes. Arch Dermatol
Res. 2007;299(9):441–7.
63. Kurokawa I, Mayer-da-Silva A, Gollnick H, Orfanos CE. Monoclonal antibody labeling for
cytokeratins and filaggrin in the human pilosebaceous unit of normal, seborrhoeic and acne
skin. J Invest Dermatol. 1988;91(6):566–71.
64. Burkhart CG, Burkhart CN. Expanding the microcomedone theory and acne therapeutics:
propionibacterium acnes biofilm produces biological glue that holds corneocytes together to
form plug. J Am Acad Dermatol. 2007;57(4):722–4.
65. Holmes RL, Williams M, Cunliffe WJ. Pilo-sebaceous duct obstruction and acne. Br J Dermatol.
1972;87(4):327–32.
Chapter 2
Clinical Presentation of Acne
Guy F. Webster
2.1 Introduction
Acne is a disease of the pilosebaceous unit that typically begins on the face and has
a spectrum of lesions and severity (Table 2.1). The classic lesion is a pustule, but
inflammatory papules and nodules are common. The primary lesion, from which all
others develop, is the microcomedo, an impaction and distention of the follicle with
sebum and improperly desquamated keratinocytes from the follicular epithelium.
When microcomedones become visible, they are described as open or closed
comedones. An open comedo has a visible pore that appears as a dark spot (Fig. 2.1).
The pigment is not dirt, but is oxidized lipid and melanin. Closed comedones have
a pore too small to see and appear as white bumps (Fig. 2.2).
In patients who are hypersensitive to Propionibacterium acnes that colonizes
follicles, [1] inflammatory lesions may develop from the microcomedones. Papules
and pustules may be superficial or deep and scarring depending on the vigor of
hypersensitivity (Fig. 2.3). Nodules are inflammatory lesions >0.5–1 cm in size.
Nodules may develop into abscesses, which have incorrectly been termed “cysts”
(Fig. 2.4). The term “nodulocystic acne” is incorrect, but probably too deeply
ensconced to readily fall from use. Conglobate lesions are intensely inflamed
neighboring nodules that merge into a loculated abscess. Secondary lesions such as
scars, keloids, sinus tracts, and true cysts may follow in the most inflammatory
disease.
Acne usually begins with the onset of puberty [2, 3]. Androgens stimulate sebaceous
secretion and microcomedones inflate with sebum and become visible. Typically initial
lesions are noninflammatory and centrofacial (Fig. 2.5). As maturation progresses
2 Clinical Presentation of Acne 15
inflammatory lesions may appear, and the distribution spreads across the face and
perhaps to the trunk (Fig. 2.6). Severity of acne is clearly correlated with the stage of
puberty. Lucky et al. [4] has shown that early in puberty comedonal acne is common,
but inflammatory acne is rare. In later stages of puberty, inflammatory acne may reach
a 50 % incidence in boys [5]. Mourlatos and colleagues [6] demonstrated that both
sebaceous secretion and follicular P. acnes colonization are elevated early in those
children destined to develop acne.
In some patients, adrenarche is a sufficient stimulus and acne may appear in
7–11-year-olds. Lucky [4] has shown that early acne in girls reflects rising levels of
adrenal dehydroepiandrosterone sulfate (DHEAS). She also found that such patients
tended to have more severe acne as teens, though a sign of difficult acne to come,
such as preadolescent acne, is not a cause for medical concern. However, acne that
occurs between 1 and 7 years, termed mid-childhood acne, is much more unusual
and may reflect an underlying medical condition such as endocrinopathy or tumor.
Work-up by a pediatric endocrinologist is indicated [3].
16 G.F. Webster
Grading acne is a surprisingly difficult task. The severity of acne varies widely among
patients and even during the course of disease in a single patient. The traditional grad-
ing method in clinical trials involves counting inflammatory and noninflammatory
2 Clinical Presentation of Acne 17
References
3.1 Introduction
Topical treatment is a key component of acne therapy. Acne treatments target factors
contributing to acne formation and act to normalize follicular keratinization and
decrease sebaceous gland activity and follicular bacterial populations [1]. With early
treatment, acne scarring, post-inflammatory hyperpigmentation, and psychological
distress may be reduced or prevented [2]. Topical therapies for acne come in pre-
scription and over-the-counter formulations. This chapter will focus on prescription
products only.
M.-M. Kober, M.D. (*) • W.P. Bowe, M.D. • A.R. Shalita, M.D.
Department of Dermatology, SUNY Downstate Medical Center,
450 Clarkson Road, Brooklyn, NY, USA
e-mail: [email protected]; [email protected]; [email protected]
Tretinoin is the oldest of the retinoids and binds with equal affinity to all RAR
receptors. Tretinoin demonstrates superiority to benzoyl peroxide (BP), sulfur-
resorcinol, and vehicle in the reduction of inflammatory and noninflammatory acne
lesions as well as the global severity of acne [7, 8].
Although there may be some improvement noted after 2–3 weeks, the most
significant reduction in acne lesions is noted after 3–4 months of consistent use [9].
The histopathologic findings seen in skin receiving tretinoin include acanthosis,
parakeratosis, and thinning of the stratum corneum [8], which correlates with the
observation that the tretinoin-induced thinning of the stratum corneum allows for
improved efficacy of topical antimicrobial products [10].
Ultraviolet light and oxidants such as benzoyl peroxide degrade tretinoin upon
exposure [9]. Advances in formulation technology have allowed for the develop-
ment of new tretinoin containing topicals that combat these limitations. One such
example is the development of microsphere formations (Retin-A Micro®) that
significantly decreases degradation [11]. Microsphere formulations also provide a
gradual time release of tretinoin, increasing transdermal penetration and decreasing
the irritation [12].
Tazarotene and adapalene only bind to RAR-β (beta) and RAR-γ (gamma) receptor
subtypes, leading to reduced side effects compared to tretinoin.
Although similar in efficacy to tretinoin in the reduction of inflammatory lesions,
tazarotene has proven to be superior to tretinoin in reducing the number of nonin-
flammatory lesions [13]. Tazarotene also shows superiority compared to adapalene
in clearing post-inflammatory hyperpigmentation caused by acne [13].
Adapalene shows similar efficacy to tretinoin in reducing inflammatory and non-
inflammatory lesions and the global severity of acne [14]. However, when compared
to tazarotene, adapalene is less effective in decreasing the number of inflammatory
and noninflammatory lesions [15]. In general, adapalene demonstrates the best
tolerability profile of the topical retinoids [16, 17].
Both tazarotene and adapalene have increased stability in the presence of ultra-
violet light and BP as a result of replacing the labile, light sensitive double bonds
of tretinoin with naphthoic acid rings [18]. This allows for the development of
stable combination products containing third-generation retinoids with other active
ingredients, such as BP.
3 Topical Therapies for Acne 21
3.3.1 Antibiotics
as part of combination therapy [33]. Several combination products are available that
combine a topical antibiotic with BP into a single product. Combination gels have
reduced antibiotic-resistant strains of P. acnes by 97 % [34] and have shown to
improve patient adherence to treatment regimens [35].
Side effects of topical antibiotics are limited. The most common adverse effects
include reactions at the site of application (erythema, scaling, stinging, or burning);
these reactions have been shown to be at least in part dependent on the vehicle
formulation [23, 24].
The antimicrobial properties of azelaic acid make it another topical treatment option
for acne [33]. Trials comparing azelaic acid to BP or topical clindamycin show
similar reductions in the number or acne lesions [36]. Reduction in the number of
lesions can be seen as early as 4–8 weeks; however, maximum benefit typically
occurs after 16 weeks. Cutaneous side effects of stinging or burning may occur in
10–20 % of patients upon initial use; however, these side effects typically resolve
with continued use and rarely necessitate the cessation of treatment [37, 38].
It should be noted, however, that one of the authors found little or no efficacy with
the original formulation of azelaic acid and one laboratory found no antibacterial
effect on P. acnes.
Topical clindamycin, erythromycin, and azelaic acid appear to be safe during
pregnancy (Category B), with no reports of teratogenicity to date.
3.3.3 Dapsone
Topical dapsone has been approved for the treatment of acne vulgaris, although it
may result in a more modest reduction in acne lesions compared to other therapies.
It has an excellent tolerability profile, but may result in a temporary yellow discol-
oration of the skin when combined with BP [34]. Topical dapsone appears to work
well as an adjuvant to topical retinoids, reducing the irritation and desquamation
commonly associated with topical retinoids as well as significantly increasing their
efficacy [34].
Novel topical acne therapies focus on targeting drug delivery to the follicle.
3 Topical Therapies for Acne 23
3.4.1 Liposomes
Liposomes are phospholipid bilayers that create a spherical vesicle with a central
aqueous cavity and possess both hydrophobic and lipophilic properties. Clindamycin
in liposomes has shown a statistically significant decrease in acne lesions compared
to liposome-free clindamycin products [39]; similar efficacy has been seen with
liposome-derived tea tree oil, salicylic acid, retinoids, and BP [40].
3.4.2 Micronization
Smaller particles permit deeper penetration into the pilosebaceous unit. Micronizing
formulations create products of smaller particle sizes, with particles frequently less
than 10 μm. A micronized formulation of tretinoin gel, Atralin®, has demonstrated
enhanced moisture capacity and tolerability with similar lesion reduction compared
to generic tretinoin gel [41].
Mild acne responds to treatment with a topical retinoid with possible addition of BP
and/or a topical antibiotic if inflammatory lesions are present. Mild-to-moderate
acne typically requires combination therapy using one of the following regimens:
a topical retinoid combined with BP and an oral antibiotic or a topical retinoid
combined with a topical BP/antibiotic product [42]. Moderate-to-severe acne may
be treated with topical retinoids in combination with BP, topical, and/or oral anti-
biotics; however, acne this severe may require systemic isotretinoin, discussed
elsewhere.
3.6 Conclusion
References
21. Loureiro KD, Kao KK, Jones KL, et al. Minor malformations characteristic of the retinoic acid
embryopathy and other birth outcomes in children of women exposed to topical tretinoin
during early pregnancy. Am J Med Genet A. 2005;136(2):117–21.
22. Zhang JZ, Li LF, Tu YT, et al. A successful maintenance approach in inflammatory acne with
adapalene gel 0.1% after an initial treatment in combination with clindamycin topical solution
1% or after monotherapy with clindamycin topical solution 1%. J Dermatol Treat. 2004;
15(6):372–8.
23. Leyden JJ, Del Rosso JQ, Webster GF. Clinical considerations in the treatment of acne vulgaris
and other inflammatory skin disorders: focus on antibiotic resistance. Cutis. 2007;79 suppl
6:9–25.
24. Gollnick H, Cunliffe W, Berson D, et al. Management of acne: a report from the global alliance
to improve outcomes in acne. J Am Acad Dermatol. 2003;49 suppl 1:S1–38.
25. Leyden JJ. The evolving role of Propionibacterium acnes in acne. Semim Cutan Med Surg.
2001;20:139–43.
26. Shalita AR, Myers JA, Krochman L, et al. The safety and efficacy of clindamycin phosphate
foam 1% versus clindamycin phosphate topical gel 1% for the treatment of acne vulgaris.
J Drugs Dermatol. 2005;4:46–8.
27. Ross JL, Snelling AM, Carneige E, et al. Antibiotic resistant acne lesions: lessons from
Europe. Br J Dermatol. 2003;148:467–78.
28. Dreno B, Reynaud A, Moyse D, et al. Erythromycin-resistance of cutaneous bacterial flora in
acne. Eur J Dermatol. 2001;11:549–53.
29. Esperson F. Resistance to antibiotics used in dermatologic practice. Br J Dermatol. 1998;
139:4–8.
30. Vowels BR, Feingold DS, Sloughfy C, et al. Effects of topical erythromycin on ecology of
aerobic cutaneous bacterial flora. Antimicrob Agents Chemother. 1996;40:2598–604.
31. Naidoo J. Interspecific co-transfer of antibiotic resistance plasmids in staphylococci in vivo.
J Hyg (Lond). 1984;93:59–66.
32. Del Rosso JQ, Leyden JJ, Thiboutot D, et al. Antibiotic use in acne vulgaris and rosacea:
clinical considerations and resistance issues of significance to dermatologist. Cutis. 2008;
82(2S[ii]):5–12.
33. Del Rosso JQ. Topical antibiotics. In: Shalita AR, Del Rosso JQ, Webster GF, editors. Acne
vulgaris. New York, NY: Informa Healthcare; 2011. p. 95–104.
34. Bowe WP, Glick JB, Shalita AR. Solodyn and updates on topical and oral therapies for acne.
Solodyn and updates on topical and oral acne therapies for acne. Curr Dermatol Rep. 2012;
1(3):97–107.
35. Yentzer BA, Ade RA, Fountain JM, et al. Simplifying regimens promotes greater adherence
and outcomes with topical acne medications: a randomized controlled trial. Cutis. 2010;
86:103–8.
36. Thiboutot D. Versatility of azelaic acid 15% gel in the treatment of inflammatory acne vulgaris.
J Drugs Dermatol. 2008;7:13–6.
37. Cunliffe WJ, Hollan KT. Clinical and laboratory studies on treatment with 20% azelaic acid
cream for acne. Acta Derm Venereol Suppl (Stockh). 1989;143:45–8.
38. Katsambas A, Graupe K. Azelaic acid for the treatment of acne vulgaris- a clinical comparison
with vehicle and topical tretinoin. Acta Derm Venereol Suppl (Stockh). 1989;143:35–9.
39. Honzak L, Sentjurc M. Development of liposome encapsulated clindamycin for treatment of
acne vulgaris. Pflugers Arch. 2000;440:44–5.
40. Castro GA, Ferreira LA. Novel vesicular and particular drug delivery systems for topical treat-
ment of acne. Expert Opin Drug Deliv. 2008;5:665–79.
41. Torok HM, Pillai R. Safety and efficacy of micronized tretinoin gel (0.05%) in treating adolescent
acne. J Drug Dermatol. 2011;10:647–51.
42. Villasenor J, Berson DS, Kroshinsky D. Combination therapy. In: Shalita AR, Del Rosso JQ,
Webster GF, editors. Acne vulgaris. New York, NY: Informa Healthcare; 2011. p. 105–12.
Chapter 4
Systemic Therapies for Acne
4.1 Introduction
4.2.1 Macrolides
Erythromycin is not commonly used given the incidence of treatment failure perhaps
due to the development of P. acnes antibiotic resistance or the low level of anti-
inflammatory activity. Erythromycin is usually recommended when tetracycline
derivatives are contraindicated [4]. A common acne dose is 500 mg twice daily. Side
effects may include gastrointestinal (GI) distress, which might be reduced if admin-
istered with food.
Azithromycin has been reported as effective in the treatment of acne, though
there is little consensus for its use and optimum dosing [5–7]. Given its role in the
treatment of respiratory infections and as an alternative treatment for patients
allergic to beta-lactam antibiotics, there is concern with increasing resistance.
Azithromycin does not usually cause GI upset and should be taken on an empty
stomach to enhance GI absorption.
4.2.2 Tetracyclines
The tetracyclines are probably the most commonly used antibiotics, and they have
both antibiotic and anti-inflammatory properties. The newer generation tetracy-
clines (doxycycline and minocycline) are often preferred over tetracycline, due to
decreased rates of resistance and better tolerability [8]. Evidence suggests a reduced
incidence of bacterial resistance with minocycline- and tetracycline-resistant
P. acnes cross-resistant to doxycycline, but sensitive to minocycline [9, 10]. Despite
this, minocycline may be used as first-line therapy due to cost, toxicities, and no
clear evidence of superior efficacy [11, 12].
Tetracycline is typically dosed at 500 mg twice daily, although 250 mg once or
twice daily may also be effective. Patients should be advised to take tetracycline on
an empty stomach as absorption is inhibited by food and with concurrent ingestion
of calcium, magnesium, and aluminum. The absorption of doxycycline and minocy-
cline is not inhibited by food, though concomitant administration with iron supple-
ments may decrease absorption. Doxycycline is commonly prescribed at 50–100 mg
once or twice daily. Although the data is limited, there may be a role for subantimi-
crobial dosing of doxycycline (20 mg twice daily) by which the anti-inflammatory
mechanism is maintained, but the antibacterial properties are lost [13]. Until more
data becomes available, subantimicrobial doses are usually only recommended
when deemed to be in the best interest of the patient. While dosing of minocycline
is usually 50–100 mg twice daily, an extended-release, once daily formulation of
minocycline is FDA-approved at 1 mg/kg/day [14, 15].
The class-related side effects may include GI distress, including esophagitis;
photosensitivity, particularly with doxycycline and tetracycline; and idiopathic
intracranial hypertension (pseudotumor cerebri). To minimize GI side effects, doxy-
cycline and minocycline can be administered with food and water. All tetracyclines
should be avoided in pregnant women and children due to the potential for reduced
4 Systemic Therapies for Acne 29
bone growth and discoloration of developing teeth. Minocycline has been associated
with acute vestibular adverse events; brown, gray, or blue pigmentation of the skin;
lupus-like syndrome; and drug hypersensitivity syndrome [16]. Although it has
been thought that continued use of systemic tetracycline antibiotics results in
colonization with tetracycline-resistant Staphylococcus aureus, this may not be the
case. Evidence suggests that S. aureus may remain sensitive to tetracycline even
after prolonged use [17].
4.3 Isotretinoin
Androgens stimulate sebaceous glands to produce sebum and may also affect
follicular hyperkeratinization. As a result, increased levels of serum androgens and
heightened sensitivity of sebaceous glands to circulating androgens may increase
the risk of acne [34]. Even in the absence of androgen excess, hormonal therapies may
be effective through inhibition of androgen action on the pilosebaceous units [35].
Hormonal therapies are typically reserved for adolescent and adult females with
moderate-to-severe acne and should be considered in those with inflammatory
lesions along jaw line and neck that flare prior to menses, concomitant hirsutism, or
menstrual irregularity.
4.4.2 Spironolactone
Other antiandrogens have been used including cyproterone acetate and flutamide but
only in selected cases [2, 52]. Oral corticosteroids may be used at low doses to suppress
adrenal hyperactivity in individuals with adrenal androgen overproduction, seen in
late-onset CAH. Frequently recommended doses are 2.5–5 mg of prednisone daily or
0.25–0.75 mg of dexamethasone [53]. Careful monitoring is always recommended.
4.5 Conclusion
References
1. Del Rosso JQ, Kim G. Optimizing use of oral antibiotics in acne vulgaris. Dermatol Clin.
2009;27:33–42.
2. Gollnick H, Cunliffe W, Berson D, et al. Management of acne: a report from a Global Alliance
to Improve Outcomes in Acne. J Am Acad Dermatol. 2003;49:S1–37.
3. Leyden JJ. A review of the use of combination therapies for the treatment of acne vulgaris.
J Am Acad Dermatol. 2003;49(3):S200–10.
4. Strauss JS, Krowchuk DP, Leyden JJ, et al. Guidelines of care for acne vulgaris management.
J Am Acad Dermatol. 2007;56:651–63.
5. Rafiei R, Yaghoobi R. Azithromycin versus tetracycline in the treatment of acne vulgaris.
J Dermatolog Treat. 2006;17:217–21.
6. Kus S, Yucelten D, Aytug A. Comparison of efficacy of azithromycin vs. doxycycline in the
treatment of acne vulgaris. Clin Exp Dermatol. 2005;30:215–20.
7. Antonio JR, Pegas JR, Cestari T, Do Nascimento LV. Azithromycin pulses in the treatment of
inflammatory and pustular acne: efficacy, tolerability and safety. J Dermatolog Treat. 2008;
19:210–5.
8. Leyden JJ, Del Rosso JQ, Webster GF. Clinical considerations in the treatment of acne vulgaris
and other inflammatory skin disorders: focus on antibiotic resistance. Cutis. 2007;79:9–25.
9. Eady EA, Cove JH, Holland KT, Cunliffe WJ. Superior antibacterial action and reduced
incidence of bacterial resistance in minocycline compared to tetracycline-treated acne patients.
Br J Dermatol. 1990;122:233–44.
10. Eady EA, Jones CE, Gardner KJ, et al. Tetracycline-resistant propionibacteria from acne
patients are cross-resistant to doxycycline but sensitive to minocycline. Br J Dermatol. 1993;
128:556–60.
11. Garner SE, Eady EA, Popescu C, et al. Minocycline for acne vulgaris: efficacy and safety.
Coch Data Syst Rev. 2003; Issue 1: CD002086.
12. McManus P, Iheanacho I. Don’t use minocycline as first line oral antibiotic in acne. BMJ.
2007;334:154.
13. Skidmore R, Kovach R, Walker C, et al. Effects of subantimicrobial-dose doxycycline in the
treatment of moderate acne. Arch Dermatol. 2003;139:459–64.
4 Systemic Therapies for Acne 33
14. Stewart DM, Torok HM, Weiss JS, et al. Dose-ranging efficacy of new once-daily
extended-release minocycline for acne vulgaris. Cutis. 2006;78:11–20.
15. Fleischer Jr AB, Dinehart S, Stough D, et al. Safety and efficacy of a new extended-release
formulation of minocycline for acne vulgaris. Cutis. 2006;78:21–31.
16. Sturkenboom MC, Meier CR, Jick H, Striker BH. Minocycline and lupus-like syndrome in
acne patients. Arch Intern Med. 1999;159:493–7.
17. Fanelli M, Kupperman E, Lautenbach E, Edelstein PH, Margolis DJ. Antibiotics, acne, and
staphylococcus aureus colonization. Arch Dermatol. 2011;147:917–21.
18. Bhambri S, Del Rosso JQ, Desai A. Oral trimethoprim/sulfamethoxazole in the treatment of
acne vulgaris. Cutis. 2007;79(6):430–4.
19. Amnesteem® (Isotretinoin Capsules USP) [package insert]. Morgantown, WV: Mylan
Pharmaceuticals, Inc. 2010.
20. Strauss JS, Rapini RP, Shalita AR, et al. Isotretinoin therapy for acne: results of a multicenter
dose-response study. J Am Acad Dermatol. 1984;10:490–6.
21. Ellis CN, Krach KJ. Uses and complications of isotretinoin therapy. J Am Acad Dermatol.
2001;45(5):S150–7.
22. White GM, Chen W, Yao J, Wolde-Tsadik G. Recurrence rates after the first course of
isotretinoin. Arch Dermatol. 1998;134:376–8.
23. Azoulay L, Oraichi D, Bérard A. Isotretinoin therapy and the incidence of acne relapse: a
nested case-control study. Br J Dermatol. 2007;157(6):1240–8.
24. Bershad S, Rubinstein A, Paterniti JR, et al. Changes in plasma lipids and lipoproteins during
isotretinoin therapy for acne. N Engl J Med. 1985;313:981–5.
25. Lammer EJ, Chen DT, Hoar RM, et al. Retinoic acid embryopathy. N Engl J Med. 1985;
313:837–41.
26. Adams J, Lammer EJ. Neurobehavioral teratology of isotretinoin. Reprod Toxicol. 1993;
7:175–7.
27. Margolis DJ, Attie M, Leyden JJ. Effects of isotretinoin on bone mineralization during routine
therapy with isotretinoin therapy for acne vulgaris. Arch Dermatol. 1996;132:769–74.
28. Reddy D, Siegel CA, Sands BE, et al. Possible association between isotretinoin and inflamma-
tory bowel disease. Am J Gastroenterol. 2006;101:1569–73.
29. Azoulay L, Blais L, Koren G, et al. Isotretinoin and the risk of depression in patients with acne
vulgaris: a case-crossover study. J Clin Psychiatry. 2008;69:526–32.
30. Crockett SD, Gulati A, Sandler RS, et al. A causal association between isotretinoin and
inflammatory bowel disease has yet to be established. Am J Gastroenterol. 2009;104:
2387–93.
31. Bigby M. Does isotretinoin increase the risk of depression? Arch Dermatol. 2008;144:
1197–9.
32. Marqueling AL, Zane LT. Depression and suicidal behavior in acne patients treated with
isotretinoin: a systematic review. Semin Cutan Med Surg. 2007;26:210–20.
33. Chia CY, Lane W, Chibnall J, et al. Isotretinoin therapy and mood changes in adolescents with
moderate to severe acne: a cohort study. Arch Dermatol. 2005;141:557–60.
34. Lolis MS, Bowe WP, Shalita AR. Acne and systemic disease. Med Clin North Am. 2009;
93:1161–81.
35. Kelekci KH, Kelekci S, Incki K, et al. Ovarian morphology and prevalence of polycystic ovary
syndrome in reproductive aged women with or without mild acne. Int J Dermatol. 2010;
49:775–9.
36. Haider A, Shaw JC. Treatment of acne vulgaris. JAMA. 2004;292:726–35.
37. Junkins-Hopkins JM. Hormone therapy for acne. J Am Acad Dermatol. 2010;62(3):486–8.
38. Arowojolu AO, Gallo MF, Lopez LM, Grimes DA, Garner SE. Combined oral contraceptive
pills for treatment of acne. Coch Data Syst Rev. 2009; Issue 3: CD004425.
39. Thorneycroft H, Gollnick H, Schellschmidt I. Superiority of a combined contraceptive
containing drospirenone to a triphasic preparation containing norgestimate in acne treatment.
Cutis. 2004;74:123–30.
40. Yaz® (Drospirenone/Ethinyl estradiol) [package insert]. Wayne, NJ: Bayer HealthCare
Pharmaceuticals, Inc.: 2011.
34 M.K. Garshick et al.
41. Castelli WP. Cardiovascular disease: pathogenesis, epidemiology, and risk among users of oral
contraceptives who smoke. Am J Obstet Gynecol. 1999;180:S349–56.
42. Helms SE, Bredle DL, Zajic J, Jatjoura D, Brodell RT, Krishnarao I. Oral contraceptive failure
rates and oral antibiotics. J Am Acad Dermatol. 1997;36:705–10.
43. Dickinson BD, Altman RD, Nielsen NH, Sterling ML, Council on Scientific Affairs, American
Medical Association. Interactions between oral contraceptives and antibiotics. Obstet Gynecol.
2001;98:853–60.
44. Berenson AB, Odom SD, Breitkopf CR, Rahman M. Physiologic and psychologic symptoms
associated with use of injectable contraception and 20 microg oral contraceptive pills. Am J
Obstet Gynecol. 2008;199:351.e1–e12.
45. Goodfellow A, Alaghband-Zadeh J, Carter G, et al. Oral spironolactone improves acne vulgaris
and reduces sebum excretion. Br J Dermatol. 1984;111:209–14.
46. Brown J, Farquhar C, Lee O, et al. Spironolactone versus placebo or in combination with
steroids for hirsutism and/or acne. Coch Data Syst Rev. 2009; Issue 2:CD000194.
47. Saint-Jean M, Ballanger F, Nguyen J, Khammari A, Dréno B. Importance of spironolactone in
the treatment of acne in adult women. J Eur Acad Dermatol Venereol. 2010;25:1480–1.
doi:10.1111/j.1468-3083.2010.03926.x.
48. Muhlemann MF, Carter GD, Cream JJ, Wise P. Oral spironolactone: an effective treatment for
acne vulgaris in women. Br J Dermatol. 1986;115:227–32.
49. Burke BM, Cunliffe WJ. Oral spironolactone therapy for female patients with acne, hirsutism
or androgenic alopecia. Br J Dermatol. 1985;112:124–5.
50. Shaw JC. Low-dose adjunctive spironolactone in the treatment of acne in women: a retrospec-
tive analysis of 85 consecutively treated patients. J Am Acad Dermatol. 2000;43:498–502.
51. Katsambas AD, Dessinioti C. Hormonal therapy for acne: why not as first line therapy? Facts
and controversies. Clin Dermatol. 2010;28:17–23.
52. Van Wayjen R, van den Ende A. Experience in the long term treatment of patients with
hirsutism and/or acne with cyproterone acetate-containing preparations: efficacy, metabolic,
and endocrine effects. Exp Clin Endocrinol Diabetes. 1995;103:241–51.
53. Thiboutot D. Acne: hormonal concepts and therapy. Clin Dermatol. 2004;22:419–28.
Chapter 5
Laser and Light Based Therapies for Acne
5.1 Introduction
Conventional topical and systemic therapeutics are effective for treating acne vulgaris,
but are not perfect solutions. Topical salicylic acid, benzoyl peroxide, and retinoid
preparations can be irritating and present challenges with compliance. Systemic
antibiotics also have issues with tolerability and increasing bacterial resistance, and
isotretinoin has attracted negative attention due to its purported links to inflamma-
tory bowel disease and depression. Laser and light-based treatments can potentially
offer a viable adjunct or even alternative to these conventional options.
There is a dearth of large, randomized controlled trials of lasers and lights for
acne [3]. One exception was an industry-sponsored evaluation of 266 severe acne
patients treated with blue light (417 ± 5 nm) and 20 % aminolevulinic acid (ALA)
(BLU-U® and Levulan® Kerastick, DUSA Pharmaceuticals, Inc.) versus blue light
and vehicle alone [4]. In this study, vehicle or ALA was applied for 45 min incuba-
tion followed by 16 min and 40 s of blue light (10 J/cm2 total dose), one treatment
every 3 weeks for four treatments. Interestingly the investigators found a decrease
in inflammatory lesions of 37.5 % for the ALA group versus 41.7 % reduction in the
vehicle group. Noninflammatory lesion counts were not performed. The study
indicates that blue light alone is as effective as blue light plus ALA.
Blue light penetrates to a depth of approximately 90–150 μm. Although it is
unlikely that significant amount of this wavelength reaches the sebaceous gland,
exposure to blue light at a wavelength of 405–420 nm destroys P. acnes. Furthermore,
blue light may inhibit the production of interleukin-1 alpha and intercellular adhesion
molecule-1 [5], as well as tumor necrosis factor alpha and matrix metalloproteinase 2,
all inflammatory mediators implicated in the pathogenesis of acne [6].
Within the visible light spectrum, a longer wavelength of light correlates with
deeper skin penetration, as there is less melanin absorption and less scattering by
connective tissue. Red light penetrates deeper than blue light, to approximately
2 mm of depth. It effectively reaches the sebaceous glands and activates porphyrins
via the Q band at 630 nm. One study evaluated 21 patients treated with 3 h incuba-
tion of 16.8 % methylaminolevulinic acid (Metvix®, Photocure) followed by 9 min
of 630 nm red light (Aktilite® CL 128, Photocure), for a total dose of 37 J/cm2 [7].
After two treatments spaced 2 weeks apart, investigators noted a 68 % reduction in
inflammatory lesions and no change in the 15 control patients. There was no reduc-
tion in noninflammatory lesions in either group. One study found that the combina-
tion of blue and red light was superior to either blue light or 5 % benzoyl peroxide
alone in reduction of the number of inflammatory acne lesions [8].
Limitations of using an exogenous photosensitizer include intra-procedure pain
and post-procedure erythema, edema, and blistering. Of note patients must avoid
direct sunlight exposure for 48 h after the procedure, which can be challenging in
the teenage acne population.
The pulsed dye laser (PDL, 585–595 nm yellow light), potassium titanyl
phosphate (KTP, 532 nm green light), and broadband intense pulsed light (IPL,
500–1,200 nm) with cutoff filters to employ predominately short wavelengths all act
on acne by activating endogenous P. acnes porphyrins and reducing sebaceous
gland function. A 2003 study found a significant reduction of inflammatory acne
lesions (49 %) in 31 patients receiving a single PDL treatment versus 10 patients
receiving “sham” treatment (10 %) [9]. However, a different, split-face trial of 40
patients receiving 1–2 sub-purpuric PDL treatments found no significant difference
in mean papule count versus the untreated control side [10]. A study of the KTP
laser found a 35.9 % reduction in inflammatory lesions 1 month after 4 biweekly
treatments [11]. Anecdotally the authors find these devices to be more useful
for improving the appearance of erythematous “stains” and scars which routinely
appear following the resolution of acne lesions.
5 Laser and Light Based Therapies for Acne 37
Mid-infrared light has also been utilized for acne treatment due to its purported
ability to shrink sebaceous glands and/or alter their secretion [12]. One study of 32
acne patients treated in a split-face fashion with 3 monthly 1,450 nm diode laser
treatments found an equal reduction in inflammatory lesions on both the treatment
and control side [13]. The authors state that this may present evidence of a systemic
anti-inflammatory effect of the laser, though the mechanism is unclear. Another
evaluation of 20 patients treated with a 1,540 nm erbium to glass nonablative
fractional laser monthly for 4 months found that acne lesions improved in 85 % of
patients and 80 % noted a reduction in sebum production [14].
Improvement of comedonal acne with lasers and lights has been a challenge to
achieve; however the newly developed photopneumatic therapy devices show poten-
tial. Photopneumatic therapy combines vacuum suction to mechanically clear pores
and concomitant broadband pulsed light. Cutoff filters limit the emission spectrum to
500–1,200 nm, wavelengths that activate endogenous porphyrins to destroy P. acnes,
as well as short-wavelength visible light that reduces perilesional erythema.
Published data supporting the efficacy of photopneumatic therapy is limited to
small, uncontrolled trials. A 2008 study of 11 patients receiving photopneumatic
therapy every 3 weeks for four treatments found a statistically significant reduction
in both inflammatory (78.8 %) and noninflammatory (57.8 %) lesions at the 3-month
follow-up visit post-therapy. Nine of the 11 patients were moderately to very satis-
fied with their results [15]. Other similar studies corroborate these findings [16, 17].
In the authors’ experience, photopneumatic therapy is an effective adjunctive
acne treatment.
Each week, the subject administered two alternate exposures: 20 min for blue and
30 min for red light, with 2–3-day intervals between the alternating light treatments.
Treatment was associated with statistically significant reduction from baseline in
the inflammatory lesion count of 69 % and reduction of 12 % of noninflammatory
lesions [19]. It is important to note that treatment time (20 min for blue light and
30 min for red) for the study covered an area of 5 cm × 6 cm. Consequently, a con-
siderably longer time would be required for full-face treatment. Therefore OTC
acne devices are best suited for spot treatment and are not routinely recommended
by the authors for full-face treatment of acne.
The future of laser and light therapy for acne is based on the foundation of modern
laser therapy—the principle of selective photothermolysis [20]. None of the afore-
mentioned laser and light sources emit wavelengths of light that are preferentially
absorbed by the sebaceous gland. One approach recently presented involved the
application of gold-coated nanoshells specifically constructed to absorb light at
800 nm. After application to periauricular human skin and subsequent treatment
with an 800 nm diode laser, histologic examination showed partial sebaceous gland
destruction without damage to the surrounding epidermis or dermis [21]. A recent
work investigated the optimal wavelength to target the sebaceous gland found that
sebum preferentially absorbs 1,210 and 1,720 nm light [22]. The investigators also
estimated that the optimal pulse duration of laser light delivered to confine heat to
the target sebaceous gland (thermal relaxation time) to be approximately 100 ms.
With these working parameters devices are in development, and perhaps 1 day laser
practitioners will be able to satisfyingly specifically target acne the way we currently
treat pigmented lesions. Until that day, lasers and lights can provide useful adjuncts
to conventional first-line acne therapy.
References
1. Sakomoto FH, Lopes JD, Anderson RR. Photodynamic therapy for acne vulgaris: a critical
review from basics to clinical practice. Part I. J Am Acad Dermatol. 2010;63:183–93.
2. Sakomoto FH, Torezan L, Anderson RR. Photodynamic therapy for acne vulgaris: a critical
review from basics to clinical practice. Part II. J Am Acad Dermatol. 2010;63:195–211.
3. Hamilton FL, Car J, Lyons M, et al. Laser and other light therapies for the treatment of acne
vulgaris: systematic review. Br J Dermatol. 2009;160:1273–85.
4. http://www.clinicaltrials.gov/ct2/show/results/NCT00706433?term=DUSA+Pharmaceuticals
&rank=1
5. Shnitkind E, Yaping E, Geen S, et al. Anti-inflammatory properties of narrow-band blue light.
J Drugs Dermatol. 2006;5(7):605–10.
6. Fan X, Xing Y-Z, Liu L-H, et al. Effects of 420-nm intense pulsed light in an acne animal
model. J Eur Acad Dermatol Venereol. 2012; doi: 10.1111/j.1468-3083.2012.04487.x
5 Laser and Light Based Therapies for Acne 39
7. Wiegell SR, Wulf HC. Photodynamic therapy of acne vulgaris using methyl aminolevulinic
acid: a blinded, randomized, controlled trial. Br J Dermatol. 2006;154:969–76.
8. Papageorgiou P, Katsambas A, Chu A. Phototherapy with blue (415 nm) and red (660 nm)
light in the treatment of acne vulgaris. Br J Dermatol. 2000;142:973–8.
9. Seaton ED, Charakida A, Mouser PE, et al. Pulsed-dye laser treatment for inflammatory acne
vulgaris: randomised controlled trial. Lancet. 2003;362:1347–52.
10. Orringer JS, Kang S, Hamilton T, et al. Treatment of acne vulgaris with a pulsed dye laser:
a randomized controlled trial. JAMA. 2004;291:2834–9.
11. Bowes L, Manstein D, Anderson R. Effects of 532 nm KTP laser on acne and sebaceous
glands. Laser Med Sci. 2003;18 Suppl 1:S6–7.
12. Perez-Maldonado A, Runger TM, Kreju-Papa N. The 1,450-nm diode laser reduces sebum
production in facial skin: a possible mode of action of its effectiveness for the treatment of
acne vulgaris. Lasers Surg Med. 2007;39:189–92.
13. Darne S, Hiscutt EL, Seukeran DC. Evaluation of the clinical efficacy of the 1450 nm laser in
acne vulgaris: a randomized split-face, investigator-blinded clinical trial. Br J Dermatol. 2011;
165:1256–62.
14. Isarria MJ, Cornejo P, Munos E, et al. Evaluation of clinical improvement in acne scars and
active acne in patients treated with the 1540-nm non-ablative fractional laser. J Drugs Dermatol.
2011;10(8):907–12.
15. Gold M, Biron J. Treatment of acne with pneumatic energy and broadband light. J Drugs
Dermatol. 2008;7(7):639–42.
16. Shamban AT, Enokibori M, Narurkar V, et al. Photopneumatic technology for the treatment of
acne vulgaris. J Drugs Dermatol. 2008;7(2):139–45.
17. Wanitphakdeedcha R, Tanzi E, Alster T. Photopneumatic therapy for the treatment of acne.
J Drugs Dermatol. 2009;8(3):239–41.
18. Metelitsa AI, Green JB. Home-use laser and light devices for the skin—an update. Semin
Cutan Med Surg. 2011;30:144–7.
19. Sadick NS. Handheld LED, array device in the treatment of acne vulgaris. J Drugs Dermatol.
2008;7:347–50.
20. Anderson RR, Parrish JA. Selective photothermolysis: precise microsurgery by selective
absorption of pulsed radiation. Science. 1983;220(4596):524–7.
21. Kauvar A, Lloyd J, Cheung W, et al. Selective photothermolysis of the sebaceous follicle with
gold-coated nanoshells for treatment of acne. Presented at 2012 American Society for Laser
Medicine and Surgery Annual Meeting, Orlando, Florida.
22. Sakamoto FH, Doukas AG, Farinelli WA, et al. Selective photothermolysis to target sebaceous
glands: theoretical estimation of parameters and preliminary results using a free electron laser.
Lasers Surg Med. 2012;44:175–83.
Part II
Infectious Diseases Mimicking
Acne Vulgaris
Chapter 6
Bacterial Folliculitis
6.1 Introduction
6.2 Background
Bacterial folliculitis affects both males and females in children and adults [1–3].
An accurate incidence is difficult to determine as many affected patients in the general
population do not seek medical attention for this condition. Predisposing factors include
occlusion, maceration, hyperhydration, nasal carriage of Staphylococcus aureus
(S. aureus), application of topical steroids, and exposure to oils and certain other chem-
icals, all of which increase bacterial colonization on the skin [1–3]. Environments that
promote occlusion or moisture such as hot or humid weather or prolonged exposure to
heated water (i.e., hot tub, heated swimming pool) are predisposing factors as well.
Fig. 6.1 Follicular-based, erythematous papules where pustules had initially existed and subse-
quently ruptured (Photo credit: Joshua A. Zeichner, M.D.)
6 Bacterial Folliculitis 45
with a central pustule that becomes necrotic within a few days [1]. Furuncles are
more common in young adult males and favor the face, back of neck, axillae, breast,
buttocks, and thighs [1]. Predisposing factors for furunculosis include diabetes
mellitus, HIV infection, malnutrition, crowded living conditions such as incarcera-
tion, and chronic staphylococcus colonization [1].
Bacterial sycosis is another form of deep folliculitis in which a subacute or
chronic staphylococcal infection involves the entire hair follicle [1, 2]. This condi-
tion typically occurs in postpubertal males, often in the third to fourth decade, and
frequently affects the beard region [1, 2]. Early in the clinical course, the condition
presents with an edematous, inflammatory papule or pustule in the beard region
with a burning sensation. This is followed by development of numerous papules and
pustules involving the surrounding follicles that then coalesce into plaques studded
with pustules [1]. In severe or chronic cases persisting for years, the follicles may
be destroyed by scarring or a granulomatous appearance may occur. This has been
termed lupoid sycosis for its morphologic resemblance to lupus vulgaris [1, 2].
The distribution of folliculitis favors hair-bearing areas, especially the scalp and
beard region, but also the upper trunk, buttocks, thighs, axillae, and groin [2]. Areas of
involvement vary among different populations. In infants and children, infection favors
the face, buttocks, and axillae [1]. Folliculitis in adolescent females is commonly
located on the legs, while it affects the flexural areas in adolescent males [1].
6.4 Work-Up
6.5 Treatment
axillae, groin, anus, and inframammary areas as well [3], while others recommend
repeating treatment for 5 days each month to reduce recurrence [8]. Topical fusidic
acid and retapamulin can also be used to eradicate MRSA colonization [6, 8].
References
7.1 Introduction
7.2 Background
7.2.1 Pathophysiology
7.2.2 Epidemiology
Fig. 7.1 Monomorphic follicular papules in the infranasal area and chin of a teenage boy with
acne who developed gram-negative folliculitis after chronic treatment with doxycycline for acne
(Photo credit: Joshua A. Zeichner, M.D.)
treatment, and isolation of gram-negative rods from pustules of facial skin and
mucous membranes of the nose [2] (Fig. 7.1).
Two main subtypes of gram-negative folliculitis have been described [5]. Type I
is much more common and seen in approximately 80 % of patients. These patients
develop papules and papulopustules flaring out from the nares and involving the
infranasal and perioral skin. In type II, seen in approximately 20 % of patients, deep
fluctuant cystic lesions resembling acne conglobata are the key presenting features.
The distribution of lesions in both types extends from the infranasal area to the chin
and cheeks.
Type I lesions are most commonly associated with a lactose-fermenting, gram-
negative rods including Klebsiella, Escherichia, and Serratia species [5]. However,
cases associated with Citrobacter, which is another organism of the Enterobacteriaceae
family, have been reported [6, 7]. Proteus species is associated with type II lesions;
these organisms are motile granting them the ability to invade more deeply, producing
large suppurative abscesses resulting in deeper cystic lesions [5].
Gram-negative folliculitis has also been described in patients following recurrent
staphylococcal pyoderma [6] as well as after long-term topical antibacterial therapy
[8]. Batholow and Maibach described a patient with acne who was treated with topi-
cal clindamycin followed by benzoyl peroxide and erythromycin and subsequently
developed gram-negative folliculitis secondary to E. coli [8].
7.4 Work-Up
folliculitis. Swabs for bacteriological analysis should be taken from pustules and
from the nasal mucosa. The pustule that is sampled should be fresh and preferably on
an erythematous base. Gram-negative organisms are sensitive to desiccation and thus
samples must be taken rapidly and transported to the laboratory as soon as possible.
It is recommended to culture pustules in any patient with acne who is in their late
teens or older and develops a pustular eruption while on antibiotics. It is important
to note that gram-negative organisms are not recoverable from every pustule.
Selective medium-containing dyes such as methylene blue allow selective growth of
gram-negative organisms while inhibiting the growth of gram-positive organisms.
Inadequate sampling, dried-out swabs, and long delay between culturing the
pustules/nasal mucosa and the arrival of the specimen in the laboratory may lead to
an underestimation of the frequency of gram-negative folliculitis in patients with
acne and/or rosacea [9].
7.5 Treatment
References
8.1 Introduction
8.2 Background
8.2.1 Pathophysiology
Pseudomonas folliculitis, but other serotypes have also been reported. Serotype
O:11 is thought to be more invasive and better adapted to survive in halogenated
water than the other serotypes, explaining the prevalence of this serotype in hot-tub
folliculitis [8]. As water temperatures rise, free chlorine levels in the water fall, even
though the total chlorine levels appear adequate. This provides an ideal environment
in which the bacteria can proliferate. Hot water, high pH (>7.8), and low chlorine
level (<0.5 mg/L) all predispose to an increased risk for infection.
8.2.2 Epidemiology
The onset of the rash of hot-tub folliculitis is usually 1–4 days after exposure to
contaminated water. However, skin lesions can develop as early as 8 h after exposure
and as late as 14 days later [9]. Most lesions begin as pruritic, follicular, erythema-
tous macules that progress to papules and pustules. Individual lesions range in size
from 2 to 10 mm in diameter and often demonstrate a central pinpoint vesiculopus-
tule. The rash is most prevalent in the intertriginous areas or under bathing suits
secondary to compression of contaminated water in those areas. It can also be seen
on the trunk and extremities, where the skin is exposed to contaminated water. The
face, neck, palms, and soles are usually spared [10]. Other associated complaints are
minimal and include earache, sore throat, fever, and malaise. Systemic infections
are rare. The rash usually clears spontaneously in 7–14 days without therapy, rarely
recurs, and heals without scarring. Sometimes, it may cause desquamation or leave
behind post inflammatory hyperpigmented macules which can resolve over time.
The rash is not unique in appearance and is most often confused with insect bites.
8.4 Work-Up
8.5 Treatment
The folliculitis usually self-resolves within 7–14 days without therapeutic inter-
vention. However, in patients who present with fever, constitutional symptoms, or
prolonged disease, a third-generation cephalosporin or fluoroquinolone like cipro-
floxacin or ofloxacin can be useful [11]. Proper maintenance and chlorination of
pools, hot tubs, whirlpools, and spas are essential to decrease the population of
Pseudomonas species. Showering after exposure to contaminated water does not
seem to prevent Pseudomonas folliculitis.
References
1. Gregory DW, Schaffner W. Pseudomonas infections associated with hot tubs and other
environments. Infect Dis Clin North Am. 1987;1(3):635–48.
2. Highsmith AK, Le PN, Khabbaz RF, Munn VP. Characteristics of Pseudomonas aeruginosa
isolated from whirlpools and bathers. Infect Control. 1985;6(10):407–12.
3. McCausland WJ. Pseudomonas aeruginosa rash associated with whirlpool. JAMA. 1976;
236(22):2490–1.
4. Price D, Ahearn DG. Incidence and persistence of Pseudomonas aeruginosa in whirlpools.
J Clin Microbiol. 1988;26(9):1650–4.
5. Ratnam S, Hogan K, March SB, Butler RW. Whirlpool-associated folliculitis caused
by Pseudomonas aeruginosa: report of an outbreak and review. J Clin Microbiol. 1986;
23(3):655–9.
6. Rose HD, Franson TR, Sheth NK, Chusid MJ, Macher AM, Zeirdt CH. Pseudomonas
pneumonia associated with use of a home whirlpool spa. JAMA. 1983;250(15):2027–9.
7. Lacour JP, el Baze P, Castanet J, Dubois D, Poudenx M, Ortonne JP. Diving suit dermatitis
caused by Pseudomonas aeruginosa: two cases. J Am Acad Dermatol. 1994;31(6):1055–6.
8. Maniatis AN, Karkavitsas C, Maniatis NA, Tsiftsakis E, Genimata V, Legakis NJ. Pseudomonas
aeruginosa folliculitis due to non-O:11 serogroups: acquisition through use of contaminated
synthetic sponges. Clin Infect Dis. 1995;21(2):437–9.
9. Berger RS, Seifert MR. Whirlpool folliculitis: a review of its cause, treatment, and prevention.
Cutis. 1990;45(2):97–8.
10. James W, Berger T, Elston D. Andrews’ clinical dermatology. 9th ed. Elsevier, Philadelphia; 2000.
11. Böni R, Nehrhoff B. Treatment of gram-negative folliculitis in patients with acne. Am J Clin
Dermatol. 2003;4(4):273–6.
Chapter 9
Malassezia Folliculitis
9.1 Introduction
9.2 Background
The Malassezia genus of yeast is considered to be normal skin flora and is found in
the stratum corneum and hair follicles of up to 90 % of individuals [1]. This lipo-
philic dimorphic yeast can be pathogenic and has been implicated in common skin
disorders such as seborrheic dermatitis and pityriasis versicolor [2]. Malassezia
folliculitis was first described by Weary et al. in 1969 as an acneiform eruption in
the setting of antibiotic therapy with tetracyclines [3]. It is characterized as an
invasion of hair follicles by large numbers of Malassezia yeast. Malassezia furfur,
9.4 Work-Up
Malassezia folliculitis should be suspected in patients who have papular lesions on the
trunk and upper arms unresponsive to acne treatments, in those who have taken broad-
spectrum antibiotics, or who are immunocompromised. Direct microscopic examina-
tion is preferable to culture since Malassezia is normal skin flora. The organisms can be
seen on direct microscopic examination at high power (40×) when mounted with
10–15 % potassium hydroxide [23]. Examination of the follicular contents after
62 P.K. Farris and A. Murina
extraction with a comedo extractor may provide superior results to simple skin scrapings
[7]. Biopsy specimens will show abundant round budding yeasts with occasional hyphae
(Fig. 9.2). PAS stain can further highlight the organisms (Fig. 9.3). An inflammatory
infiltrate surrounding the hair follicle consisting of lymphocytes, histiocytes, and
neutrophils is seen on histology [24, 25]. This inflammatory response may be due in part
to the fact that Malassezia yeasts have been shown to hydrolyze triglycerides into free
fatty acids [21, 23, 24]. Hair follicles tend to be dilated, full of keratinous material, and
may have focal rupture. The diagnosis can be made from clinical presentation, a positive
direct microscopic examination, histopathology demonstrating yeast engorged hair
follicles, and a positive response to antifungal treatment.
9.5 Treatment
9.6 Conclusion
References
1. Roberts SO. Pityrosporum orbiculare: incidence and distribution on clinically normal skin.
Br J Dermatol. 1969;81:264–9.
2. Gupta AK, Batra R, Bluhm R, Boekhout T, Dawson TL. Skin diseases associated with malas-
sezia species. J Am Acad Dermatol. 2004;51(5):785–98.
3. Weary PE, Russell CM, Butler HK, Hsu YT. Acneiform eruption resulting from antibiotic
administration. Arch Dermatol. 1969;100(2):179–83.
4. Tragiannidis A, Bisping G, Koehler G, Groll AH. Minireview: Malassezia infections in immu-
nocompromised patients. Mycoses. 2009;53:187–95.
5. Hill MK, Goodfield MJ, Rodgers FG, Crowley JL, Saihan EM. Skin surface electron micros-
copy in Pityrosporum folliculitis: the role of follicular occlusion in disease and the response to
oral ketoconazole. Arch Dermatol. 1990;126:1071–4.
6. Sweeney K. Pityrosporum folliculitis: diagnosis and management in 6 female adolescents with
acne vulgaris. Arch Pediatr Adolesc Med. 2005;159(1):64–7.
7. Yu H, Lee S, Son SJ, Kim YS, Yang HY, Kim JH. Steroid acne vs. pityrosporum folliculitis:
the incidence of Pityrosporum ovale and the effect of antifungal drugs in steroid acne. Int
J Dermatol. 1998;37(10):772–7.
8. Ferrandiz C, Ribera M, Barranco JC, Clotet B, Lorenzo JC. Eosinophilic pustular folliculitis
in patients with acquired immunodeficiency syndrome. Int J Dermatol. 1992;31(3):193–5.
9. Nervi SJ, Schwartz RA, Dmochowski M. Eosinophilic pustular folliculitis: a 40 year retrospect.
J Am Acad Dermatol. 2006;855(2):285–9.
10. Cholongitas E, Pipili C, Ioannidou D. Malassezia folliculitis presented as acneiform eruption
after cetuximab administration. J Drugs Dermatol. 2009;8(3):274–5.
11. Duvic M. EGFR inhibitor-associated acneiform folliculitis. Am J Clin Dermatol. 2008;
9(5):285–94.
12. Archer-Dubon C, Icaza-Chivez M, Orozco-Topete R, Reyes E, Baez-Martinez R, de León SP.
An epidemic outbreak of malassezia folliculitis in three adult patients in an intensive care unit:
a previously unrecognized nosocomial infection. Int J Dermatol. 1999;38(6):453–6.
13. Yohn JJ, Lucas J, Camisa C. Malassezia folliculitis in immunocompromised patients. Cutis.
1985;35(6):536–8.
14. Fearfield LA, Rowe A, Francis N, Bunker CB, Staughton RC. Itchy folliculitis and human
immunodeficiency virus infection: clinicopathological and immunological features, pathogen-
esis and treatment. Br J Dermatol. 1999;141(1):3–11.
15. Helm KF, Lookingbill DP. Pityrosporum folliculitis and severe pruritus in two patients with
Hodgkin’s disease. Arch Dermatol. 1993;129(3):380–1.
16. Rhie S, Turcios R, Buckley H, Suh B. Clinical features and treatment of malassezia folliculitis
with fluconazole in orthotopic heart transplant recipients. J Heart Lung Transplant. 2000;
19(2):215–9.
17. Koranda FC, Dehmel EM, Kahn G, Penn I. Cutaneous complications in immunosuppressed
renal homograft recipients. JAMA. 1974;229(4):419–24.
9 Malassezia Folliculitis 65
18. Bufill JA, Lum LG, Caya JG, Chitambar CR, Ritch PS, Anderson T, et al. Pityrosporum
folliculitis after bone marrow transplantation. Ann Intern Med. 1988;108(4):560.
19. Gueho EF, Faegemann J, Lyman C, Anaissie EJ. Malassezia and Trichosporin: two emerging
pathogenic basidiomycetous yeast-like fungi. J Med Vet Mycol. 1994;32:367–78.
20. Bäck O, Faergemann J, Hörnqvist R. Pityrosporum folliculitis: a common disease of the young
and middle-aged. J Am Acad Dermatol. 1985;12(1, Part 1):56–61.
21. Faergemann J, Johansson S, Back O, Scheynius A. An immunologic and cultural study of
pityrosporum folliculitis. J Am Acad Dermatol. 1986;14(3):429–33.
22. Jacinto-Jamora S, Tamesis J, Katigbak ML. Pityrosporum folliculitis in the philippines: diagnosis,
prevalence, and management. J Am Acad Dermatol. 1991;24(5 Pt 1):693–6.
23. Potter BS, Burgoon Jr CF, Johnson WC. Pityrosporum folliculitis: report of seven cases and
review of the pityrosporum organism relative to cutaneous disease. Arch Dermatol. 1973;
107(3):388–91.
24. Faergemann J, Bergbrant I, Dohsé M, Scott A, Westgate G. Seborrhoeic dermatitis and
pityrosporum (malassezia) folliculitis: characterization of inflammatory cells and mediators in
the skin by immunohistochemistry. Br J Dermatol. 2001;144(3):549–56.
25. Sina B, Kauffman CL, Samorodin CS. Intrafollicular mucin deposits in pityrosporum folliculitis.
J Am Acad Dermatol. 1995;32(5 Pt 1):807–9.
26. Levy A, Feuilhade de Chauvin M, Dubertret L, Morel P, Flageul B. Malassezia folliculitis:
characteristics and therapeutic response in 26 patients. Ann Dermatol Venereol. 2007;
134:823–8.
27. Parsad D, Saini R, Negi KS. Short-term treatment of pityrosporum folliculitis: a double blind
placebo-controlled study. J Eur Acad Dermatol Venereol. 1998;11(2):188–90.
28. Faergemann J. Treatment of pityriasis versicolor with a single dose of fluconazole. Acta Derm
Venereol. 1992;72(1):74–5.
29. Friedman SJ. Pityrosporum folliculitis: treatment with isotretinoin. J Am Acad Dermatol.
1987;16:632–3.
30. Lee J, Kim B, Kim M. Photodynamic therapy: new treatment for recalcitrant malassezia
folliculitis. Lasers Surg Med. 2010;42(2):192–6.
Chapter 10
Tinea Barbae
10.1 Introduction
10.2 Background
Tinea barbae is most frequently seen in tropical countries with high temperatures
and high humidity. The incidence has decreased in recent years due to the increased
availability of disposal razors, as the infection was regularly transmitted by barbers
using unsanitary razors. Today, tinea barbae is more often found among rural
inhabitants exposed to zoophilic dermatophytes from sources such as cattle, horses,
cats, and dogs (Table 10.1). The species commonly isolated include T. mentagrophytes
and T. verrucosum. Microsporum canis and T. mentagrophytes var. erinacei are
less common. In certain geographic regions, other anthropophilic dermatophytes,
Tinea barbae, as the name suggests, is a dermatophyte infection of the bearded areas
of the face and neck of adult males [1]. The clinical presentation maybe either one
of two types: inflammatory or noninflammatory, depending on the infecting fungus
and the patient’s immune response. The zoophilic dermatophyte infections typically
cause severe, deep inflammation, characterized commonly by inflammatory nodules
and multiple follicular pustules [4]. Abscesses, sinus tracts, kerion-like plaques, and
bacterial superinfections may also occur. Furthermore, patients may have constitu-
tional symptoms such as malaise and lymphadenopathy. The involved hairs can
become loose or broken, and this variety of tinea barbae may lead to permanent,
scarring alopecia [2].
The second type of tinea barbae is clinically similar to tinea corporis, as it is
superficial and less inflammatory and is usually caused by T. rubrum. The charac-
teristic lesion is an annular, erythematous plaque with an active, papular border
(Fig. 10.1). Vesicles and overlying crust are other features that are not uncommon.
Reversible alopecia in the center of the lesion may also be present [5].
A third variant of chronic tinea barbae may present similar to sycosis (inflamma-
tion of the hair follicles), with papules and pustules in a follicular distribution [4].
The differential diagnosis of tinea barbae includes bacterial folliculitis, pseudofol-
liculitis barbae, atypical rosacea, periorbital dermatitis, acne vulgaris, cervicofacial
actinomycosis, and viral infections such as herpes simplex or herpes zoster [6].
10.4 Work-up
Fig. 10.1 Scaly papules affecting the skin within the beard of the anterior neck of a young man.
Fungal culture revealed Trichophyton species (Photo credit: Joshua A. Zeichner, M.D.)
from infected hair and pustules. The dermatophyte Microsporum canis may also be
visualized with Wood’s light, and a biopsy can be diagnostic as well. Fungal culture
is performed on agar with cycloheximide to identify the causative organism [7].
10.5 Treatment
Tinea barbae should be treated with systemic antifungal therapy, as these drugs are
able to penetrate the infected hair shaft, whereas topical therapies cannot.
Debridement and shaving of the affected hairs is also recommended [2]. Griseofulvin
up to 1 g daily, FDA approved for the treatment of tinea capitis, is dosed daily for
6–12 weeks [8]. However, some authors advocate a 4-week course of terbinafine
250 mg daily [7]. Terbinafine should not be the first line of therapy if the causative
organism is unknown, as M. canis is resistant to terbinafine. Treatment with itracon-
azole 200 mg or fluconazole 200 mg daily for 4–6 weeks is also effective [2].
Adjuvant therapy with antifungal shampoos, such as ketoconazole 2 % shampoo
and selenium sulfide 2.5 % shampoo, used at least three times weekly, will decrease
shedding of the infectious fungal elements [9]. Furthermore, elimination of the
source of the infection, whether by treatment of infected animals or treatment of
onychomycosis, is very important [4].
70 L. Kole and B. Elewski
References
1. Burns T, Breathnach S, Cox N, Griffiths C, editors. Rook’s textbook of dermatology. 8th ed. San
Francisco: Wiley-Blackwell; 2010.
2. Bolognia JL, Jorizzo JL, Rapini RP, editors. Dermatology. 2nd ed. St. Louis: Mosby Elsevier;
2008.
3. Drake LA, Dinehart SM, Farmer ER, et al. Guidelines of care for superficial mycotic infections
of the skin: tinea capitis and tinea barbae. Guidelines/Outcomes Committee. American Academy
of Dermatology. J Am Acad Dermatol. 1996;34:290–4.
4. Xavier MH, Torturella DM, Rehfeldt FVS, et al. Sycosiform tinea barbae caused by Trichophyton
rubrum. Dermatol Online J. 2008;14:10.
5. Szepietowski JC, Matusiak L. Trichophyton rubrum autoinoculation from infected nails is not
such a rare phenomenon. Mycoses. 2008;51:345–6.
6. Habif TP. Clinical dermatology. 5th ed. St. Louis: Mosby Elsevier; 2010.
7. Baran W, Szepietowski JC, Schwartz RA. Tinea barbae. Acta Dermatovenerol Alp Panonica
Adriat. 2004;13:91–4.
8. Gupta AK, Dlova N, Taborda P, et al. Once weekly fluconazole is effective in children in the
treatment of tinea capitis: a prospective, multicenter study. Br J Dermatol. 2000;142:965–8.
9. Elewski BE. Treatment of tinea capitis: beyond griseofulvin. J Am Acad Dermatol. 1999;
40:S27–30.
Chapter 11
Flat Warts
11.1 Introduction
Verruca plana, or flat warts, are virally induced papules caused by the human
papillomavirus (HPV) that can mimic a closed comedone or an acneiform papule. This
chapter will discuss the etiology, risk factors, diagnosis, and treatment of flat warts.
Table 11.1 summarizes the similarities and differences between flat warts and acne.
11.2 Background
HPV includes greater than 100 genotypes of a double-stranded DNA virus [1].
Cutaneous HPV types comprise a group of viruses that infect the skin and induce
common warts, palmar and plantar warts, flat warts, and butcher’s warts.
Classification of warts is based upon morphology, anatomic localization, and HPV
typing.
Common warts are hyperkeratotic, exophytic papules, or nodules typically asso-
ciated with HPV-1, HPV-2, or HPV-4 [2]. They are most commonly located on fin-
gers, the dorsal surfaces of hands, and other sites prone to trauma. They typically
spare the face but can occur at any anatomical location. HPV types 2, 27, and 57
caused the majority of palmoplantar warts [2]. Myrmecia are large, deep burrowing
warts on the plantar surface caused by HPV-1. Most commonly, flat warts that have
been analyzed by polymerase chain reaction (PCR) studies contain HPV-3 and, less
often, types 10, 28, 29, and 41 [2–6].
Table 11.1 Similarities and differences between acne and flat warts
Features Flat warts Acne
Affected age group—childhood and young adult Yes Yes
Koebner phenomenon Yes Occasionallya
Follicular-based papules No Yes
Flat-topped papules Yes No
Virally induced Yes No
Ultraviolet light influenced Yes—worsened Yes—most often improved
Can heal with a scar Occasionallyb Yes
Hormonally influenced No Yes
Cellular immunity related Yes No
Spontaneously resolve Yes Yes
Response to retinoid therapy Yes Yes
a
Acne can be triggered by minor trauma/friction (e.g., acne mechanica)
b
Treatments can lead to permanent scarring
Flat warts are 2–4 mm in diameter, slightly elevated, flat-topped, smooth papules
(Fig. 11.1). They can be skin colored or slightly erythematous on pale skin and
hyperpigmented on darker skin. They are generally multiple and grouped. The fore-
head, cheeks, and nose, perioral region, and dorsal hands are classic locations [9].
Hyperpigmented lesions may be confused with lentigines or ephelides. Skin-colored
to erythematous lesions located on the central face may be confused with closed
11 Flat Warts 73
Fig. 11.1 Adolescent woman who presented for treatment of “acne” with typical facial flat warts
11.4 Work-up
Multiple flat warts are commonly seen in healthy individuals but may also be a sign
of a compromised immune system. When there is extensive involvement in an adult
with no obvious risk factors, or in a treatment-refractory case, the patient’s cellular
immune function should be evaluated. Extensive flat warts have been reported in
patients with human immunodeficiency virus infection (HIV) [11]. Widespread flat
warts have also been reported in immune-reconstitution syndrome in HIV patients
on HAART (Highly Active Anti-Retroviral Treatment) [12]. Similarly, the trans-
plant population and primary immunodeficiency diseases, such as common variable
immunodeficiency, severe combined immunodeficiency, Hyper-IgM, Hyper-
immunoglobulin E syndrome, idiopathic CD4+ T-cell lymphocytopenia [13], and
WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) [14] have
a higher incidence of viral warts.
Epidermodysplasia verruciformis (EDV) is a rare, autosomal recessive disorder
with widespread skin colonization by HPV. The clinical presentation is of dissemi-
nated flat warts or large pityriasis versicolor-like patches induced most commonly by
HPV-3 and HPV-10 [15]. One-third to one-half of patients will develop squamous
cell carcinoma, associated most frequently with HPV subtypes 5, 8, and 47 [2, 15].
74 T. Rosen and S. Risner-Rumohr
A flat wart can be easily discerned from an acneiform lesion with a histological
evaluation. Flat warts show hyperkeratosis and acanthosis, no papillomatosis, only
slight elongation of the rete ridges, and no areas of parakeratosis. In the upper stra-
tum malpighii, including the thickened granular layer, there is diffuse vacuolization
of the cells [16]. The nuclei of the vacuolated cells lie at the centers of the cells, and
some appear deeply basophilic. The dermis appears normal. In spontaneously
regressing warts, there is often a superficial lymphocytic infiltrate in the dermis with
exocytosis and apoptosis of cells in the epidermis [16].
11.5 Treatment
Of all clinical HPV infections, flat warts have the highest rate of spontaneous remis-
sion; they almost universally resolve without treatment over several months to years
[9]. This makes evaluation of any treatment modality difficult [17]. Among immu-
nosuppressed patients, the warts typically persist longer and are more resistant to
treatment [11, 14].
Wart regression is probably the result of cell-mediated immune mechanisms,
although the process is not completely understood [18]. Considerations related to
treatment of flat warts include the patient’s age, immune status, skin integrity, and
potential cosmetic outcome. Despite the fact that a wide range of local treatments
for nongenital cutaneous warts exists, there is only limited reliable evidence regard-
ing the efficacy of various potential interventions [17]. Viral wart survival is based
on evading the immune detection apparatus within the epidermis. Inflammation of
the skin may stimulate a concurrent immunologic response against HPV.
Keratolytics, such as many over-the-counter products containing with salicylic acid
or prescription strength retinoids, are frequently used for flat warts. These cause
chemical irritation and debridement of the wart’s keratotic surface. In reality, topical
salicylic acid products have the highest grade clinical evidence regarding their supe-
riority to placebo [17]. Topical retinoids are potent keratolytics but also assist in the
treatment of warts by the disruption of epidermal growth and differentiation, as well
as via modest immunomodulatory effects. One study investigating tretinoin 0.05 %
cream resulted in an 85 % clearance in a series of children with flat warts compared
to 32 % spontaneous clearance in the controls [19]. Astringent chemicals such as
trichloroacetic acid and cantharidin are also frequently used in the office setting.
Cantharidin is a chemical derived from the blister beetle, Cantharis vesicatoria.
When applied it causes dermal blistering and local necrosis. Responses are variable,
and this material can cause scarring or spread of warts around the index, treated
lesion. Podophyllin, topical 5 % 5-fluorouracil, and intralesional bleomycin are
other topical therapies that have varying degrees of antimitotic effect; however, they
tend to be more beneficial for mucosal lesions and exophytic warts.
Destructive techniques, such as curettage, electrodesiccation, laser therapy, pho-
todynamic therapy, or cryotherapy using liquid nitrogen, have all been utilized with
good results. Nonetheless, destructive techniques are considered second-line
11 Flat Warts 75
therapy as they carry the risk of scar formation and recurrence in or around the treat-
ment site. This is especially true for smaller, less exophytic warts, such as verruca
plana. Prolonged occlusive duct tape treatment followed by periodic manual debride-
ment has been shown to be superior or at least comparable to cryotherapy for com-
mon warts but has not been reported for flat warts [20].
Many laser therapies have been employed. The carbon dioxide (CO2) laser emits
an infrared light emission (10,600 nm) that causes a nonselective thermal tissue
destruction; the lost skin then heals by secondary intent [21]. The erbium:yttrium/
aluminum/garnet (Er:YAG) emits an infrared light emission (2,940 nm) which is
absorbed more efficiently by epidermal water, thus allowing for a more precise
ablation and a smaller zone of thermal damage than the CO2 laser [22]. No random-
ized studies on the efficacy of laser therapy for flat warts have been published.
Infectious hazards have also been demonstrated for both laser and electrocoagula-
tion, as HPV DNA has been identified in the vaporized laser plume [23]. Other laser
therapies such as pulse dye laser have been used to selectively destroy the dilated
capillaries within the warts, producing less pain and scarring than CO2 laser. This
technique has been utilized more for flat warts in children and for facial lesions in
all ages due to a preferable safety profile [24].
Contact immunotherapy can be an effective treatment. Dinitrochlorobenzene
diphenylcyclopropenone or squaric acid have been used as non-mutagenic sensitiz-
ers eliciting a contact allergy that theoretically concomitantly heightens the immune
response against the virus. Intralesional immunotherapy has little role in flat warts
due to their thin nature. A topical immune response modifier, imiquimod, has been
FDA approved for external anogenital warts. Imiquimod has been shown to interact
with Toll-like receptors 7 and 8, resulting in activation of cytokine secretion from
monocytes/macrophages (including α-interferon, interleukin-12, and TNF-α), as
well as stimulation of antigen-presenting dendritic cells [2, 25]. No randomized
controlled studies have been performed with imiquimod cream at any of the avail-
able concentrations (5, 3.75, and 2.5 %) for the management of flat warts.
Nonetheless, imiquimod has been widely utilized in children and for facial flat
warts in all ages as it generally elicits less discomfort compared to many of the other
available treatments. In 2006, the FDA approved an ointment extract of green tea
(Camellia sinensis) containing 15 % sinecatechins for treatment of genital warts.
No studies have been published to date regarding efficacy in flat warts, but it may
well represent a novel topical approach for recalcitrant lesions [26].
Systemic therapies including garlic supplements (Allium sativum) have been
shown to inhibit cellular proliferation of virally infected cells [27]. Oral cimetidine
(20–40 mg/kg/day) and oral zinc sulfate supplementation (10 mg/kg/day) have been
used in an attempt to enhance cell-mediated immunity [28, 29]. Systemic retinoids,
both isotretinoin and acitretin, have been used for extensive flat warts. In immuno-
compromised patients (both EDV and HIV populations), systemic and 1–3 % topical
cidofovir, an antiviral that inhibits DNA synthesis, has been used effectively [30].
A combination of therapies is often pursued due to the frequent resilience of
warts. There is no single gold standard wart treatment. The specific treatment regi-
men should be individualized to the needs and desires of the patient and
76 T. Rosen and S. Risner-Rumohr
subsequently altered or modified based upon the clinical response obtained. With the
exception of topical tretinoin, none of the typical repertoire of topical or systemic
acne interventions would benefit flat warts.
References
22. Park JH, Hwang ES, Kim SN, Kye YC. Er:YAG laser treatment of verrucous epidermal nevi.
Dermatol Surg. 2004;30:378–81.
23. Sawchuk WS, Weber PJ, Lowy DR, Dzubow LM. Infectious papillomavirus in the vapor of
warts treated with carbon dioxide laser or electrocoagulation: detection and protection. J Am
Acad Dermatol. 1989;21:41–9.
24. Vargas H, Hove CR, Dupree ML, Milliams EF. The treatment of facial verrucae with pulsed
dye laser. Laryngoscope. 2002;112:1573–6.
25. Gaspari A, Tyring SK, Rosen T. Beyond a decade of 5% imiquimod topical therapy. J Drugs
Dermatol. 2009;8:467–74.
26. Viera MH, Amini S, Huo R, et al. Herpes simplex virus and human papillomavirus genital
infections: new and investigational therapeutic options. Int J Dermatol. 2010;49:733–49.
27. Seki R, Tsuji K, Hayato Y, et al. Garlic and onion oils inhibit proliferation and induce differ-
entiation of HL-60 cells. Cancer Lett. 2000;160:29–35.
28. Al-Gurairi FT, Al-Waiz M, Sharquie KE. Oral zinc sulphate in the treatment of recalcitrant
viral warts: randomized placebo-controlled clinical trial. Br J Dermatol. 2002;146:423–31.
29. Rogers CJ, Gibney MD, Siegfried EC, et al. Cimetidine therapy for recalcitrant warts in adults:
is it any better than placebo? J Am Acad Dermatol. 1999;41:123–7.
30. Grone D, Treudler R, de Villiers EM, et al. Intravenous cidofovir treatment for recalcitrant
warts in the setting of a patient with myelodysplastic syndrome. J Eur Acad Dermatol Venereol.
2006;20:202–5.
Chapter 12
Molluscum Contagiosum
12.1 Introduction
Y. Clark, MPAS-C
Department of Dermatology, Sharp Rees-Stealy, La Mesa, CA, USA
e-mail: [email protected]
L.F. Eichenfield, M.D. (*)
Division of Pediatric and Adolescent Dermatology, Rady Children’s Hospital, San Diego,
8010 Frost Street, Suite 620, San Diego, CA 92123, USA
e-mail: [email protected]
12.2 Background
12.4 Work-Up
12.5 Treatment
available, patient’s age, pain tolerance, and accessibility to clinic should be taken
into consideration when choosing a remedy. Destructive methods can be painful to
younger children but well tolerated in older children and adults. To date there are no
standard treatment protocols or reliable evidence-based recommendations for
treating molluscum contagiosum [13].
The most commonly utilized treatment amongst pediatric dermatologist is can-
tharidin [14]. Cantharidin has been available and used to treat molluscum since 1950.
Cantharidin is derived from the blistering beetle, Cantharis vesicatoria, and causes
vesiculation of the epidermis. Application in office by a physician or other qualified
and well-trained professional is recommended. Side effects include blistering, tem-
porary burning, localized pain, erythema, and pruritus. Occlusion of treated areas is
not recommended and a typical wash-off period can range from 2 to 6 h or sooner if
blistering or discomfort is noted. Cantharidin is a powerful toxin that should not be
ingested or absorbed; therefore application to mucosal areas is not recommended.
Cantharidin generally is not used on the face or on occluded areas such as under a
diaper [15]. Facial lesions may be treated with topical tretinoin cream. Systemic
therapy with oral cimetidine has been advocated by some as monotherapy or in con-
junction with cantharidin for extensive lesions or recalcitrant lesions to cantharidin,
though there is a minimal evidence to support its utility (Table 12.1).
References
5. Homepage | CDC Molluscum Contagiosum. Centers for Disease Control and Prevention.
2011. Web 11 Dec 2011. http://www.cdc.gov/ncidod/dvrd/molluscum/.
6. American Academy of Pediatrics. Summaries of infectious disease. In: Pickering LK, Baker
CJ, Kimberlin DW, Long SS, editors. Red Book: 2009 Report of the committee on infectious
diseases. 28th ed. Elk Grove Village, IL: American Academy of Pediatrics; 2009. p. 466.
7. Dohil M, Lin P, Lee J, Lucky A, Paller A, Eichenfield L. The epidemiology of molluscum
contagiosum in children. J Am Acad Dermatol. 2006;54(1):47–54.
8. Krishnamurthy J, Nagappa DK. The cytology of molluscum contagiosum mimicking skin
adnexal tumor. J Cytol. 2010;27(2):74.
9. Ianhez M, Sda Cestari C, Enokihara MY, Seize MB. Dermoscopic patterns of molluscum
contagiosum: a study of 211 lesions confirmed by histopathology. An Bras Dermatol.
2011;86(1):74–9.
10. Trama JP, Adelson ME, Mordechai E. Identification and genotyping of molluscum contagio-
sum from genital swab samples by real-time PCR and pyrosequencing. J Clin Virol. 2007;
40(4):325–9.
11. Maronn M, Salm C, Lyon V, Galbraith S. One-year experience with Candida antigen
immunotherapy for warts and molluscum. Pediatr Dermatol. 2008;25:189–92. doi:10.1111
/j.1525-1470.2008.00630.x.
12. Enns LL, Evans MS. Intralesional immunotherapy with Candida antigen for the treatment of
molluscum contagiosum in children. Pediatr Dermatol. 2011;28:254–8. doi:10.1111/
j.1525-1470.2011.01492.x.
13. van der Wouden JC, van der Sande R, van Suijlekom-Smit LWA, Berger M, Butler CC, Koning
S. Interventions for cutaneous molluscum contagiosum. Cochrane Database Syst Rev. 2009;4,
CD004767. doi:10.1002/14651858.CD004767.pub3.
14. Coloe J, Morrell DS. Cantharidin use among pediatric dermatologists in the treatment of mol-
luscum contagiosum. Pediatr Dermatol. 2009;26:405–8. doi:10.1111/j.1525-1470.2008.00860.x.
15. Silverberg N, Sidbury R, Mancini AJ. Childhood molluscum contagiosum: experience with
cantharidin therapy in 300 patients. J Am Acad Dermatol. 2000;43(3):503–7.
Chapter 13
Herpes Simplex Virus
13.1 Introduction
Our understanding of human herpes simplex virus (HSV) has increased tremen-
dously since the early descriptions of disease provided by Hippocrates [1, 2].
Notable advances include the correlation of herpetic lesions with genital infections
in the eighteenth century [3] and Vidal’s recognition of human-to-human transmis-
sion in 1893 [2]. Antigenic differences between HSV subtypes, suspected on clini-
cal observations by Lipschitz [4], were confirmed in 1968 [5]. In modern day, there
is successful antiviral treatment available for most HSV infections [6]. Insight into
the viral life cycle and gene expression has been a driving force behind the develop-
ment of antiviral treatment, including new vaccines and gene therapy [4].
13.2 Background
stress, immunosuppression, UV light, fever, and tissue damage [4, 7]. The severity
of the initial infection correlates with the chance of recurrence. Rarely, life-
threatening infections occur in cases of severe immune compromise, pregnancy, and
neonatal disseminated HSV. “Primary infections” are considered first-time events,
but their occurrence in seropositive individuals indicates that latent infection can be
established without prior symptoms [6].
HSV infections are ubiquitous, even in remote areas [8]. More than 57 % of the
US population between the ages of 14 and 49 are seropositive for HSV1 [9].
Incidence correlates with age in a linear fashion, globally reaching 60–90 % in older
adults [10]. HSV1 is most commonly transmitted in childhood and adolescence.
The overall incidence of HSV1 is significantly higher than that of HSV2 [10], which
occurs more commonly in women and in subpopulations with high-risk sexual
behavior [4, 10]. Humans are the only reservoir for HSV infection, and there have
been no reported animal vectors [4].
Infections around the mouth are the most common sites and reservoirs of HSV.
Primary infection is usually asymptomatic [11]. Symptomatic cases may include
extensive orolabial vesiculo-ulcerative lesions, gingivostomatitis, fever, and local-
ized lymphadenopathy [4]. Adolescents can present with acute pharyngitis and
mononucleosis-like symptoms [12, 13]. Dehydration from poor oral intake is the
most frequent reason for hospitalization [6].
The incubation period for HSV infections is 4 days on average, and clinical
symptoms may persist for 2–3 weeks [4, 14]. Vesicles form within 1–2 days of pro-
dromal symptoms, progress to ulcers in another 1–2 days, and heal within 8–10
days. Pain is most severe with the appearance of lesions [15]. The frequency of
recurrences varies among individuals [4], but is estimated to occur in 20–40 % of
adults [6], most commonly on the vermillion border. Viral shedding increases with
active lesions [16], during episodes of the common cold, oral trauma, and false
prodromes [17]. Shedding persists in the absence of symptoms in an estimated 7 %
of the healthy population [18] (Figs. 13.1 and 13.2).
The degree of immune suppression correlates with the risk of developing HSV
outbreaks. Organ transplant recipients and individuals with HIV/AIDS are at high
risk for severe infections and frequent recurrences [4, 20]. The most frequent com-
plication is progressive, chronic mucocutaneous infection with subsequent tissue
necrosis [6]. Progressive disease involving the esophagus, respiratory, and gastroin-
testinal tract has been reported [4].
88 R. Gordon and S. Tyring
Individuals with a damaged epithelial barrier, most commonly from atopic dermati-
tis [23], are susceptible to eczema herpeticum. This presents as a vesiculopustular
eruption in areas of underlying skin disease; lesions may erode and become second-
arily infected. Recurrent episodes are generally less severe [24]. In wrestlers, herpes
gladiatorum occurs in areas of close contact, usually the face and neck [25]. Facial
HSV folliculitis presents as folliculocentric vesiculopapules and is confirmed histo-
pathologically by HSV changes limited to the pilosebaceous unit [26] (Fig. 13.3).
13 Herpes Simplex Virus 89
13.4 Work-Up
Classically, detection is performed by viral culture in media that will show the HSV
cytopathic effect, followed by typing with monoclonal antibodies [27, 28]. Viral
swabs may be obtained from vesicles or other involved sites: oral mucosa, cerebro-
spinal fluid, or conjunctiva [4]. Polymerase chain reaction (PCR) has been the gold
standard for diagnosing CNS infections and may become the standard test to diag-
nose HSV infections in other sites [29]. It has shown greater sensitivity than culture
in detecting virus from oral [30] and genital lesions [31]. Serologic assays using
enzyme-linked immunosorbent assay (ELISA) distinguish between HSV1 and
HSV2 infections and detect infection in the absence of symptoms. Utilization of
Western blot is restricted to research labs [4].
While not as sensitive as PCR [32], Tzanck smears allow for the cytopatho-
logic detection of HSV. They are prepared by scraping the periphery of an ulcer,
smearing the material on a glass slide, fixing immediately in cold ethanol, and
then staining with Giemsa, Papanicolaou, or Wright stain [4]. Herpetic giant cells
have multiple nuclei with molding and a ground-glass appearance, and intranu-
clear inclusion bodies [33]. Intranuclear inclusion bodies are not specific and may
be seen in VZV infections as well [4]. Biopsy samples may show intraepidermal
vesicles containing acantholytic keratinocytes with these cytologic changes [34]
(Fig. 13.4).
Fig. 13.4 HSV-infected cells with ground-glass nuclei, marginated peripheral chromatin, and a
multinucleated giant cell with molding
90 R. Gordon and S. Tyring
13.5 Treatment
Most HSV infections are treated with acyclovir, its prodrug valacyclovir, and
famciclovir (prodrug of penciclovir). These nucleoside analogues are activated by
viral thymidine kinase and selectively inhibit viral DNA production by competing
as substrate for DNA polymerase [6, 35].
In moderate to severe primary disease in children, acyclovir 15 mg/kg five times per
day for 7 days has been shown to decrease the duration of lesions from 10 to 4 days
[11, 36]. Appropriate regimens for treatment of primary infections in adults include
acyclovir 400 mg TID for 7–10 days, valacyclovir 1 g BID for 7–10 days, famciclo-
vir 500 mg BID for 7 days, or 250 mg TID for 7–10 days [11, 37].
The ability to change the course of recurrent disease by administering antiviral
therapy at the onset of symptoms has long been recognized [4, 15, 38]. Beginning
treatment in this narrow therapeutic window is possible with patient-initiated epi-
sodic therapy, which reduces healing time to a greater extent than physician-initiated
therapy [39]. First-line therapy in recurrent herpes labialis consists of famciclovir
1,500 mg once a day or valacyclovir 2 g twice daily for 1 day initiated at the first
prodromal symptom [40, 41]. Suppressive treatment is not frequently practiced
[40], but is effective at reducing recurrences [41, 42]. Valacyclovir 500 mg once
daily is the simplest regimen [4].
Topical therapy includes docosanol 10 % cream (Abreva®) which demonstrated
an 18-h reduced median time to healing in treated patients compared to placebo
[43]. Penciclovir 1 % cream (Denavir®) and acyclovir 5 % cream (Zovirax®) both
demonstrate therapeutic efficacy in early- and late-stage lesions [44].
Acyclovir is effective in the prevention and treatment of HSV infections [4, 45]. For
adults intravenous dosing is 5 mg/kg over 1 h, every 8 h for 7 days. In children, the
13 Herpes Simplex Virus 91
dose is 250 mg/m2 with the same schedule. For limited disease, topical 5 % acyclovir
ointment can be used every 3 h for 7 days [4]. One recent review found no evidence
that valacyclovir is more efficacious than acyclovir [45]. In patients with HIV, epi-
sodic treatment with famciclovir 500 mg BID for 7 days or valacyclovir 500 mg to
1 g BID for 7 days is appropriate. The same regimen can be used off-label for
chronic suppressive therapy [4].
Antiviral-resistant herpes virus, usually secondary to mutations in viral thymi-
dine kinase, is a special concern in this population as suppressive therapy has
become standard. In such cases, treatment with the pyrophosphate analogue foscar-
net and the nucleotide analogue cidofovir are appropriate [46]. Standard drug-
sensitivity tests take more than 10 days for a result, but newer, faster methods are
being developed [47].
Without controlled studies, it is intuitive that primary and recurrent cutaneous infec-
tions may be treated with valacyclovir or famciclovir at doses used to treat primary
and recurrent herpes genitalis [4]. Prophylactic use of valacyclovir (500 mg or 1 g
once or twice daily) is effective at preventing recurrences of herpes gladiatorum
during the wrestling season [48].
References
1. Beswick TS. The origin and the use of the word herpes. Med Hist. 1962;6:214–32.
2. Roizman B, Whitley RJ. The nine ages of herpes simplex virus. Herpes. 2001;8:23–7.
3. Hutfield DC. History of herpes genitalis. Br J Vener Dis. 1966;42:263–8.
4. Tyring SK, Yen-Moore A, Lupi O. Mucocutaneous manifestations of viral diseases. London:
Informa Healthcare; 2010. p. 65–97.
5. Nahmias AJ, Dowdle WR. Antigenic and biologic differences in herpesvirus hominis. Prog
Med Virol. 1968;10:110–59.
6. Arvin A, Roizman B, Whitley R, Mocarski E, Campadelli-Fiume G, Yamanishi K, editors.
Human herpes-virus: biology, therapy & immunoprophylaxis. Cambridge: Cambridge
University Press; 2007.
92 R. Gordon and S. Tyring
7. El Hayderi L, Raty L, Failla V, Caucanas M, Paurobally D, Nikkels AF. Severe herpes simplex
virus type-I infections after dental procedures. Med Oral Patol Oral Cir Bucal. 2011;
16(1):e15–8.
8. Lin H, He N, Su M, Feng J, Chen L, Gao M. Herpes simplex virus infections among rural resi-
dents in eastern China. BMC Infect Dis. 2011;11:69.
9. Xu F, Sternberg MR, Kottiri BJ, McQuillan GM, Lee FK, Nahmias AJ, Berman SM, Markowitz
LE. Trends in herpes simplex virus type 1 and type 2 seroprevalence in the United States.
JAMA. 2006;296(8):964–73.
10. Smith JS, Robinson NJ. Age-specific prevalence of infection with herpes simplex virus types
2 and 1: a global review. J Infect Dis. 2002;186 Suppl 1:S3–28.
11. Wheeler Jr CE. The herpes simplex problem. J Am Acad Dermatol. 1988;18:163–8.
12. Glezen WP, Fernald GW, Lohr JA. Acute respiratory disease of university students with special
reference to the etiologic role of Herpesvirus hominis. Am J Epidemiol. 1975;101(2):111–21.
13. McMillan JA, Weiner LB, Higgins AM, Lamparella VJ. Pharyngitis associated with herpes
simplex virus in college students. Pediatr Infect Dis J. 1993;12(4):280–4.
14. Young SK, Rowe NH, Buchanan RA. A clinical study for the control of facial mucocutaneous
herpes virus infections. I. Characterization of natural history in a professional school popula-
tion. Oral Surg Oral Med Oral Pathol. 1976;41:498–507.
15. Spruance SL, Overall Jr JC, Kern ER, Krueger GG, Pliam V, Miller W. The natural history of
recurrent herpes simplex labialis: implications for antiviral therapy. N Engl J Med. 1977;
297:69–75.
16. Daniels CA, LeGoff SG. Shedding of infectious virus/antibody complexes from vesicular
lesions of patients with recurrent herpes labialis. Lancet. 1975;2:524–8.
17. Spruance SL. Pathogenesis of herpes simplex labialis: excretion of virus in the oral cavity.
J Clin Microbiol. 1984;19(5):675–9.
18. Buddingh GJ, Schrum DI, Lanier JC, Guidry DJ. Studies of the natural history of herpes sim-
plex infections. Pediatrics. 1953;11:595–610.
19. Simmons A. Clinical manifestations and treatment considerations of herpes simplex virus
infection. J Infect Dis. 2002;186 Suppl 1:S71–7.
20. Shiley K, Blumberg E. Herpes viruses in transplant recipients: HSV, VZV, human herpes
viruses, and EBV. Infect Dis Clin North Am. 2010;24:373–93.
21. Corey L, Wald A. Maternal and neonatal herpes simplex virus infections. N Engl J Med.
2009;361:1376–85.
22. Brown ZA, Selke S, Zeh J, Kopelman J, Maslow A, Ashley RL, et al. The acquisition of herpes
simplex virus during pregnancy. N Engl J Med. 1997;337:509–15.
23. Mooney MA, Janniger CK, Schwartz RA. Kaposi’s varicelliform eruption. Cutis. 1994;53:
243–5.
24. Kramer SC, Thomas CJ, Tyler WB, Elston DM. Kaposi’s varicelliform eruption: a case report
and review of the literature. Cutis. 2004;73:115–22.
25. Anderson BJ. The epidemiology and clinical analysis of several outbreaks of herpes gladiato-
rum. Med Sci Sports Exerc. 2003;35:1809–14.
26. Al-Dhafiri SA, Molinari R. Herpetic folliculitis. J Cutan Med Surg. 2002;6:19–22.
27. Scott DA, Coulter WA, Lamey PJ. Oral shedding of herpes simplex virus type 1: a review.
J Oral Pathol Med. 1997;26:441–7.
28. Aarnaes SL, Peterson EM, de la Maza LM. Evaluation of a new herpes simplex virus typing
reagent for tissue culture confirmation. Diagn Microbiol Infect Dis. 1989;12(3):269–70.
29. Strick LB, Wald A. Diagnostics for herpes simplex virus: is PCR the new gold standard? Mol
Diagn Ther. 2006;10:17–28.
30. Tateishi K, Toh Y, Minagawa H, Tashiro H. Detection of herpes simplex virus (HSV) in the
saliva from 1,000 oral surgery outpatients by the polymerase chain reaction (PCR) and virus
isolation. J Oral Pathol Med. 1994;23:80–4.
31. Cone RW, Hobson AC, Palmer J, Remington M, Corey L. Extended duration of herpes simplex
virus DNA in genital lesions detected by the polymerase chain reaction. J Infect Dis. 1991;
164:757–60.
13 Herpes Simplex Virus 93
32. Nahass GT, Goldstein BA, Zhu WY, Serfling U, Penneys NS, Leonardi CL. Comparison of
Tzanck smear, viral culture, and DNA diagnostic methods in detection of herpes simplex and
varicella-zoster infection. JAMA. 1992;268:2541–4.
33. Douglas D, Richman RJW, Hayden FG, editors. Clinical virology. Washington, DC: ASM
Press; 2002.
34. Weedon D. Skin pathology. Edinburgh: Churchill Livingstone; 2002.
35. Derse D, Cheng YC, Furman PA, St Clair MH, Elion GB. Inhibition of purified human and
herpes simplex virus-induced DNA polymerases by 9-(2-hydroxyethoxymethyl)guanine
triphosphate. Effects on primer-template function. J Biol Chem. 1981;256:11447–51.
36. Amir J, Harel L, Smetana Z, Varsano I. Treatment of herpes simplex gingivostomatitis with
aciclovir in children: a randomised double blind placebo controlled study. BMJ. 1997;
314(7097):1800–3.
37. Thomas Kreig DRB, Miyachi Y, editors. Therapy of skin diseases: a worldwide perspective on
therapeutic approaches and their molecular basis. London: Springer; 2010.
38. Spruance SL, Schnipper LE, Overall Jr JC, Kern ER, Wester B, Modlin J, et al. Treatment of her-
pes simplex labialis with topical acyclovir in polyethylene glycol. J Infect Dis. 1982;146:85–90.
39. Reichman RC, Badger GJ, Mertz GJ, Corey L, Richman DD, Connor JD, Redfield D, Savoia
MC, Oxman MN, Bryson Y, et al. Treatment of recurrent genital herpes simplex infections
with oral acyclovir. A controlled trial. Treatment of recurrent genital herpes simplex infections
with oral acyclovir. JAMA. 1984;251(16):2103–7.
40. Modi S, Van L, Gewirtzman A, Mendoza N, Bartlett B, Tremaine AM, Tyring S. Single-day
treatment for orolabial and genital herpes: a brief review of pathogenesis and pharmacology.
Ther Clin Risk Manag. 2008;4(2):409–17.
41. Rooney JF, Straus S, Mannix ML, Wohlenberg CR, Alling DW, Dumois JA, Notkins AL. Oral
acyclovir to suppress frequently recurrent herpes labialis. A double-blind, placebo-controlled
trial. Ann Intern Med. 1993;118(4):268–72.
42. Baker D, Eisen D. Valacyclovir for prevention of recurrent herpes labialis: 2 double-blind,
placebo-controlled studies. Cutis. 2003;71(3):239–42.
43. Sacks SL, Thisted RA, Jones TM, Barbarash RA, Mikolich DJ, Ruoff GE, Jorizzo JL, Gunnill
LB, Katz DH, Khalil MH, Morrow PR, Yakatan GJ, Pope LE, Berg JE, Docosanol 10% Cream
Study Group. Clinical efficacy of topical docosanol 10% cream for herpes simplex labialis: a
multicenter, randomized, placebo-controlled trial. J Am Acad Dermatol. 2001;45(2):222–30.
44. Spruance SL, Nett R, Marbury T, Wolff R, Johnson J, Spaulding T. Acyclovir cream for treat-
ment of herpes simplex labialis: results of two randomized, double-blind, vehicle-controlled,
multicenter clinical trials. Antimicrob Agents Chemother. 2002;46(7):2238–43.
45. Glenny AM, Fernandez Mauleffinch LM, Pavitt S, Walsh T. Interventions for the prevention
and treatment of herpes simplex virus in patients being treated for cancer. Cochrane Database
Syst Rev. 2009;1, CD006706.
46. Piret J, Boivin G. Resistance of herpes simplex viruses to nucleoside analogues: mechanisms,
prevalence, and management. Antimicrob Agents Chemother. 2011;55:459–72.
47. Shiota T, Lixin W, Takayama-Ito M, Iizuka I, Ogata M, Tsuji M, et al. Expression of herpes
simplex virus type 1 recombinant thymidine kinase and its application to a rapid antiviral
sensitivity assay. Antiviral Res. 2011;91:142–9.
48. Anderson BJ. The effectiveness of valacyclovir in preventing reactivation of herpes gladiatorum
in wrestlers. Clin J Sport Med. 1999;9:86–90.
Chapter 14
Varicella Zoster Virus
14.1 Introduction
Varicella zoster virus causes two clinical syndromes. Chicken pox, or varicella, is a
self-limiting disease of childhood characterized by a highly pruritic rash. Shingles,
or herpes zoster, is a reactivation of the virus typically seen in adults. Heberden first
distinguished these illness as separate entities in 1767 [1], and Osler emphasized the
distinction in his book on clinical medicine [2]. Their relation was suggested by von
Bokay in 1892 when he noted that children developed varicella after coming into
contact with herpes zoster patients [3]. Weller et al. proved a common etiological
agent [3–5], and Garland and Hope-Simpson were the first to propose a reactivation
of latent varicella as the cause of herpes zoster [3]. In 1995, the FDA approved the
first vaccine for the prevention of varicella, significantly changing the epidemiology
of this disease [6].
14.2 Background
Varicella virus is transmitted both by direct contact and through airborne spread
when a patient breathes or sneezes. Transmission may occur more easily in temper-
ate environments as compared to tropical climates, though seasonal peaks are seen
in both climates [2]. In the United States, April has the highest incidence of VZV
infection [7]. Before the advent of vaccination, primary varicella was commonplace
and approximately 11,000 hospitalizations per year in the United States resulted
from complications of the illness in otherwise healthy children [2]. Vaccination
decreased the incidence of disease in all age groups with the greatest decline in
children aged 12 months to 4 years [8]. By 2004, mortality associated with varicella
had decreased 82 % from the pre-vaccine era [9].
While the incidence of varicella has decreased by 76–90 % as a result of vaccina-
tion [10, 11], roughly 90–95 % of adults in the United States are seropositive for
varicella [12, 13]. Therefore, most people are susceptible to herpes zoster, the life-
time risk of which has been estimated to be 10–30 % [10, 14]. Age is a major risk
factor for herpes zoster, and its prevalence increases dramatically every 5 years; the
sharpest spike is between 50 and 60 years of age [15]. Other risk factors include
HIV [16], malignancy (especially lymphoproliferative cancers), immunosuppres-
sive therapy, history of organ transplant [17], and possibly trauma [18]. Genetics
may also be a risk factor as 39 % of herpes zoster patients recall a blood relative
with a history of shingles compared to 11 % of control patients without herpes
zoster who report shingles in a blood relative [19]. Recurrent zoster is rare, but may
occur in 1.7–6 % of patients [15, 20].
VZV is a DNA alpha-herpesvirus that adheres to respiratory mucosa with the
glycoproteins on its outer lipid-containing envelope. Viral replication occurs in
regional lymph nodes and the reticuloendothelial system, followed respectively by
viremia at 4–6 and again at 14 days [21]. Involvement of the skin is due to transmis-
sion of the virus from infected CD4+ T cells expressing skin-homing factors to
dermal fibroblasts and keratinocytes [22]. During primary infection, the virus
spreads to the cranial or dorsal root ganglia, possibly by retrograde transport, infects
ganglia tissue, and then enters a latent state [23]. VZV resides in both neurons and
satellite cells, unlike herpes simplex virus, which is restricted to neurons [24].
Primary infection occurs typically in childhood when susceptible individuals
inhale airborne virus or directly contact the rash of primary varicella or herpes zos-
ter. VZV is highly contagious and 61–100 % of contacts develop clinical infection
after exposure. Transmission by airborne respiratory droplets occurs 1–2 days before
the development of lesions. Virus can be transmitted from the rash up to a week after
lesions appear [2]. Once lesions are crusted, they are no longer contagious [21].
Herpes zoster results from VZV reactivation following an unknown trigger, pos-
sibly when cell-mediated immunity decreases below a crucial level [15]. Replication
in affected ganglia and anterograde spread via secondary afferent sensory neurons
are associated with inflammation and necrosis of neuronal and non-neuronal cells
[2, 3]. The neuropathic pain associated with shingles is caused by damage to neu-
rons and altered central nervous system (CNS) signal processing [2, 25].
14 Varicella Zoster Virus 97
Primary varicella infection presents with fever and a rash starting on the head and
extending to the trunk and extremities. Adults and adolescents may have a pro-
drome of fever, malaise, and headache 1–2 days before the rash, while in children
the fever and rash develop at the same time. The rash begins as macules and then
evolves through stages of papules, vesicles, and pustules before forming scabs [2].
Pruritus is universal [26]. A unique characteristic is that new lesions appear as older
ones crust; thus lesions will be at different stages [27]. Lesions may involve muco-
sal surfaces [28], and scarring is unusual [2].
Varicella is usually a self-limiting disease, but complications exist and morbidity
and mortality are significantly higher in adults. In 1990–1994, adults had a risk of
dying from varicella 25 times greater than children 1–4 years old, and most people
who died were previously healthy [29]. One severe complication, varicella pneumo-
nia, develops within 6 days of rash onset [2]. In a study of adult men, the mortality
rate was 10 % or 30 % in immunocompetent and immunocompromised people,
respectively [30]. The most common complication is bacterial superinfection (usu-
ally with staphylococcus or streptococcus), which frequently scars, but rarely leads
to septicemia. CNS complications occur in less than 1 out of 1,000 cases and include
encephalitis, meningoencephalitis, acute cerebellar ataxia, and Guillain-Barre
syndrome [2]. Since introduction of the vaccine, the most common neurological
complication is meningitis [31].
If primary varicella is acquired during pregnancy, there is a 10 % risk of devel-
oping pneumonia [32]. Risk to the fetus is highest in the first trimester with a 2.2 %
chance of embryopathy [33]. Defects include hypoplastic limbs, cortical atrophy,
ocular abnormalities, psychomotor retardation, and low birth weight (37). It is
recommended that susceptible women be vaccinated before pregnancy [32]
(Figs. 14.1 and 14.2).
midline, but bilateral zoster has been reported in both immunocompromised and
immunosuppressed patients [2]. Symptoms of zoster along with its serologic or
virologic evidence presenting without cutaneous signs are called zoster sine herpete;
it rarely occurs.
14 Varicella Zoster Virus 99
Fig. 14.3 Herpes zoster in V1 distribution of the trigeminal nerve with contralateral edema
The neuralgia of herpes zoster usually resolves as the lesion crusts fall off [2].
However, 20 % of all patients will experience the most common complication of
herpes zoster post-herpetic neuralgia (PHN) [15]. Under the age of 40, PHN is
uncommon [2], but more than one third of patients over 60 years [2, 34], and 75 %
of those over 75 years will develop PHN [35]. In addition to age, risk factors include
female sex, immunosuppression, and severity of rash and acute pain [36]. The inten-
sity of PHN typically lessens in 1–6 months, but the duration varies [2] (Fig. 14.3).
Reactivation in the ophthalmic division of the trigeminal nerve (V1) occurs in
10–20 % of zoster cases and is known as herpes zoster ophthalmicus (HZO) [37].
The development of ocular disease occurs in 20–70 % of these cases [2], validating
referral to an ophthalmologist [15]. Ocular complications include scleritis, acute
epithelial keratitis, uveitis, chorioretinitis, oculomotor palsies, optic neuritis, and
panophthalmitis secondary to bacterial infection [2, 38]. Impaired corneal sensation
may result in ulceration. Involvement of the second or third divisions of the trigemi-
nal nerve may result in lesions in the mouth, pharynx, larynx, or ears. In Ramsay-
Hunt syndrome, involvement of the facial or auditory nerves may cause symptoms
of vertigo, loss of taste, tinnitus, and otalgia [2]. Cutaneous and visceral dissemina-
tion is rare in immunocompetent individuals (Figs. 14.4, 14.5, and 14.6).
14.4 Work-Up
Most cases of varicella and herpes zoster can be diagnosed on the basis of clinical
history and exam. However, herpes simplex virus (HSV) can be a very good imita-
tor of VZV, especially when it occurs outside of its typical distribution. Differentiation
100 R. Gordon et al.
of VZV from HSV with viral culture is most specific, but it takes 1–2 weeks for
results and the sensitivity is 60–75 % [2, 15]. Serologic testing is limited by possible
preexistence of antibodies to VZV as well as to HSV 1 or 2. In addition, commercial
ELISA tests are usually not sufficiently sensitive to identify the level of immunity
that develops in vaccines [10]. PCR is the most sensitive and specific test [15] and
has been used to detect VZV from skin lesions, peripheral blood, CSF, and other
14 Varicella Zoster Virus 101
tissues from infected patients. Because of their rapid results and high sensitivity,
direct immunofluorescence and PCR are currently the preferred methods of diagno-
sis. Histopathology and cytopathology are not clinically useful in the differentiation
of varicella from herpes zoster or VZV [2].
14.5 Treatment
14.6 Conclusion
References
5. Weller TH, Witton HM, Bell EJ. The etiologic agents of varicella and herpes zoster; isolation,
propagation, and cultural characteristics in vitro. J Exp Med. 1958;108:843–68.
6. Creed R, Satyaprakash A, Ravanfar P. Varicella zoster vaccines. Dermatol Ther. 2009;22:143–9.
7. Shah AP, Smolensky MH, Burau KD, Cech IM, Lai D. Seasonality of primarily childhood and
young adult infectious diseases in the United States. Chronobiol Int. 2006;23:1065–82.
8. Seward JF, Watson BM, Peterson CL, Mascola L, Pelosi JW, Zhang JX, et al. Varicella disease
after introduction of varicella vaccine in the United States, 1995–2000. JAMA. 2002;287:606–11.
9. Roush SW, Murphy TV. Vaccine-preventable disease table working G. Historical comparisons
of morbidity and mortality for vaccine-preventable diseases in the United States. JAMA.
2007;298:2155–63.
10. Gershon AA. Varicella-zoster virus infections. Pediatr Rev. 2008;29:5–10. quiz 1.
11. Guris D, Jumaan AO, Mascola L, Watson BM, Zhang JX, Chaves SS, et al. Changing varicella
epidemiology in active surveillance sites–United States, 1995–2005. J Infect Dis. 2008;197
Suppl 2:S71–5.
12. Choo PW, Donahue JG, Manson JE, Platt R. The epidemiology of varicella and its complica-
tions. J Infect Dis. 1995;172:706–12.
13. Johnson RW. Herpes zoster and postherpetic neuralgia. Expert Rev Vaccines. 2010;9:21–6.
14. Ragozzino MW, Melton 3rd LJ, Kurland LT, Chu CP, Perry HO. Population-based study of
herpes zoster and its sequelae. Medicine (Baltimore). 1982;61:310–6.
15. Lapolla W TS. Clinical decision support in dermatology: herpes zoster. Heyman et al., Elsevier
2011.
16. Buchbinder SP, Katz MH, Hessol NA, Liu JY, O'Malley PM, Underwood R, et al. Herpes
zoster and human immunodeficiency virus infection. J Infect Dis. 1992;166:1153–6.
17. Gnann Jr JW, Whitley RJ. Clinical practice. Herpes zoster. N Engl J Med. 2002;347:340–6.
18. Lee MR, Ryman W. Herpes zoster following cryosurgery. Australas J Dermatol. 2005;46:42–3.
19. Hicks LD, Cook-Norris RH, Mendoza N, Madkan V, Arora A, Tyring SK. Family history as a
risk factor for herpes zoster: a case–control study. Arch Dermatol. 2008;144:603–8.
20. Yawn BP, Wollan PC, Kurland MJ, St Sauver JL, Saddier P. Herpes zoster recurrences more
frequent than previously reported. Mayo Clin Proc. 2011;86:88–93.
21. Heininger U, Seward JF. Varicella. Lancet. 2006;368:1365–76.
22. Taylor SL, Moffat JF. Replication of varicella-zoster virus in human skin organ culture. J Virol.
2005;79:11501–6.
23. Eshleman E, Shahzad A, Cohrs RJ. Varicella zoster virus latency. Future Virol. 2011;6:341–55.
24. Abendroth A, Arvin A. Varicella-zoster virus immune evasion. Immunol Rev. 1999;168:
143–56.
25. Kost RG, Straus SE. Postherpetic neuralgia–pathogenesis, treatment, and prevention. N Engl J
Med. 1996;335:32–42.
26. McCrary ML, Severson J, Tyring SK. Varicella zoster virus. J Am Acad Dermatol. 1999;41:1–
14. quiz 5–6.
27. Cohen JI, Brunell PA, Straus SE, Krause PR. Recent advances in varicella-zoster virus infec-
tion. Ann Intern Med. 1999;130:922–32.
28. Chen TM, George S, Woodruff CA, Hsu S. Clinical manifestations of varicella-zoster virus
infection. Dermatol Clin. 2002;20:267–82.
29. Meyer PA, Seward JF, Jumaan AO, Wharton M. Varicella mortality: trends before vaccine
licensure in the United States, 1970–1994. J Infect Dis. 2000;182:383–90.
30. Weber DM, Pellecchia JA. Varicella pneumonia: study of prevalence in adult men. JAMA.
1965;192:572–3.
31. Pahud BA, Glaser CA, Dekker CL, Arvin AM, Schmid DS. Varicella zoster disease of the
central nervous system: epidemiological, clinical, and laboratory features 10 years after the
introduction of the varicella vaccine. J Infect Dis. 2011;203:316–23.
32. Gnann Jr JW. Varicella-zoster virus: atypical presentations and unusual complications. J Infect
Dis. 2002;186 Suppl 1:S91–8.
33. Pastuszak AL, Levy M, Schick B, Zuber C, Feldkamp M, Gladstone J, et al. Outcome after
maternal varicella infection in the first 20 weeks of pregnancy. N Engl J Med. 1994;330:901–5.
104 R. Gordon et al.
34. Burgoon Jr CF, Burgoon JS, Baldridge GD. The natural history of herpes zoster. J Am Med
Assoc. 1957;164:265–9.
35. Vander Straten M, Carrasco D, Lee P, Tyring SK. Reduction of postherpetic neuralgia in
herpes zoster. J Cutan Med Surg. 2001;5:409–16.
36. Jung BF, Johnson RW, Griffin DR, Dworkin RH. Risk factors for postherpetic neuralgia in
patients with herpes zoster. Neurology. 2004;62:1545–51.
37. Liesegang TJ. Herpes zoster ophthalmicus natural history, risk factors, clinical presentation,
and morbidity. Ophthalmology. 2008;115:S3–12.
38. Liesegang TJ. The varicella-zoster virus: systemic and ocular features. J Am Acad Dermatol.
1984;11:165–91.
39. Lai J, Fay KE, Bocchini JA. Update on childhood and adolescent immunizations: selected
review of US recommendations and literature: part 2. Curr Opin Pediatr. 2011;23:470–81.
40. Kimberlin DW, Whitley RJ. Varicella-zoster vaccine for the prevention of herpes zoster.
N Engl J Med. 2007;356:1338–43.
41. Whitley RJ, Volpi A, McKendrick M, Wijck A, Oaklander AL. Management of herpes zoster
and post-herpetic neuralgia now and in the future. J Clin Virol. 2010;48 Suppl 1:S20–8.
42. Andrei G, Snoeck R. Emerging drugs for varicella-zoster virus infections. Expert Opin Emerg
Drugs. 2011;16(3):507–35.
43. Lapolla W, Digiorgio C, Haitz K, Magel G, Mendoza N, Grady J, et al. Incidence of posther-
petic neuralgia after combination treatment with gabapentin and valacyclovir in patients with
acute herpes zoster: open-label study. Arch Dermatol. 2011;147(8):901–7.
Part III
Variants of Acne Vulgaris
Chapter 15
Acne Conglobata
15.1 Introduction
The term acne conglobata is reserved for the most severe form of inflammatory
acne. While uncommon, the condition presents as a highly inflammatory, extensive
nodulocystic eruption. Acne conglobata is differentiated from acne fulminans by
the former’s lack of systemic symptoms, including fever [1]. Severe disfigurement
and scarring are a common result leading to potential psychological impairment,
including anxiety and depression. Acne conglobata comprises one part of the
follicular occlusion triad, along with perifolliculitis capitis abscedens et suffodiens
(dissecting cellulitis of the scalp) and hidradenitis suppurativa [2].
15.2 Background
in some patients with this most severe form of inflammatory acne, including expo-
sure to halogenated hydrocarbons or ingestion of halogens [3]. Chromosomal
defects have been found in some patients with acne conglobata, namely, an XXY
karyotype in individuals that do not exhibit Klinefelter’s syndrome [5]. PAPA
syndrome, consisting of pyoderma gangrenosum, acne conglobata, and pyogenic
aseptic arthritis, has been mapped to a locus on the long arm of chromosome 15 and
may be associated with mutations CD 2 binding protein 1 (CD2BP1) [6, 7].
Fig. 15.1 Hypertrophic scars on the chest of a young man with acne conglobata (Photo credit:
Joshua A. Zeichner, M.D.)
15 Acne Conglobata 109
15.4 Work-Up
15.5 Treatment
Isotretinoin in a dosage of 0.5–1 mg/kg per day is the treatment of choice for fully
developed acne conglobata [3]. Its effects on remission of nodulocystic lesions are
unparalleled in acne therapeutics. Because isotretinoin can increase inflammation
early in the course of therapy, in the absence of contraindications, the prudent prac-
titioner may also add systemic corticosteroids (prednisone/prednisolone) 40–60 mg
daily with taper over the first 2–4 weeks of therapy. By reducing the overall inflam-
mation and severity of nodules and cysts, corticosteroids may alleviate discomfort
and reduce ultimate scarring. If cultures are positive for pathogenic organisms, anti-
biotics are also indicated, the choice being guided by sensitivity testing. During
isotretinoin therapy, tetracycline antibiotics are relatively contraindicated due to
risks of pseudotumor cerebri. If isotretinoin is contraindicated or refused by the
patient, less optimal therapeutic options include oral antibiotics. Tetracycline, doxy-
cycline, minocycline, clarithromycin, and sulfamethoxazole/trimethoprim would be
good initial choices. Dapsone in dosages of 50–150 mg daily can be used as adjuvant
therapy in patients showing suboptimal responses to either isotretinoin or antibiot-
ics [13]. For patients on antibiotic therapy, topical retinoid treatment may help with
comedonal and microinflammatory lesions.
Procedures that may be indicated to assist with resolution of acne conglobata
include aspiration of cysts/sinuses and injection of intralesional corticosteroids.
Both may be performed in association with either isotretinoin or systemic antibiotic
therapy, either alone or together. Triamcinolone 2.5–3 mg/ml is the authors’ agent
of choice for intralesional injection. Incision and drainage of cysts or excision of
110 J.S. Weiss and E. Wilder
nodules have been previously advocated but should be avoided during isotretinoin
treatment due to poor healing tendencies under the influence of systemic retinoids
[14]. Cryotherapy of nodules has also been advocated by some authors [15]. After
resolution, fractional laser resurfacing, CO2 laser, dermabrasion, and chemical
peeling have been utilized to reduce the appearance of scarring [16].
Psychological implications of acne conglobata cannot be overlooked. Depression,
feelings of shame, and anxiety over one’s appearance all contribute to a decreased
quality of life. Depression can also be a rare but highly publicized side effect of
isotretinoin therapy. At the very least, the treating practitioner should question
patients and/or their family members regarding their psychological state and the
effects that the condition and therapies are having on their lives. Referral to appro-
priate mental health professionals should always be considered.
References
16.1 Introduction
16.2 Background
A patient with acne excoriée most commonly presents as a young, Caucasian female
with excoriated acne and scars. As a result of the self-inflicting nature of the condi-
tion, patients tend to pick at skin regions most easily accessible. Therefore, the
distribution of scars or excoriations over the body can provide a useful clue to clini-
cians. The patient with acne excoriée can have a distribution of lesions resembling
the shape of butterfly wings on the back, referred to as the “butterfly sign” [3]. In the
butterfly sign, there is sparing of the upper, lateral sides of the back bilaterally
resulting from the fact that the patient cannot reach these areas. Similarly, there
tends to be more involvement of the extensor arm as compared to the medial arm
and more involvement of the anterior thigh as compared to the posterior thigh.
Often, patients report a sense of tension immediately prior to picking at their skin
and a sense of relief after the behavior is complete [6].
Patients can present with severe psychosocial impairment. Comorbidities of acne
excoriée include mood and anxiety disorders. Mood disorders are found in 48–68 %
of patients, which include major depression, dysthymia, and bipolar disorders [7, 8].
Anxiety disorders are found in 41–65 % of patients and include generalized anxiety
disorder, agoraphobia, panic disorder, social and more specific phobia, obsessive-
compulsive disorder, and post-traumatic stress disorder [7, 8]. Additionally, if a
patient has a mood or anxiety disorder, he or she frequently has comorbid psychiatric
disorders related to the mood or anxiety disorder, particularly a compulsive-impulsive
spectrum disorder [9], including body dysmorphic disorder, eating disorder, sub-
stance use disorder, or an impulse control disorder, which includes kleptomania,
compulsive buying, and trichotillomania [7, 8, 10]. For very rare patients, acne
excoriée may even present as a manifestation of a delusional disorder [2].
Significant functional impairment is a common occurrence. Patients are often
embarrassed to admit their behavior to a physician. Many report impairment in social
functioning including avoidance of activities that expose their skin to the public, such
as sexual activity, going to the beach, and attending sports and community events [4,
11]. Patients often use cosmetics, bandages, and clothing to hide their excoriations.
16.4 Work-Up
16.5 Treatment
Due to the nature of the disorder, therapy targeting the psyche can help decrease
destructive behavior involved in acne excoriée. For a patient with depression as the
underlying cause of the acne excoriée, an antidepressant with psychotherapy can be
provided [1, 13, 14]. A patient with anxiety as the underlying source for acne
excoriée can use an anti-anxiolytic medication combined with psychotherapy.
Patients with obsessive thoughts and compulsive urges to damage the skin may find
relief through an anti-OCD medication such as paroxetine (Paxil®) and fluoxetine
(Prozac®) along with behavioral therapy to reduce the obsessions and compulsions
[4, 15]. Behavioral therapy is generally thought to be more efficacious for the treat-
ment of OCD than insight-oriented psychotherapy [16]. For mixed depression-OCD
patients, selective serotonin reuptake inhibitors (SSRIs) are the preferred choice of
therapy because of their dual antidepressant and anti-OCD properties [2]. In gen-
eral, SSRIs are commonly used to treat patients in dermatology with psychiatric
comorbidity, especially major depressive disorder [1, 11]. It is important to under-
stand that pharmacologic therapy alone may not be effective if the patient is not
motivated to control the compulsive urges such as a case in which a teenager is
brought in by his/her parents. If the clinician perceives a “power struggle” between
the teenager and the parent over the issue of excoriation, it is often helpful to see the
teenager alone and first try to build therapeutic rapport with the teenage patient.
In this situation, it is important to establish rapport so that the dermatologist is not
seen as another “authority figure” to rebel against.
Case reports have also documented efficacy of pulsed dye laser irradiation along
with cognitive psychotherapy. Treatment of hypertrophic scars and acne lesions
with laser was first introduced with argon laser [17]. In a case series, 585-nm
flashlamp-pumped pulsed dye with concomitant cognitive psychodynamic therapy
16 Acne Excoriée 115
was used to stop skin picking and scar formation in two OCD patients with acne
excoriée [18]. In the cases, practical behavior modification techniques, including
removal of mirrors in the home and avoidance of situations that would induce stress
or conflict, were helpful.
Finally, biofeedback techniques and hypnosis have also been documented to
improve acne excoriée and other dermatoses with a psychological component [19].
Posthypnotic suggestion has also been used to treat the condition. In two case
reports, patients were instructed to remember the word “scar” when they felt the
urge to pick at the face and to say “scar” to refrain from picking. In both cases, acne
excoriée resolved [19]. Aversion therapy techniques and habit reversal have been
noted in case reports as successful strategies for cognitive-behavioral therapy [20–22].
Aversion therapy occurs when self-destructive behavior is linked to an aversive
stimulus. Habit reversal treatment involves making the patient aware of the scratch-
ing behavior, teaching the patient about the negative social impact of the habit, and
developing competing response of isometric exercise using fist clenching to prevent
scratching.
It is important to recognize the clinical presentation and work-up of acne
excoriée, as the underlying source of the condition can be found in the psychopa-
thology rather than in the skin itself. Working closely with the patient to serve his or
her specific needs and establishing a solid therapeutic alliance can significantly
improve outcomes with all treatments.
Acknowledgements Figures 16.1 and 16.2 are courtesy of Joseph Bikowski, M.D.
References
1. Gupta MA, Gupta AK. The use of antidepressant drugs in dermatology. J Eur Acad Dermatol
Venereol. 2001;15:512–8.
2. Koo JYM, Lee CS, editors. Psychocutaneous medicine. New York: Marcel Dekker; 2003.
3. Koo J. Psychodermatology: a practical manual for clinicians. Current problems in dermatol-
ogy 1995; Nov/Dec: 204–32.
4. Arnold LM, Auchenbach MB, McElroy SL. Psychogenic excoriation: clinical features, proposed
diagnostic criteria, epidemiology and approaches to treatment. CNS Drugs. 2001;15:351–9.
5. Shah KN, Fried RG. Factitial dermatosis in children. Curr Opin Pediatr. 2006;18:403–9.
6. Bach M, Bach D. Psychiatric and psychometric issues in acne excoriée. Psychother Psychosom.
1993;60:207–10.
7. Arnold LM, McElroy SL, Mutasim DF, Dwight MM, Lamerson CL, Morris EM. Characteristics
of 34 adults with psychogenic excoriation. J Clin Psychiatry. 1998;59:509–14.
8. Wilhelm S, Keuthen NJ, Deckersbach T, Engelhard IM, Forker AE, Baer L, O’Sullivan RL,
Jenike MA. Self-injurious skin picking: clinical characteristics and comorbidity. J Clin
Psychiatry. 1999;60:454–9.
9. McElroy SL, Phillips KA, Keck PE. Obsessive compulsive spectrum disorder. J Clin
Psychiatry. 1994;55(10 Suppl):33–53.
10. Oldham JM, Hollander E, Skodol AE. Impulsivity and compulsivity. Washington, DC:
American Psychiatric Press; 1996.
11. Simeon D, Stein DJ, Gross S, Islam N, Schmeidler J, Hollander E. A double-blind trial of
fluoxetine in pathologic skin picking. J Clin Psychiatry. 1997;58:509–14.
116 J.W. Millsop and J.Y.M. Koo
12. American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 4th
ed. Washington, DC: American Psychiatric Association; 2000.
13. Kalivas J, Kalivas L, Gilman D, Hayden CT. Sertraline in the treatment of neurotic excoria-
tions and related disorders. Arch Dermatol. 1996;132:589–90.
14. Klobenzer CS. Psychocutaneous disease. New York, NY: Grune and Stratton; 1987.
15. Kearney CA, Silverman WK. Treatment of an adolescent with obsessive-compulsive disorder
by alternating response prevention and cognitive therapy: an empirical analysis. J Behav Ther
Exp Psychiatry. 1990;21:39–47.
16. Mancebo MC, Elsen JL, Sibrava NJ, Dyck IR, Rasmussen SA. Patient utilization of cognitive-
behavioral therapy for OCD. Behav Ther. 2011;42:399–412.
17. Alster TS, Kurban AK, Grove GL, Grove MJ, Tan OT. Alteration of argon laser-induced scars
by the pulsed dye laser. Lasers Surg Med. 1993;13:368–73.
18. Bowes LE, Alster TS. Treatment of facial scarring and ulceration resulting from acne excoriée
with 585-nm pulsed dye laser irradiation and cognitive psychotherapy. Dermatol Surg. 2004;
30:934–8.
19. Shenefelt PD. Biofeedback, cognitive-behavioral methods, and hypnosis in dermatology: is it
all in your mind? Dermatol Ther. 2003;16:114–22.
20. Ratliffe R, Stein N. Treatment of neurodermatitis by behavior therapy: a case study. Behav Res
Ther. 1968;6:397–9.
21. Rosenbaum MS, Ayllon T. The behavioral treatment of neurodermatitis through habit reversal.
Behav Res Ther. 1981;19:313–8.
22. Kent A, Drummond LM. Acne excoriée- a case report of treatment using habit reversal. Clin
Exp Dermatol. 1989;14:163–4.
Chapter 17
Acne Fulminans
17.1 Introduction
Acne fulminans is a rare, severe form of acne vulgaris associated with the acute
onset of systemic symptoms. This entity was first described in 1959 when Burns
and Colville reported a case of acne conglobata with septicemia in a 16-year-old
boy with an acute febrile illness [1]. In 1971, Kelly and Burns described two patients
with a syndrome they termed “acute febrile ulcerative conglobate acne with polyar-
thralgia.” The features of this syndrome included the sudden onset of severe ulcer-
ative acne conglobata without cyst formation, fever, polyarthralgia, failure to
respond to usual antibiotic therapy, and a favorable response to debridement in com-
bination with steroid therapy [2]. In 1975, Plewig and Kligman named this disease
acne fulminans [3]. This rare entity may occur in isolation or as part of the SAPHO
syndrome (synovitis, acne, pustulosis, hyperostosis, and osteitis).
17.2 Background
A. Schram, B.S.
School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
M. Rosenbach, M.D. (*)
Department of Dermatology, University of Pennsylvania, 3600 Spruce Street,
2nd Floor, Maloney Building, Philadelphia, PA 19014, USA
e-mail: [email protected]
unlikely to be the causative mechanism. One theory suggests that acne fulminans is
the manifestation of a severe immunologically mediated hypersensitivity reaction to
Propionibacterium acnes (P. acnes) antigens. Cases of acne fulminans precipitated
by isotretinoin therapy have been described, leading some to postulate that
isotretinoin-induced fragility of the pilosebaceous unit increases the immune
system’s contact with P. acnes antigens [4, 5]. This theory is consistent with data
suggesting that isotretinoin may initially increase superoxide ion and myeloperoxi-
dase release from neutrophils in the early stages of acne treatment [6]. Genetically
determined changes in neutrophil activity may put some individuals at increased
risk for this aberrant immune response. Interestingly, a pair of siblings has been
reported in whom one developed isotretinoin-induced eruptive pyogenic granulo-
mas and the other isotretinoin-induced acne fulminans [5]. There have been reports
of monozygotic twins and HLA-matched siblings who presented at the same age
with similar clinical features, lending support to the role of hereditary factors in the
pathogenesis of this entity [7–9].
Circulating immune complexes have been documented in patients with acne
fulminans, causing some to postulate that the mechanism is that of an autoimmune
disease. Other factors that favor the autoimmune hypothesis include the increase in
γ-globulins and decrease in complement (C3) levels seen in some patients [10–12].
Additionally, patients often respond rapidly to systemic steroids.
Acne fulminans develops almost exclusively in adolescent boys, raising the
question of whether hormonal factors are relevant in the pathogenesis of this condi-
tion. One report describes acne fulminans in three boys treated with testosterone for
excessively tall stature [13]. Additionally, a 21-year-old body builder developed
acne fulminans after taking 4 weeks of testosterone and anabolic steroids; however,
on measurement his serum testosterone levels were found to be within normal limits [14].
Androgenic steroids augment sebum excretion in postpubertal men, leading to an
increase in P. acnes. In some individuals, this may lead to an immunological reac-
tion that manifests as acne fulminans.
The precise etiopathogenesis of this rare entity is uncertain and may involve a
combination of factors including genetic susceptibility (potentially with abnormal
hormone levels or immune response) and a vigorous reaction to bacterial antigens.
Acne fulminans occurs primarily in adolescent, Caucasian males, aged 13–19
years old. While it may occur in women, it is exceptionally rare. The disease usually
begins as mild cystic acne that becomes fulminant after 1–2 years on average,
although de novo presentations with acne fulminans as the initial presentation of
acne have been reported [15].
Patients usually present with a history of mild to moderate acne that suddenly
erupts into spreading, ulcerative acne lesions with intense inflammation and necro-
sis. The lesions most often occur on the chest, and back, or face, and rarely the
17 Acne Fulminans 119
Fig. 17.1 Severe depressed scarring and post-inflammatory erythema in a patient with resolving
acne fulminans
scalp and thighs [15]. The face is usually less severely affected than the trunk, and
isolated lesions on the face are uncommon [4]. The highly inflammatory lesions
initially resemble acne conglobata; however, they quickly form hemorrhagic
nodules and plaques that undergo suppurative degeneration. The resultant lesions
appear as ragged ulcerations with gelatinous, necrotic debris at their bases [10]
(Figs. 17.1, 17.2, and 17.3). In contrast to acne vulgaris, open and closed comedo-
nes are uncommon. These fulminant eruptions are extremely tender and painful.
A broad spectrum of systemic reactions can be seen in patients with acne fulmi-
nans. The majority of cases are accompanied by systemic signs and symptoms, such
as fever, fatigue, malaise, arthralgias, and myalgias. Other systemic findings of
arthritis, myositis, erythema nodosum, hepatosplenomegaly, and aseptic bone
lesions are less common [11, 15–17]. Musculoskeletal pain is often located in the
chest, shoulder girdle, lower back, and large joints [15]. Patients may experience
painful joint swelling in the iliosacral, iliac, and knee joints, resulting in a bent-over
posture when walking. If present, erythema nodosum usually occurs on the shins
[11, 16, 17]. Additionally, a patient has been reported with concurrent posterior
scleritis and pyoderma gangrenosum-like eruptions on the lower legs [18].
17.4 Work-Up
Laboratory findings in acne fulminans are not consistent and may include leukocy-
tosis, thrombocytosis, anemia, proteinuria, microscopic hematuria, and elevated
erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), and liver enzymes.
Occasionally, a leukemoid reaction is present with an increased percentage of
120 A. Schram and M. Rosenbach
17.5 Treatment
The mainstay of treatment in patients with acne fulminans includes local wound
care, supportive systemic care, and systemic corticosteroids. Patients often feel sick
and require bed rest or hospitalization. The first step in management is gentle surgi-
cal debridement and frequent application of warm compresses with 20–40 % urea
solutions to prevent the accumulation of crusts. Urea is also beneficial for its deodor-
ant and antiseptic properties. Other topical treatments, including local antimicrobial
agents, may be used as well. High-potency topical corticosteroids can effectively
decrease inflammation when applied to the ulcerated nodules twice daily for 7–10
days [10]. Pulsed laser has also been reported to improve local control [24].
122 A. Schram and M. Rosenbach
References
1. Burns RE, Colville JM. Acne conglobata with septicemia. Arch Dermatol. 1959;79:361–3.
2. Kelly AP, Burns RE. Acute febrile ulcerative conglobate acne with polyarthralgia. Arch
Dermatol. 1971;104:182–7.
3. Plewig G, Kligman AM. Acne: morphogenesis and treatment. Berlin: Springer; 1975. p. 196.
4. Zaba R, Schwartz RA, Jarmuda S, Czarnecka-Operacz M, Silny W. Acne fulminans: explosive
systemic form of acne. J Eur Acad Dermatol Venereol. 2011;25(5):501–7.
5. Blanc D, Zultak M, Wendling D, et al. Eruptive pyogenic granulomas and acne fulminans in
two siblings treated with isotretinoin: a possible common pathogenesis. Dermatologica.
1988;177(1):16–8.
6. Perkins W, Crocket KV, Hodgins MB, Mackie RM, Lackie JM. The effect of treatment with
13-cis-retinoic acid on the metabolic burst of peripheral blood neutrophils from patients with
acne. Br J Dermatol. 1991;124:429–32.
7. Darley CR, Currey HL, Baker H. Acne fulminans with arthritis in identical twins treated with
isotretinoin. J R Soc Med. 1984;77:328–30.
17 Acne Fulminans 123
18.1 Introduction
18.2 Background
Acne mechanica can occur in several settings. The most commonly recognized
cause of acne mechanica is use of a chinstrap in football [5]. The sweat that accu-
mulates beneath the plastic chin cup macerates the skin that is then subject to con-
tinuous rubbing from speaking, mastication, and facial movement. However, acne
mechanica can be seen on all body areas where football pads are worn, including the
upper back and shoulders [6]. The fact that the pads contacting the skin are caus-
ative has been proven by the initiation of acne mechanica with football activities
and its spontaneous resolution at the end of the season. Acne mechanica needs to be
separated from hormonally induced acne; however, the presentation is similar and
both may occur simultaneously. It should be recognized that acne mechanica can
occur in the absence of traditional acne.
Another common setting for acne mechanica is under the chinstrap in persons
who wear a helmet for equestrian activities [7]. Any helmet or hat that has a chin-
strap could potentially cause problems. Acne mechanica is even seen under the chin
in violin players from rubbing on the chin rest, a condition known as fiddler’s neck
[8]. A variety of acne mechanica has been reported from the repeated trauma of a
comb and brush on the face [9]. Other causes include rubbing from a backpack or
riding in a vehicle for prolonged periods.
18.4 Work-Up
18.5 Treatment
References
19.1 Introduction
N. Zilieris, D.O.
Department of Internal Medicine, Baptist Hospital, Miami Beach, FL, USA
C.J. Gustafson, M.D. (*)
Department of Dermatology, Wake Forest University Baptist Medical Center,
4618 Country Club Road, Winston-Salem, NC 27104, USA
e-mail: [email protected]
S.R. Feldman, M.D., Ph.D.
Department of Dermatology, Pathology, and Public Health Sciences, Wake Forest University
Baptist Medical Center, Winston-Salem, NC, USA
19.2 Background
disparities have been identified [5, 6]. Cushing syndrome occurs in 30 % of people
with Carney complex, a familial form of micronodular hyperplasia of the adrenal
gland that results in an ACTH-independent form of Cushing’s syndrome [7].
Iatrogenic Cushing’s syndrome, due to the exogenous administration of
glucocorticoids, accounts for the majority of Cushing’s syndrome cases. However,
traditional estimates regarding the prevalence of different forms of Cushing’s
syndrome do not include iatrogenic cases. This is largely due to the lack of a uni-
formly accepted definition and/or gold standard diagnostic tests. Despite this fact,
exogenous hypercortisolism accounts for more causes of Cushing’s syndrome than
all causes of endogenous hypercortisolism combined. Although there is no widely
accepted definition, most clinicians agree that exogenous Cushing’s syndrome is
present when symptoms of Cushing’s syndrome develop in individuals treated with
supraphysiological doses of glucocorticoids [8].
Endogenous Cushing’s syndrome results from excessive production of cortisol.
Overproduction of cortisol may occur in three ways: (1) Excess production of
ACTH from the pituitary (Cushing’s disease) accounts for 70 % of these cases.
Although pituitary microadenomas are one of the main sources of excess ACTH
from the pituitary, 40–60 % of patients with Cushing’s disease have no identified
tumor [8]. (2) Ectopic corticotropin-producing tumors (typically bronchogenic or
pancreatic cancer) are the underlying etiology in 15 % of Cushing’s syndrome
patients. However, in infants, adrenal carcinoma is the leading cause of Cushing’s
syndrome [9]. (3) Primary adrenal sources of excess cortisol production (e.g., adre-
nal hyperplasia, adrenal adenomas) account for the remaining 15 % of endogenous
Cushing’s syndrome cases.
described as a fine “cigarette paper” wrinkling and primarily involves the dorsal
surfaces of the hands and elbows [11]. In severe cases, the epidermis can peel off
after being covered with adhesive tape, a finding referred to as the Liddle sign [12].
Likewise, the dermis becomes thin and loose in areas of reduced subcutaneous fat
[13]. Overall, the skin becomes friable and easily damageable with markedly
impaired wound healing. Subsequently, other complications may result, such as
infections (e.g., dermatophyte, bacterial, fungal, and opportunistic) and ulcerations
[14]. Another common cutaneous manifestation of Cushing’s syndrome is plethora
of the face, neck, and chest, which is not accompanied by an increase in red blood
cell concentration.
Acneiform eruptions occur with Cushing’s syndrome because sebaceous gland
activity and sebum production are intricately involved acne formation and are hor-
monally regulated (Fig. 19.1). The hormones specifically related to Cushing’s syn-
drome that affect the sebaceous glands include androgens, CRH, ACTH, and
glucocorticoids [15]. Androgen receptors have been localized to the basal layer of
the sebaceous gland and the outer root sheath of the hair follicle. When activated,
these receptors stimulate the sebaceous gland to secrete sebum, which can poten-
tially clog the follicle and result in pimples [15]. When glucocorticoid excess is
accompanied by androgen excess, acne and oily skin can develop. In Cushing’s
syndrome, acneiform eruptions are typically monomorphic, perifollicular papules
produced by hyperkeratosis of follicular openings typically on the face, chest, and
back. Mild pustule formation can be seen; however, deep cystic lesions and comedo
19 Cushing’s Syndrome 133
19.4 Work-Up
The overnight dexamethasone suppression test is the initial screening test of choice
as it has a sensitivity of 97 % and a specificity of 80 %, if the cortisol level is >3 μg/
dl [3]. A 24-h urine free cortisol level can be used as an alternative screening test. A
level >140 nmol is suggestive of Cushing’s syndrome with 90–95 % specificity and
sensitivity [18]. Definitive diagnosis is confirmed with a standard low-dose dexa-
methasone suppression test done over 48 h. The test is positive when urinary corti-
sol stays >25 nmol/L or plasma cortisol remains >140 nmol/L.
Determining the etiology of Cushing’s syndrome is complicated secondary to the
fact that tests lack specificity. Additionally, tumors producing this syndrome are
prone to spontaneous and often dramatic changes in hormone secretion (periodic
hormonogenesis) [3]. Hence, a combination of laboratory and imaging tests is often
needed to determine the specific cause.
Plasma ACTH levels can be useful in determining the specific etiology of
Cushing’s syndrome, particularly in regard to differentiating ACTH-dependent from
ACTH-independent causes. Evaluating the response of cortisol output upon admin-
istration of high-dose dexamethasone can help distinguish ACTH-secreting pituitary
134 N. Zilieris et al.
19.5 Treatment
References
9. Fauci AS, Braunwald E, Kasper DL, Hauser SL, Longo DL, Jameson JL, Loscalzo J.In:
Disorders of the adrenal cortex (chapter 336). Fauci AS, Braunwald E, Kasper DL, Hauser SL,
Longo DL, Jameson JL, Loscalzo J, editors. Harrison’s Principles of internal medicine, 17th ed.
10. Freinkel RK. Cutaneous manifestations of endocrine diseases. In: Fitzpatrick TB, Eisen AZ,
Wolff K, et al., editors. Dermatology in general medicine. 4th ed. New York: McGraw-Hill;
1993. p. 2113–31.
11. Yanovski JA, Cutler GB. Glucocorticoid action and the clinical features of Cushing’s
syndrome. Endocrinol Metab Clin North Am. 1994;23(3):487–509.
12. Orth DN, Kovacs WJ. The adrenal cortex. In: Wilson JD, Foster DW, Kronenberg HM, et al.,
editors. William’s textbook of endocrinology. 9th ed. Philadelphia: Saunders; 1998. p. 565–9.
13. Rokowski K, Sheehy J, Cutroneo KR. Glucocorticoid-mediated selective reduction of func-
tioning collagen messenger ribonucleic acid. Arch Biochem Biophys. 1981;210:74–81.
14. Findling JW, Raff H. Diagnosis and differential diagnosis of Cushing’s syndrome. Endocrinol
Metab Clin North Am. 2001;30(3):729–47.
15. Lolis MS, Bowe WP, Shalita AR. Acne and systemic disease. Med Clin N Am. 2009;93:
1161–81.
16. Ross EJ, Linch DC. Cushing’s syndrome-killing disease: discriminatory value of signs and
symptoms aiding early diagnosis. Lancet. 1982;2:646–9.
17. Shome B, Saffran M. Peptides of the hypothalamus. J Neurochem. 1966;13(5):433–48.
18. Meier CA, Biller BM. Clinical and biochemical evaluation of Cushing’s syndrome. Endocrinol
Metab Clin North Am. 1997;26:741–62.
19. Winkelmann RK, Scheen RS, Underhal LO. Acanthosis nigricans and endocrine disease.
JAMA. 1960;174(9):1145–52.
Chapter 20
PAPA Syndrome
Abbreviations
20.1 Introduction
The pyogenic sterile arthritis, pyoderma gangrenosum, and acne (PAPA) syndrome
was first recognized in 1997 when ten family members in three generations mani-
fested with variable expression of juvenile-onset arthritis and subsequent presenta-
tion of pyoderma gangrenosum and cystic acne in adolescence and adulthood [1].
This inherited disorder stems from dysregulation of the innate immune system,
presenting with symptoms of seemingly unprovoked episodes of synovial tissue
and skin inflammation [1]. Recurrent inflammation seen in PAPA syndrome
typifies that classically seen in autoinflammatory disorders. Autoinflammatory
disorders, though lacking clear diagnostic criteria, are marked by chronic and
intermittent episodes of fever, cytokine dysregulation, and organ inflammation;
skin and joint involvement is a prominent feature [2, 3]. Unlike autoimmune
diseases, autoinflammatory disorders lack high-titer autoantibodies and autoreac-
tive T lymphocytes [2]. Genes associated with inherited syndromes of autoinflam-
mation encode for protein mediators of apoptosis, inflammation, and cytokine
processing (Table 20.1) [3].
20.2 Background
Five years after the first cases of PAPA was described, a gene mutation associated
with this syndrome was localized to the CD2-binding protein 1 (CD2BP1) gene on
chromosome 15q. CD2BP1 and its murine ortholog, proline-serine-threonine phos-
phatase interacting protein 1(PSTPIP1), are cellular signaling adaptors known to
play integral roles in cellular distribution of proteins, including actin reorganization
[2]. Indeed, cultured macrophages from patients with PAPA syndrome demonstrate
abnormal podosome structures and focal adhesion complexes, leading to defects in
chemotaxis and migration [4].
CD2BP1 also activates innate immune responses by regulating inflammatory cyto-
kine production, especially interleukin-1-beta (IL-1β). Key members of the signaling
cascade upstream of IL-1β activation include pyrin (the susceptibility gene for
Familial Mediterranean fever, FMF) and the Nalp3 inflammasome, a multi-protein
signaling complex comprised of cryopyrin (also known as nucleotide oligomerization
domain-like receptor family pyrin domain containing 3, NLRP3), apoptosis-associ-
ated speck-like protein with a caspase recruitment domain (ASC) and caspase 1[4, 5].
Mutations in CD2BP1 seen in association with PAPA syndrome result in
enhanced IL-1β production through its effects on critical upstream regulatory path-
ways. Specifically, known mutations of CD2BP1 abrogate binding to proline-
glutamic acid-serine-threonine-rich family of protein tyrosine phosphatases
(PTP-PEST), an interaction essential for CD2BP1 dephosphorylation [3, 6].
Hyperphosphorylation of CD2BP1 in turn augments its interaction with pyrin,
downregulating the inhibitory effect of pyrin on activation of the Nalp3 inflamma-
some, the multi-protein signaling complex that mediates the catalytic cleavage of
pro-IL-1β to its active form [4, 7]. While the direct mechanism by which pyrin
interacts with the inflammasome remains unclear, mutations in CD2BP1 associated
with PAPA syndrome may enhance recruitment of CD2BP1 to ASC aggregates,
potentially augmenting inflammasome signaling [8]. An alternative hypothesis pos-
tulates that mutations in CD2BP1 activate pathways leading to cell death and cyto-
kine release, including IL-1β [4, 9]. Taken in sum, mutations in CD2BP1 impact
many important aspects of immunity: immune cell adhesion, migration, activation,
signaling, cytokine production, and apoptosis.
The importance of the IL-1 regulatory pathway in inflammation is highlighted by
the emerging knowledge that several inherited autoinflammatory syndromes are asso-
ciated with IL-1 dysregulation, including FMF, cryopyrin-associated periodic syn-
dromes (CAPS), and deficiency of IL-1 receptor antagonist (DIRA). It is unknown
whether enhanced IL-1 production seen in autoinflammatory diseases results from
aberrant activation (i.e., activation of the pathway in the absence of physiologic trig-
gers such as infection) or an inability to turn off normal inflammatory responses due
to mutations in key signaling or regulatory components [10]. The aforementioned
IL-1 signaling cascade is highly expressed in both skin and synovial tissue, possibly
explaining the common involvement of these tissues in autoinflammatory disorders.
140 F. Liu and K. Shinkai
To date there are eight families with PAPA syndrome reported in the literature,
with a total of 37 affected family members (Table 20.2) [1, 11–17]. The syndrome
is inherited in an autosomal dominant pattern, with clinical manifestations
typically presenting in childhood. The most common clinical feature of PAPA
syndrome is chronic, recurrent, sterile arthritis with prominent neutrophilic syno-
vial infiltrate with onset in childhood. Skin involvement is variable and presents in
adolescence or adulthood after joint signs have resolved or subsided; cutaneous
manifestations include pyoderma gangrenosum, severe acne, and ulcerations
(Figs. 20.1 and 20.2) [1, 16].
Patients with PAPA syndrome present with asymmetric, destructive, and inflamma-
tory poly-arthropathy with onset usually by 5 years of age. The natural history of
this arthritis is chronic and intermittent. Some patients recall an initiating monoar-
ticular traumatic event [1, 13–17]. Recurrent episodes of synovial inflammation
fluctuate with periods of inactivity and eventually enter remission by late adoles-
cence. While the most commonly affected joints are the ankle, knee, and elbow,
cases of jaw and axial involvement, specifically the cervical spine, have been
reported [14]. Diagnostic imaging often reveals diffuse joint space narrowing,
osteophyte formation, subchondral sclerosis, cyst formation, periosteal prolifera-
tion, and ankylosis (Fig. 20.3) [1, 14].
Table 20.2 Clinical features of 37 patients with PAPA syndrome reported in the literature
Family
Family member Arthritis Pyoderma gangrenosum Acne Other
PAPA Syndrome
a a
Family 4 [13] 1 16 Knee, ankle Yes Mild; peri-nasal None
a a a
2 Yes Yes Mild; peri-nasal None
a a a
3 Yes Yes Mild; peri-nasal None
a a a
4 Yes Yes Mild; peri-nasal None
a a a
5 Yes Yes Mild; peri-nasal None
Family 5 [14] 1 5 Cervical spine, Probable Legs Yes Mild Psoriasis
PAPA Syndrome
jaw, hip,
knee, ankle
2 5 Jaw, shoulder, None None Yes Severe Pustular rosacea
hip, knee,
foot
a
3 2 Cervical spine, Yes Yes Severe None
jaw, elbow,
hand, hip,
knee
a
4 Before age 5 Jaw, shoulder, Probable Yes Mild, cystic None
elbow, wrist,
hand, hip,
knee, ankle
5 Before age 5 Jaw, elbow, None None Yes Mild None
wrist, hand,
knee, ankle,
foot
Family 6 [15] 1 Adolescence Elbow, knee, Before age 42 Neck, leg Adolescence Severe; face, upper None
ankle back
a
Family 7 [16] 1 22 Knee 12 Legs 16 Nodulocystic; face
Family 8 [17] 1 3 Ankle None None None None Pustulosis
a
2 33 Ankle None None 18 Abscess,
hidradenitis
suppurativa
a a
3 26 None None 16 Abscess
143
a
Data not reported
144 F. Liu and K. Shinkai
20.4 Work-Up
The unique constellation of clinical features and natural history of PAPA syndrome
often make it a clinical diagnosis. However, fever, arthralgias, elevated markers of
systemic inflammation, acneiform or neutrophilic dermatoses, and ulcers may be
20 PAPA Syndrome 145
Fig. 20.3 X-ray of the hands of a patient with PAPA syndrome demonstrating advanced arthritis
with joint space narrowing, osteophyte formation, and subchondral sclerosis [14]
20.5 Treatment
Given PAPA syndrome is so rare, there is limited evidence supporting the efficacy
of treatments for this condition. Treatments for pyogenic arthritis have been reported
in the literature with variable success. These therapies include aspirin, ibuprofen,
methotrexate, leflunomide, sulfasalazine, hydroxychloroquine, antibiotics, osteot-
omy, and bone grafting. High-dose intra-articular and systemic corticosteroids may
be effective in controlling arthritic flares [1, 11, 13]. Disease-modifying antirheu-
matic drugs (DMARDs) have also been used early in the treatment course in an
attempt to prevent full expression of the PAPA syndrome with inconclusive results.
Patients with aggressive joint disease may require joint replacements [14].
20 PAPA Syndrome 147
Fig. 20.4 Patient with PAPA syndrome and pyoderma gangrenosum of the neck before (a) and
1 month after (b) anakinra therapy [15]
documented in the literature, the available reports suggest that these agents are less
consistently efficacious for acne. There are reports of successful treatment of severe
nodulocystic acne in patients with PAPA with systemic isotretinoin [16, 17].
References
1. Lindor NM, Arsenault TM, Solomon H, Seidman CE, McEvoy MT. A new autosomal domi-
nant disorder of pyogenic sterile arthritis, pyoderma gangrenosum, and acne: PAPA syndrome.
Mayo Clin Proc. 1997;72(7):611–5.
2. Wise CA, Gillum JD, Seidman CE, Lindor NM, Veile R, Bashiardes S, Lovett M. Mutations in
CD2BP1 disrupt binding to PTP PEST and are responsible for PAPA syndrome, an autoinflam-
matory disorder. Hum Mol Genet. 2002;11(8):961–9.
3. Hull KM, Shoham N, Chae JJ, Aksentijevich I, Kastner DL. The expanding spectrum of sys-
temic autoinflammatory disorders and their rheumatic manifestations. Curr Opin Rheumatol.
2003;15(1):61–9.
4. Smith EJ, Allantaz F, Bennett L, Zhang D, Gao X, Wood G, Kastner DL, Punaro M,
Aksentijevich I, Pascual V, Wise CA. Clinical, molecular, and genetic characteristics of PAPA
syndrome: a review. Curr Genomics. 2010;11(7):519–27.
5. McDermott MF. A common pathway in periodic fever syndromes. Trends Immunol. 2004;
25(9):457–60.
6. Veillette A, Rhee I, Souza CM, Davidson D. PEST family phosphatases in immunity, autoim-
munity, and autoinflammatory disorders. Immunol Rev. 2009;228(1):312–24.
7. Goldfinger S. The inherited autoinflammatory syndrome: a decade of discovery. Trans Am
Clin Climatol Assoc. 2009;120:413–8.
8. Waite AL, Schaner P, Richards N, Balci-Peynircioglu B, Masters SL, Brydges SD, Fox M,
Hong A, Yilmaz E, Kastner DL, Reinherz EL, Gumucio DL. Pyrin modulates the intracellular
distribution of PSTPIP1. PLoS One. 2009;4(7):e6147.
9. Yu JW, Fernandes-Alnemri T, Datta P, Wu J, Juliana C, Solorzano L, McCormick M, Zhang Z,
Alnemri ES. Pyrin activates the ASC pyroptosome in response to engagement by autoinflam-
matory PSTPIP1 mutants. Mol Cell. 2007;28(2):214–27.
10. Shinkai K, McCalmont TH, Leslie KS. Cryopyrin-associated periodic syndromes and autoin-
flammation. Clin Exp Dermatol. 2008;33(1):1–9.
11. Wise CA, Bennett LB, Pascual V, Gillum JD, Bowcock AM. Localization of a gene for familial
recurrent arthritis. Arthritis Rheum. 2000;43(19):2041–5.
12. Cortis E, De Benedetti F, Insalaco A, Cioschi S, Muratori F, D’Urbano LE, Ugazio AG.
Abnormal production of tumor necrosis factor (TNF) alpha and clinical efficacy of the TNF
inhibitor etanercept in a patient with PAPA syndrome. J Pediatr. 2004;145(6):851–5.
13. Dierselhuis MP, Frenkel J, Wulffraat NM, Boelens JJ. Anakinra for flares of pyogenic arthritis
in PAPA syndrome. Rheumatology. 2005;44(3):406–8.
14. Tallon B, Corkill M. Peculiarities of PAPA syndrome. Rheumatology. 2006;45(9):1140–3.
15. Brenner M, Ruzicka T, Plewig G, Thomas P, Herzer P. Targeted treatment of pyoderma gangreno-
sum in PAPA (pyogenic arthritis, pyoderma gangrenosum and acne) syndrome with the recombi-
nant human interleukin-1 receptor antagonist anakinra. Br J Dermatol. 2009;161(5):1199–201.
16. Hong JB, Su YN, Chiu HC. Pyogenic arthritis, pyoderma gangrenosum, and acne syndrome
(PAPA syndrome): report of a sporadic case without an identifiable mutation in the CD2BP1
gene. J Am Acad Dermatol. 2009;61(3):533–5.
17. Schellevis MA, Stoffels M, Hoppenreijs EP, Bodar E, Simon A, van der Meer JW. Variable
expression and treatment of PAPA syndrome. Ann Rheum Dis. 2011;70(6):1168–70.
18. Lolis MS, Bowe WP, Shalita AR. Acne and systemic diseases. Med Clin North Am. 2009;
93(6):1161–81.
19. Stichweh DS, Punaro M, Pascual V. Dramatic improvement of pyoderma gangrenosum with
infliximab in a patient with PAPA syndrome. Pediatr Dermatol. 2005;22(3):262–5.
Chapter 21
Polycystic Ovary Syndrome
Joslyn Kirby
21.1 Introduction
Polycystic ovary syndrome (PCOS) was first described in 1935 by Stein and
Leventhal [1]. They described several women with amenorrhea, hirsutism, obesity,
and polycystic ovaries. Today, PCOS is one of the most common endocrine
diseases in women. It is also one of the most common causes of infertility and
menstrual irregularity. Acne is common in women with PCOS and may be the
initial reason women with occult PCOS seek medical attention. Inquiring about
menstrual irregularity and/or hirsutism in women with acne may facilitate referral
to a gynecologist or endocrinologist for early intervention on fertility issues and
medical comorbidities.
21.2 Background
Fig. 21.2 Typical appearance of acne along the chin of the a woman with polycystic ovary syndrome
(Photo credit: Joshua A. Zeichner, M.D.)
Given the high prevalence of obesity, it is not surprising women with PCOS have
an elevated prevalence of other systemic metabolic diseases. Impaired glucose
tolerance is found in 30–40 % [16]. Women are also more likely to develop endo-
metrial hyperplasia and carcinoma [17], lipid abnormalities (including hypertri-
glyceridemia, elevated LDL, and low HDL) [18], obstructive sleep apnea [19], and
cardiovascular disease [20].
152 J. Kirby
21.4 Work-Up
21.5 Treatment
Typical therapies such as topical retinoids, benzoyl peroxide, and topical antibiotics
(singly or fixed-combination products), as well as oral antibiotics, are utilized to
treat acne for women with PCOS. In addition, due to the hyperandrogenism under-
lying the cutaneous findings of PCOS (namely, acne, androgenetic alopecia, and
hirsutism), therapy is aimed at reducing androgen production and the efficacy of
their hormones at their targets (e.g., hair follicle, sebaceous glands) [13].
One of the most common treatments is hormonal birth control. Combination
hormonal birth control, containing both an estrogen and progestin, has been shown
to decrease ovarian production of androgens and increase levels of sex-hormone-
binding globulin [24]. Oral contraceptives with 30–35 mcg of ethinyl estradiol and
a progestin with minimal androgenicity such as norethindrone norgestimate,
desogestrel, or drospirenone are favored [25].
21 Polycystic Ovary Syndrome 153
References
10. Azziz R, Carmina E, Dewailly D, et al. The Androgen Excess and PCOS Society criteria for the
polycystic ovary syndrome: the complete task force report. Fertil Steril. 2009;91(2):456–88.
11. Revised 2003 consensus on diagnostic criteria and long-term health risks related to polycystic
ovary syndrome (PCOS). Hum Reprod. 2004;19(1):41–7.
12. Azziz R, Ehrmann D, Legro RS, et al. Troglitazone improves ovulation and hirsutism in the
polycystic ovary syndrome: a multicenter, double blind, placebo-controlled trial. J Clin
Endocrinol Metab. 2001;86(4):1626–32.
13. Chuan SS, Chang RJ. Polycystic ovary syndrome and acne. Skin Therapy Lett. 2010;
15(10):1–4.
14. Kelekci KH, Kelekci S, Incki K, Ozdemir O, Yilmaz B. Ovarian morphology and prevalence
of polycystic ovary syndrome in reproductive aged women with or without mild acne. Int
J Dermatol. 2010;49(7):775–9.
15. Timpatanapong P, Rojanasakul A. Hormonal profiles and prevalence of polycystic ovary syn-
drome in women with acne. J Dermatol. 1997;24(4):223–9.
16. Legro RS, Kunselman AR, Dodson WC, Dunaif A. Prevalence and predictors of risk for type
2 diabetes mellitus and impaired glucose tolerance in polycystic ovary syndrome: a prospec-
tive, controlled study in 254 affected women. J Clin Endocrinol Metab. 1999;84(1):165–9.
17. Hardiman P, Pillay OC, Atiomo W. Polycystic ovary syndrome and endometrial carcinoma.
Lancet. 2003;361(9371):1810–2.
18. Talbott E, Guzick D, Clerici A, et al. Coronary heart disease risk factors in women with
polycystic ovary syndrome. Arterioscler Thromb Vasc Biol. 1995;15(7):821–6.
19. Gopal M, Duntley S, Uhles M, Attarian H. The role of obesity in the increased prevalence of
obstructive sleep apnea syndrome in patients with polycystic ovarian syndrome. Sleep Med.
2002;3(5):401–4.
20. Orio Jr F, Palomba S, Spinelli L, et al. The cardiovascular risk of young women with polycystic
ovary syndrome: an observational, analytical, prospective case-control study. J Clin Endocrinol
Metab. 2004;89(8):3696–701.
21. Azziz R, Carmina E, Dewailly D, et al. Positions statement: criteria for defining polycystic
ovary syndrome as a predominantly hyperandrogenic syndrome: an Androgen Excess Society
guideline. J Clin Endocrinol Metab. 2006;91(11):4237–45.
22. Derksen J, Nagesser SK, Meinders AE, Haak HR, van de Velde CJ. Identification of virilizing
adrenal tumors in hirsute women. N Engl J Med. 1994;331(15):968–73.
23. Codner E, Villarroel C, Eyzaguirre FC, et al. Polycystic ovarian morphology in postmenarchal
adolescents. Fertil Steril. 2011;95(2):702–6. e701-702.
24. Azzizz R. The evaluation and management of hirsutism. Obstet Gynecol. 2003;101(5):
995–1007.
25. Arowojolu AO, Gallo MF, Lopez LM, Grimes DA, Garner SE. Combined oral contraceptive
pills for treatment of acne. Cochrane Database Syst Rev. 2009;3, CD004425.
26. Goodfellow A, Alaghband-Zadeh J, Carter G, et al. Oral spironolactone improves acne
vulgaris and reduces sebum excretion. Br J Dermatol. 1984;111(2):209–14.
27. Krunic A, Ciurea A, Scheman A. Efficacy and tolerance of acne treatment using both spirono-
lactone and a combined contraceptive containing drospirenone. J Am Acad Dermatol. 2008;
58(1):60–2.
28. Yemisci A, Gorgulu A, Piskin S. Effects and side-effects of spironolactone therapy in women
with acne. J Eur Acad Dermatol Venereol. 2005;19(2):163–6.
29. Cusan L, Dupont A, Gomez JL, Tremblay RR, Labrie F. Comparison of flutamide and spirono-
lactone in the treatment of hirsutism: a randomized controlled trial. Fertil Steril. 1994;
61(2):281–7.
30. Carmina E, Lobo RA. A comparison of the relative efficacy of antiandrogens for the treatment
of acne in hyperandrogenic women. Clin Endocrinol (Oxf). 2002;57(2):231–4.
31. Harborne L, Fleming R, Lyall H, Sattar N, Norman J. Metformin or antiandrogen in the
treatment of hirsutism in polycystic ovary syndrome. J Clin Endocrinol Metab. 2003;
88(9):4116–23.
Chapter 22
Pomade Acne
22.1 Introduction
Pomade acne (aka acne venenata) is considered a clinically distinct entity from the
more common acne vulgaris. It is caused by the use of pomades applied to the hair
and scalp in individuals of African descent with tight curly hair. Pomades are oil- or
ointment-based hair care products used to lubricate the scalp and improve manage-
ability of the hair. Pomade acne consists mainly of uniform, closely set comedones
predominantly of the forehead and temple region. Involvement of the cheeks and
ears has been documented as well [1]. It is said to be associated with little to no
inflammation [2] which is in stark contrast with the comedones of acne vulgaris in
which Halder et al. histologically found a marked inflammatory response in indi-
viduals with skin of color [3]. These lesions are primarily follicular and considered
to be of very slow onset. Characteristically, pomade acne is limited to the come-
donal stage but inflammatory papules, pustules, miliary cyst, and an erythematous
and edematous phase have all been documented [1, 2].
22.2 Background
distinguished from the adolescent type of acne by the age of onset, the uniformity
of the eruption, the overwhelming predominance of comedones, and finally the lack
of seborrhea and lack of involvement of back and chest. Improvement was seen
upon discontinuation of the use of paraffin oil to the scalp as well as the use of a
comedo extractor and benzine to dissolve the oil [1].
In 1970, Plewig et al. described a similar eruption. This was seen in African
American men who applied various grooming substances to the face and scalp. The
observations in this study were confined to Negro male prisoners. The mean age
was 30 but ranged from 21 to 53 years of age. Six products were found to be in com-
mon use among them: Noxzema, Wildroot, Dixie Peach, Royal Crown, and mineral
oil. Common to these are high melting hydrocarbons. In the small study conducted,
five of these pomades were applied to the backs of three African Americans and
three Caucasian subjects for a period of 8 weeks. The formulation was applied every
other day to a 4 cm square area. Occlusion was primarily maintained via the use of
a polyethylene film. The control site consisted of a similar occlusive dressing with-
out application of any agents or vehicles. After 8 weeks, 6 mm full thickness punch
biopsies were obtained. Clinically, only two of the six subjects displayed follicular
papules. The observed lesions were not dense in number and a few were mildly
inflammatory. In decreasing order, Dixie Peach, Wildroot, and Noxzema had the
highest frequency of pomade acne occurrence. Not surprisingly, histologic accumu-
lation of horny material in the follicular lumen was most evident in the three afore-
mentioned formulations [2].
Plewig et al. showed that a number of chemicals can induce an acneiform
eruption upon contact with human skin [2]. Some of these agents contain various
mixtures of petrolatum; lanolin; and vegetable, mineral, or animal oils [2, 4–6].
It has been shown that cultural practices play a role in the development of pomade
acne. Due to the tight and curly nature of the hair of some individuals of skin of
color, hair pomades or “hair grease” and other products are placed on the scalp and
hair regularly for more manageability. These agents are usually rubbed into the
scalp, hair shaft, and at times directly applied to the face. In addition, the many
artistic hairstyles being worn by the same population, requires gels and other prod-
ucts to help keep such styles in place. In one study, skin manifestations related to
cultural practices were seen in more than 20 % of patients and most prevalent were
alopecia and hypertrophic scars followed by pomade acne [7].
The underlying etiopathogenesis is believed to be occlusion of sebaceous and fol-
licular openings [7]. Pomades which have been implicated are usually thought to be
cheaper products in which the acneigenic components are impurities [2]. The question
remains whether other factors such as differences in gland size, sebaceous activity, or
follicular hyperkeratinization among races are predisposing factors. The true etiology
is likely multifactorial. The pomades that typically cause pomade acne are weak
acneigens as it generally takes a year or more of daily use to produce the eruption
clinically [2]. In addition, human skin is not considered conducive or the optimal host
for testing comedogenecity. Plewig et al. were able to produce clinically significant
comedones with all six agents in the ear canal of rabbits [2].
22 Pomade Acne 157
The true prevalence of pomade acne is not known. It is, however, less common
than acne vulgaris and typically limited to patients of skin of color. The eruption
occurs in all age groups and is not restricted to the adolescent or young adult [2].
A survey conducted by Taylor et al. showed that 46.2 % of black individuals admit-
ted to hair pomade use. Of them, 70.3 % presented with lesions clinically consistent
with pomade acne [8]. Likewise, Arfan-aul bari et al. reported pomade acne in 103
South African patients or 3.4 % of their study population [7].
Pomade acne occurs characteristically in African American adults who apply vari-
ous grooming substances to the scalp and hair. The lesions are typically uniform
follicular-based comedones that are in close proximity to one another (Fig. 22.1).
Expression of the contents of lesions with an acne extractor yields a firm material
with a cheesy consistency [2]. Some comedonal lesions show inflammatory changes.
The main ingredients are thought to be materials/hydrocarbons with a high melting
point. These pomades are considered to be weak acneigens as a clinically apparent
eruption isn’t evident after limited use. It generally takes a year or more of daily use
to produce the eruption [2] and this may be in part due to the fact that the finished
products using comedogenic ingredients are not necessarily comedogenic [7].
In addition, human skin is not a very suitable host for testing comedogenicity [2].
The disease is considered to be relatively mild and typically leaves no scars [2]. On
the contrary, due to the inflammatory nature of some of the comedones, pigmentary
alterations have been identified.
22.4 Treatment
Discontinuation of the offending agent can lead to resolution in 4–6 months [2]. So
therefore, a discussion of the use of current hair and skin care products should be
done to determine if these practices are involved in the etiology of pomade acne and
should be avoided [4]. Lighter hair moisturizing agents that are less occlusive, such
as silicone-based products (dimethacone) should replace the thicker pomades [4,
9–12]. In addition, hair mositurizers should be applied to the hair shaft only, avoid
scalp application and the hair should be kept off of the face.
For patients with skin of color, optimal treatment for comedones should include
an agent with anti-inflammatory effects to help combat possible pigmentary altera-
tions. As mentioned earlier, in a study conducted by Halder et al., marked inflam-
mation with infiltrates of polymorphonuclear leukocytes was noted in comedonal
lesions of African Americans. Of note, these findings were not seen in the Caucasian
group that was studied [3]. Topical retinoids are seemingly the drug of choice as it
combats follicular hyperkeratosis and concurrently treats post-inflammatory hyper-
pigmentation (PIH). Other topical regimens solo or in combination with retinoids
include azelaic acid, topical dapsone, salicylic acid, benzoyl peroxide, and topical
antibiotics. Hydroquinone-containing medications should also be considered in the
treatment of acne-induced PIH [10, 12].
Comedone extraction and superficial chemical peels, in particular salicylic acid
peels, are useful adjunctive methods of treating open and closed comedones in
patients with pomade acne [13].
References
1. Berlin C. Acne comedo in children due to paraffin oil applied on the head. AMA Arch Derm
Syphilol. 1954;69(6):683–7.
2. Plewig G, Fulton JE, Kligman AM. Pomade acne. Arch Dermatol. 1970;101(5):580–4.
3. Halder RM, Holmes YC, Bridgeman-Shah S, Kligman AM. A clinicohistopathologic study of
acne vulgaris in black females (abstract). J Invest Dermatol. 1996;106:888.
4. Draelos ZD. Black individuals require special products for hair care. Cosmet Dermatol. 1993;
6:19–20.
22 Pomade Acne 159
23.1 Introduction
While acne has been traditionally considered an adolescent disorder, recent studies
show a significant prevalence among adults. The mean age of presentation for acne
treatment is 24 years with 21 % of acne office visits by patients 25–34 years old and
15 % of these visits by patients 35 years and older [1]. In adults, acne occurs more
frequently among women than men [2]. The characteristic clinical picture is mild to
moderate deep-seated inflammatory papules and nodules on the face, especially the
chin, jawline, and neck. Most commonly, this is “persistent” acne, which began in
adolescence and has continued to adulthood, although a “late-onset” form also
occurs. Recognizing acne in this population and providing treatment is essential
since acne scarring can be correlated with duration of disease [3].
23.2 Background
23.3 Pathophysiology
The reason acne persists into adulthood is multifactorial. The most common form of
adult acne is persistence of acne that began in adolescence, and this form appears to
share the same pathogenic features of increased sebum production, follicular hyper-
keratinization, inflammation, and increased Propionibacterium acnes colonization
as seen in adolescent acne [7]. Women with post-adolescent acne have significantly
higher sebum excretion rate compared to women without acne [8]. Since androgens
play a role in stimulating the sebaceous gland, the roles of both systemic and local
tissue-derived androgens have been explored. While most women with post-
adolescent acne have androgen levels in the normal range, several studies report
lower levels of sex hormone-binding globulin (SHBG) and higher levels of free
testosterone and dehydroepiandrosterone sulfate (DHEA-S) in adult female acne
patients compared to controls [9–11]. However, the severity of the acne is not
positively correlated with these levels [9–12]. In patients with signs of hyperan-
drogenism, the presence of an underlying endocrine disorder such as polycystic
ovarian syndrome or late-onset adrenal hyperplasia should be explored.
The skin and sebaceous gland can produce and metabolize androgens. The
presence of several key steroidogenic enzymes, 3β-hydroxysteroid dehydroge-
nase (3β-HSD), 17β-HSD, and 5α-reductase, enables the conversion of inactive
adrenal precursors to potent androgens in the sebaceous gland. Excess local tissue
androgens produced through increased sebaceous steroidogenic enzyme activity
and therefore increased androgen synthesis has been proposed as a cause of the
elevated sebum production seen in adult acne patients [13–15].
Other etiological factors include genetic predisposition, smoking, and stress. The
role of heredity in acne vulgaris is well established by several twin and cross-
sectional studies [16, 17]. The post-adolescent variant also appears to have a genetic
component since 50 % of patients were found to have a first-degree relative also
with post-adolescent acne [6]. While the correlation between smoking and acne in
the general population remains controversial [18, 19], there appears to be a strong
correlation between smoking and the comedonal post-adolescent (CPAA) variant of
acne in adult females [20]. Nicotine has been shown to have a hyperkeratizing effect
by stimulating the acetylcholine receptors on epidermal keratinocytes while also
being anti-inflammatory and vasoconstrictive. This correlates with the clinical pic-
ture of CPAA, a predominance of micro- and macrocomedones with few inflamma-
tory lesions [21]. Chronic stress activating the hypothalamic-pituitary-adrenal
(HPA) axis has also been purported to exacerbate acne. Activation of HPA axis
leads to both enhanced secretion of adrenal androgens and neuropeptides such as
corticotrophin-releasing hormone (CRH). Recent studies demonstrate that CRH
promotes lipid synthesis in the sebaceous gland and that the CRH system is abun-
dantly expressed in acne-involved skin [22].
While antibacterial resistance of P. acnes and use of cosmetics were initially pro-
posed as potential etiological factors, the current evidence suggests that they do
not play a significant role in the pathophysiology of post-adolescent female acne.
23 Post-adolescent Female Acne 163
When the cutaneous microbiology and serum P. acne antibody levels of patients with
either persistent or late-onset post-adolescent acne were compared to adolescent acne
patients, no differences were found between the acne variants [23]. Initially the use of
cosmetics was the attributed etiological factor in the majority of adult females pre-
senting with a mild acneiform eruption, leading Kligman to coin the condition “acne
cosmestica” [24]. While a variety of cosmetic ingredients are known to be comedo-
genic including lanolin, petrolatum, types of vegetable oils, butyl stearate, lauryl
alcohol, and oleic acid, many cosmetic companies now replace these ingredients with
non-comedogenic alternatives [24, 25]. Recent studies suggest that external factors
such as cosmetics, drugs, and occupation are less important etiological factors [6].
Classically post-adolescent acne has been divided into two clinical types: persistent
and late onset. Persistent acne represents a continuation of adolescent acne into
adulthood. It is the more prevalent of the two types occurring in 82 % of cases [2].
Late-onset acne occurs for the first time after age 25. The characteristic clinical
picture for both types is mild to moderate deep-seated, tender inflammatory papules
predominantly involving the lower third of the face, jaw line and neck. The shoul-
ders and back may also be affected. Comedonal lesions may occur on the forehead
or lateral margins of the face but they are not prominent. Acne flares that occur
premenstrually or at times of increased psychological stress are common.
Several clinical variants have been described. Comedonal post-adolescent acne
(CPAA) has been documented in darker skin types (type III and IV) and is charac-
terized by the predominance of retention lesions and micro- and macrocomedones,
with fewer than 5 % inflammatory lesions. The comedones are homogenously dis-
tributed over the entire face [20]. While acne scarring is usually attributed to inflam-
matory lesions [26], ice pick scars occur in CPAA patients, and in severe cases
numerous ice pick scars can coalesce into carters [20]. “Chin acne” is a clinical
variant in mature females, which is characterized by premenstrual flares of inflam-
matory papules on the chin and perioral region (see Fig. 23.1). “Sporadic acne”
consists of unpredictable sudden outbreaks of inflammatory papules and pustules in
usually one but sometimes several locations in middle aged and older adults. These
outbreaks usually coincide with a systemic illness or surgical operation, although
inciting events are not always found [27].
23.5 Work-Up
While routine endocrinologic evaluation is not indicated for the majority of acne
patients, this evaluation can be helpful in adult women with recalcitrant or late-
onset acne or those with symptoms of hyperandrogenism including irregular
164 G. Heinecke and D. Berson
Fig. 23.1 Pustules and inflammatory papules on the cheeks and chin of an adult woman. She has
been suffering from acne since her teenage years
23.6 Treatment
The general principles of treating female post-adolescent acne are not significantly
different from treating other acne populations, though there are some subtle differ-
ences that should be considered. Older skin tends to be more sensitive to the poten-
tial irritant effects of topical retinoids but more resistant to the irritant effects of
benzoyl peroxide [27]. Acne is a chronic condition for many of these patients with
81 % of women reporting failures with systemic antibiotics and 15–30 % having
23 Post-adolescent Female Acne 165
Table 23.1 Typical results of screening laboratory tests in women with endocrine abnormalities
[30–32, 35]
Endocrine abnormality Typical screening test abnormalities
Polycystic ovarian syndrome LH/FSH ratio greater than 2–3
Serum total testosterone 70–120 ng/dL
Androstenedione 3–5 ng/mL
50 % of women also have elevation in DHEA-S
Late-onset congenital adrenal DHEAS 4,000–8,000 ng/mL
hyperplasia 17-Hydroxyprogesterone greater than 200 ng/dL
Cushing’s syndrome Overnight dexamethasone suppression test >5 μg/100 mL
Ovarian tumor Serum total testosterone greater than 150–200 ng/dL
Normal serum DHEAS
Adrenal tumor DHEAS >8,000 ng/mL
Serum total testosterone greater than 150–200 ng/dL
had recurrence after isotretinoin treatment [30]. Hormonal therapies can be a useful
therapeutic approach and can be effective even in patients with normal serum andro-
gen levels [31]. These include androgen receptor blockers, which block the effect of
the androgens on the sebaceous gland, and inhibitors of androgen production either
by oral contraceptives which block ovarian production or glucocorticoids which
block adrenal production (Table 23.2). Glucocorticoids are most commonly used to
treat patients with late-onset adrenal hyperplasia.
166 G. Heinecke and D. Berson
23.7 Conclusion
Acne vulgaris is a disease that affects not only teenagers but also adults. It can be a
source of significant psychological impairment and interfere with interpersonal and
professional activities. A full work-up is important in evaluating these patients to
rule out potential endocrine abnormalities. While many of the same treatments can
be used in adult women as other patients with acne, an additional option includes
the use of hormonal and anti-androgen therapies.
References
1. McConnell RC, Fleischer Jr AB, Willford PM, Feldman SR. Most topical tretinoin treatment
is for acne vulgaris through the age of 44 years: an analysis of the national ambulatory medical
care survey, 1990–1994. J Am Acad Dermatol. 1998;38:221–6.
2. Goulden V, Stables GI, Cunliffe WJ. Prevalence of facial acne in adults. J Am Acad Dermatol.
1999;41:577–80.
3. Layton AM, Henderson K, Cunliffe WJ. A clinical assessment of acne scarring. Clin Exp
Dermatol. 1994;4:303–8.
4. Cunliffe WJ, Gould DJ. Prevalence of facial acne vulgaris in late adolescence and in adults. Br
Med J. 1979;1:1109–10.
5. Collier CN, Harper JC, Cafardi JA, et al. The prevalence of acne in adults 20 years and older.
J Am Acad Dermatol. 2008;58:56–9.
6. Goulden V, Clark SM, Cunliffe WJ. Post-adolescent acne: a review of clinical features. Br
J Dermatol. 1997;136:66–70.
7. Gollnick H, Cunliffe W, Berson D, et al. Management of acne: a report from a global alliance
to improve outcomes in acne. J Am Acad Dermatol. 2003;49:S1–37.
8. McGeown CH, Goulden V, Holland DB, et al. Sebum excretion rate in post-adolescent acne
compared to controls and adolescence acne. J Invest Dermatol. 1997;108:386.
9. Aizawa H, Niimura M. Adrenal androgen abnormalities in women with late onset and persis-
tent acne. Arch Dermatol Res. 1993;284:451–5.
168 G. Heinecke and D. Berson
10. Darley CR, Kirby JD, Besser GM et al. Circulating testosterone, sex hormone binding globulin
and prolactin in women with late onset or persistent acne vulgaris
11. Seirafi H, Farnaghi F, Vasheghani-Farahani A, et al. Assessment of androgens in women with
adult-onset acne. Int J Dermatol. 2007;46:1188–91.
12. Cibula D, Hill M, Vohradnikova O, Kuzel D, Fanta M, Zivny J. The role of androgens in
determining acne severity in adult women. Br J Dermatol. 2000;143:399–404.
13. Lookingbill DP, Horton R, Demers LM, Egan N, Marks Jr JG, Santen RJ. Tissue production of
androgens in women with acne. J Am Acad Dermatol. 1985;12:481–7.
14. Carmina E, Lobo RA. Evidence for increased androsterone metabolism in some normandro-
genic women with acne. J Clin Endocrinol Metab. 1993;76:1111–4.
15. Carmina E, Godwin AJ, Stanczyk FZ, Lippman JS, Lobo RA. The association of serum
androsterone glucuronide with inflammatory lesions in women with adult acne. J Endocrinol
Invest. 2002;25:765–8.
16. Cunliffe WJ. Natural history of acne. In: Plewig G, Marks R, editors. Acne and related
disorders. London: Martin Dunitz; 1989. p. 4–6.
17. Ballanger F, Baudry P, N’Guyen JM, Khammari A, Dréno B. Heredity: a prognostic factor for
acne. Dermatology. 2006;212:145–9.
18. Schäfer T, Nienhaus A, Vieluf D, Berger J, Ring J. Epidemiology of acne in the general
population: the risk of smoking. Br J Dermatol. 2001;145:100–4.
19. Mills CM, Peters TJ, Finlay AY. Does smoking influence acne? Clin Exp Dermatol.
1993;18:100–1.
20. Capitanio B, Sinagra JL, Bordignon V, Cordiali Fei P, Picardo M, Zouboulis CC. Underestimated
clinical features of post-adolescent acne. J Am Acad Dermatol. 2010;63:782–8.
21. Misery L. Nicotine effects on the skin: are they positive or negative? Exp Dermatol. 2004;
13:665–70.
22. Ganceviciene R, Graziene V, Fimmel S, Zouboulis CC. Involvement of the corticotropin-releasing
hormone system in the pathogenesis of acne vulgaris. Br J Dermatol. 2009;160:345–52.
23. Till AE, Goulden V, Cunliffe WJ, Holland KT. The cutaneous microflora of adolescence,
persistent and late-onset acne patients does not differ. Br J Dermatol. 2000;142:885–92.
24. Kligman AM, Mills Jr OH. Acne Cosmetica. Arch Dermatol. 1975;106:843–50.
25. Nelson FP, Rumsfield J. Cosmetics. Content and function. Int J Dermatol. 1988;27:665–72.
26. Rivera AE. Acne scarring: a review and current treatment modalities. J Am Acad Dermatol.
2008;59:659–76.
27. Marks R. Acne and its management beyond the age of 35 years. Am J Clin Dermatol. 2004;
5:459–62.
28. Strauss JS, Krowchuk DP, Leyden JJ, et al. Guidelines of care for acne vulgaris management.
J Am Acad Dermatol. 2007;56:651–63.
29. Goudas VT, Dumesic DA. Polycystic ovary syndrome. Endocrinol Metab Clin North Am.
1997;26:893–912.
30. Ebede TL, Arch EL, Berson D. Hormonal treatment of acne in women. J Clin Aesthetic
Dermatol. 2009;2:16–22.
31. Thiboutot D. Acne: hormonal concepts and therapy. Clin Dermatol. 2004;22:419–28.
32. Lobo RA. Hyperandrogenism: physiology, etiology, differential diagnosis, management.
In: Katz VL, Lentz GM, Lobo RA, Gershenson DM, editors. Comprehensive gynecology. 5th
ed. Philadelphia, PA: Elsevier; 2007. p. 983–92.
33. Goodfellow A, Alaghband-Zadeh J, Carter G, et al. Oral spironolactone improves acne vulgaris
and reduces sebum excretion. Br J Dermatol. 1984;111:209–14.
34. Saint-Jean M, Ballanger F, Nguyen JM, Khammari A, Dreno B. Importance of spironolactone
in the treatment of acne in adult women. J Eur Acad Dermatol Venereol. 2011;25:1480–1.
35. George R, Clarke S, Thiboutot D. Hormonal therapy for acne. Semin Cutan Med Surg. 2008;
27:188–96.
36. Frangos JE, Alavian CN, Kimball AB. Acne and oral contraceptives: update on women’s
health screening guidelines. J Am Acad Dermatol. 2008;58:781–6.
Chapter 24
SAPHO Syndrome
24.1 Introduction
SAPHO syndrome is a chronic rheumatologic disorder that involves the skin, bone,
and joints. The name SAPHO is an acronym for the primary manifestations
associated with this disorder: synovitis, acne, pustulosis, hyperostosis, and osteitis.
It is listed by the National Institutes of Health as a rare disease, but it is an important
one for physicians to include in their differential diagnosis for acne as it is often
misdiagnosed and under-recognized.
24.2 Background
The clinical course of the SAPHO syndrome has great variability between patients.
Proposed diagnostic criteria require one of the three following criteria [8]:
1. Chronic recurrent multifocal osteomyelitis
(a) Usually sterile
(b) Spine may be involved
(c) With or without skin condition
24 SAPHO Syndrome 171
though rarely, involves the pelvis and the long bones. In children and young adults,
osteomyelitis has a preference for the anterior chest wall, particularly the clavicle,
and often displays multiple, symmetric lesions along the metaphyses of long bones.
Chronic recurrent multifocal osteomyelitis (CRMO), a chronic nonbacterial
osteomyelitis, manifests as recurrent flares of inflammatory bone pain, typically
involving the metaphyses of long bones, and may present with or without fever.
Some consider CRMO to be the pediatric equivalent of SAPHO syndrome or at
least in the same spectrum of disease [11, 12].
Though usually confined to the skin, bone, and joints, systemic symptoms such
as fever sometimes occur [13]. Inflammatory bowel disease, both Crohn’s disease
and ulcerative colitis, have been associated [3, 14].
24.4 Work-Up
Laboratory blood tests are often useful but rarely diagnostic in SAPHO syndrome.
The erythrocyte sedimentation rate (ESR) is usually elevated, indirectly indicative
of the inflammation associated with synovitis, acne, pustulosis, and osteitis. The
ESR is a screening test and cannot be used to diagnose SAPHO syndrome but may
be used for monitoring the degree of inflammation in patients. Similarly, elevated
C-reactive protein (CRP) values may also indicate degree of inflammation in the
condition.
Definitive diagnosis is difficult and often requires confirmation by scintigraphy
or histopathology of bony lesions. Bone metastasis cannot completely be ruled out
by scintigraphy alone, and fluorodeoxyglucose positron emission tomography
(FDG-PET) is used for definitive exclusion of metastatic bone tumors [15, 16].
Bone scans, CT scans, and even full-body MRI are also helpful diagnostically in
establishing sites of bone involvement. Radiological features of SAPHO syndrome
are mainly hyperostosis and osteitis. Multiple skeletal sites may be involved and the
lesions can occur simultaneously or successively. Hyperostosis is characterized by
a chronic periosteal reaction and cortical thickening that results in narrowing of the
medullary canal and leads to hypertrophy.
Bone biopsy may be performed to confirm a diagnosis of CRMO or to distin-
guish unifocal CRMO from a tumor or infection. Histopathological analysis of the
biopsy specimen may show Paget-like hyperostosis and inflammatory cell infiltra-
tion. Inflammation characterizes the acute phase, which shows polymorphonuclear
leukocytes, plasma cells, edema, and prominent periostitis. The later phase has less
evident inflammation but displays enlarged and sclerotic trabeculae and increased
marrow fibrosis.
Skin biopsy will reveal the histopathologic findings characteristic of the associ-
ated skin finding such as acne or pustular psoriasis. Polymorphonuclear leukocytes
characteristically predominate the various skin lesions associated with SAPHO
syndrome.
24 SAPHO Syndrome 173
24.5 Treatment
Treatment for SAPHO syndrome is based on the severity of presentation and gener-
ally aimed at reducing the associated inflammation in the affected organ system,
bone, or skin. Treatment options and effects are listed in Table 24.2.
If severe acne is seen, prompt and aggressive systemic treatment with oral tetra-
cyclines or isotretinoin should be started to prevent scarring. The addition of oral
corticosteroids may be necessary to control inflammation when starting isotretinoin.
Etanercept has been used in severe refractory cases.
For the psoriatic manifestations (palmoplantar pustulosis, pustular psoriasis, or
pustular psoriasis), topical corticosteroids may be used to control mild and limited
presentations. The addition of phototherapy with narrowband UVB is typically con-
sidered second-line therapy for most patients with psoriatic skin lesions. For exten-
sive, severe, or refractory disease, oral retinoids, methotrexate, or anti-TNF-α agents
or PUVA could be employed depending on the age of the patient and comorbid
factors as well as the severity of joint disease. Combinations of these therapies may
be required in those patients unresponsive to monotherapy.
For the joint disease, a variety of standard therapies are used. Nonsteroidal
anti-inflammatory drugs (NSAIDs), such as aspirin, ibuprofen, and naproxen are
first-line drugs used to provide relief from bone pain. Corticosteroids, including oral
or intralesional injection, as well as topical cold applications, are also used to treat
the inflammatory symptoms. Successful prolonged antibiotic therapy has also been
reported for cases where bone biopsy is positive for P. acnes, but flaring occurs with
discontinuation [17]. Sulfasalazine and methotrexate are used for more severe cases
or for patients with persistent joint pain. Etanercept, an anti-TNF-α biological agent,
has shown success in several case reports as well [18, 19]. Of note, life-threatening
disseminated tuberculosis was observed as a complication of TNF-α blockade in a
17-year-old female with SAPHO, highlighting the need for careful screening of
patients prior to initiating systemic biologic therapies [20]. Combination therapy is
often required to control the disease. Pamidronate or other bisphosphonates have
demonstrated success in some uncontrolled series and case reports in preventing
bone loss [21].
24.6 Conclusion
References
1. Karadag-Saygi E, et al. SAPHO syndrome: misdiagnosed and operated. Acta Reumatol Port.
2008;33(4):460–3.
2. Van Doornum S, et al. SAPHO: rare or just not recognized? Semin Arthritis Rheum.
2000;30(1):70–7.
3. Hayem G, et al. SAPHO syndrome: a long-term follow-up study of 120 cases. Semin Arthritis
Rheum. 1999;29(3):159–71.
4. Hurtado-Nedelec M et al. Genetic susceptibility factors in a cohort of 38 patients with SAPHO
syndrome: a study of PSTPIP2, NOD2, and LPIN2 genes. J Rheumatol. 2010;37(2):401–9.
5. Ferguson PJ, et al. Neutrophil dysfunction in a family with a SAPHO syndrome-like pheno-
type. Arthritis Rheum. 2008;58(10):3264–9.
6. Colina M, et al. Propionibacterium acnes and SAPHO syndrome: a case report and literature
review. Clin Exp Rheumatol. 2007;25(3):457–60.
7. Hurtado-Nedelec M, et al. Characterization of the immune response in the synovitis, acne,
pustulosis, hyperostosis, osteitis (SAPHO) syndrome. Rheumatology. 2008;47(8):1160–7.
8. Kahn M-F, Khan MA. The SAPHO syndrome. Baillière's Clin Rheumatol. 1994;8(2):333–62.
9. Bachmeyer C, et al. Overlapping neutrophilic dermatosis in a patient with SAPHO syndrome.
Arch Dermatol. 2007;143(2):275–6.
24 SAPHO Syndrome 175
10. Davies AM, et al. SAPHO syndrome: 20-year follow-up. Skeletal Radiol. 1999;28(3):159–62.
11. Earwaker JW, et al. SAPHO: syndrome or concept? Imaging findings. Skeletal Radiol. 2003;
32(6):311–27.
12. Tlougan BE, et al. Chronic recurrent multifocal osteomyelitis (CRMO) and synovitis, acne,
pustulosis, hyperostosis, and osteitis (SAPHO) syndrome with associated neutrophilic dermato-
ses: a report of seven cases and review of the literature. Pediatr Dermatol. 2009;26(5):497–505.
13. Beretta-Piccoli BC, et al. Synovitis, acne, pustulosis, hyperostosis, osteitis (SAPHO) syndrome
in childhood: a report of ten cases and review of the literature. Eur J Pediatr. 2000; 159(8):
594–601.
14. Colina M, et al. Clinical and radiologic evolution of synovitis, acne, pustulosis, hyperostosis,
and osteitis syndrome: a single center study of a cohort of 71 subjects. Arthritis Rheum.
2009;61(6):813–21.
15. Quirico Rodriguez M et al. The importance of bone scintigraphy in the diagnosis of SAPHO
syndrome. Rev Esp Med Nucl. 2010;29(3):127–30.
16. Salles M et al. The SAPHO syndrome: a clinical and imaging study. Clin Rheumatol.
2011;30(2):245–9.
17. Assmann G, et al. Efficacy of antibiotic therapy for SAPHO syndrome is lost after its
discontinuation: an interventional study. Arthritis Res Ther. 2009;11(5):R140.
18. Vilar-Alejo J et al. SAPHO syndrome with unusual cutaneous manifestations treated success-
fully with etanercept. Acta Derm Venereol. 2010;90(5):531–2.
19. Ben Abdelghani K et al. Tumor necrosis factor-alpha blockers in SAPHO syndrome.
J Rheumatol. 2010;37(8):1699–704.
20. Hess S et al. Life-threatening disseminated tuberculosis as a complication of TNF-α blockade
in an adolescent. Eur J Pediatr. 2011;170(10):1337–42.
21. Ichikawa J, et al. Successful treatment of SAPHO syndrome with an oral bisphosphonate.
Rheumatol Int. 2009;29(6):713–5.
Part IV
Genetic Syndromes Mimicking
Acne Vulgaris
Chapter 25
Apert Syndrome
25.1 Introduction
25.2 Background
25.4 Work-Up
25.5 Treatment
continuous positive airway pressure (CPAP) machines and placement of ear tubes
must be performed. Finally behavioral and psychological interventions may be
necessary for social and intellectual impairments.
For cutaneous disease, topical therapy may suffice in mild to moderate cases of
acne. In resistant or more severe patients, oral therapies may be added. Oral isotreti-
noin has been reported to be successful in treating severe, nodulocystic acne in
patients with Apert syndrome [18–22]. One published report describes a female
patient with Apert syndrome put on an oral contraceptive pill to treat a large ovarian
cyst, who experienced a significant improvement of her acne [23].
References
1. Cunningham ML, Seto ML, Ratisoontorn C, Heike CL, Hing AV. Syndromic craniosynostosis:
from history to hydrogen bonds. Orthod Craniofac Res. 2007;10(2):67–81.
2. Wheaton SW. Two specimens of congenital cranial deformity in infants associated with fusion
of the fingers and toes. Trans Pathol Soc Lond. 1894;45:238.
3. Steffen C. The man behind the eponym. Eugene Apert (1868–1940). Am J Dermatopathol.
1984;6:223–4.
4. Boulet SL, Rasmussen SA, Honein MA. A population-based study of craniosynostosis in
metropolitan Atlanta, 1989–2003. Am J Med Genet A. 2008;146A:984–91.
5. Blank CE. Apert’s syndrome (a type of acrocephalosyndactyly)—observations on British
series of 39 cases. Ann Hum Genet. 1950;24:151–63.
6. Melnik BC. Role of FGFR2-signaling in the pathogenesis of acne. Dermatoendocrinol.
2009;1(3):141–56.
7. Melnik BC. FoxO1- the key for the pathogenesis and therapy of acne? J Dtsch Dermatol Ges.
2010;8:105–14.
8. Melnik BC. Is nuclear deficiency of FoxO1 due to increased growth factor⁄PI3K⁄Akt-signalling
in acne vulgaris reversed by isotretinoin treatment? Br J Dermatol. 2010;162:1398–400.
9. Wilkie AO, Slaney SF, Oldridge M, et al. Apert syndrome results from localized mutations of
FGFR2 and is allelic with Crouzon syndrome. Nat Genet. 1995;9:165–72.
10. Melnik BC, Schmitz G, Zouboulis CC. Anti-acne agents attenuate FGFR2 signal transduction
in acne. J Investig Dermatol. 2009;129:1868–77.
11. Chen W, Obermayer-Pietsch B, Hong JB, et al. Acne-associated syndromes; models for better
understanding of acne pathogenesis. J Eur Acad Dermatol Venereol. 2011;25:637–46.
12. Henderson CA, Knaggs H, Clark A, Highet AS, Cunliffe WJ. Apert’s syndrome and androgen
receptor staining of the basal cells of sebaceous glands. Br J Dermatol. 1995;132:139–43.
13. Freiman A, Tessler O, Barankin B. Apert syndrome. Int J Dermatol. 2006;45:1341–3.
14. Cohen MM, Kreiborg S. Cutaneous manifestations of Apert syndrome. Am J Med Genet.
1995;58:94–6.
15. Cohen Jr MM, Kreiborg S. Visceral anomalies in the Apert syndrome. Am J Med Genet.
1993;45:758–60.
16. Solomon LM, Fretzin D, Pruzansky S. Pilosebaceous abnormalities in Apert’s syndrome. Arch
Dermatol. 1970;102:381–5.
17. Atherton DJ, Rebello T, Wells RS. Apert’s syndrome with severe acne vulgaris. Proc Roy Soc
Med. 1976;69:517–8.
18. Downs AMR, Condon CA, Tan R. Isotretinoin therapy for antibiotic-refractory acne in Apert’s
syndrome. Clin Exp Dermatol. 1999;24:461–3.
19. Robinson D, Wilms NA. Successful treatment of the acne of Apert syndrome with isotretinoin.
J Am Acad Dermatol. 1989;21:315–6.
182 Y. Albahrani and J.A. Zeichner
20. Campanati A, Marconi B, Penna L, Paolinelli M, Offidani A. Pronounced and early acne in
Apert’s syndrome: a case successfully treated with oral isotretinoin. Eur J Dermatol. 2002;
12:496–8.
21. Gilaberte M, Puig L, Alomar A. Isotretinoin treatment of acne in a patient with Apert
syndrome. Pediatr Dermatol. 2003;20:443–6.
22. Parker TL, Roth JG, Esterly NB. Isotretinoin for acne in Apert syndrome. Pediatr Dermatol.
1992;9:298–300.
23. Hsieh T, Ho N. Resolution of acne following therapy with an oral contraceptive in a patient
with Apert syndrome. J Am Dermatol. 2005;53(1):173–4.
Chapter 26
Birt-Hogg-Dubé Syndrome
26.1 Introduction
26.2 Background
The gene locus for BHD was identified in 2001, localized to chromosome 17p11.2
by linkage analysis [5, 6]. Nickerson and colleagues later described a truncating
germline mutation in a novel gene, the FLCN (BHD) gene, coding for a protein of
unknown function called folliculin (FLCN) [7]. Chromosome 17p11.2 contains 14
exons and encodes folliculin. This evolutionary conserved protein of 579 amino
acids that has no major homology to any other human protein and its function is
unknown. Tumor suppressor function for FLCN was noted in BHD-associated
renal tumors, consistent with somatic second-hit mutations [8], and loss of FLCN
mRNA expression was found in renal tumors from patients with BHD [9].
Interestingly, Van Steensel and colleagues did not detect loss of FLCN mRNA in
fibrofolliculomas, suggesting that mechanisms of tumorigenesis might differ in
renal and skin tumors [10].
Pathogenesis of Birt-Hogg-Dubé syndrome has also been associated with the
energy-sensing mammalian target of rapamycin (mTOR) pathway [11, 12]. A 130-
kDa FLCN-interacting protein, FNIP1, interacts with 5′AMP-activated protein
kinase (AMPK), a protein involved in the mTOR pathway [13]. Hasumi and col-
leagues and Tagaki and colleagues showed an interaction between an FNIP homo-
logue, FNIP2, with FLCN and AMPK [14, 15].
Some clinical overlap between BHD and tuberous sclerosis exists, such as skin
hamartomas, pulmonary cysts, pneumothorax, and renal tumors [4]. The tuberous
sclerosis complex genes TSC1 and TSC2 encode proteins that regulate the mTOR
pathway. It has been shown that downregulation of FLCN leads to mTOR inhibition
and downregulation of TSC proteins leads to mTOR activation [16]. Therefore, it
seems likely that folliculin has several functions and its role in the mTOR pathway
is still under investigation [4].
Birt-Hogg-Dubé is a rare condition and its epidemiology is largely unknown.
Some authors suggest that it may be underdiagnosed [17].
Fig. 26.1 Monomorphic, flesh-colored to white, dome-shaped papules on the face of a patient
with Birt-Hogg-Dubé syndrome. Histologically these lesions reveal fibrofolliculomas and
trichodiscomas, which can be difficult to distinguish clinically
The risk of a renal neoplasm in patients with BHD syndrome ranges from 20 to
30 %, with equal male and female distribution [20, 21]. This differs from general
population, where renal cell carcinoma is more prevalent in men. The average age
of presentation is around 50 year old, which is younger than patients with spontane-
ous renal cell carcinoma usually present [22]. Multiple renal cysts have also been
reported in patients with BHD and these are usually seen after the age of 40 [23].
186 K. Goldenberg and G. Goldenberg
FLCN is thought to be a tumor suppressor gene in the kidneys [18]. Somatic mutations
or loss of heterozygosity of the FLCN gene causes different renal tumors in patients with
BHD [8]. Therefore, BHD differs from other hereditary renal cancer syndromes in the
diversity of histological subtypes of renal cell tumors these patients develop.
The most common and characteristic tumor types in BHD syndrome are hybrid
chromophobe/oncocytic (50 %) and chromophobe (33 %) renal cancer, clear cell
carcinomas (9 %), oncocytoma (5 %), and papillary carcinomas [24]. These tumors
can be solitary and unilateral or multifocal and bilateral.
26.4 Work-Up
26.5 Treatment
The treatment options for patients with fibrofolliculomas and trichodiscomas are
limited. Treatment is largely surgical, with excision, shave and cautery, electrodes-
sication, and curettage reported [4]. Laser ablation with erbium-YAG or fractional
CO2 laser has also been reported [26, 27]. Although the treatment is not curative, the
psychological burden of multiple facial lesions should be conserved and treatment
offered to the patients.
Patients with known BHD syndrome should undergo an annual renal MRI, with
renal ultrasound also an option [18]. Surveillance for renal tumors is also indi-
cated for carriers of FLCN germline mutations and for relatives of patients with
known BHD syndrome. Once renal cancer is diagnosed, staging and treatment
should follow standard procedure [28]. It has also been suggested that rapamycin
and its derivatives can be used, since FLCN mutation can result in deregulation on
mTOR [18].
26.6 Conclusion
Skin changes may be the first manifestation of the Birt-Hogg-Dubé syndrome. This
condition affects several organ systems in the body, and recognition of the unique
presenting skin changes can lead to early diagnosis and work-up. Given the poten-
tially severe renal and pulmonary manifestations, full work-ups should be per-
formed for patients in which this condition is suspected. Referral to the appropriate
specialists, including genetic counselors, is important to properly manage this
syndrome.
188 K. Goldenberg and G. Goldenberg
References
1. Toro JR, Glenn G, Duray P, Darling T, Weirich G, Zbar B, Linehan M, Turner ML. Birt-Hogg-
Dubé syndrome: a novel marker of kidney neoplasia. Arch Dermatol. 1999;135(10):1195–202.
2. Warwick G, Izatt L, Sawicka E. Renal cancer associated with recurrent spontaneous pneumo-
thorax in Birt-Hogg-Dubé syndrome: a case report and review of the literature. J Med Case
Rep. 2010;4:106.
3. Birt AR, Hogg GR, Dube WJ. Hereditary multiple fibrofolliculomas with trichodiscomas and
acrochordons. Arch Dermatol. 1977;113:1674–7.
4. Menko FH, van Steensel MA, Giraud S, Friis-Hansen L, Richard S, Ungari S, Nordenskjöld
M, Hansen TV, Solly J, Maher ER, European BHD Consortium. Birt-Hogg-Dubé syndrome:
diagnosis and management. Lancet Oncol. 2009;10(12):1199–206.
5. Khoo SK, Bradley M, Wong FK, Hedblad M-AM, Nordenskjöld BT. The Birt-Hogg-Dubé
syndrome: mapping of a novel hereditary neoplasia gene to chromosome 17p12–q11.2.
Oncogene. 2001;20:5239–42.
6. Schmidt LS, Warren MB, Nickerson ML, et al. Birt-Hogg-Dubé syndrome, a genodermatosis
associated with spontaneous pneumothorax and kidney neoplasia, maps to chromosome
17p11.2. Am J Hum Genet. 2001;69:876–82.
7. Nickerson ML, Warren MB, Toro JR, et al. Mutations in a novel gene lead to kidney tumors,
lung wall defects, and benign tumors of the hair follicle in patients with the Birt-Hogg-Dubé
syndrome. Cancer Cell. 2002;2:157–64.
8. Vocke CD, Yang Y, Pavlovich CP, et al. High frequency of somatic frameshift BHD gene muta-
tions in Birt-Hogg-Dubé-associated renal tumors. J Natl Cancer Inst. 2005;97:931–5.
9. Warren MB, Torres-Cabala CA, Turner ML, et al. Expression of Birt-Hogg-Dubé gene mRNA
in normal and neoplastic human tissues. Mod Pathol. 2004;17:998–1011.
10. van Steensel MA, Verstraeten VL, Frank J, et al. Novel mutations in the BHD gene and absence
of loss of heterozygosity in fibrofolliculomas of Birt-Hogg-Dubé patients. J Invest Dermatol.
2007;127:588–93.
11. Inoki K, Corradetti MN, Guan K-L. Dysregulation of the TSC-mTOR pathway in human dis-
ease. Nat Genet. 2005;37:19–24.
12. van Steensel MA, van Geel M, Badeloe S, Poblete-Gutiérrez P, Frank J. Molecular pathways
involved in hair follicle tumor formation: all about mammalian target of rapamycin. Exp
Dermatol. 2009;18:185–91.
13. Baba M, Hong S-B, Sharma N, et al. Folliculin encoded by the BHD gene interacts with a
binding protein, FNIP1, and AMPK, and is involved in AMPK and mTOR signaling. Proc Natl
Acad Sci U S A. 2006;103:15552–7.
14. Hasumi H, Baba M, Hong SB, et al. Identification and characterization of a novel folliculin-
interacting protein FNIP2. Gene. 2008;415:60–7.
15. Takagi Y, Kobayashi T, Shiono M, et al. Interaction of folliculin (Birt-Hogg-Dubé gene prod-
uct) with a novel Fnip1-like (FnipL/Fnip2) protein. Oncogene. 2008;27:5339–47.
16. Hartman TR, Nicolas E, Klein-Szanto A, et al. The role of the Birt-Hogg-Dubé protein in
mTOR activation and renal tumorigenesis. Oncogene. 2009;28:1594–604.
17. Lindor NM, et al. Birt-Hogg-Dube syndrome: an autosomal dominant disorder with predispo-
sition to cancers of the kidney, fibrofolliculomas, and focal cutaneous mucinosis. Int
J Dermatol. 2001;40(10):653–6.
18. Reese E, Sluzevich J, Kluijt I, Teertstra HJ, De Jong D, Horenblas S, Ryu J. Birt-Hogg-Dubé
syndrome. In: Riegert-Johnson DL, Boardman LA, Hefferon T, Roberts M, editors. Cancer syn-
dromes [Internet]. Bethesda, MD: National Center for Biotechnology Information (US); 2009.
19. Vincent A, et al. Birt-Hogg-Dube syndrome: a review of the literature and the differential
diagnosis of firm facial papules. J Am Acad Dermatol. 2003;49(4):698–705.
20. Pavlovich CP, et al. Renal tumors in the Birt-Hogg-Dube syndrome. Am J Surg Pathol.
2002;26(12):1542–52.
26 Birt-Hogg-Dubé Syndrome 189
21. Toro JR, et al. BHD mutations, clinical and molecular genetic investigations of Birt-Hogg-
Dube syndrome: a new series of 50 families and a review of published reports. J Med Genet.
2008;45(6):321–31.
22. Schmidt LS, et al. Germline BHD-mutation spectrum and phenotype analysis of a large cohort
of families with Birt-Hogg-Dube syndrome. Am J Hum Genet. 2005;76(6):1023–33.
23. Gupta P, et al. Radiological findings in Birt-Hogg-Dube syndrome: a rare differential for
pulmonary cysts and renal tumors. Clin Imaging. 2007;31(1):40–3.
24. Linehan WM, et al. Identification of the genes for kidney cancer: opportunity for disease-
specific targeted therapeutics. Clin Cancer Res. 2007;13(2 Pt 2):671s–9.
25. Ayo DS, et al. Cystic lung disease in Birt-Hogg-Dube syndrome. Chest. 2007;132(2):679–84.
26. Gambichler T, Wolter M, Altmeyer P, Hoffman K. Treatment of Birt-Hogg-Dubé syndrome
with erbium: YAG laser. J Am Acad Dermatol. 2000;43:856–8.
27. Kahle B, Hellwig S, Schulz T. Multiple mantelome bei Birt-Hogg-Dubé-Syndrom.
Erfolchreiche therapie mit dem CO2-laser. Hautarzt. 2001;52:43–6.
28. Ng CS, Wood CG, Silverman PM, Tannir NM, Tamboli P, Sandler CM. Renal cell carcinoma:
diagnosis, staging, and surveillance. Am J Roentgenol. 2008;191:1220–32.
Chapter 27
Brooke-Spiegler Syndrome
27.1 Introduction
27.2 Background
More than 50 mutations have been described within exons 9–20 of the CYLD
gene. The CYLD tumor suppressor protein product functions by negatively regulating
the nuclear factor (NF)-kB and c-Jun N-terminal kinase pathways [1, 4]. Approximately
86 % of these mutations lead to a truncated CYLD protein, while the remaining 14 %
are missense mutations leading to a dysfunctional CYLD protein [4]. CYLD is
thought to be involved in regulating many cellular functions, including cell prolifera-
tion [4]. CYLD expression has found to be exceptionally high in the outer root sheath
of human scalp hair follicles [5]. Recently, animal models have been developed to
study the effects of the CYLD gene mutation, including those in “immunity, lipid
metabolism, spermatogenesis, antimicrobial defense, and inflammation” [4].
Dysfunction of the CYLD gene in BSS is limited to the skin adnexa, with resulting
appendageal tumors. Cylindromas and spiradenomas are thought to develop from the
secretory portion of the sweat gland, while trichoepitheliomas from hair follicles [2].
27.4 Work-Up
All patients with multiple, fixed facial papules of unclear etiology should be worked
up, as definite diagnosis by clinical inspection is difficult. While dermoscopic
evaluation may provide a consistent appearance with telangiectasia and blue struc-
tures [16], histopathology is often necessary [2]. Trichoepitheliomas represent an
over proliferation of follicular germinative, basaloid cells organized in a cribriform
pattern surrounded by a coarse, fibrous stroma [1, 17, 18]. As opposed to BCCs,
there is no clefting between basaloid cells and the stroma [19]. Cylindromas are
well-circumscribed nodules in the dermis or subcutis comprised of compact lobules
of basaloid cells that form a “jigsaw puzzle” arrangement [1]. Spiradenomas appear
as dermal or subcutaneous multinodular growths with pale-staining cells in
trabecular pattern, with basophilic cells at the periphery. A clue to the diagnosis is
the presence of lymphocytes scattered throughout the lesion [1].
Patients with multiple trichoepitheliomas, spiradenomas, and/or cylindromas
should be referred to genetics to be screened for CYLD gene mutations. The
differential diagnosis for trichoepitheliomas and trichoepitheliomas includes
Bazex and Rombo syndromes, but the patients also develop epidermoid cysts,
milia, hypotrichosis, BCCs, atrophoderma vermiculatum, and peripheral
cyanosis [1]. Patients should also be referred to otolaryngology for evaluation
for salivary or parotid tumors.
27.5 Treatment
Most of the cutaneous neoplasm in BSS are benign in nature and pose more of a
cosmetic challenge than medical necessity. However, close monitoring for malig-
nant transformation is prudent. Biopsies should be performed lesions that are
growing or ulcerating [1, 9]. When lesions grow so large that they cause functional
derangements, they may require treatment. Individual lesions may be surgically
excised, but surgery may not be a realistic option for BSS patients with large num-
bers of lesions [1, 20], Alternative therapies include electrosurgery, cryosurgery,
photodynamic therapy, imiquimod, or laser ablation that can be used alone or in
combination [20].
27.6 Conclusion
References
1. Bolognia JL, Jorizzo JL, Schaffer JV. Dermatology. 3rd ed. Saunders; June 4, 2012. Ch 111;
1834–1835.
2. Lee DA, Grossman ME, Schneiderman P, Celebi JT. Genetics of skin appendage neoplasms
and related syndromes. JMEd Genet. 2005;42:811–9.
3. Welch JP, Wells RS, Kerr CB. Ancell-Spiegler cylindromas (turban tumors) and Brooke-
Fordyce trichoepitheliomas: evidence for a single genetic entity. J Med Genet. 1968;5:29–35.
4. Blake PW, Toro JR. Update of cylindromatosis gene (CYLD) Mutations in Brooke-Spiegler
syndrome: novel insights into the role of deubiquitination in cell signaling. Hum Mutat.
2009;30(7):1025–36.
5. Massoumi R, Podda M, Fassler R, Paus R. Cylindroma as tumor of hair follicle origin. J Invest
Dermatol. 2006;126:1182–4.
6. Michal M, Lamovec J, Mukensnabl P, Pizinger K. Spiradenocylindromas of the skin: tumors
with morphological features of spiradenoma and cylindroma in the same lesion: report of 12
cases. Pathol Int. 1999;49:419–25.
7. Nerad JA, Folberg R. Multiple cylindromas. “The turban tumor”. Arch Opthalmol. 1987;105:1137.
8. Uede K, Yamamoto Y, Furukawa F. Brooke-Spiegler syndrome associated with cylindroma,
trichoepithelioma, spiradenoma, and syringoma. J Dermatol. 2004;31:32–8.
9. Gerretsen A, van der Putte S, Deenstra W. Cutaneous cylindroma with malignant transforma-
tion. Cancer. 1993;72:1618–23.
10. Durani BK, Kurzen H, Jaeckel A, Kuner N, Naeher H, Hartschuh W. Malignant transformation
of multiple dermal cylindromas. Br J Dermatol. 2001;145(4):653–6.
11. Pizinger K, Michal M. Malignant cylindroma in Brooke-Spiegler Syndrome. Dermatology.
2000;201(3):255–7.
12. Ayhan M, Adanali G, Senen D, Gorgu M, Erdogan B. Rarely seen cutaneous lesions in an
elderly patient: malignant transformation of multiple trichoepithelioma. Ann Plast Surg. 2001;
47:98–9.
13. Ishikawa M, Nakanishi Y, Yamazaki N, Yamamoto A. Malignant eccrine spiradenoma: a case
report and review of the literature. Dermatol Surg. 2001;27:67–70.
14. Lo JS, Snow SN, Reizner GT, Mohs FE, Larson PO, Hruza GJ. Metastatic basal cell carcinoma:
report of twelve cases with a review of the literature. J Am Acad Dermatol. 1991;24: 715–9.
15. Jungehulsing M, Wagner M, Damm M. Turban tumor with involvement of the parotid gland.
J Laryngol Otol. 1999;113:779–83.
16. Jarret R, Walker L, Bowling J. Dermoscopy of Brooke-Spiegler syndrome. Arch Dermatol.
2009;145(7):854.
17. Ackerman AB, Reddy VB, Soyer HP. Neoplasms with follicular differentiation. 2nd ed. New
York: Ardor Scribendi; 2001. p. 1109.
18. Elston D, Ferringer T, Ko C, Peckham S, High W, DiCaudo D, Bhuta S. Requisites in
dermatology: dermatopathology. New York, Saunders Elsevier; 2008. p. 70–1.
19. Bettencourt MS, Preito VG, Shea CR. Trichoepithelioma: a 19-year clinicopathologic
re-evaluation. J Cutan Pathol. 1999;26:398–404.
20. LoPiccolo M, Sage R, Kouba D. Comparing ablative fractionated resurfacing, photodynamic
therapy, and topical imiquimod in the treatment of trichoblastomas of Brooke-Spiegler
syndrome: a case study. Dermatol Surg. 2011;37:1047–50.
Chapter 28
Cowden Syndrome
28.1 Introduction
28.2 Background
Cowden syndrome affects various organs and increases the risk for overgrowth of
different tissues. Mucocutaneous lesions are found in nearly all cases of CS [10].
While the age of onset of these characteristic lesions is quite variable, the lesions most
commonly associated with CS include trichilemmomas, oral papillomas, and acral
keratoses. Trichilemmomas are benign hamartomatous lesions of the follicle outer
root sheath that end to be slow growing and skin colored and most commonly found
on the face or neck (Fig. 28.1) [11]. Oral papillomas are benign lesions that can occur
on the oral mucosa, primarily on the tongue (Fig. 28.2) [12]. Acral keratoses are clini-
cally seen as verrucous papules on the dorsal surfaces of the hands and feet (Fig. 28.3).
Fig. 28.3 Acral keratoses on the dorsal hands, which clinically appear as warty, stuck on papules
(Photo credit: Joshua A. Zeichner, M.D.)
28.4 Work-Up
Patients with fixed facial papules or oral lesions of unclear diagnosis should be
evaluated with a biopsy for histologic evaluation, as they may be a presenting sign
of CS. The United States National Comprehensive Cancer Network (NCCN) has
published screening criteria, outlined in Table 28.1. Clinical diagnostic criteria for
CS are met if the patient has adult L’hermitte-Duclos disease or a requisite number
of mucocutaneous lesions, macrocephaly plus one other major criterion, one major
and three minor criteria, or four minor criteria. Any patient who fits the screening
criteria, should be referred to genetics for PTEN gene testing [24]. Patients should
also be evaluated for the presence of internal malignancies, including breast and
thyroid disease.
28.5 Treatment
28.6 Conclusion
References
6. Starlink T, Cander Veen J, Artwert F, Erikson AW. The Cowden syndrome: a clinical and
genetic study in 21 patients. Clin Genet. 1986;29:222–33.
7. Salem OS, Steck WD. Cowden’s disease. A case report and review of the English literature.
J Am Acad Dermatol. 1983;8(5):686–96.
8. Pilarski R, Eng C. Will the real Cowden syndrome please stand up (again)? Expanding muta-
tional and clinical spectra of the PTEN hamartoma tumour syndrome. J Med Genet. 2004;
41(5):323–6.
9. Guldberg P, Thor Straten P, Birck A, Ahrenkiel V, Kirkin AF, Zeuthen J. Disruption of the
MMAC1/PTEN gene by deletion of mutation is a frequent event in malignant melanoma.
Cancer Res. 1997;57(17):3660–3.
10. Fistarol SK, Marc D, Anliker MS, Peter H. Cowden disease or multiple hamartoma syndrome—
cutaneous clue to internal malignancy. Eur J Dermatol. 2002;12(5):411–21.
11. Brownstein MH, Mehregan AH, Bilowski JB. Trichilemmomas on Cowden’s disease. J Am
Med Assoc. 1977;238(1):26.
12. Devlin MF, Barrie R, Ward-Booth RP. Cowden’s disease: a rare but important manifestation of
oral papillomatosis. Br J Oral Maxillofacial Surg. 1992;30:335–6.
13. Blumenthal GM, Dennis PA. PTEN hamartoma tumor syndromes. Eur J Human Genet.
2008;16:1289–300.
14. Nusbaum R, Vogel JK, Ready K. Susceptibility to breast cancer: hereditary syndromes and low
penetrance genes. Breast Dus. 2007;27:21–50.
15. Schrager CA, Schneider D, Gruener AC. Clinical and pathological features of breast disease in
Cowden’s syndrome: an under-recognized syndrome with an increased risk of breast cancer.
Human Pathol. 1998;29:47–53.
16. Brownstein MH, Wolf M, Bikowski JB. Cowden’s disease: a cutaneous marker of breast
cancer. Cancer. 1978;41:2393–8.
17. Farooq A, Walker LJ, Bowling J, Audisio RA. Cowden syndrome. Cancer Treat Rev. 2010;8:
577–83.
18. Pilariski R. Cowden syndrome: a critical review. J Genet Counsel. 2009;18:13–27.
19. Lok C, Viseux V, Avril MF, Richard MA, Gondry-Jouet C, Deramond H, et al. Brain magnetic
resonance imaging in patients with Cowden syndrome. Medicine. 2005;84(2):129–36.
doi:10.1097/01.md.0000158792.24888.d2.
20. Bagan JV, Penarrocha M, Vera-Sempere F. Cowden syndrome: clinical and pathological
considerations in two new cases. J Oral Maxillofacial Surg. 1989;47(3):291–4. doi:10.1016/
0278-2391(89)90234-6.
21. Starink TM. Cowden’s disease: analysis of fourteen new cases. J Am Acad Dermatol.
1984;11(6):1127–41. doi:10.1016/S0190-9622(84)70270-2.
22. Salem OS, Steck WD (1983) Cowden’s disease (multiple hamartoma and neoplasia syndrome).
A case report and review of the English literature. J Am Acad Dermatol. 8(5):686–96.aa
23. Parisi M, Dinulos MB, Leppid KA, Sybert VP, Eng C, Hudgins L. The spectrum and evolution of
phenotypic findings in PTEN mutation positive cases of Bannayan-Riley-Ruvalcaba syndrome.
J Med Genet. 2001;38:52–7.
24. The NCCN 1.2009 Cowden syndrome clinical practice guidelines in oncology. National
Comprehensive Cancer Network, 2009. Available from: http://www.nccn.org. Adapted with
permission from NCCN [accessed 10.06.09]. (To view the most recent and complete version
of the guideline, go online to www.nccn.org).
Chapter 29
Gardner Syndrome
29.1 Introduction
29.2 Background
FAP and GS are caused by mutations in the adenomatous polyposis coli (APC)
tumor suppressor gene on chromosome 5q21–22 [2]. There is a correlation between
the site of the mutation on the APC gene and the clinical phenotype [3]. Specifically,
APC mutations between codon 1395 and 1493 are frequently associated with
features of GS [4]. APC gene mutations result in accumulation of β-catenin which
bypasses the growth-regulating effects of Wnt signaling resulting in important
implications for cell differentiation, proliferation, and adhesion [5, 6]. GS is inherited
as an autosomal dominant disorder with high penetrance and variable expressivity,
though approximately 25 % of patients have de novo mutations and no family
history [6]. The estimate of incidence of GS ranges from approximately 1 in 6,850 to
1 in 31,250 births [7–9].
Cutaneous lesions and bony abnormalities in GS often appear during childhood and
adolescence, frequently before polyposis develops. Epidermoid cysts are the most
common cutaneous finding in GS, affecting approximately one-third of patients [10].
Epidermoid cysts are firm, well-circumscribed dermal nodules that contain soft
keratin secretions. They may be present at birth but often occur at puberty, occur in
multiplicity in more than 50 % of patients, and tend to occur in unusual locations
such as the face, scalp, and extremities [10]. Cysts in GS typically occur earlier than
ordinary cysts, as seen in patient with acne. Though the majority of cysts are asymp-
tomatic, they can be pruritic, inflamed, or ruptured [10]. Cutaneous cysts showing
unusual histological features—namely, a mixture of epidermoid, trichilemmal, and/
or pilomatricoma-like features—are considered a hallmark of GS [6].
Desmoid tumors are benign fibrous mesenchymal neoplasms that result from the
proliferation of well-differentiated fibroblasts seen in 3–30 % of patients with GS
[2, 6, 11], with average onset around age 30 and a marked female predominance
(70–85 %) [6, 11, 12]. Desmoid tumors in GS are associated with mutations occurring
downstream of codon 1400 of the APC gene [6, 12] and can be located in the abdomi-
nal region (intra-abdominally or within the abdominal wall) or extra-abdominally in
the shoulder girdle, chest well, and inguinal regions [12, 13]. Physical exam may
reveal a well-circumscribed, firm, flesh-colored tumor [13]. These tumors may occur
spontaneously or at incision sites, commonly developing after colectomy which is a
leading cause of morbidity in GS [12]. Although cytologically benign and non-meta-
static, desmoid tumors are locally aggressive and can be invasive, recur after excision,
and even lead to death [11].
Fibromas are another feature of GS and may occur in the skin, subcutaneous
tissues, mesentery, or even retroperitoneum [11]. Nuchal fibromas classically
present with diffuse swelling and induration of the back of the neck [2, 6]. “Gardner
fibromas” arise in the first decade of life, favor the trunk or paraspinal region, and
can serve as a desmoid precursor [11]. Lipomas, leiomyomas, trichoepitheliomas,
and neurofibromas are other skin findings less commonly observed in GS [2, 6, 11].
None of the cutaneous lesions in GS have been reported to progress to malignancy.
childhood and often predate intestinal polyposis. These tumors are benign and
painless but may be multiple or grow to reach significant enough size to be detected
on physical exam [11]. Other skeletal abnormalities in GS include exostoses,
endostoses, and cortical thickening of the long bones [11].
Dental anomalies are well described in GS and are seen in approximately 20 %
of patients. These include odontomas and odontogenic cysts; absent, supernumer-
ary, or rudimentary teeth; and multiple dental caries [2, 6].
Congenital hypertrophy of the retinal pigment epithelium (CHRPE) affects
approximately two-thirds of patients with GS and can be an early sign of the syn-
drome [14]. CHRPE is a patch of darkly pigmented retinal epithelium that can be
detected on ophthalmologic exam at birth [13]. While unilateral solitary CHRPE is
not specific for FAP, multiple or bilateral lesions are considered highly specific for
FAP [2, 6, 15]. In GS, CHRPE is associated with mutations between codons 767
and 1513 [16].
Importantly, patients with GS are at increased risk for the development of several
extracolonic malignancies. These include duodenal cancer, papillary thyroid carcinoma
(mostly in females), brain tumors (glioblastoma, astrocytoma, medulloblastoma), hepa-
toblastoma (mostly in children), pancreatic and biliary tract carcinomas, adrenal ade-
noma, and various sarcomas (including fibrosarcoma and osteosarcoma) [2, 5, 6, 11, 17].
29.4 Work-Up
29.5 Treatment
29.6 Conclusion
Gardner syndrome affects multiple body systems, including the skin, soft tissue, bone,
gastrointestinal system, as well as other solid organs. Recognition of cutaneous signs
may provide an early diagnosis and can be lifesaving with proper management.
References
17. Tsao H. Update on familial cancer syndromes and the skin. J Am Acad Dermatol. 2000;
42(6):939–69.
18. Jasperson KW, Tuohy TM, Neklason DW, Burt RW. Hereditary and familial colon cancer.
Gastroenterology. 2010;138(6):2044–58.
19. Pujol RM, Casanova JM, Egido R, Pujol J, de Moragas JM. Multiple familial pilomatricomas:
a cutaneous marker for Gardner syndrome? Pediatr Dermatol. 1995;12(4):331–5.
20. Coffin CM, Hornick JL, Zhou H, Fletcher CD. Gardner fibroma: a clinicopathologic and
immunohistochemical analysis of 45 patients with 57 fibromas. Am J Surg Pathol. 2007;
31(3):410–6.
21. Naylor EW, Lebenthal E. Early detection of adenomatous polyposis coli in the Gardner’s
syndrome. Pediatrics. 1979;63(2):222–7.
22. de Camargo VP, et al. Clinical outcomes of systemic therapy for patients with deep fibromatosis
(desmoid tumor). Cancer. 2010;116(9):2258–65.
23. Hughes-Fulford M, Boman B. Growth regulation of Gardner’s syndrome colorectal cancer
cells by NSAIDs. Adv Exp Med Biol. 1997;407:433–41.
24. Boardman LA. Heritable colorectal cancer syndromes: recognition and preventive management.
Gastroenterol Clin North Am. 2002;31(4):1107–31.
25. Gallagher MC, Phillips RK, Bulow S. Surveillance and management of upper gastrointestinal
disease in familial adenomatous polyposis. Fam Cancer. 2006;5(3):263–73.
Chapter 30
Gorlin Syndrome
30.1 Introduction
30.2 Background
BCNS results from mutations in the PTCH1 tumor suppressor gene which is found
on chromosome 9q22.3-q31 [8]. The product of the PTCH1 gene normally represses
gene transcription in the Hedgehog signaling pathway by acting as a transmem-
brane receptor for the Sonic Hedgehog (SHH) ligand [9]. The Hedgehog signaling
pathway was first described as a developmental pathway during research on the fruit
fly, Drosophila melanogaster [10]. Post-developmentally in humans, the Hedgehog
signaling pathway is typically only active in hair follicles and skin cells [11].
The neoplasms of the syndrome require two hits to the PTCH1 gene, with a muta-
tion in the second normal allele [12]. However, some clinical features of BCNS,
such as palmar pits, bifid rib, and macrocephaly, only require the inherited defective
allele [3].
Women and men appear to be equally affected by BCNS. However, the prevalence
of BCNS ranges from 1/57,000 to 1/256,000 depending on the studied population
[13, 14]. It should be noted that certain clinical manifestations of BCNS, mainly
BCCs, are less prevalent in darker skin individuals [15].
More than 100 different clinical features have been found in association with BCNS
[16]. The most frequent and characteristic features of BCNS have been listed chron-
ologically by age of presentation.
Facial Dysmorphism: Most individuals with BCNS have one or more of the
following “coarse” facial features: macrocephaly, frontal bossing, broad nasal
bridge, prognathism, high arched eyebrows and/or palate, and cleft lip/palate [4,
17]. Patients with BCNS sometimes have a history of being delivered by cesarean
section due to macrocephaly which is the most common facial dysmorphism asso-
ciated with BCNS [18].
Skeletal Abnormalities: In addition to abnormal skull findings, other skeletal
abnormalities typically present at birth include bifid ribs, vertebral abnormalities,
Sprengel shoulder, pectus deformity, short 4th metacarpal, and syndactyly of the
digits [7].
Ocular Abnormalities: Other congenital abnormalities associated with BCNS
include severe eye defects such as strabismus, hypertelorism, telecanthus, cataracts,
glaucoma, microphthalmia, orbital cyst, and coloboma [6, 14, 17].
Cardiac Fibromas: Less than 5 % of patients with BCNS develop cardiac fibromas
which typically appear in the first year of life [6]. The detection of this benign tumor
qualifies as one of the minor criteria for the diagnosis. In general, cardiac fibromas
are asymptomatic, but they can cause conduction defects (arrhythmias) and ven-
tricular outflow obstruction [19].
Medulloblastoma: Less than 5 % of patients with BCNS also present with medul-
loblastomas. Compared to sporadic medulloblastomas which usually occur between
the ages of 6 and 10, medulloblastomas in association with BCNS are more common
in male patients and typically present by 2 years of age [13, 20].
Odontogenic (Jaw) Keratocysts: Approximately 90 % of patients with BCNS
develop multiple jaw keratocysts which occur mostly in the mandible [14]. These
keratocysts have been reported as early as 4 years old, but typically occur during
30 Gorlin Syndrome 209
the second and third decade of life [21]. The presentation of these keratocysts
ranges from asymptomatic to painless jaw swelling, tooth disruption, and rarely jaw
fracture [22].
BCCs: BCCs in patients with BCNS occur in more than 75 % of Caucasians
(Fig. 30.1) but less often in African American and Asian patients [15, 23]. BCCS in
BCNS has been reported as early as 3 years of age, but the peak incidence occurs
at 20 years of age [6]. They typically occur in sun-exposed area. While only a small
percentage are locally invasive, they are challenging to treat and surgical manage-
ment is potentially disfiguring [17].
Palmoplantar Pitting: Approximately 80 % of patients with BCNS develop
palmoplantar pitting typically beginning in the second decade of life [24] (Fig. 30.2).
Intracranial Calcification: In a study of 105 individuals with BCNS (mean age of
32.5 years), approximately 65 % of these patients had intracranial calcification,
particularly of the falx cerebri [4].
Ovarian Fibromas: Approximately 15–25 % of female patients with BCNS have
ovarian fibroma which is often bilateral and calcified [4, 6]. Most are asymptomatic
and are usually discovered incidentally, but some can undergo torsion [3].
Lymphomesenteric Cysts: The exact incidence and age of presentation for lympho-
mesenteric cysts is unknown. They are also usually asymptomatic [23].
210 M. Haddican and J. Spencer
Fig. 30.2 1–2 mm depressed papules on the palms (Photo credit: Joshua A. Zeichner, M.D.)
30.4 Work-Up
30.5 Treatment
There is no cure for BCNS and patients are managed based on syndrome-related
findings [3]. The different clinical manifestations are treated similarly in patients
without the disorder. However, surveillance guidelines for the detection of different
tumors and odontogenic cysts are important; and thus, patients with BCNS need to
be treated under the care of several specialists including medical geneticists, derma-
tologists, oral surgeons, and plastic surgeons [25]. Fortunately, the life span of
patient with BCNS is not significantly different from the average [3], but exposure
to ionizing and UV radiation should be minimized to reduce the total number of
BCCs [28].
For the management of BCCs, a regular complete skin examination by a
dermatologist every 2–4 months is recommended for patients with BCNS [3, 25].
Due to the increased number of BCCs typically present during a lifetime, there is a
tendency to reserve surgical treatment for aggressive or invasive tumors in order to
prevent disfigurement [3, 29]. Although the majority of BCCs are nodular in BCNS,
topical application of 0.1 % tretinoin cream, 5-fluorouracil cream, and imiquimod
5 % cream is effective for treating superficial BCCs and has been used for these
BCCs as well [3, 29, 30]. One of the more novel therapeutic approaches for BCC
involves photodynamic therapy (PDT) which is the topical application of an agent,
such as 5-aminolevulinic acid (ALA, Levulan®, DUSA Pharmaceuticals, Inc.,
Wilmington, MA, USA) or methyl ester form, methyl 5-aminolevulinate (MAL,
Metvixia TM, Galderma, Princeton, NJ, USA) [31, 32]. Both ALA and MAL are
converted to a photosensitizer protoporphyrin IX in tumor cells. MAL-PDT is
unique in not only demonstrating a high efficacy in the treatment of superficial
BCC; it has also been shown to be effective in the treatment of nodular BCC [33].
Vismodegib, an oral Hedgehog pathway inhibitor, has also been shown to be an
important option in the treatment of BCCs. However, the side effects of oral vismo-
degib, including muscle spasms, hair loss, altered taste sensation, weight loss,
fatigue, nausea, decreased appetite, and diarrhea, may not be well tolerated in some
patients [34, 35]. Additionally, a topical inhibitor of the Hedgehog pathway has
shown promising results in a randomized trial for patients with BCNS [36].
While great care is taken to treat BCC’s, prophylactic therapy may be used to
prevent growth of new cancers. Given these patients’ high risk for continually
developing BCCs, therapy to prevent them can save disfiguring treatments. Acitretin
has been shown to reduce the size of clinically apparent skin cancers and may been
used as chemoprevention [37]. While the efficacy of acitretin has been described for
squamous cell carcinomas (SCCs), the drug may be used for patients with BCNS.
Nonsteroidal anti-inflammatory drugs (NSAIDs) are also used as chemoprevention
for SCCs. Oral celecoxib has been shown to be effective in suppression BCCs in
both mice and humans with PTCH1 gene mutations [38].
For the detection of odontogenic keratocysts, annual dental panoramic X-rays are
recommended beginning at the age of eight [27]. These cysts are typically removed
by wide excision/enucleation with curettage but have a high rate of recurrence [23,
212 M. Haddican and J. Spencer
39]. There is some evidence that oral vismodegib is also an important treatment
option for keratocysts; however, more studies are needed in patients with BCNS [40].
Notably in pediatric patients, surveillance for the development of medulloblas-
toma and cardiac fibroma is important. An annual cranial MRI is recommended
until 8 years of age for the detection of a medulloblastoma [25]. For the treatment of
a medulloblastoma, radiation therapy should be avoided because it causes multiple
BCCs to develop in the field of radiation [28]. To monitor for the presence of a car-
diac fibroma, an echocardiogram should be completed in the first year of life.
Asymptomatic patients with a cardiac fibroma should be closely followed by a
pediatric cardiologist [25].
30.6 Conclusion
References
1. Gorlin RJ, Goltz RW. Multiple nevoid basal-cell epithelioma, jaw cysts, and bifid rib. A syndrome.
N Engl J Med. 1960;262:908.
2. Anderson D, Taylor W, Falls H, Davidson R. The nevoid basal cell carcinoma syndrome. Am
J Hum Genet. 1967;19:12–22.
3. Lo Muzio L. Nevoid basal cell carcinoma syndrome (Gorlin syndrome). Orphanet J Rare Dis.
2008;3:32.
4. Kimonis VE, et al. Clinical manifestations in 105 persons with nevoid basal cell carcinoma
syndrome. Am J Med Gen. 1997;69:300.
5. Shanley S, et al. Nevoid basal cell carcinoma syndrome: review of 118 affected individuals.
Am J Med Genet. 1994;50:282.
6. Evans DG, Ladusans EJ, Rimmer S, Burnell LD, Thakker N, Farndon PA. Complications of
the naevoid basal cell carcinoma syndrome: results of a population based study. J Med Genet.
1993;30:460–4.
7. Ratcliffe JF, Shanley S, Chenevix-Trench G. The prevalence of cervical and thoracic congeni-
tal skeletal abnormalities in basal cell naevus syndrome; a review of cervical and chest radio-
graphs in 80 patients with BCNS. Br J Radiol. 1995;68:596–9.
8. Farndon PA, Del Mastro RG, Evans DG, Kilpatrick MW. Location of gene for Gorlin
syndrome. Lancet. 1992;339:581–2.
9. Booth DR. The hedgehog signaling pathway and its role in basal cell carcinoma. Cancer
Metastasis Rev. 1999;18:261.
10. Hahn H, Wicking C, Zaphiropoulous PG, Gailani MR, Shanley S, Chidambaram A,
Vorechovsky I, Holmberg E, Unden AB, Gillies S, et al. Mutations of the human homolog of
Drosophila patched in the nevoid basal cell carcinoma syndrome. Cell. 1996;85:841–51.
30 Gorlin Syndrome 213
11. Gailani M, Bale A. Developmental genes and cancer: role of patched in basal cell carcinoma
of the skin. J Natl Cancer Inst. 1997;89:1103.
12. Levanat S, Gorlin R, Fallet S, et al. A two-hit model for developmental defects in Gorlin
syndrome. Nat Genet. 1996;12:85.
13. Evans DG, Farndon PA, Burnell LD, Gattamaneni HR, Birch JM. The incidence of Gorlin
syndrome in 173 consecutive cases of medulloblastoma. Br J Cancer. 1991;64:959–61.
14. Lo Muzio L, Nocini PF, Savoia A, Consolo U, Procaccini M, Zelante L, Pannone G, Bucci P,
Dolci M, Bambini F, et al. Nevoid basal cell carcinoma syndrome. Clinical findings in 37
Italian affected individuals. Clin Genet. 1999;55:34–40.
15. Goldstein AM, Pastakia B, DiGiovanna JJ, Poliak S, Santucci S, Kase R, Bale AE, Bale SJ.
Clinical findings in two African-American families with the nevoid basal cell carcinoma
syndrome (NBCC). Am J Med Genet. 1994;50:272–81.
16. Farndon PA. Gorlin (naevoid basal cell carcinoma) syndrome. In: Eeles R, Easton DF, Ponder
BAJ, Eng C, editors. Genetic predisposition to cancer. London: Hodder Arnold; 2004.
p. 193–213.
17. Gorlin RJ. Nevoid basal-cell carcinoma syndrome. Medicine (Baltimore). 1987;66:98.
18. Hogge WA, Blank C, Roochvarg LB, Hogge JS, Wulfsberg EA, Raffel LJ. Gorlin syndrome
(naevoid basal cell carcinoma syndrome): prenatal detection in a fetus with macrocephaly and
ventriculomegaly. Prenat Diagn. 1994;14:725–7.
19. Jones KL, Wolf PL, Jensen P, et al. The Gorlin syndrome: a genetically determined disorder
associated with cardiac tumor. Am Heart J. 1986;111:1013.
20. Lacombe D, Chateil JF, Fontan D, Battin J. Medulloblastoma in the nevoid basal-cell carcinoma
syndrome: case reports and review of the literature. Genet Couns. 1990;1:273.
21. Woolgar JA, Rippin JW, Browne RM. The odontogenic keratocyst and its occurrence in the
nevoid basal cell carcinoma syndrome. Oral Surg Oral Med Oral Pathol. 1987;64:727–30.
22. Myoung H, Hong SP, Hong SD, et al. Odontogenic keratocyst: review of 256 cases for recur-
rence and clinicopathologic parameters. Oral Surg Oral Med Oral Pathol Oral Radiol Endon.
2001;91:328.
23. Gorlin RJ. Nevoid basal cell carcinoma syndrome. Dermatol Clin. 1995;13:113–25.
24. Gutierrez MM, Mora RG. Nevoid basal cell carcinoma syndrome. A review and case report of
a patient with unilateral basal cell nevus syndrome. J Am Acad Dermatol. 1986;15:1023–30.
25. Bree AF, Shah MR, BCNS Colloquium Group. Consensus statement from the first interna-
tional colloquium on basal cell nevus syndrome. Am J Med Genet A. 2011;155:2091–7.
26. Soekarman D, Fryns JP, Casaer P, Berghe H. Van Den: increased head circumference and
facial cleft as presenting signs of the nevoid basal-cell carcinoma syndrome. Genet Couns.
1991;2:157–62.
27. Lo Muzio L, Nocini P, Bucci P, Pannone G, Consolo U, Procaccini M. Early diagnosis of
nevoid basal cell carcinoma syndrome. J Am Dent Assoc. 1999;130:669–74.
28. Frentz G, Munch-Petersen B, Wulf HC, et al. The nevoid basal cell carcinoma syndrome:
sensitivity to ultraviolet and X-ray irradiation. J Am Acad Dermatol. 1987;17:637.
29. Van der Geer S, et al. Treatment of basal cell carcinomas in patients with nevoid basal cell
carcinoma syndrome. J Eur Acad Dermatol Venereol. 2008;23:308.
30. Gollnick H, Barona CG, Frank RG, et al. Recurrence rate of superficial basal cell carcinoma
following treatment with imiquimod 5 % cream: conclusion of a 5-year long-term follow-up
study in Europe. Eur J Dermatol. 2008;18:677–82.
31. Hsi RA, Rosenthal DI, Glatstein E. Photodynamic therapy in the treatment of cancer:current
state of the art. Drugs. 1999;57:725–34.
32. Morton CA, MacKie RM, Whitehurst C, Moore JV, McColl JH. Photodynamic therapy for
basal cell carcinoma: effect of tumor thickness and duration of photosensitizer application on
response. Arch Dermatol. 1998;134:248–9.
33. Horn M, Wolf P, Wulf HC, et al. Topical methyl aminolaevulinate photodynamic therapy in
patients with basal cell carcinoma prone to complications and poor cosmetic outcome with
conventional treatment. Br J Dermatol. 2003;149:1242–9.
214 M. Haddican and J. Spencer
34. LoRusso PM, Rudin CM, Reddy JC, Tibes R, Weiss GJ, Borad MJ, Hann CL, Brahmer JR,
Chang I, Darbonne WC, Graham RA, Zerivitz KL, Low JA, Von Hoff DD. Phase I trial of
hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally
advanced or metastatic solid tumors. Clin Cancer Res. 2011;17:2502–11.
35. http://www.roche.com/media/media_releases/med-cor-2011-06-20.html
36. Skvara H, Kalthoff F, Meingassner JG, et al. Topical treatment of Basal cell carcinomas in
nevoid basal cell carcinoma syndrome with a smoothened inhibitor. J Invest Dermatol. 2011;
131:1735.
37. Lebwohl M, Tannis C, Carrasco D. Acitretin suppression of squamous cell carcinoma: case
report and literature review. J Dermatolog Treat. 2003;14 Suppl 2:3–6. Review.
38. Tang JY, Aszterbaum M, Athar M, et al. Basal cell carcinoma chemoprevention with nonste-
roidal anti-inflammatory drugs in genetically predisposed PTCH1+/− humans and mice.
Cancer Prev Res (Phila). 2010 Jan;3(1):25–34.
39. Dominguez FV, Keszler A. Comparative study of keratocysts, associated and non-associated
with nevoid basal cell carcinoma syndrome. J Oral Pathol. 1988;17:39–42.
40. Goldberg LH, Landau JM, Moody MN, et al. Resolution of odontogenic keratocysts of the jaw
in basal cell nevus syndrome with GDC-0449. Arch Dermatol. 2011;147:839.
Chapter 31
Muir-Torre Syndrome
31.1 Introduction
31.2 Background
arise from the alimentary system, with colorectal carcinoma being the most common
[3, 18]. MTS-associated colorectal carcinoma usually develops proximal to the
splenic flexure and typically appears by age 50. In contrast, colorectal carcinoma in
the general population is diagnosed, on average, 10 years later (age range 55–65)
and is commonly located in the distal colon [7, 13]. Other visceral malignancies
linked to MTS include genitourinary, breast, hematologic, head and neck, small
intestine, and stomach [19].
While MTS-associated malignancies are usually less aggressive than their
sporadic counterparts, approximately 60 % of patients affected by the syndrome
will develop metastatic disease [20]. Few studies have examined survival rates
among individuals with MTS, however, MTS-associated neoplasms may be more
surgically responsive compared to sporadic carcinomas [2].
31.4 Work-Up
31.5 Treatment
31.6 Conclusion
References
1. Schwartz RA, Torre DP. The Muir-Torre syndrome: a 25-year retrospect. J Am Acad Dermatol.
1995;33(1):90–104.
2. Ponti G, Ponz de Leon M. Muir-Torre syndrome. Lancet Oncol. 2005;6(12):980–7.
3. Curry ML, Eng W, Lund K, Paek D, Cockerell CJ. Muir-Torre syndrome: role of the dermato-
pathologist in diagnosis. Am J Dermatopathol. 2004;26(3):217–21.
4. Muir EG, Bell AJ, Barlow KA. Multiple primary carcinomata of the colon, duodenum, and
larynx associated with kerato-acanthomata of the face. Br J Surg. 1967;54(3):191–5.
5. Torre D. Multiple sebaceous tumors. Arch Dermatol. 1968;98(5):549–51.
6. Fahmy A, Burgdorf WH, Schosser RH, Pitha J. Muir-Torre syndrome: report of a case and
reevaluation of the dermatopathologic features. Cancer. 1982;49(9):1898–903.
7. Higgins HJ, Voutsalath M, Holland JM. Muir-Torre syndrome: a case report. J Clin Aesthet
Dermatol. 2009;2(8):30–2.
31 Muir-Torre Syndrome 219
8. Cohen PR, Kohn SR, Kurzrock R. Association of sebaceous gland tumors and internal
malignancy: the Muir-Torre syndrome. Am J Med. 1991;90(5):606–13.
9. South CD, Hampel H, Comeras I, Westman JA, Frankel WL, De la Chapelle A. The frequency
of Muir-Torre syndrome among Lynch syndrome families. J Natl Cancer Inst. 2008;
100(4):277–81.
10. Eisen DB, Michael DJ. Sebaceous lesions and their associated syndromes: part II. J Am Acad
Dermatol. 2009;61(4):563–80.
11. Mangold E, Rahner N, Friedrichs N, et al. MSH6 mutation in Muir-Torre syndrome: could this
be a rare finding? Br J Dermatol. 2007;156(1):158–62.
12. Lynch HT, Fusaro RM. The Muir-Torre syndrome in kindreds with hereditary nonpolyposis
colorectal cancer (Lynch syndrome): a classic obligation in preventive medicine. J Am Acad
Dermatol. 1999;41(5):797–9.
13. Hare HH, Mahendraker N, Sarwate S, Tangella K. Muir-Torre syndrome: a rare but important
disorder. Cutis. 2008;82(4):252–6.
14. Marazza G, Masouyé I, Taylor S, et al. An illustrative case of Muir-Torre syndrome: contribution
of immunohistochemical analysis in identifying indicator sebaceous lesions. Arch Dermatol.
2006;142(8):1039–42.
15. Bayliss SJ, Berk DR. Muir-Torre syndrome. In: Bolognia JL, Jorizzo JL, Schaffer JV, editors.
Dermatology. 3rd ed. St. Louis, MO: Elsevier; 2012.
16. Eisen DB, Michael DJ. Sebaceous lesions and their associated syndromes: part I. J Am Acad
Dermatol. 2009;61(4):549–62.
17. Dores GM, Curtis RE, Toro JR, Devesa SS, Fraumeni Jr JF. Incidence of cutaneous sebaceous
carcinoma and risk of associated neoplasms: insight into Muir-Torre syndrome. Cancer.
2008;113(12):3372–81.
18. Akhtar S, Oza KK, Khan SA, Wright J. Muir-Torre syndrome: case report of a patient with
concurrent jejunal and ureteral cancer and a review of the literature. J Am Acad Dermatol.
1999;41(5):681–6.
19. Cohen PR. Muir-Torre syndrome in patients with hematologic malignancies. Am J Hematol.
1992;40(1):64–5.
20. Poleksic S. Keratoacanthoma and multiple carcinomas. Br J Dermatol. 1974;91(4):461–3.
21. Pardo FS, Wang CC, Albert D, Stracher MA. Sebaceous carcinoma of the ocular adnexa:
radiotherapeutic management. Int J Radiat Oncol Biol Phys. 1989;17(3):643–7.
22. Hata M, Koike I, Omura M, Maegawa J, Ogino I, Inoue T. Noninvasive and curative radiation
therapy for sebaceous carcinoma of the eyelid. Int J Radiat Oncol Biol Phys. 2012;82(2):
605–11.
23. Spielvogel RL, DeVillez RL, Roberts LC. Oral isotretinoin therapy for familial Muir-Torre
syndrome. J Am Acad Dermatol. 1985;12(3):475–80.
24. Pettey AA, Walsh JS. Muir-Torre syndrome: a case report and review of the literature. Cutis.
2005;75(3):149–55.
25. Cohen PR, Kohn SR, Davis DA, Kurzrock R. Muir-Torre syndrome. Dermatol Clin. 1995;
13(1):79–89.
Chapter 32
Reed’s Syndrome
32.1 Introduction
32.2 Background
The coexistence of benign smooth muscle growths in the skin and in the uterus is
known as Reed’s syndrome [1]. In 1973, Reed et al reported on two families in
which members of successive generations demonstrated cutaneous leiomyomas,
Reed’s syndrome manifests during the second to fourth decade of life, with solitary
or multiple cutaneous leiomyomas, which appear as firm skin-colored or pink-brown
papules or nodules ranging from 0.2 to 2 cm in diameter [5, 7, 11] (Figs. 32.1 and
32.2). A pseudo-Darier sign may be present, which is a transient piloerection or
elevation of a lesion induced by rubbing. The clinical presentation of multiple lesions
has been described in various patterns or distributions, such as bilateral and symmetric,
clustered, linear, zosteriform, dermatomal like, or disseminated [4, 21]. Although
commonly distributed over the trunk, the face may also be affected.
It is important to consider a leiomyoma in any indolent nodular or papular growth
associated with pain. In 89–92 % of cases, patients experience localized photosen-
sitivity and pain upon contact, injury or exposure to change in temperature [20]. An
acute increase in size and/or number of lesions and/or an increase in pain should be
suggestive of worsening disease or the rare possibility of malignant degeneration.
224 K. Pacific and J. Emer
32.4 Work-Up
32.5 Treatment
References
7. Badeloe S, Frank J. Clinical and molecular genetic aspects of hereditary multiple cutaneous
leiomyomatosis. Eur J Dermatol. 2009;19(6):545–51.
8. Malhotra P, Walia H, Singh A, Ramesh V. Leiomyoma cutis: a clinicopathological series of 37
cases. Indian J Dermatol. 2010;55(4):337–41.
9. Wei MH, Toure O, Glenn GM, et al. Novel mutations in FH and expansion of the spectrum of
phenotypes expressed in families with hereditary leiomyomatosis and renal cell cancer. J Med
Genet. 2006;43(1):18–27.
10. Kiuru M, Launonen V. Hereditary leiomyomatosis and renal cell cancer (HLRCC). Curr Mol
Med. 2004;4(8):869–75.
11. García Muret MP, Pujol RM, Alomar A, et al. Familial leiomyomatosis cutis et uteri (Reed’s
syndrome). Arch Dermatol Res. 1988;280:S29–32.
12. Toro JR, Nickerson ML, Wei MH, et al. Mutations in the fumarate hydratase gene cause hered-
itary leiomyomatosis and renal cell cancer in families in North America. Am J Hum Genet.
2003;73(1):95–106.
13. Alam NA, Olpin S, Leigh IM. Fumarate hydratase mutations and predisposition to cutaneous
leiomyomas, uterine leiomyomas and renal cancer. Br J Dermatol. 2005;153(1):11–7.
14. Tomlinson IP, Alam NA, Rowan AJ, et al. Germline mutations in FH predispose to dominantly
inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat Genet.
2002;30(4):406–10.
15. Kiuru M, Launonen V, Hietala M, et al. Familial cutaneous leiomyomatosis is a two-hit
condition associated with renal cell cancer of characteristic histopathology. Am J Pathol. 2001;
159(3):825–9.
16. Alam NA, Bevan S, Churchman M, et al. Localization of a gene (MCUL1) for multiple
cutaneous leiomyomata and uterine fibroids to chromosome 1q42.3–q43. Am J Hum Genet.
2001;68(5):1264–9.
17. Alam NA, Rowan AJ, Wortham NC, et al. Genetic and functional analyses of FH mutations in
multiple cutaneous and uterine leiomyomatosis, hereditary leiomyomatosis and renal cancer,
and fumarate hydratase deficiency. Hum Mol Genet. 2003;12(11):1241–52.
18. Badeloe S, van Geel M, van Steensel MA, et al. Diffuse and segmental variants of cutaneous
leiomyomatosis: novel mutations in the fumarate hydratase gene and review of the literature.
Exp Dermatol. 2006;15(9):735–41.
19. Sudarshan S, Pinto PA, Neckers L, Linehan WM. Mechanisms of disease: hereditary leiomyo-
matosis and renal cell cancer—a distinct form of hereditary kidney cancer. Nat Clin Pract Urol.
2007;4(2):104–10.
20. Mitchum MD, Adams EG, Holcomb KZ. JAAD Grand Rounds quiz. A 46-year-old man with
agminated papules on the buttock. Reed syndrome. J Am Acad Dermatol. 2012;66(2):337–9.
21. Smith CG, Glaser DA, Leonardi C. Zosteriform multiple leiomyomas. J Am Acad Dermatol.
1998;38(2 Pt 1):272–3.
22. Naverson DN, Trask DM, Watson FH, Burket JM. Painful tumors of the skin: “LEND AN
EGG”. J Am Acad Dermatol. 1993;28(2 Pt 2):298–300.
23. Apatenko AK, Turusov VS. The painful tumours of the skin. Neoplasma. 1968;15(2):
187–202.
24. Lehtonen HJ. Hereditary leiomyomatosis and renal cell cancer: update on clinical and molecular
characteristics. Fam Cancer. 2011;10(2):397–411.
25. Kilpatrick SE, Mentzel T, Fletcher CD. Leiomyoma of deep soft tissue. Clinicopathologic
analysis of a series. Am J Surg Pathol. 1994;18(6):576–82.
26. Spencer JM, Amonette RA. Tumors with smooth muscle differentiation. Dermatol Surg.
1996;22(9):761–8.
27. Miettinen M, Lehto VP, Virtanen I. Antibodies to intermediate filament proteins. The differential
diagnosis of cutaneous tumors. Arch Dermatol. 1985;121(6):736–41.
28. McGinley KM, Bryant S, Kattine AA, et al. Cutaneous leiomyomas lack estrogen and proges-
terone receptor immunoreactivity. J Cutan Pathol. 1997;24(4):241–5.
32 Reed’s Syndrome 227
29. Pithukpakorn M, Toro JR. Hereditary leiomyomatosis and renal cell cancer. In: Pagon RA,
Bird TD, Dolan CR, Stephens K, editors. Genereviews. Seattle, Washington: University of
Washington; 1993–2006.
30. Rongioletti F, Fausti V, Ferrando B, et al. A novel missense mutation in fumarate hydratase in
an Italian patient with a diffuse variant of cutaneous leiomyomatosis (Reed’s syndrome).
Dermatology. 2010;221(4):378–80.
31. Fisher WC, Helwig EB. Leiomyomas of the skin. Arch Dermatol. 1963;88:510–20.
32. Montgomery H, Winkelmann RK. Smooth-muscle tumors of the skin. AMA Arch Derm.
1959;79(1):32–40.
33. George S, Pulimood S, Jacob M, Chandi SM. Pain in multiple leiomyomas alleviated by
nifedipine. Pain. 1997;73(1):101–2.
34. Thompson JA. Jr Therapy for painful cutaneous leiomyomas. J Am Acad Dermatol. 1985;
13(5 Pt 2):865–7.
35. Batchelor RJ, Lyon CC, Highet AS. Successful treatment of pain in two patients with cutane-
ous leiomyomata with the oral alpha-1 adrenoceptor antagonist, doxazosin. Br J Dermatol.
2004;150(4):775–6.
36. Thyresson HN, Su WP. Familial cutaneous leiomyomatosis. J Am Acad Dermatol. 1981;
4(4):430–4.
37. Haugen RN, Tharp MD. The use of gabapentin for recurrent painful attacks with multiple
piloleiomyomas. J Drugs Dermatol. 2008;7(4):401–2.
38. Sifaki MK, Krueger-Krasagakis S, Koutsopoulos A, et al. Botulinum toxin type A-treatment
of a patient with multiple cutaneous piloleiomyomas. Dermatology. 2009;218(1):44–7.
39. Onder M, Adien E. A new indication of botulinum toxin: leiomyoma-related pain. J Am Acad
Dermatol. 2009;60(2):325–8.
40. Christenson LJ, Smith K, Arpey CJ. Treatment of multiple cutaneous leiomyomas with CO2
laser ablation. Dermatol Surg. 2000;26(4):319–22.
41. Aoki KR. Review of a proposed mechanism for the antinociceptive action of botulinum toxin
type A. Neurotoxicology. 2005;26(5):785–93.
42. Aoki KR. Pharmacology and immunology of botulinum toxin type A. Clin Dermatol. 2003;
21(6):476–80.
43. Hindley JT, Law PA, Hickey M, et al. Clinical outcomes following percutaneous magnetic
resonance image guided laser ablation of symptomatic uterine fibroids. Hum Reprod. 2002;
17(10):2737–41.
Chapter 33
Tuberous Sclerosis
33.1 Introduction
33.2 Background
TSC is caused by a mutation in either the TSC1 or TSC2 genes with resultant loss of
cell growth control. While TSC is an autosomal dominant disorder, the majority
of cases are not inherited. Instead, 65–85 % arise from spontaneous mutations [2].
The two genes are distinct in location and function but produce nearly indistinguish-
able clinical presentation. Mutations in TSC2 are found in more than half of the total
number of patients. In about one fifth of patients, no mutation is identified in either
of the TSC1 or TSC2 genes [3]. The range of prevalence estimates vary widely from
1 in 5,800 to 1 in 25,000 in the Caucasian population [4, 5].
The biological roles of hamartin (the protein product of TSC1) and tuberin (the
protein product of TSC2) have been fairly well defined. They form a multimeric
complex that functions in the mTOR pathway. The mTOR pathway plays an
important role in cell growth and proliferation hence its effect on cell growth.
The loss of function of either gene leads to increased activation of this pathway. The
TSC complex interacts with Rheb, a member of the RAS subfamily, and stimulates
conversion of Rheb to its inactive form. Loss of function TSC complex mutation
leads to an increase in active Rheb, which in turn increases downstream mTOR
expression.
Presentation for TSC in the skin varies widely from a few subtle angiofibromas to
diffuse angiofibromatosis with shagreen patches and ash-leaf macules, with clinical
manifestations depending largely on the genetic expression in a given patient.
Ash-leaf macules and shagreen patches are more likely to be present either at birth
or shortly thereafter. Angiofibromas, however, may appear at any time but most
often appear in late childhood or early adolescence frequently appearing much like
the papules of acne vulgaris. Angiofibromas are several millimeter in diameter,
fleshy pink to red papules. These can occur singly or in clusters of hundreds often
in a malar distribution (Fig. 33.1). Koenen tumors are subungual fibromas that are
pathognomonic for TSC (Fig. 33.2). Ash-leaf macules are hypopigmented macules
that generally have a single broad side that tapers into a narrow tip, while shagreen
patches are hamartomas of connective tissue that are typically raised, irregularly
shaped, and firmer than surrounding tissue.
Fig. 33.1 Thousands of angiofibromas cover the face of a patient with Tuberous Sclerosis (Photo
credit: Joshua A. Zeichner, M.D.)
33 Tuberous Sclerosis 231
Fig. 33.2 Reddish to flesh-colored, smooth, firm subungual papules. Koenen tumors may also appear
in a periungual distribution, emerging along the nail folds (Photo credit: Joshua A. Zeichner, M.D.)
In addition to the multiple skin findings, TSC also has effects on internal organ
systems as well. The presence of skin changes should prompt a more thorough
work-up to examine for possible renal lesions, neurologic findings, and pulmonary
manifestations. Sequelae from internal organ involvement can cause serious morbidity
and mortality [6].
33.4 Work-Up
Diagnosis of TSC is based upon diagnostic criteria and therefore cannot simply be
made upon any one clinical finding such as the presence of angiofibromas. Diagnosis
of TSC requires associated lesions of two or more organ systems or at least two
different lesions in the same organ to confirm diagnosis [7]. In fact, some of the
so-called pathognomonic features such as intellectual disability and epilepsy are so
common in the general population that they are not specific enough to contribute to
diagnosis [8]. For this reason a list of criteria was set forth to establish consensus as
to a reliable and consistent method of diagnosis [9]. Complete diagnosis and the
evaluation of TSC go beyond the scope of this text; however, one or more angiofi-
bromas, without other appropriate findings, do not suffice for diagnosis nor does it
imply a work-up for TSC is warranted. Angiofibromas occur in over 90 % of indi-
viduals with TSC. However, a variant of angiofibromas occur commonly in the
general population and are known as fibrous papules of the nose or simply fibrous
papules. These may occur singly or multiply but only rarely in as high numbers or
as diffusely as is manifest in the angiofibromas of TSC [10].
232 O. Pacha and A. Hebert
33.5 Treatment
The treatment of TSC depends on the patient’s clinical status and radiologic findings.
Diagnosis and treatment should include appropriate referral to a TSC clinic if pos-
sible with adequate follow-up and screening. Cutaneous lesions can be monitored or
treated depending on patient preference and overall status using mechanical removal
or Argon and /or CO2 laser technologies. More inexpensively, radiofrequency
ablation, chemical peel, and dermabrasion may also be effective [11, 12].
With the discovery of the role of the TSC complex in the mTOR pathway,
utilization of mTOR inhibitors was identified as a potential treatment of all tumors
in TSC patients. One mTOR inhibitor is sirolimus, also known as rapamycin, an
immunosuppressant FDA-approved for use in transplant patients. Sirolimus has
shown promise in clinical trials both systemically and topically for the treatment of
angiofibromas [13, 14]. This therapeutic strategy appears to inhibit angiogenesis by
decreasing production of vascular endothelial growth factor [15]. Ongoing clinical
trials are underway to study sirolimus as an effective and safe for mainstream
treatment.
References
1. Hofbauer GFL, Marcollo-Pini A, Corsenca A, et al. The mTOR inhibitor rapamycin signifi-
cantly improves facial angiofibroma lesions in a patient with tuberous sclerosis. Br J Dermatol.
2008;159:473–5.
2. Osborne JP, Fryer A, Webb D. Epidemiology of tuberous sclerosis. Ann N Y Acad Sci. 1991;
615:125–7.
3. Sancak O, Nellist M, Goedbloed M, et al. Mutational analysis of the TSC1 and TSC2 genes in
a diagnostic setting: genotype–phenotype correlations and comparison of diagnostic DNA
techniques in tuberous sclerosis complex. Eur J Hum Genet. 2005;13:731–41.
4. Morrison PJ. Tuberous sclerosis: epidemiology, genetics and progress towards treatment.
Neuroepidemiology. 2009;33:342–3.
5. OSBORNE JP, FRYER A, WEBB D. Epidemiology of tuberous sclerosis. Ann N Y Acad Sci.
1991;615:125–7. doi:10.1111/j.1749-6632.1991.tb37754.x.
6. Weiner DM, Ewalt DH, Roach ES, Hensle TW. The tuberous sclerosis complex: a comprehensive
review. J Am Coll Surg. 1998;187:548–61.
7. Roach ES, Sparagana SP. Diagnosis of tuberous sclerosis complex. J Child Neurol. 2004;
19:643.
8. Józwiak S, Schwartz RA, Janniger CK, Michałowicz R, Chmielik J. Skin lesions in children
with tuberous sclerosis complex: their prevalence, natural course, and diagnostic significance.
Int J Dermatol. 1998;37:911–7. doi:10.1046/j.1365-4362.1998.00495.x.
9. Roach ES, Gomez MR, Northrup H. Tuberous sclerosis complex consensus conference:
revised clinical diagnostic criteria. J Child Neurol. 1998;13:624–8.
10. Sand M, Sand D, Thrandorf C, Paech V, Altmeyer P, Bechara FG. Review Cutaneous lesions
of the nose 2010. Head Face Med. 2010;6:7.
11. Swaroop MR, Nischal KC, Rajesh Gowda CM, Umashankar NU, Basavaraj HB, Sathyanarayana
BD. Radiofrequency ablation of adenoma sebaceum. J Cutan Aesthet Surg. 2008;1:89.
33 Tuberous Sclerosis 233
12. Kaufman AJ, Grekin RC, Geisse JK, Frieden IJ. Treatment of adenoma sebaceum with the
copper vapor laser. J Am Acad Dermatol. 1995;33:770–4.
13. Salido R, Garnacho-Saucedo G, Cuevas-Asencio I, Ruano J, Galán-Gutierrez M, Vélez A,
Moreno-Giménez J. Sustained clinical effectiveness and favorable safety profile of topical
sirolimus for tuberous sclerosis—associated facial angiofibroma. J Eur Acad Dermatol
Venereol. 2011. doi:10.1111/j.1468-3083.2011.04212.x.
14. Kaufman ME, Curtis AR, Fleischer AB. Successful treatment of angiofibromata of tuberous
sclerosis complex with rapamycin. J Dermatol Treat. 2012;23(1):46–8.
15. Guba M, Von Breitenbuch P, Steinbauer M, et al. Rapamycin inhibits primary and metastatic
tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor. Nat Med.
2002;8:128–35.
Part V
Other Mimickers of Acne Vulgaris
Chapter 34
Acne Scarring
Neal Bhatia, Consuelo Veronica David, Salar Hazany, and Aman Samrao
34.1 Introduction
The inflammatory lesions caused by acne such as papules, pustules, nodules, and
cysts can result in two types of changes to the skin—temporary pigment changes
and true scarring. These lesions are common and can be just as must a source of
psychologically distress to patients as active acne lesions. While pigment changes are
temporary and resolve on their own over months to years, true scars are permanent.
The appearance of true scars may be improved with various modalities including
intralesional cortisone injections and laser therapies. Pigmentary alteration will be
covered in another chapter, and in the following we will review true scars.
34.2 Background
True scars result from permanent changes to the epidermis, dermis, and subcutaneous
tissue. Following the initial injury, tissue undergoes three stages of wound healing:
inflammation, granulation, and tissue remodeling [1]. During tissue remodeling,
keratinocytes and fibroblasts produce enzymes that dictate the proportion of matrix
metalloproteinases (MMPs) to MMP inhibitors. Exuberance of the wound healing
response, as well as a predominance of either MMPs or MMP inhibitors, determines
how a scar will heal [1]. Acne scars are classified as atrophic or hypertrophic, and a
summary of their clinical appearances is summarized (Table 34.1). Although
research supports a genetic predisposition for the development of keloids, such a
predisposition has not been suggested for atrophic scars.
N. Bhatia, M.D. (*) • C.V. David, M.D. • S. Hazany, M.D. • A. Samrao, M.D.
Division of Dermatology, Harbor-UCLA Medical Center, 1000 W. Carson Street,
Box 259, Torrence, CA 90502, USA
e-mail: [email protected]
In general, patients with darker skin types more commonly develop keloids than
patients of other skin types, although the exact prevalence is not known. It is widely
accepted that keloids develop as a result of a genetic predisposition with an environ-
mental influence. It is unclear whether the predisposition to keloids results in single
Mendelian or polygenetic disorder. Both autosomal dominant with incomplete pen-
etrance and autosomal recessive modes have been reported [2, 3]. Chromosomes
2q23 and 7p11 mutations and HLA-DRB1*15, HLA-DQA1*0104, DQ-B1*0501,
and DQB1*0503 all may play a role [2].
Hypertrophic scars and keloids are two types of scars resulting from over-exuberant
wound healing responses with excess collagen deposition and decreased collage-
nase activity [1]. Hypertrophic scars are lesions that elevate above the skin surface,
but remain within the boundaries of the original area of damage [6]. These lesions
may spontaneously involute. Keloids on the other hand are lesions with collagen
deposition beyond the border of the original wound (Fig. 34.2). Histologically,
keloids are characterized by thick, hyalinized collagen bundles [6, 7]. Keloids can
be painful, itchy, and burn. Importantly, keloids due to acne should not be confused
with acne keloidalis nuchae, a primary cicatricial alopecia that has no association
with acne vulgaris [8].
34.4 Work-Up
The diagnosis of acne scarring is based on clinical appearance, and little work-up is
necessary. Patients should be educated on the difference between post-inflammatory
erythema or pigmentation and true acne scarring. It is important to set realistic
expectations on outcomes, treatment options, and the time course for improvement.
Psychosocial issues should also be addressed, as acne scars can have a large impact
on self esteem and be a source of depression [9].
Scars have distinct histologic appearances. The various atrophic scar subtypes
are similar, with an atrophic epidermis and effacement of the rete ridges. Collagen
fibers are arranged parallel to the epidermis, while blood vessels are oriented
34 Acne Scarring 241
34.5 Treatment
34.6 Conclusion
References
1. Thiboutot D, Gollnick H, Bettoli V, Dreno B, Kang S, Leyden JJ, et al. New insights into the
management of acne: an update from the global alliance to improve outcomes in acne group.
J Am Acad Dermatol. 2009;60:S1–50.
2. Shih B, Bayat A. Genetics of keloid scarring. Arch Dermatol Res. 2010;302:319–39.
3. Halim AS, Emami A, Salahshourifar I, Kannan TP. Keloid scarring: understanding the genetic
basis, advances, and prospects. Arch Plast Surg. 2012;39:184–9.
4. Rivera AE. Acne scarring: a review and current treatment modalities. J Am Acad Dermatol.
2008;59:659–76.
5. Jacob CI, Dover JS, Kaminer MS. Acne scarring: a classification system and review of treat-
ment options. J Am Acad Dermatol. 2001;45:109–17.
6. Rapini R. Practical dermatopathology. Houston: Elsevier-Mosby; 2012.
7. Verhaegen PD, van Zuijlen PP, Pennings NM, van Marle J, Niessen FB, van der Horst CM,
et al. Differences in collagen architecture between keloid, hypertrophic scar, normotrophic
scar, and normal skin: an objective histopathological analysis. Wound Repair Regen. 2009;
17:649–56.
8. James W, Berger T, Elston D. Andrews’ diseases of the skin clinical dermatology. Philadelphia:
Elsevier-Saunders; 2011.
9. Levy LL, Zeichner JA. Management of acne scarring, part II: a comparative review of
non-laser-based, minimally invasive approaches. Am J Clin Dermatol. 2012;13:331–40.
10. Fabbrocini G, Annunziata MC, D’Arco V, De Vita V, Lodi G, Mauriello MC, et al. Acne scars:
pathogenesis, classification and treatment. Dermatol Res Pract. 2010;2010:893080.
34 Acne Scarring 243
11. Fife D. Practical evaluation and management of atrophic acne scars: tips for the general
dermatologist. J Clin Aesthet Dermatol. 2011;4:50–7.
12. Orentreich D, Orentreich N. Acne scar revision update. Dermatol Clin. 1987;5:359–68.
13. Tsao SS, Dover JS, Arndt KA, Kaminer MS. Scar management: keloid, hypertrophic, atrophic,
and acne scars. Semin Cutan Med Surg. 2002;21:46–75.
14. Johnson WC. Treatment of pitted scars: punch transplant technique. J Dermatol Surg Oncol.
1986;12:260–5.
15. Solotoff SA. Treatment for pitted acne scarring–postauricular punch grafts followed by
dermabrasion. J Dermatol Surg Oncol. 1986;12:1079–84.
16. Al-Waiz MM, Al-Sharqi AI. Medium-depth chemical peels in the treatment of acne scars in
dark-skinned individuals. Dermatol Surg. 2002;28:383–7.
17. Kim S, Cho KH. Clinical trial of dual treatment with an ablative fractional laser and a nonabla-
tive laser for the treatment of acne scars in Asian patients. Dermatol Surg. 2009;35:1089–98.
18. Ong MW, Bashir SJ. Fractional laser resurfacing for acne scars: a review. Br J Dermatol.
2012;166:1160–9.
19. Roenigk Jr HH. Dermabrasion for miscellaneous cutaneous lesions (exclusive of scarring from
acne). J Dermatol Surg Oncol. 1977;3:322–8.
20. Yarborough Jr JM. Ablation of facial scars by programmed dermabrasion. J Dermatol Surg
Oncol. 1988;14:292–4.
Chapter 35
Eosinophilic Pustular Folliculitis
35.1 Introduction
First described in Japan by Ofuji in 1970 [1], eosinophilic pustular folliculitis (EPF) is
a noninfectious inflammatory skin disease that manifests with coalescing papulopus-
tular plaques. The disease is histologically characterized by eosinophilic infiltration of
hair follicles. There are three known variants of EPF: classic (as originally described
by Ofuji and predominantly affecting Japanese individuals), HIV-associated, and
infantile.
35.2 Background
The majority of reported EPF cases are related to immunosuppression, while the
classic and infantile EPF are far less common [2]. The classic variant of EPF is
found predominantly in Japanese patients, though any race may be affected [3].
Males are more commonly affected than women, in a ratio generally reported as
4.8:1 [2, 4], though a study in Singapore found a ratio of 1.6:1 [5]. The incidence of
classic EPF is concentrated in the third and fourth decades of life, with the average
age of onset at 30 years [6].
The immunosuppression-related variant chiefly affects HIV-infected individuals,
though cases have also been seen in intravenous drug abusers [7, 8]. There is also a
tremendous male predominance; only six cases have been reported in female
patients [9]. Most cases of HIV-related EPF have been seen in Caucasian patients,
and the majority of cases reported are from the United States and Great Britain.
Patients tend to present to the doctors’ offices later in the course of their disease,
often once they have established acquired immunodeficiency syndrome [10].
The pathophysiology of EPF is largely unknown. Theories on its etiology take
into account the prominent eosinophilic cellular infiltrate, the folliculocentric char-
acter of the lesions, and the distribution of the eruption. Hypersensitivity reactions
to infection or medication, as well as an autoimmune etiology have been proposed
as possible mechanisms of EPF [2]. For example, EPF can present similarly to a
fungal folliculitis with respect to both its clinical and histological features; it is
hypothesized that the folliculitis may be attributable to immune hyperreactivity to
dermatophytes or other fungi [11]. This theory is supported by the positive thera-
peutic response in patients treated with antifungal agents [2]. Other possible
antigenic sources investigated include mites (Demodex folliculorum and Demodex
brevis) and bacteria (Leptotrichia buccalis) [2]. In addition, medications such as
carbamazepine, minocycline, allopurinol, timepidium bromide [3, 12, 13], and
silicone tissue injections [14] have been reported as possible causes of EPF skin
reactions.
Given the high incidence of eosinophilic pustular reactions in the setting of
coexisting immune dysfunction, it has been suggested that immune dysregulation
may also be a contributing factor to EPF [15]. Furthermore, it has been proposed
that the HIV-associated variant of EPF may also be an autoimmune disorder, with a
component of sebum behaving as the target antigen [10, 16]. Of note, there are also
cases of EPF in HIV-negative immunocompromised states, such as in lymphoma
patients after autologous peripheral blood stem cell and allogeneic bone marrow
transplantation [3, 17]. It is likely that this variant of EPF is a nonspecific manifesta-
tion that presents in the setting of a compromised immune system and may be pro-
voked by various antigens [2]. Given these reported findings, it is critical to evaluate
EPF patients for coexisting systemic disease [2]. Though many hypotheses have
been proposed, more research is required to further elucidate the mechanism and
conclusively determine the pathophysiology of EPF.
Though HIV-related EPF and classic EPF (a.k.a. Ofuji’s disease) appear similar
histologically, they differ clinically. The classic form arises in healthy patients and
generally presents with chronic, recurring, sterile, folliculocentric papules and pus-
tules [10, 18]. These lesions usually develop into confluent annular plaques with
central clearing [2]. The classic distribution includes seborrheic areas such as the
face (85 %), back, and trunk (59 %), as well as non-hair-bearing areas such as
the palms and soles (20 %) [2, 16, 18]. The lesions typically resolve without scarring,
but may develop postinflammatory hyperpigmentation. Furthermore, pruritus affects
<50 % of patients [10].
In HIV-associated EPF, immunocompromised patients universally present with a
defining feature: chronic, severe, intractable pruritus [10]. Rather than confluent
35 Eosinophilic Pustular Folliculitis 247
Fig. 35.1 Flesh colored and excoriated papules on face (Photo credits: Joshua A. Zeichner, M.D.)
Fig. 35.2 Post-inflammatory pigmentation and excoriations on the chest. The patient complained
of extreme itching, yet no primary lesions are visible. The patient is HIV positive (Photo credits:
Joshua A. Zeichner, M.D.)
35.4 Work-Up
Patients with suspected EPF should have a complete blood count with differential, as
peripheral eosinophilia has been found in up to 35 % of classic EPF patients [1, 6]
and 50 % of HIV-associated EPF patients [8, 10, 21, 22]. In the classic form, a mild
to moderate leukocytosis may be present, whereas a leukopenia may be present in
the HIV-associated variant [18]. Serum IgE levels have also been reported to be
elevated significantly in a large number of HIV-associated EPF patients, as com-
pared to levels in HIV controls [21, 23]. Patients tend to have a CD4 count less than
250–300 cells/ml [21].
The EPF variants share common findings on histopathologic analysis: noninfec-
tious infiltration of eosinophils. Eosinophilic spongiosis and pustulosis are seen on
pathology, particularly in the infundibulum of the hair follicle [18]. The infiltrate
often extends to the adjacent sebaceous gland and duct [18]. In addition, there is a
perivascular and perifollicular infiltrate consisting of lymphocytes [18]. When tak-
ing the biopsy, it is necessary o acquire an entire papule or pustule with a contiguous
follicle for adequate histopathological diagnosis. In addition, because EPF is
folliculocentric, serial sections may be necessary to visualize the inflamed follicle
and confirm the diagnosis [2, 10]. Routine hematoxylin and eosin stain should be
performed, as well as specific stains for fungi and bacteria.
If a microbial infection is suspected in the differential diagnosis, skin swabs
for microscopy and culture and scrapings for mycologic examination should be performed.
It is clinically difficult to distinguish between the different HIV-associated
folliculitides, including EPF and infective folliculitis. However, routine bacterial
and fungal cultures are usually negative in HIV-associated EPF [21, 24]. Furthermore,
histology is diagnostic and can be used to differentiate between infective folliculitis
and EPF [10].
35.5 Treatment
A myriad of treatment options have been described with variable results. Topical
corticosteroids are the first-line treatment for all forms of EPF [6]. The dose is
modified depending on the age of the patient: mild to moderate potency for children
and moderate to high potency for adults [2, 6]. Topical tacrolimus also appears to be
an effective first-line agent [6, 25].
Classic EPF is often treated with nonsteroidal anti-inflammatory drug deriva-
tives, most commonly oral indomethacin [3, 5, 6, 10]. Throughout the many years
of use by Japanese dermatologists, it appears to be the most effective treatment for
the classic form of the disease. One case series of 25 patients found indomethacin
to be effective in 92 % of patients [26].
If topical corticosteroids and oral indomethacin are unsuccessful in treating HIV-
associated EPF, a variety of other treatment options can be utilized. These include
35 Eosinophilic Pustular Folliculitis 249
35.6 Conclusion
References
36.1 Introduction
36.2 Background
36.4 Work-Up
36.5 Treatment
regimens. Avoidance of risk factors such as excessive sun exposure, long term ste-
roid use, and tobacco should be discussed. The importance of proper sun protection
should also be addressed to help limit the potential actinic damage. When consider-
ing therapeutic options, physicians must take into consideration their patient’s skin
type, predominant lesion, likelihood of recurrence, and willingness to undergo
high-risk procedures. Even though this condition is not life threatening, it can result
in significant psychological, emotional, and social distress. Quality of life must
therefore be taken into consideration when treating this relatively benign skin
condition.
References
37.1 Introduction
37.2 Background
HS typically begins during the second and third decade of life, most often between
the ages of 21 and 23, although cases of prepubertal HS and postmenopausal HS
occasionally occur. The prevalence rate varies between 0.03 and 4 %, but true preva-
lence is probably 1 % in the general population [1, 2]. Women are more likely to be
affected than men and disease distribution varies by gender. The groin and inframa-
mmary regions are more commonly affected in women, while the buttocks, perineum,
and perianal area are more commonly affected in men.
The pathogenesis of HS is unclear and may include genetic, infectious, hormonal,
behavioral, and even host defense factors. Once thought to be a disorder of the apo-
crine gland, HS actually represents follicular plugging as the primary event. As such,
inflammation of the apocrine gland is not essential and apocrinitis is a secondary
phenomenon.
Approximately 40 % of HS patients have familial HS, inherited as an autosomal
dominant trait with 100 % penetrance. The likely defect may be a dysfunctional γ
(gamma)-secretase-Notch pathway with mutations in Presenilin-1 (PSEN1), Presenilin
Enhancer-2 (PSENEN), and Nicastrin (NCSTN) genes, which encode proteins integral
to γ (gamma)-secretase enzyme [3]. Gamma-secretase cleaves type 1 transmembrane
proteins such as Notch, and mice without Notch-1 have occluded hair follicles, thought
to be the initiating pathogenic event in HS.
HS is a component of the follicular occlusion tetrad which includes acne conglo-
bata, dissecting cellulitis of the scalp, and pilonidal cysts. Known associations include
acne vulgaris (30–70 %), obesity (51–75 %), smoking (70–89 %), and Crohn’s
disease. HS occasionally occurs with some genetic disorders, specifically keratosis-
ichthyosis-deafness (KID) syndrome, Dowling-Degos Disease, SAPHO syndrome,
PAPA syndrome, pachyonychia congenita, and Fox-Fordyce disease [1, 4].
to axillary disease; most patients with perianal and perineal involvement have
recurrent disease and poorer quality of life [5].
During the course of the disease, additional nodules and plaques develop de novo
or from extension of existing nodules, either on affected or adjacent skin or occa-
sionally at distant sites. Fifty percent of patients experience a prodrome consisting
of burning, stinging, pain, and pruritus, with or without hyperhidrosis, occurring
12–48 h before disease onset. The subcutaneous coalescence of neighboring cysts
or the lateral extension of proliferating pilosebaceous material with rupture to the
surface produces deep dermal abscesses and interlinked sinus tracts, which may
lead to the formation of honeycombed fistulous tracts with chronic infection.
Drainage from these tracts is common and may be serous, purulent, bloody, or a
mixture and is often malodorous.
Ultimately, over time, disease becomes quiescent, invariably healing with scarring.
Occasionally, indolent cysts may develop on the face, behind the ears, at the nape of
the neck, on the trunk, and in the genital area. These cysts are more common in men.
Sinus tracts can coalesce to form hypertrophic, fibrous fistulae, and subcutaneous
sinus networks can form characteristic hypertrophic scars or dense, ropelike fibrotic
bands. Fifty percent of patients develop the “tombstone comedone” of permanently
dilated pores within an old “burned out” area of disease.
Most patients with chronic or untreated HS develop complications, e.g., anal,
urethral, and rectal strictures, fistulae, and fecal incontinence from genitofemoral
involvement or contractures and compromised mobility, especially in the axillae
and thighs, from axillary or perianal disease. Other sequelae include spondyloar-
thropathy, pyogenic granuloma, lymphedema, lymphangiectasia [6], scrotal and
vulvar edema, secondary infections including cellulitis and even septicemia,
epidural abscesses, and sacral osteomyelitis. Metabolic sequelae are uncommon,
262 D.M. Sikorski and K.J. Tomecki
37.4 Work-Up
The diagnosis is made clinically and biopsy is rarely necessary. The differential
diagnosis includes infectious and inflammatory diseases such as acne, carbuncles,
furuncles, inflamed cysts, fistulous abscess, granuloma inguinale, lymphogranuloma
venereum, tuberculous abscess, noduloulcerative syphilis, actinomycosis, blastomyco-
sis, nocardiosis, and cat scratch disease. HS can resemble Crohn’s disease; if suggestive,
GI and surgical evaluation may be necessary. Drainage warrants bacterial, fungal, and
mycobacterial culture, which may reveal Staphylococcus aureus or Gram-negative
organisms. Disease severity is variable and is quantified by the older Hurley scale or
newer Sartorius scale.
37.5 Treatment
There is no uniformly successful therapy for HS. Standard treatment depends on the
extent and severity of disease. Purely medical treatment is invariably prolonged
since permanent cure is uncommon. Regardless of severity, all patients deserve
information and education regarding preventative measures including the impor-
tance of good daily hygiene coupled with antimicrobial cleansers and reduction of
heat, friction, and excessive sweating, including use of loose-fitting clothing.
Adjunctive therapy should include smoking cessation, weight reduction, and support
group referral.
37 Hidradenitis Suppurativa 263
Medical therapies include topical and systemic antibiotics, topical and systemic
retinoids, systemic and intralesional corticosteroids, anti-androgens, dapsone,
cyclosporine, and anti-TNF-alpha inhibitors. Mild HS is often treated with intral-
esional corticosteroids and/or short courses of topical or oral antibiotics, with
selection based on sensitivities of cultured organisms, most commonly clindamy-
cin or a tetracycline. Complete remission with antibiotic treatment is rare, but
combination therapy with oral clindamycin and rifampicin [8] or with rifampicin,
moxifloxacin, and metronidazole has produced remissions in patients with mild to
moderate HS. Anti-androgen therapy with ethinylestradiol, cyproterone acetate,
and finasteride has proven to be helpful in many patients. TNF-alpha inhibitors
such as infliximab [9], etanercept [10], and adalimumab [11] have been effective in
some patients with moderate to severe disease, but long-term results have been
poor and the side effect profile of these drugs and their cost have limited their use.
Despite successful use in acne, oral retinoids have not been shown to be effective
in treating HS.
Surgical interventions with botulinum toxin, radiotherapy, carbon dioxide laser,
long-pulsed neodymium: yttrium-aluminum-garnet (YAG) laser [12], and photody-
namic [13] therapy have shown success in some patients. Incision and drainage has
little benefit since recurrence is so common. Surgical excision remains one of the most
successful therapies for recalcitrant HS, despite its appreciable morbidity and compli-
cations such as infection, scarring, graft failure, ischemic necrosis of myocutaneous
flaps, and recurrence. A recent review of 72 surgical HS patients showed a recurrence
rate of 54 % following excision to fascia with primary closure, but 19 % recurrence
with myocutaneous flap and 13 % recurrence with split thickness skin graft [14].
References
11. Blanco R, Martínez-Taboada VM, Villa I, et al. Long-term successful adalimumab therapy in
severe hidradenitis suppurativa. Arch Dermatol. 2010;145:580–84.
12. Mahmoud BH, Tierney E, Hexsel CL, Pui J, Ozog DM, Hamzavi IH. Prospective controlled
clinical and histopathologic study of hidradenitis suppurativa treated with the long-pulsed
neodymium:yttrium-aluminium-garnet laser. J Am Acad Dermatol. 2010;62:637–45.
13. Syed ZU, Hamzavi IH. Photomedicine and phototherapy considerations for patients with skin
of color. Photodermatol Photoimmunol Photomed. 2011;27:10.
14. Rambhatla PV, Lim HW, Hamzavi I. A systematic review of treatments for hidradenitis
suppurativa. Arch Dermatol. 2012;148:439–46.
Chapter 38
Perioral Dermatitis
38.1 Introduction
Perioral dermatitis (PD), a common acneiform facial eruption, was first described in
the 1950s. The terms “light-sensitive seborrheid” and “steroid-induced rosacea-like
dermatitis” were previously used to describe this condition [1, 2]. Given the now
recognized periorbital, perinasal, and perioral distribution of lesions, the term peri-
orificial dermatitis may be most appropriate [3].
38.2 Background
The exact etiology and pathogenesis of PD has not been elucidated. However, an
association with the use/misuse of topical corticosteroids has been well established.
Corticosteroid exposure through intranasal, inhaled, and systemic modes of delivery
has also been reported to incite PD [4–8]. Not all cases of PD demonstrate a history
of corticosteroid exposure [9].
Other specific environmental factors have not been identified, and patients do not
exhibit photosensitivity. Suggestions of microbiologic pathogenesis due to Candida
species, Demodex folliculorum, or fusiform bacteria have not been substantiated.
Allergic contact dermatitis to constituents of dentifrices (i.e., fluoride) and cosmet-
ics has been reported as a contributing factor in some cases although rechallenge
with these same products after resolution of PD may not consistently incite recur-
rence of PD.
Although PD has been considered by some to be a variant of acne rosacea, the
distribution of inflammatory lesions and absence of persistent background erythema
and telangiectasia differentiate PD from acne rosacea.
PD affects both adults and children. Adult PD disproportionately affects females
of reproductive age (25–40 years old). Pediatric PD affects both girls only slightly
more often than boys with onset typically before 5 years old. Pediatric PD in
children as young as 6 months has been reported [10]. There appears to be no racial
predilection in pediatric or adult PD. PD has been shown to occur in siblings, but
genetic predisposition has not been further elucidated [11].
Fig. 38.1 Erythematous papules and pustules on a background of erythema and fine scaling
involving the perioral and perinasal skin. Sparing of the skin immediately surrounding the vermil-
ion border
Fig. 38.2 (a) A patient at baseline before therapy for perioral dermatitis (b) 6 weeks after therapy
with oral doxycycline and topical metronidazole for perioral dermatitis (c) 12 weeks after therapy
with oral doxycycline and topical metronidazole for perioral dermatitis
38 Perioral Dermatitis 267
38.4 Work-Up
38.5 Treatment
There are no FDA-approved therapies for PD. While the therapeutic armamentarium
for the treatment of PD is large, the level of evidence supporting many of the available
agents is poor. Two systematic, evidence-based reviews of the literature suggest that
some of the strongest evidence supports “zero therapy” for PD, i.e., withdrawal of any
offending or exacerbating agents, such as topical or inhaled corticosteroids or cosmet-
ics [18, 19]. Multiple trials have demonstrated that avoidance of topical corticosteroids
and cosmetics results in improvement in 4 weeks and resolution within an average of
2–3 months. It is well recognized that continued use of topical corticosteroids and
cosmetics prolongs PD regardless of treatment employed.
Cutaneous irritation and sensitivity may limit the utility of topical agents for PD
though use of a gentle non-medicated cleanser and moisturizer may reduce this
tendency. Topical metronidazole, as monotherapy or in combination with oral
antibiotics, has demonstrated efficacy for PD (Fig. 38.2a–c). Use of topical metro-
nidazole gel 0.75 % twice daily in children with PD demonstrated significant
improvement after 8 weeks and resolution after 14 weeks [20]. A prospective,
randomized, double-blind study comparing metronidazole cream 1 % twice daily
with oral tetracycline 250 mg twice daily demonstrated significant reduction in pap-
ule number in the metronidazole group and complete resolution in the tetracycline
group after 8 weeks [21].
Topical erythromycin may also provide benefit [22]. Topical erythromycin
emulsion 2 % twice daily was superior to placebo in clearing papules with an
average time to clearance of 7 weeks; topical erythromycin demonstrated similar
efficacy to oral tetracycline [23]. A case series reported that 98 % of 700 patients
cleared on topical erythromycin [24].
38 Perioral Dermatitis 269
Pimecrolimus has been shown to expedite resolution of PD but may offer limited
long-term benefit. Two randomized, placebo-controlled trials of adults with PD
independently demonstrated that pimecrolimus cream 1 % twice daily significantly
reduced erythema, scaling, and papule number after 2 weeks compared with vehicle.
After 4 weeks, however, there was no significant difference between pimecrolimus
and vehicle [25, 26].
Oral tetracycline remains the gold standard for the treatment of PD. Randomized
controlled trials and large case series support the use of oral tetracycline as a first-
line agent [27]. Oral tetracycline (250 mg twice daily) has been shown to be more
effective for PD than topical metronidazole or topical erythromycin [21, 23]. Severe
perioral dermatitis recalcitrant to erythromycin ointment 2 % has been shown to
respond to oral tetracycline [24]. The use of doxycycline or minocycline in PD is
supported by case reports and small series [7, 28, 29].
Additional therapeutic options for PD are supported by small, uncontrolled trials
or case series which fail to assess whether an agent is more efficacious than zero
therapy. Open-label studies of azelaic acid cream 20 % twice daily reported complete
clearance within 5–6 weeks [30, 31]. A split-face study using photodynamic therapy
(PDT) with 5-aminolevulinic acid showed greater lesion reduction with PDT than
topical clindamycin gel 1 % at 1 month (92 % vs. 80 %); photosensitivity and post-
inflammatory hyperpigmentation complicated PDT treatment [32]. A retrospective
review of 25 patients treated with sodium sulfacetamide/sulfur 10/5 % reported
clearance in 14 of 25 (56 %) after an average of 1.2 months of treatment [33]. Limited
case reports detail the efficacy adapalene gel 0.1 % and isotretinoin [34, 35].
38.6 Conclusion
References
1. Frumess GM, Lewis HM. Light-sensitive seborrheid. AMA Arch Dermatol. 1957;
75(2):245–8.
2. Mihan R, Ayres Jr S. Perioral dermatitis. Arch Dermatol. 1964;89:803–5.
3. Kihiczak GG, Cruz MA, Schwartz RA. Periorificial dermatitis in children: an update and
description of a child with striking features. Int J Dermatol. 2009;48(3):304–6.
4. Hafeez ZH. Perioral dermatitis: an update. Int J Dermatol. 2003;42(7):514–7.
270 B. Gammon and B.J. Schlosser
30. Jansen T. Azelaic acid as a new treatment for perioral dermatitis: results from an open study.
Br J Dermatol. 2004;151(4):933–4.
31. Jansen T, Melnik BC, Schadendorf D. Steroid-induced periorificial dermatitis in children–
clinical features and response to azelaic acid. Pediatr Dermatol. 2010;27(2):137–42.
32. Richey DF, Hopson B. Photodynamic therapy for perioral dermatitis. J Drugs Dermatol.
2006;5(2 Suppl):12–6.
33. Bendl BJ. Perioral dermatitis: etiology and treatment. Cutis. 1976;17(5):903–8.
34. Jansen T. Perioral dermatitis successfully treated with topical adapalene. J Eur Acad Dermatol
Venereol. 2002;16(2):175–7.
35. Smith KW. Perioral dermatitis with histopathologic features of granulomatous rosacea:
successful treatment with isotretinoin. Cutis. 1990;46(5):413–5.
Chapter 39
Photocontact Dermatitis
39.1 Introduction
39.2 Background
The exact prevalence of PCD in the general population is not known. However,
estimates of its frequency have ranged from 7 to 15 % for phototoxicity and 4 to 8 %
for photoallergy [3–6].
Common phototoxic agents include coal-tar derivatives, dyes, antiarrhythmics,
diuretics, non-steroidal anti-inflammatory drugs (NSAIDs), phenothiazines,
Fig. 39.1 Erythematous, tender plaques on the face that developed after application of a sunscreen
and exposure to UV light outside. The robust photocontact dermatitis became secondarily infected,
with the development of yellow crusted plaques (Photo credit: Joshua A. Zeichner, M.D.)
276 N. Gulati and E. Guttman-Yassky
39.4 Work-Up
39.5 Treatment
Individuals with acute PCD reactions including erythema and edema may find relief
with ice-cold compresses of Burow’s solution in a 1:10 dilution. For acute photo-
toxic reactions, aspirin or other NSAIDs can be used. Alternatively, for generalized
cases, systemic corticosteroid therapy may provide prompt symptomatic relief.
The mainstay of treatment of PCD is avoidance of the offending agents as well as
appropriate photoprotection. Protective clothing such as long-sleeved shirts and hats
should be worn and sunscreens should be applied (provided they are not the offend-
ing agent) [29]. As needed, changes in lifestyle including avoidance of midday sun
as well as alterations in leisure and occupational activities may be recommended.
Special UV-absorbing plastic can be installed over windows and windscreens to
improve photoprotection. For chronic cases that only respond to systemic corticoste-
roids and which have high morbidity, immunosuppressive agents such as azathioprine
can be used [30].
39.6 Conclusion
References
8. Polat M, Oztas P, Dikilitas MC, Alli N. Phytophotodermatitis due to Ficus carica. Dermatol
Online J. 2008;14(12):9.
9. Lagey K, Duinslaeger L, Vanderkelen A. Burns induced by plants. Burns. 1995;21(7):542–3.
10. Bylaite M, Grigaitiene J, Lapinskaite GS. Photodermatoses: classification, evaluation and
management. Br J Dermatol. 2009;161 Suppl 3:61–8.
11. Schauder S, Ippen H. Contact and photocontact sensitivity to sunscreens. Review of a 15-year
experience and of the literature. Contact Derm. 1997;37(5):221–32.
12. Szczurko C, Dompmartin A, Michel M, Moreau A, Leroy D. Photocontact allergy to oxyben-
zone: ten years of experience. Photodermatol Photoimmunol Photomed. 1994;10(4):144–7.
13. Darvay A, White IR, Rycroft RJ, et al. Photoallergic contact dermatitis is uncommon. Br J
Dermatol. 2001;145(4):597–601.
14. Diaz RL, Gardeazabal J, Manrique P, et al. Greater allergenicity of topical ketoprofen in
contact dermatitis confirmed by use. Contact Derm. 2006;54(5):239–43.
15. Agin PP, Ruble K, Hermansky SJ, McCarthy TJ. Rates of allergic sensitization and irritation
to oxybenzone-containing sunscreen products: a quantitative meta-analysis of 64 exaggerated
use studies. Photodermatol Photoimmunol Photomed. 2008;24(4):211–7.
16. Ferguson J. Photosensitivity due to drugs. Photodermatol Photoimmunol Photomed. 2002;
18(5):262–9.
17. Kochevar IE, Harber LC. Photoreactions of 3,3’,4’,5-tetrachlorosalicylanilide with proteins.
J Invest Dermatol. 1977;68(3):151–6.
18. Pendlington RU, Barratt MD. Molecular basis of photocontact allergy. Int J Cosmet Sci.
1990;12(2):91–103.
19. Atarashi K, Kabashima K, Akiyama K, Tokura Y. Stimulation of Langerhans cells with
ketoprofen plus UVA in murine photocontact dermatitis to ketoprofen. J Dermatol Sci. 2007;
47(2):151–9.
20. Miyachi Y, Takigawa M. Mechanisms of contact photosensitivity in mice: II. Langerhans cells
are required for successful induction of contact photosensitivity to TCSA. J Invest Dermatol.
1982;78(5):363–5.
21. Gerberick GF, Ryan CA, Von Bargen EC, Stuard SB, Ridder GM. Examination of tetrachloro-
salicylanilide (TCSA) photoallergy using in vitro photohapten-modified Langerhans cell-
enriched epidermal cells. J Invest Dermatol. 1991;97(2):210–8.
22. Gerberick GF, Ryan CA, Fletcher ER, Howard AD, Robinson MK. Increased number of
dendritic cells in draining lymph nodes accompanies the generation of contact photosensitivity.
J Invest Dermatol. 1991;96(3):355–61.
23. Kurita M, Shimauchi T, Kobayashi M, et al. Induction of keratinocyte apoptosis by photosen-
sitizing chemicals plus UVA. J Dermatol Sci. 2007;45(2):105–12.
24. Epstein JH. Phototoxicity and photoallergy. Semin Cutan Med Surg. 1999;18(4):274–84.
25. Lankerani L, Baron ED. Photosensitivity to exogenous agents. J Cutan Med Surg. 2004;
8(6):424–31.
26. Stein KR, Scheinfeld NS. Drug-induced photoallergic and phototoxic reactions. Expert Opin
Drug Saf. 2007;6(4):431–43.
27. Hölzle E, Lehmann P, Neumann N. Phototoxic and photoallergic reactions. J Dtsch Dermatol
Ges. 2009;7(7):643–9.
28. Anon X. Photopatch testing—methods and indications. British Photodermatology Group. Br J
Dermatol. 1997;36(3):371–6.
29. Fourtanier A, Moyal D, Seité S. Sunscreens containing the broad-spectrum UVA absorber,
Mexoryl SX, prevent the cutaneous detrimental effects of UV exposure: a review of clinical
study results. Photodermatol Photoimmunol Photomed. 2008;24(4):164–74.
30. Yap LM, Foley P, Crouch R, Baker C. Chronic actinic dermatitis: a retrospective analysis of
44 cases referred to an Australian photobiology clinic. Australas J Dermatol. 2003;
44(4):256–62.
Chapter 40
Post-inflammatory Pigment Alteration
40.1 Introduction
40.2 Background
the enzyme tyrosinase, variations in number, size, and groupings of the melano-
somes, as well as the efficiency of melanosome transfer to keratinocyte [7–10].
A key characteristic in skin of color is the tendency for melanocytes to exhibit
labile responses to inflammation and injury [11, 12], which contributes to the high
prevalence of post-inflammatory hyperpigmentation in darker skin types.
Inflammatory mediators, including prostaglandins and leukotrienes, can stimulate
increased melanin synthesis, which can lead to increased pigment in the epidermis
alone or also in the dermis [13]. When there is dermal involvement, there is disrup-
tion of the basal layer that leads to macrophages engulfing the melanin and the for-
mation of melanophages. In addition, a study by Halder et al. examined biopsies in
thirty black patients with acne vulgaris and found the presence of inflammation
histologically even in clinically noninflammatory lesions such as comedones. In
addition, papules and pustules displayed considerable inflammation histologically
that extended significantly beyond the margins of each lesion [14]. While subclini-
cal inflammation is likely a feature of acne vulgaris in all skin types, it may contrib-
ute to the tendency toward dyschromias in acne patients with skin of color [2].
Fig. 40.1 An African-American woman with Fitzpatrick skin type VI and severe post-
inflammatory hyperpigmentation associated with acne vulgaris
40 Post-inflammatory Pigment Alteration 281
is prolonged and can last several months to years. Hypopigmentation can also be seen
in acne patients as a sequela of irritant dermatitis from topical acne therapies. It can
also be seen with the use of skin lightening agents to lighten PIH, typically presenting
with perilesional halos of hypopigmentation.
40.4 Work-Up
40.5 Treatment
If considered, skin type should always be taken into account given the high risk of
post-inflammatory hyperpigmentation [32]. While fractional nonablative lasers may
be useful for hyperpigmentation, optimal treatment parameters for dyschromias in
skin of color have not been well established and treatment outcomes are unpredict-
able; however, lower treatment levels and prophylactic use of hydroquinone both
before and after laser treatment are recommended to minimize post-treatment
hyperpigmentation [53–55].
Post-inflammatory hyperpigmentation secondary to acne vulgaris can be an
extremely challenging process to manage, and thus the prevention of such lesions is
paramount with early intervention and meticulous sun protection. Anti-inflammatory
agents (including systemic therapies where appropriate) should be initiated as early
as possible in patients prone to post-inflammatory hyperpigmentation [56].
Combination therapies with the addition of lightening agents have shown to be ben-
eficial, and chemical peels can be considered as second-line approaches. New thera-
peutic agents, however, are needed including alternatives to hydroquinone and
improved strategies for the treatment of resistant dermal pigmentation.
References
1. Davis EC, Callender VD. A review of acne in ethnic skin: pathogenesis, clinical manifesta-
tions, and management strategies. J Clin Aesthet Dermatol. 2010;3(4):24–38. PubMed PMID:
20725545. Pubmed Central PMCID: 2921746. Epub 2010/08/21. eng.
2. Alexis AF. Acne in patients with skin of color. J Drugs Dermatol. 2011;10(6):s13–6. PubMed
PMID: 21780341. Epub 2011/07/26. eng.
3. Shah SK, Alexis AF. Acne in skin of color: practical approaches to treatment. J Dermatolog
Treat. 2010;21(3):206–11. PubMed PMID: 20132053. Epub 2010/02/06. eng.
4. Halder RM. Hair and scalp disorders in blacks. Cutis. 1983;32(4):378–80. PubMed PMID:
6226494. Epub 1983/10/01. eng.
5. Alexis AF, Sergay AB, Taylor SC. Common dermatologic disorders in skin of color: a com-
parative practice survey. Cutis. 2007;80(5):387–94. PubMed PMID: 18189024. Epub
2008/01/15. eng.
6. Taylor SC, Cook-Bolden F, Rahman Z, Strachan D. Acne vulgaris in skin of color. J Am Acad
Dermatol. 2002;46(2 Suppl Understanding):S98–106. PubMed PMID: 11807471. Epub
2002/01/25. eng.
7. Iozumi K, Hoganson GE, Pennella R, Everett MA, Fuller BB. Role of tyrosinase as the deter-
minant of pigmentation in cultured human melanocytes. J Invest Dermatol. 1993;100(6):806–
11. PubMed PMID: 8496620. Epub 1993/06/01. eng.
8. Staricco RJ, Pinkus H. Quantitative and qualitative data on the pigment cells of adult human
epidermis. J Invest Dermatol. 1957;28(1):33–45. PubMed PMID: 13416674. Epub 1957/01/01.
eng.
9. Seiji M, Shimao K, Birbeck MS, Fitzpatrick TB. Subcellular localization of melanin biosyn-
thesis. Ann N Y Acad Sci. 1963;100:497–533. PubMed PMID: 13992624. Epub 1963/02/15.
eng.
10. Jimbow K, Quevedo Jr WC, Fitzpatrick TB, Szabo G. Some aspects of melanin biology: 1950–
1975. J Invest Dermatol. 1976;67(1):72–89. PubMed PMID: 819593. Epub 1976/07/01. eng.
11. Ruiz-Maldonado R, Orozco-Covarrubias ML. Postinflammatory hypopigmentation and
hyperpigmentation. Semin Cutan Med Surg. 1997;16(1):36–43. PubMed PMID: 9125764.
Epub 1997/03/01. eng.
40 Post-inflammatory Pigment Alteration 285
12. Grimes PE, Stockton T. Pigmentary disorders in blacks. Dermatol Clin. 1988;6(2):271–81.
PubMed PMID: 3378372. Epub 1988/04/01. eng.
13. Grimes PE. Management of hyperpigmentation in darker racial ethnic groups. Semin Cutan
Med Surg. 2009;28(2):77–85. PubMed PMID: 19608057. Epub 2009/07/18. eng.
14. Halder R, Holmes Y, Bridgeman-Shah S, Kligman A. A clinical pathological study of acne
vulgaris in black females. J Invest Dermatol. 1996;106(4):888.
15. Gilchrest BA, Fitzpatrick TB, Anderson RR, Parrish JA. Localization of malanin pigmentation
in the skin with Wood's lamp. Br J Dermatol. 1977;96(3):245–8. PubMed PMID: 857837.
Epub 1977/03/01. eng.
16. Pathak MA, Riley FC, Fitzpatrick TB. Melanogenesis in human skin following exposure to
long-wave ultraviolet and visible light. J Invest Dermatol. 1962;39:435–43. PubMed PMID:
13941837. Epub 1962/11/01. eng.
17. Summers P, Bena J, Arrigain S, Alexis AF, Cooper K, Bordeaux JS. Sunscreen use: Non-
Hispanic Blacks compared with other racial and/or ethnic groups. Arch Dermatol.
2011;147(7):863–4. PubMed PMID: 21768492. Epub 2011/07/20. eng.
18. Alexis AF, Lamb A. Concomitant therapy for acne in patients with skin of color: a case-based
approach. Dermatol Nurs. 2009;1:33–6. PubMed PMID: 19283960. Epub 2009/03/17. eng.
19. Sheth VM, Pandya AG. Melasma: a comprehensive update Part II. J Am Acad Dermatol.
2011;65(4):699–714. PubMed PMID: 21920242. Epub 2011/09/17. eng.
20. Vazquez M, Sanchez JL. The efficacy of a broad-spectrum sunscreen in the treatment of
melasma. Cutis. 1983;32(1):92, 95–6. PubMed PMID: 6349944. Epub 1983/07/01. eng.
21. Bulengo-Ransby SM, Griffiths CE, Kimbrough-Green CK, Finkel LJ, Hamilton TA, Ellis CN,
et al. Topical tretinoin (retinoic acid) therapy for hyperpigmented lesions caused by inflamma-
tion of the skin in black patients. N Engl J Med. 1993;328(20):1438–43. PubMed PMID:
8479462. Epub 1993/05/20. eng.
22. Grimes P, Callender V. Tazarotene cream for postinflammatory hyperpigmentation and acne
vulgaris in darker skin: a double-blind, randomized, vehicle-controlled study. Cutis.
2006;77(1):45–50. PubMed PMID: 16475496. Epub 2006/02/16. eng.
23. Dogra S, Kanwar AJ, Parsad D. Adapalene in the treatment of melasma: a preliminary report.
J Dermatol. 2002;29(8):539–40. PubMed PMID: 12227492. Epub 2002/09/14. eng.
24. Romero C, Aberdam E, Larnier C, Ortonne JP. Retinoic acid as modulator of UVB-induced
melanocyte differentiation. Involvement of the melanogenic enzymes expression. J Cell Sci.
1994;107(Pt 4):1095–103. PubMed PMID: 8056833. Epub 1994/04/01. eng.
25. Ortonne JP. Retinoid therapy of pigmentary disorders. Dermatol Ther. 2006;19(5):280–8.
PubMed PMID: 17014483. Epub 2006/10/04. eng.
26. Taylor SC, Torok H, Jones T, Lowe N, Rich P, Tschen E, et al. Efficacy and safety of a new
triple-combination agent for the treatment of facial melasma. Cutis. 2003;72(1):67–72.
PubMed PMID: 12889718. Epub 2003/08/02. eng.
27. Grimes PE. A microsponge formulation of hydroquinone 4% and retinol 0.15% in the treat-
ment of melasma and postinflammatory hyperpigmentation. Cutis. 2004;74(6):362–8.
28. Kligman AM, Willis I. A new formula for depigmenting human skin. Arch Dermatol.
1975;111(1):40–8. PubMed PMID: 1119822. Epub 1975/01/01. eng.
29. Lynde CB, Kraft JN, Lynde CW. Topical treatments for melasma and postinflammatory hyperpig-
mentation. Skin Therapy Lett. 2006;11(9):1–6. PubMed PMID: 17075653. Epub 2006/11/01. eng.
30. Baumann L, Grimes P, Pandya AG, et al. Triple combination cream is an effective treatment
for post-inflammatory hyperpigmentation. Presented at the 65th Annual American Academy
of Dermatology Conference, Washington, DC, 3–5 Feb 2007
31. Guevara IL, Pandya AG. Melasma treated with hydroquinone, tretinoin, and a fluorinated ste-
roid. Int J Dermatol. 2001;40(3):212–5. PubMed PMID: 11422530.
32. Rendon M, Berneburg M, Arellano I, Picardo M. Treatment of melasma. J Am Acad Dermatol.
2006;54(5 Suppl 2):S272–81. PubMed PMID: 16631968. Epub 2006/04/25. eng.
33. Gupta AK, Gover MD, Nouri K, Taylor S. The treatment of melasma: a review of clinical
trials. J Am Acad Dermatol. 2006;55(6):1048–65. PubMed PMID: 17097400. Epub
2006/11/14. eng.
286 R.I. Nijhawan and A.F. Alexis
34. Kircik LH. Efficacy and safety of azelaic acid (AzA) gel 15% in the treatment of post-
inflammatory hyperpigmentation and acne: a 16-week, baseline-controlled study. J Drugs
Dermatol. 2011;10(6):586–90. PubMed PMID: 21637899. Epub 2011/06/04. eng.
35. Woolery-Lloyd H, Friedman A. Optimizing patient care with “natural” products: treatment of
hyperpigmentation. J Drugs Dermatol. 2009;8(6 Suppl):s10–3. PubMed PMID: 19562884.
Epub 2009/07/01. eng.
36. Hantash BM, Jimenez F. A split-face, double-blind, randomized and placebo-controlled pilot
evaluation of a novel oligopeptide for the treatment of recalcitrant melasma. J Drugs Dermatol.
2009;8(8):732–5. PubMed PMID: 19663110. Epub 2009/08/12. eng.
37. Khemis A, Kaiafa A, Queille-Roussel C, Duteil L, Ortonne JP. Evaluation of efficacy and
safety of rucinol serum in patients with melasma: a randomized controlled trial. Br J Dermatol.
2007;156(5):997–1004. PubMed PMID: 17388924. Epub 2007/03/29. eng.
38. Maeda K, Naganuma M. Topical trans-4-aminomethylcyclohexanecarboxylic acid prevents
ultraviolet radiation-induced pigmentation. J Photochem Photobiol B. 1998;47(2–3):136–41.
PubMed PMID: 10093913. Epub 1999/03/27. eng.
39. Briganti S, Camera E, Picardo M. Chemical and instrumental approaches to treat hyperpigmenta-
tion. Pigment Cell Res. 2003;16(2):101–10. PubMed PMID: 12622786. Epub 2003/03/08. eng.
40. Paine C, Sharlow E, Liebel F, Eisinger M, Shapiro S, Seiberg M. An alternative approach to
depigmentation by soybean extracts via inhibition of the PAR-2 pathway. J Invest Dermatol.
2001;116(4):587–95. PubMed PMID: 11286627. Epub 2001/04/05. eng.
41. Hakozaki T, Minwalla L, Zhuang J, Chhoa M, Matsubara A, Miyamoto K, et al. The effect of
niacinamide on reducing cutaneous pigmentation and suppression of melanosome transfer. Br
J Dermatol. 2002;147(1):20–31. PubMed PMID: 12100180. Epub 2002/07/09. eng.
42. Yamakoshi J, Sano A, Tokutake S, Saito M, Kikuchi M, Kubota Y, et al. Oral intake of proanthocyanidin-
rich extract from grape seeds improves chloasma. Phytother Res. 2004;18(11):895–9. PubMed
PMID: 15597304. Epub 2004/12/15. eng.
43. Rigopoulos D, Gregoriou S, Katsambas A. Hyperpigmentation and melasma. J Cosmet
Dermatol. 2007;6(3):195–202. PubMed PMID: 17760699. Epub 2007/09/01. eng.
44. Sarkar R, Kaur C, Bhalla M, Kanwar AJ. The combination of glycolic acid peels with a topical
regimen in the treatment of melasma in dark-skinned patients: a comparative study. Dermatol
Surg. 2002;28(9):828–32. PubMed PMID: 12269877. discussion 32. Epub 2002/09/25. eng.
45. Erbil H, Sezer E, Tastan B, Arca E, Kurumlu Z. Efficacy and safety of serial glycolic acid peels
and a topical regimen in the treatment of recalcitrant melasma. J Dermatol. 2007;34(1):25–30.
PubMed PMID: 17204097. Epub 2007/01/06. eng.
46. Javaheri SM, Handa S, Kaur I, Kumar B. Safety and efficacy of glycolic acid facial peel in
Indian women with melasma. Int J Dermatol. 2001;40(5):354–7. PubMed PMID: 11555002.
Epub 2001/09/14. eng.
47. Burns RL, Prevost-Blank PL, Lawry MA, Lawry TB, Faria DT, Fivenson DP. Glycolic acid
peels for postinflammatory hyperpigmentation in black patients. A comparative study.
Dermatol Surg. 1997;23(3):171–4. PubMed PMID: 9145958. discussion 5. Epub 1997/03/01.
eng.
48. Lee HS, Kim IH. Salicylic acid peels for the treatment of acne vulgaris in Asian patients.
Dermatol Surg. 2003;29(12):1196–9. PubMed PMID: 14725662. discussion 9. Epub
2004/01/17. eng.
49. Grimes PE. The safety and efficacy of salicylic acid chemical peels in darker racial-ethnic
groups. Dermatol Surg. 1999;25(1):18–22. PubMed PMID: 9935087. Epub 1999/02/06. eng.
50. Ahn HH, Kim IH. Whitening effect of salicylic acid peels in Asian patients. Dermatol Surg.
2006;32(3):372–5. PubMed PMID: 16640681. discussion 5. Epub 2006/04/28. eng.
51. Joshi SS, Boone SL, Alam M, Yoo S, White L, Rademaker A, et al. Effectiveness, safety, and
effect on quality of life of topical salicylic acid peels for treatment of postinflammatory hyper-
pigmentation in dark skin. Dermatol Surg. 2009;35(4):638–44. PubMed PMID: 19400885.
discussion 44. Epub 2009/04/30. eng.
40 Post-inflammatory Pigment Alteration 287
52. Terrell S, Aires D, Schweiger ES. Treatment of acne vulgaris using blue light photodynamic
therapy in an African-American patient. J Drugs Dermatol. 2009;8(7):669–71. PubMed
PMID: 19588644. Epub 2009/07/11. eng.
53. Sherling M, Friedman PM, Adrian R, Burns AJ, Conn H, Fitzpatrick R, et al. Consensus rec-
ommendations on the use of an erbium-doped 1,550-nm fractionated laser and its applications
in dermatologic laser surgery. Dermatol Surg. 2010;36(4):461–9. PubMed PMID: 20187898.
Epub 2010/03/02. eng.
54. Rahman Z, Alam M, Dover JS. Fractional Laser treatment for pigmentation and texture
improvement. Skin Therapy Lett. 2006;11(9):7–11. PubMed PMID: 17075654. Epub
2006/11/01. eng.
55. Robins P, Battle Jr EF, Alexis AF, Cook-Bolden F, Alqubaisy Y, McLeod MP, et al. Unique
laser techniques in patients with skin of color. J Drugs Dermatol. 2011;10(12):4–26. PubMed
PMID: 22134557. Epub 2011/12/03. eng.
56. Callender VD. Acne in ethnic skin: special considerations for therapy. Dermatol Ther.
2004;17(2):184–95. PubMed PMID: 15113286. Epub 2004/04/29. eng.
Chapter 41
Pseudofolliculitis Barbae
41.1 Introduction
41.2 Background
tightly curled such as the axilla and groin regions [4, 6]. Some authors have also
noted perimenopausal women can suffer from PFB as higher androgen levels simu-
late hair growth in the beard area [4]. A survey of patients at the Skin of Color
Center in New York found that 83 % of their patients with PFB were between ages
11 and 30 with the average age of 22, and among female patients 98 % were of
African ancestry and 2 % were Hispanic [4].
There are two types of lesions in pseudofolliculitis barbae: intrafollicular and
transfollicular lesions. The initiating event in the pathogenesis of pseudofolliculitis
barbae is generally thought to be shaving; however it should be noted that female
PFB patients report grooming via tweezing, electrolysis, waxing, and use of depila-
tory agents [4]. A better working understanding may be to consider any grooming
technique that causes trauma as a potential inciting event in PFB pathogenesis.
Since most patients develop PSB secondary to shaving, an understanding of how
shaving in particular leads to PFB is helpful. Shaving leaves an oblique cut at the
distal end of the hair strand [7]. The curved morphology of the hair and hair follicle
in patients with curly hair contributes to formation of lesions as the hair grows.
Instead of growing straight out, it curves back into the epidermis or dermis. Helical
and spiral hair types are the most likely to cause these lesions [4]. Intrafollicular
lesions occur when the growing hair penetrates the dermis while still growing within
its follicle of origin. Transfollicular lesions occur when the distal end of the hair
penetrates the skin outside the follicle. Transfollicular lesions typically penetrate
the skin 1.5–2 mm from the originating hair follicle [7].
German researchers have also discovered a genetic risk factor that partially con-
tributes to the PFB phenotype. They found that a single-nucleotide polymorphism
resulting in an abnormal Ala12Thr substitution in the 1A α(alpha)-helical segment
of the accompanying layer-specific keratin K6hf was responsible for a 6.12 odds
ratio increased risk of PFB compared to wild type [8]. In contrast, a curved hair fol-
licle was associated with a 51.27 odds ratio for expression of the PFB phenotype in
the German study population [8]. These researchers note that most other disruptive
keratin mutations are phenotypically silent, and the mutations are only unmasked
phenotypically secondary to mechanical trauma to the tissue [9]. The German
researchers postulate that the stretching of the skin involved with shaving results in
trauma that unmasks PFB, which explain why in many cases, allowing the beard to
grow normally resolves symptoms.
In pseudofolliculitis, inflammation occurs as a result of the body’s inflammatory
response to the ingrown hair. Suppurative parafollicular foci form at the sites where
infiltration occurs [10]. As lesions progress, small, foreign body granulomas and a
mixed inflammatory cell infiltrate become evident [10]. The lesions are sometimes
cleared by a process of transepithelial elimination in which the surface epithelium
grows around the lesion to encase the inflammatory response along with the inciting
ingrown hair [10].
41 Pseudofolliculitis Barbae 291
41.4 Work-Up
41.5 Treatment
PFB patients. In one study, use of one such manual razor specially made for PFB
patients conferred a 25 % or greater reduction in the number of lesions for 72.7 %
of study participants with 86.4 % of patients reporting some improvement [12].
A study of electric rotary razors found that 90 % of the 300 participants switched to
use the razors after the trial period [13].
For women with PFB, topical eflornithine is an option that can help decrease the
rate of hair growth and thickness of individual hairs. This often decreases the fre-
quency of hair removal techniques. Unfortunately, hair growth rate resumes to its
normal rate after cessation. Laser hair removal is another frequently recommended
option. This addresses both the desire to be clean shaven along with addressing the
root cause of PFB.
41.6 Conclusion
References
11. Kligman Am Jr MOH. Pseudofolliculitis of the beard and topically applied tretinoin. Arch
Dermatol. 1973;107(4):551–2.
12. Alexander AM. Evaluation of a foil-guarded shaver in the management of pseudofolliculitis
barbae. Cutis. 1981;27(5):534–7, 40–2. PubMed PMID: 7238107. Epub 1981/05/01. eng.
13. Garcia R, Henderson R. The adjustable rotary electric razor in the control of pseudofolliculitis
barbae. J Assoc Milit Dermatol. 1978;4:28.
Chapter 42
Pustular Psoriasis
42.1 Introduction
42.2 Background
Psoriasis is the most prevalent autoimmune disease in the United States. Affecting
approximately 2.2 % of the American population, psoriasis is associated with sig-
nificant patient morbidity and is estimated to cost $11.25 billion in direct and indi-
rect health care costs annually [3, 4].
S. Bernardo, M.D.
Department of Dermatology, Mt. Sinai School of Medicine,
161 West 86th Street, Apt 1a, New York, NY 10024, USA
M. Lebwohl, M.D. (*)
Department of Dermatology, Mt. Sinai School of Medicine,
5 East 98th Street, 5th Floor, New York, NY 10029, USA
e-mail: [email protected]
Fig. 42.1 Pustular psoriasis—sterile pustules on erythematous skin. Borrowed from the Mount
Sinai Dermatology collection
Fig. 42.2 Pustular psoriasis—von Zumbusch variant. Borrowed from the Mount Sinai Dermatology
collection
shed (Fig. 42.2) [27]. The acute phase is associated with fever, fatigue, headache,
chills, malaise, onycholysis, and burning [29]. With large areas of skin involved,
patients are susceptible to infection, fluid loss, hypothermia, and electrolyte abnor-
malities, a combination that represents a medical emergency that demands immediate
workup and appropriate treatment.
298 S. Bernardo and M. Lebwohl
Fig. 42.3 Pustular psoriasis—annular variant. Borrowed from the Mount Sinai Dermatology
collection
Fig. 42.4 Impetigo herpetiformis. Borrowed from “The Skin and Systemic Disease: A Color Atlas
and Text” [66]
Fig. 42.5 Palmar pustular psoriasis. Borrowed from the Mount Sinai Dermatology collection
Fig. 42.7 Plantar pustular psoriasis with involuting lesions. Borrowed from the Mount Sinai
Dermatology collection
42 Pustular Psoriasis 301
Fig. 42.8 Acrodermatitis of hallopeau. Borrowed from the Mount Sinai Dermatology collection
42.4 Workup
The diagnosis of pustular psoriasis can usually be made on the basis of its clinical fea-
tures. However, when the diagnosis is in question, pustular psoriasis has unique patho-
logical and laboratory findings that allow it to be distinguished from other psoriatic
variants. A punch biopsy allows for optimal visualization of disease histology, which is
predominantly characterized by neutrophil accumulation [2]. Migrating from sinuous,
dilated capillaries within the dermis, neutrophils collect between residual keratinocyte
plasma membranes accompanied by intercellular edema [31]. This forms a macropus-
tule in the epidermis that begins as a spongiform pustule of Kogoj and develops into a
large munro macroabscess [27]. Histological features of psoriasis vulgaris may also be
present and include parakeratosis, elongation of epidermal rete ridges, and a perivascu-
lar lymphocytic or mixed inflammatory infiltrate within the papillary dermis [27, 34].
Laboratory findings associated with pustular psoriasis include neutrophilia with WBC
counts as high as 30,000, absolute lymphopenia, hypoalbuminemia, hypocalcemia, ane-
mia, decreased creatinine clearance, and elevations in ESR, transaminases, alkaline
phosphatase, and bilirubin [9, 29, 35, 36]. Cultures of pustules are sterile. However,
since the protective functions of the skin are lost in GPP, patients have succumbed to
staphylococcal sepsis and blood cultures may be warranted.
42.5 Treatment
Although many systemic and topical treatments have been described as effec-
tive management options for patients with pustular psoriasis, no evidence-
based consensus has been reached regarding the correct therapeutic hierarchy
302 S. Bernardo and M. Lebwohl
In the acute setting, topical therapies are frequently ineffective in patients with gen-
eralized disease, and management with systemic agents in an experienced burn unit
is often necessary to prevent patient morbidity and mortality (Table 42.1). In con-
junction with starting therapy for GPP, supportive measures such as bedrest,
compresses, fluid replacement, correction of electrolyte abnormalities, as well as
antibiotics and serial blood cultures are essential in urgent cases [9, 29]. First-line
therapy includes oral retinoids such as acitretin and isotretinoin [1, 38]. Retinoid
therapy was effective in 84 % of patients in one large retrospective study [39]. Side
effects include hypertriglyceridemia, xerosis, hair loss, hepatotoxicity, and cheilitis
[6, 40, 41].
Methotrexate or cyclosporine are also first-line therapies for GPP [1]. Both have
been consistently documented as valuable treatment options for GPP and were
effective in 76.2 % and 71.2 % of patients, respectively, in a large, multicenter study
of 385 patients with GPP in Japan [1, 9, 16, 32, 39, 42–47]. Before initiating therapy
with either agent, it is necessary to obtain a detailed medical history and frequent
blood tests to prevent potentially severe side effects such as methotrexate-induced
bone marrow toxicity [1]. Advantages of methotrexate are its efficacy, affordability,
and convenient weekly oral dose [40]. It is limited by its teratogenicity and side
effects, which include myelosuppression, hepatotoxicity, and pulmonary toxicity
[48]. Cyclosporine has been used safely during pregnancy and its rapid onset makes
it effective in managing acute GPP [1, 32]. However, the side effects of nephrotoxic-
ity, hypertension, and immunosuppression limit its use [48].
The advent of biological agents has added another medication class to the
dermatologist’s armamentarium in the treatment of GPP. The efficacy of infliximab,
adalimumab, and etanercept has been documented in several reports in the
literature, including cases that were refractory to methotrexate, acitretin, and other
systemic therapies [6, 49–53]. With a faster onset of action than adalimumab or
etanercept, infliximab may be particularly useful in life-threatening cases of GPP
with systemic involvement [1]. As a class, the biologics are generally well tolerated.
They potentially increase the risk of infections and carcinogenesis so baseline test-
ing with a complete metabolic panel, CBC, LFTs, and tuberculosis screening should
be performed with appropriate follow-up [1, 50].
Localized cases may be managed with superpotent topical steroids or light therapies
but may require systemic treatment with retinoids, methotrexate, cyclosporine, or
biologics in refractory cases [46, 54–58] (Table 42.2). Topical corticosteroids can
be given as monotherapy, used under occlusion for increased penetration, or com-
bined with newer vitamin D analogues such as calcitriol or calcipotriene [1].
Unfortunately, many patients fail to respond to topical therapy and other treatment
304 S. Bernardo and M. Lebwohl
modalities are required. Both oral and bath “soak” PUVA therapies have been con-
sistently shown to be effective management options for patients with palmoplantar
pustulosis [1, 29, 59–61]. In the first 4 weeks of treatment, oral PUVA has been
found to be slightly more effective than bath PUVA but the former has more sys-
temic side effects such as nausea [59]. Combination therapy with PUVA and reti-
noids has been found to be more effective than PUVA alone and is associated with
a reduction in the number of PUVA sessions and UVA dose required for clearing
[60, 62, 63].
42.6 Conclusion
Several effective therapies for pustular psoriasis have emerged since von Zumbusch
described his case over 100 years ago. Going forward, much remains to be learned
and the development of evidence-based guidelines for treatment is imperative.
Continued research will yield a better understanding of the complex interactions
underlying disease pathogenesis and identify new targets for improved therapy.
References
1. Roosevelt L, Emer J. Pustular psoriasis: a practical review of the literature. Psoriasis Forum.
2011;17(1):33–41.
2. Van de Kerkhof PCM. Psoriasis. In: Bolognia JL, Jorizzo JL, Rapini RP, editors. Dermatology.
New York, NY: Mosby [Elsevier]; 2003. p. 125–46.
3. National Psoriasis Foundation—Psoriasis statistics, prevalence, severity, age of onset and pso-
riasis research. The National Psoriasis Foundation Website. http://www.psoriasis.org/learn_
statistics. Accessed 29 Sept 2011.
4. Stern RS, Nijsten T, Feldman SR, Margolis DJ, Rolstad T. Psoriasis is common, carries a sub-
stantial burden even when not extensive, and is associated with widespread treatment dissatis-
faction. J Investig Dermatol Symp Proc. 2004 Mar;9(2):136–9.
5. Zelickson BD, Muller SA. Generalized pustular psoriasis: a review of 63 cases. Arch Dermatol.
1991;127:1339–45.
6. de Oliveira ST, Maragno L, Arnone M, Fonseca Takahashi MD, Romiti R. Generalized pustu-
lar psoriasis in childhood. Pediatr Dermatol. 2010;27:349–54.
7. Liao PB, Rubinson R, Howard R, Sanchez G, Frieden IJ. Annular pustular psoriasis—most
common form of pustular psoriasis in children: report of three cases and review of the literature.
Pediatr Dermatol. 2002;19:19–25.
8. Gelfand JM, Stern RS, Nijsten T, et al. The prevalence of psoriasis in african americans: results
from a population-based study. J Am Acad Dermatol. 2005;52:23–6.
9. Visual Dx—adult rash: psoriasis, pustular. Visual Dx website. http://www.visualdx.com/visualdx/
visualdx6/getDiagnosisText.do?moduleId=7&diagnosisId=52229. Accessed 12 Sept 2011.
10. Gudjonsson JE, Elder JT. Psoriasis. In: Fitzpatrick TB, Wolff K, editors. Fitzpatrick’s derma-
tology in general medicine. 7th ed. New York, NY: McGraw-Hill; 2008. p. 169–93.
11. Schon MP, Boehncke WH. Psoriasis. N Engl J Med. 2005;352:1899–912.
12. Benoit S, Toksoy A, Bröcker EB, Gillitzer R, Goebeler M. Treatment of recalcitrant pustular
psoriasis with infliximab: effective reduction of chemokine expression. Br J Dermatol.
2004;150(5):1009–12.
42 Pustular Psoriasis 305
13. Yamamoto M, Tada Y, Asahina A, et al. Severe generalized pustular psoriasis accompanied by
bullae formation with increased serum vascular endothelial growth factor level. J Dermatol.
2012;39(2):183–5. 2011 July 18. [Epub ahead of print].
14. Marrkchi S, Guigue P, Renshaw BR, et al. Interleukin-36-receptor antagonist deficiency and
generalized pustular psoriasis. N Engl J Med. 2011;365(7):620–8.
15. Onoufriadis A, Simpson MA, Pink AE, et al. Mutations in Il36RN/Il1F5 are associated with
the severe episodic inflammatory skin disease known as generalized pustular psoriasis. Am J
Hum Genet. 2011;89(3):432–7.
16. Umezawa Y, Ozawa A, Kawasima T, et al. Therapeutic guidelines for the treatment of general-
ized pustular psoriasis (GPP) based on a proposed classification of disease severity. Arch
Dermatol Res. 2003;295(1):S43–54.
17. Freedberg IM, Eisen AZ, Wolff K, Austen KF, Goldsmith LA, Katz SI, editors. Dermatology
in general medicine. 6th ed. New York, NY: McGraw Hill; 2003.
18. Brenner M, Molin S, Ruebsam K, Weisenseel P, Ruzicka T, Prinz JC. Generalized pustular
psoriasis induced by systemic glucocorticosteroids: four cases and recommendations for treat-
ment. Br J Dermatol. 2009;161(4):964–6.
19. Ryan T, Baker H. Systemic corticosteroids and folic acid antagonists in the treatment of gen-
eralized pustular psoriasis evaluation and prognosis based on the study of 104 cases. Br J
Dermatol. 1969;81:134–45.
20. Rongioletti F, Fiorucci C, Parodi A. Psoriasis induced or aggravated by drugs. J Rhematol
Suppl. 2009;83:59–61.
21. Park JJ, Lee SC. A case of tumor necrosis factor-α inhibitors-induced pustular psoriasis. Ann
Dermatol. 2010;22:212–5.
22. Wenk KS, Claros JM, Ehrlich A. Flare of pustular psoriasis after initiating ustekinumab ther-
apy. J Dermatolog Treat. 2012;23(3):212–4. 2011 Jan 22 [Epub ahead of print].
23. Pourciau C, Shwayder T. Occurrence of pustular psoriasis after treatment of Crohn disease
with infliximab. Pediatr Dermatol. 2010;27(5):539–40.
24. Collamer AN, Guerrero KT, Henning JS, Battafarano DF. Psoriatic skin lesions induced by
tumor necrosis factor antagonist therapy: a literature review and potential mechanisms of
action. Arthritis Rheum. 2008;59(7):996–1001.
25. Shmidt E, Wetter DA, Ferguson SB, Pittelkow MR. Psoriasis and palmoplantar pustulosis
associated with tumor necrosis factor-α inhibitors: The Mayo Clinic experience, 1998 to 2010.
J Am Acad Dermatol. 2012;67(5):e179–85. 2011 Jul 11 [Epub ahead of print].
26. Gudjonsson JE, Thorarinsson AM, Sigurgeirsson B, Kristinsson KG, Valdimarsson H.
Streptococcal throat infections and exacerbation of chronic plaque psoriasis: a prospective
study. Br J Dermatol. 2003;149:530–4.
27. Prystowsky JH, Cohen PR. Pustular and erytherodermic psoriasis. Psoriasis. 1995;13:757–70.
28. Katz M, Seidenbaum M, Weinrauch L. Penicillin-induced generalized pustular psoriasis. J Am
Acad Dermatol. 1987;17:918–20.
29. Fitzpatrick TB, Johnson RA, Wolff K, Suurmond D, editors. Color atlas and synopsis of
clinical dermatology common and serious diseases. New York, NY: McGraw-Hill; 2001.
30. Maize JC, Burgdorf WHC, Hurt MA, et al. Cutaneous pathology. 1st ed. Philadelphia, PA:
Churchill Livingstone; 1998.
31. Barnhill RL, Crowson AN, Magro CM, Piepkorn MW. Dermatopathology. 3rd ed. China:
McGraw-Hill; 2010. p. 66–7.
32. Hazarika D. Generalized pustular psoriasis of pregnancy successfully treated with cyclospo-
rine. Indian J Dermatol Venereol Leprol. 2009;75(6):638–41.
33. Puig L, Barco D, Vilarrasa E, Alomar A. Treatment of acrodermatitis continua of hallopeau
with TNF-blocking agents: case report and review. Dermatology. 2010;220(2):154–8.
34. Zelickson BD, Muller SA. Generalized pustular psoriasis in childhood: report of thirteen
cases. J Am Acad Dermatol. 1991;24:186–94.
35. Viguier M, Allez M, Zagdanski AM, et al. High frequency of cholestasis in generalized pustu-
lar psoriasis: evidence for neutrophilic involvement of the biliary tract. Hepatology. 2004;40:
452–8.
306 S. Bernardo and M. Lebwohl
36. Borges-Costa J, Silva R, Goncalves L, et al. Clinical and laboratory features in acute general-
ized pustular psoriasis. Am J Clin Dermatol. 2011;12(4):271–6.
37. Lebwohl MG, Heymann WR, Berth-Jones J, Coulson I. Treatment of skin disease. 2nd ed.
China: Mosby-Elsevier; 2006.
38. Sofen HL, Moy RL, Lowe NJ. Treatment of generalised pustular psoriasis with isotretinoin.
Lancet. 1984;7:40.
39. Ozawa A, Ohkido M, Haruki Y, et al. Treatments of generalized pustular psoriasis: a multi-
center study in Japan. J Dermatol. 1999;26:141–9.
40. Gollnick HPM. Oral retinoids—efficacy and toxicity in psoriasis. Br J Dermatol.
1996;135(49):6–17.
41. Dogra S, Kaur I. Childhood psoriasis. Indian J Dermatol Venereol Leprol. 2010;76(4):
357–65.
42. Kalb RE, Strober B, Weinstein G, Lebwohl M. Methotrexate and psoriasis: 2009 national
psoriasis foundation consensus conference. J Am Acad Dermatol. 2009;60(5):824–37.
43. Meinardi MM, Westerhof W, Bos JD. Generalized pustular psoriasis (von zumbusch) respond-
ing to cyclosporine A. Br J Dermatol. 1987;116(2):269–70.
44. Rosmarin DM, Lebwohl M, Elewski BE, Gottlieb AB. Cyclosporine and psoriasis: 2008
national psoriasis foundation consensus conference. J Am Acad Dermatol. 2010;62(5):
838–53.
45. Dogra S, Kumaran MS, Handa S, Kanwar AJ. Methotrexate for generalized pustular psoriasis
in a 2-year-old child. Pediatr Dermatol. 2005;22(1):85–6.
46. Maza A, Montaudie H, Sbidian E, et al. Oral cyclosporine in psoriasis: a systematic review on
treatment modalities, risk of kidney toxicity and evidence for use in non-plaque psoriasis.
J Eur Acad Dermatol Venereol. 2011;25(2):19–27.
47. Paul C, Gallini A, Maza A, et al. Evidence-based recommendations on conventional systemic
treatments in psoriasis: systemic review and expert opinion of a panel of dermatologists. J Eur
Acad Dermatol Venereol. 2011;25(2):2–11.
48. Gottlieb A, Korman NJ, Gordon KB, et al. Guidelines of care for the management of psoriasis
and psoriatic arthritis: section 4: guidelines of care for the management and treatment of pso-
riasis with traditional systemic agents. J Am Acad Dermatol. 2008;58:851–64.
49. Esposito M, Mazzotta A, Casciello C, Chimenti S. Etanercept at different dosages in the treat-
ment of generalized pustular psoriasis: a case series. Dermatology. 2008;216(4):355–60.
50. Zangrilli A, Papoutsaki M, Talamonti M, Chimenti S. Long-term efficacy of adalimumab in
generalized pustular psoriasis. J Dermatolog Treat. 2008;19(3):185–7.
51. Alvarez AC, Rodriguez-Nevado I, de Argila D. Recalcitrant pustular psoriasis successfully
treated with adalimumab. Pediatr Dermatol. 2011;28(2):195–7.
52. Chandran NS, Chong WS. A dramatic response to a single dose of infliximab as rescue therapy
in acute generalized pustular psoriasis of von Zumbusch associated with a neutrophilic chol-
angitis. Australas J Dermatol. 2010;51(1):29–31.
53. Torii H, Nakagawa H. Long-term study of infliximab in Japanese patients with plaque psoria-
sis, psoriatic arthritis, pustular psoriasis and psoriatic erythroderma. J Dermatol. 2011;38(4):
321–34.
54. Visual Dx—pustulosis, palmoplantar. Visual Dx website. http://www.visualdx.com/visualdx/
visualdx6/getDiagnosisText.do?moduleId=7&diagnosisId=52240. Accessed 29 Sept 2011.
55. Thielen AM, Barde C, Marazza G, Saurat JH. Long-term control with etanercept (Enbrel) of a
severe acrodermatitis continua of hallopeau refractory to infliximab (Remicade). Dermatology.
2008;217(2):137–9.
56. Lee CS, Koo J. A review of acitretin, a systemic retinoid for the treatment of psoriasis. Expert
Opin Pharmacother. 2005;6(10):1725–34.
57. Di Lernia V, Guareschi E. Successful treatment of hand and foot psoriasis with infliximab.
Dermatol Online J. 2010;16(7):8.
58. Adişen E, Tekin O, Gülekon A, Gürer MA. A retrospective analysis of treatment responses of
palmoplantar psoriasis in 114 patients. J Eur Acad Dermatol. 2009;23(7):814–9.
42 Pustular Psoriasis 307
Joseph Bikowski
43.1 Introduction
43.2 Background
The pathophysiology of rosacea is not well elucidated, although the clinical features
of the disease are well documented. Based on recent molecular studies, current under-
standing holds that vascular and inflammatory manifestations of the disease are medi-
ated by an altered innate immune response [1, 9]. Recently, the antimicrobial peptide
cathelicidin and its activator kallikrein-5 have been found to contribute to the exacer-
bated immune response in rosacea [10]. Neutrophils have also been shown to induce
inflammation associated with rosacea and are thought to promote the release of reactive
oxygen species [11]. Other inflammatory mediators implicated in the development of
rosacea include histamine, serotonin, bradykinin, or prostaglandins [1].
Chronic UV exposure has been suggested to play a role in the pathophysiology
of rosacea, causing damage to dermal connective tissues, which facilitates and exac-
erbates the effects of vasodilation and vascular pooling [12]. A recent study involv-
ing 1,000 patients, however, failed to demonstrate an association between UV
exposure and papulopustular rosacea [13].
Environmental trigger factors may be associated with exacerbation of rosacea
and may initiate the flushing and blushing response in susceptible individuals. The
degree to which any individual is affected, if at all, by a given trigger is variable.
Commonly cited triggers include thermally hot beverages or foods, alcoholic drinks,
and/or spicy foods [14].
Fig. 43.3 Phymatous rosacea (Subtype 3) is characterized by thickened skin, nodules, and ana-
tomical enlargement
While the classification system is helpful for recognizing signs and symptoms of
rosacea and understanding disease progression, its clinical relevance may be lim-
ited. In fact, an international consensus committee has emphasized the importance
of implementing therapy based on signs and symptoms of the specific presenting
case rather than its classification [10].
Rosacea may be mistaken for acne vulgaris, though the two are distinguished by
the presence (acne) or absence (rosacea) of comedones. Furthermore, while rosa-
cea’s centrofacial distribution includes the forehead, lesions are not typically
43 Rosacea 313
43.4 Work-Up
There are no diagnostic tests for rosacea. When ocular rosacea is suspected, patients
may be referred for ophthalmic evaluation and treatment. Patients with rosacea
should be educated about ocular involvement and be encouraged to attend to their
ocular health.
Despite historic associations of rosacea with acne vulgaris, the two are of dis-
tinctly different etiologies, and there is no microorganism widely accepted as caus-
ative in rosacea. An association between rosacea and the Demodex folliculorum
mite has been identified, although no evidence has elevated this correlation to cau-
sation [19]. In fact, there is some evidence that a distinct condition termed Demodex
dermatitis may exist that is frequently inaccurately diagnosed as rosacea [20]. This
condition, which is characterized by facial erythema, dryness, scaling, and rough-
ness with or without papules/pustules, responds to treatment with topical crotami-
ton or permethrin; such therapeutic response may confirm the diagnosis of Demodex
dermatitis rather than rosacea.
43.5 Treatment
Among the treatment options for rosacea are both topical and systemic formulations
(Table 43.2). Increasingly, laser and light-based interventions are emerging for the
management of vascular and phymatous irregularities, which are generally not
314 J. Bikowski
subtype 2 [26]. These agents do not offer significant efficacy for diffuse erythema,
telangiectases, or phymatous formations.
A first-in-class topical alpha-agonist received FDA approval for the treatment of
the erythema of rosacea in August 2013. Brimonidine gel 0.33 % is a once-daily
topical agent that was shown in clinical trials to provide a significantly greater
improvement in the facial redness of rosacea than vehicle gel. The drug has vaso-
constrictive properties that are believed to confer the redness reducing effects.
Additional topical vasoconstrictors are in development for use in rosacea.
Conventional antibiotics are still used in the management of rosacea, with their
efficacy presumed to be associated with their anti-inflammatory effects. The avail-
ability of anti-inflammatory dose doxycycline (40 mg, controlled release) has modi-
fied the approach to systemic rosacea therapy [27]. Anti-inflammatory dose
doxycycline is shown to be at least as effective as conventional doxycycline doses
for the treatment of PPR but with a reduction in side effects [26] and no risk of
contributing to the development of bacterial resistance [27]. Like topical interven-
tions, oral antibiotics confer greatest efficacy in managing the signs and symptoms
of PPR rather than the other subtypes.
Use of topical and systemic agents in combination is expected to enhance thera-
peutic responses and maintain results [27, 28]. Topical and systemic agents may
also be used in combination with devices, which are increasingly used for the reduc-
tion of erythema and telangiectases associated with rosacea. Specifically, pulsed
dye laser and intense-pulsed light (IPL) devices that target hemoglobin as the target
chromophore are used to treat erythema and telangiectases [29].
Oral isotretinoin, which has both anti-inflammatory and immunomodulatory
properties, has been used as a treatment for severe rosacea, particularly phymatous
presentations [30, 31]. Surgical correction and ablative laser surgery may also be
used to manage phymas.
43.6 Conclusion
References
1. Yamasaki K, Gallo RL. The molecular pathology of rosacea. J Dermatol Sci. 2009;55:77–81.
2. Diamantis S, Waldorf HA. Rosacea: clinical presentation and pathophysiology. J Drugs
Dermatol. 2006;5:8–12.
3. Balkrishnan R, McMichael AJ, Hu JY, Camacho FT, Shew KR, Bouloc A, et al. Correlates of
health-related quality of life in women with severe facial blemishes. Int J Dermatol.
2006;45:111–5.
316 J. Bikowski
44.1 Introduction
44.2 Background
Because of the particularly ferocious nature of the disease, patients may become
depressed, anxious, and isolated because of the appearance [2, 13]. Scarring is
common and expected and can range from minimal damage to keloids [2, 14].
44.4 Work-up
The diagnosis of rosacea fulminans is largely clinical, although supportive labs and
a skin biopsy can be performed. Some patients have been documented to have a
mild anemia, leukocytosis, increased erythrocyte sedimentation rate (ESR), ele-
vated C-reactive protein (CRP), and positive rheumatoid factor and antinuclear anti-
bodies [1, 2, 4, 14, 15]. Skin biopsy reveals evidence of inflammation, with massive
neutrophil infiltration in early stages and epithelioid cell granulomas in older lesions
[4, 12, 16]. Bacterial pathogens have not been consistently recovered, and most
bacteria identified are commensal skin flora [2, 3]. Isolated cases report identifica-
tion of Staphylococcus aureus and gram-negative bacteria [3, 5].
44.5 Treatment
44.6 Conclusion
References
45.1 Introduction
45.2 Background
There are many theories surrounding the etiology of sarcoidosis, but the cause(s) of
the disease is still unknown. A multicenter trial of over 700 patients, entitled “A
Case Control Etiologic Study of Sarcoidosis (ACCESS),” was unable to identify a
specific antigen in sarcoidosis but did establish an increased risk of the disease with
exposure to mold and musty odors, agricultural chemicals and aerosols, and insec-
ticides [1]. In sarcoidosis, normal tissue function is disrupted by noncaseating gran-
ulomas. The formation of the noncaseating granuloma begins with macrophages
and dendritic cells (antigen-presenting cells) using class II major histocompatibility
(MHC) complexes to present an unknown antigen(s) to CD4+ T cells [2]. Through
interactions with the MHC and costimulatory molecules, the T cell becomes acti-
vated and ignites a TH1 inflammatory response. The T cells release IL-2 and IFN-γ
to recruit additional T cells, as well as stimulate macrophages to produce TNF-α,
leading to the differentiation of macrophages into epithelioid cells and formation of
multinucleated giant cells [2].
In the United States, there are two peaks for sarcoidosis, in people between 25
and 35 years and in women between 45 and 65 years [3]. Sarcoidosis is more com-
mon in African Americans (although all races can be affected), and this group is
more likely to present with advanced disease and have a poorer prognosis [4]. The
highest incidence of disease in the United States in African American women in
their fourth decade of life [3]. Sarcoidosis persists as a progressive disease in
30 % but has a mortality of less than 5 % [5]. The most common causes of death
are progressive pulmonary fibrosis or other pulmonary or cardiac complications
[4]. The first-degree relatives of people with sarcoidosis are at a 5-time increased
risk for developing the disease [6]. Recent research has proposed certain genetic
susceptibilities associated with the disease. An increased risk of sarcoidosis is
associated with HLA-DRB1 and a splice site mutation in the gene butyrophilin-
like 2 (BTNL2) [7].
45.4 Workup
45.5 Treatment
theoretically decreasing granuloma formation [16]. The dosing ranges and sched-
ules of corticosteroids are highly variable, and there is not adequate evidence sup-
porting their use for cutaneous sarcoidosis [11]. Historically, oral prednisone has
been reported as effective for cutaneous sarcoidosis, but randomized controlled tri-
als are needed [14, 17, 18]. Given the extensive side effects of systemic corticoste-
roids, they are generally only used for expansive disfiguring cutaneous lesions after
local steroids have failed [3]. Corticosteroids must be tapered slowly over weeks to
months to avoid disease flares.
Hydroxychloroquine is an antimalarial agent which is thought to work by
decreasing antigen presentation by APCs, resulting in decreased T-cell activa-
tion and decreased granuloma formation [19]. The efficacy of antimalarials in
cutaneous sarcoidosis has been reported since the 1960s, but there have not been
any randomized controlled trials for this indication [20, 21]. More recent studies
have reported regression of cutaneous sarcoidosis with hydroxychloroquine
(2–3 mg/kg/day for up to 12 weeks) [22] or chloroquine (500 mg daily for 14
days, then 250 mg for long-term maintenance) [23]. Antimalarial medications
require frequent eye exams (at least yearly) for ototoxicity. Methotrexate, a
dihydrofolate reductase inhibitor, has emerged as a second-line treatment for
cutaneous sarcoidosis, typically prescribed at doses of 10–30 mg/week [16].
Webster [24] reported clearing of refractory cutaneous sarcoidosis in three
patients with 15 mg/week for up to 11 months [24]. Baughman [25] reported
improvement in 16 out of 17 patients treated with methotrexate over a period of
2 years and with average doses of 28 mg/week [25]. Minocycline (200 mg/day)
has been reported in one open prospective study of 12 patients with refractory
cutaneous sarcoidosis, with 8 patients experiencing complete regression and 2
with partial regression after 12 months, but with 3 suffering relapse of disease
after stopping this therapy [26].
Other medications that have been reported for the treatment of sarcoidosis
include pentoxifylline [27], thalidomide [28–32], allopurinol [33, 34], isotretinoin
[35, 36], infliximab [37, 38], adalimumab [39], azathioprine [40], cyclophospha-
mide [40], cyclosporine [41], chlorambucil [42, 43], leflunomide [44, 45], and mel-
atonin [46], but randomized controlled trials are needed to evaluate these treatments.
Non-pharmaceutical options include lasers, dermabrasion, excision, and photother-
apy (PUVA, UVB, and photodynamic therapy).
45.6 Conclusion
References
1. Newman L, ACCESS Research Group. A case control etiologic study of sarcoidosis: environ-
mental and occupational risk factors. Am J Respir Crit Care Med. 2004;170(12):1324–30.
2. Noor A. Immunopathogenesis of sarcoidosis. Clin Dermatol. 2007;25:250–8.
3. English J. Sarcoidosis. J Am Acad Dermatol. 2001;44(5):725–43.
4. Heath C. Sarcoidosis: are there differences in your skin of color patients? J Am Acad Dermatol.
2012;66(1):121.e1–14.
5. Morgenthau A. Recent advances in sarcoidosis. Chest. 2011;139:174–82.
6. Judson M. The diagnosis of sarcoidosis. Clin Chest Med. 2008;29:415–27.
7. Valentonyte R. Sarcoidosis is associated with a truncating splice site mutation in BTNL2. Nat
Genet. 2005;37:357–64.
8. English J. Sarcoidosis. In: Callen J, editor. Dermatologic signs of internal disease. China:
Saunders Elsevier; 2009.
9. Mana J. Cutaneous involvement in sarcoidosis. Arch Dermatol. 2007;133:882–8.
10. Costabel U. Systemic evaluation of a potential cutaneous sarcoidosis patient. Clin Dermatol.
2007;25:303–11.
11. Izikson A, English JC. Cutaneous sarcoidosis. In: Williams H, editor. Evidence-based derma-
tology. 2nd ed. London: Wiley-Blackwell; 2008.
12. Skold C. Determination of cardiac involvement in sarcoidosis by magnetic resonance imaging
and Doppler echocardiography. J Intern Med. 2002;252(5):465–71.
13. Iannuzzi M. Sarcoidosis. JAMA. 2011;305(4):391–9.
14. Russo G. Cutaneous sarcoidosis: diagnosis and treatment. Complement Ther. 1994;20:418–21.
15. Wilson N. Cutaneous sarcoidosis. Postgrad Med J. 1998;74:649–52.
16. Badgwell C. Cutaneous sarcoidosis therapy updated. J Am Acad Dermatol. 2007;56:69–83.
17. Albertini J. Ulcerative sarcoidosis: case report and review of the literature. Arch Dermatol.
1997;133:215–9.
18. Veien N. Cutaneous sarcoidosis: prognosis and treatment. Clin Dermatol. 1986;4:75–87.
19. Fox R. Mechanism of action of antimalarial drugs: inhibition of antigen processing and pre-
sentation. Lupus. 1993;2 Suppl 1:S9–12.
20. Morse S. The treatment of sarcoidosis with chloroquine. Am J Med. 1961;1961:779–84.
21. Siltzbach L. Chloroquine therapy in 43 patients with intrathoracic and cutaneous lesions. Acta
Med Scand. 1964;176 Suppl 425:302–6.
22. Jones E. Hydroxychloroquine is effective therapy for control of cutaneous sarcoidal granulo-
mas. J Am Acad Dermatol. 1990;23:487–9.
23. Zic J. Treatment of cutaneous sarcoidosis with chloroquine. Arch Dermatol. 1991;
127:1034–40.
24. Webster G. Weekly low-dose methotrexate for cutaneous sarcoidosis. J Am Acad Dermatol.
1991;24:451–4.
25. Lower E. Prolonged use of methotrexate for sarcoidosis. Arch Intern Med. 1995;155:846–51.
26. Bachelez H. The use of tetracyclines for the treatment of sarcoidosis. Arch Dermatol.
2001;137:69–73.
27. Zabel P. Pentoxifylline in treatment of sarcoidosis. Am J Respir Crit Care Med. 1997;155:
1665–9.
28. Carlesimo M. Treatment of cutaneous and pulmonary sarcoidosis with thalidomide. J Am
Acad Dermatol. 1995;32:866–9.
29. Rousseau L. Cutaneous sarcoidosis successfully treated with low doses of thalidomide. Arch
Dermatol. 1998;134:1045–6.
30. Lee J. Disfiguring cutaneous manifestation of sarcoidosis treated with thalidomide: a case
report. J Am Acad Dermatol. 1998;39:835–8.
31. Baughman R. Thalidomide for chronic sarcoidosis. Chest. 2002;122:227–32.
32. Nguyen Y. Treatment of cutaneous sarcoidosis with thalidomide. J Am Acad Dermatol.
2004;50:235–41.
45 Sarcoidosis 327
33. Samuel M. Sarcoidosis: initial results on six patients treated with allopurinol. Br J Dermatol.
1984;111 Suppl 26:20.
34. Voelter-Mahlknecht S. Treatment of subcutaneous sarcoidosis with allopurinol. Arch
Dermatol. 1999;135:1560–1.
35. Waldinger T. Treatment of cutaneous sarcoidosis with isotretinoin. Arch Dermatol.
1983;119:1003–5.
36. Georgiou S. Cutaneous sarcoidosis: complete remission after oral isotretinoin therapy. Acta
Derm Venereol. 1998;78:457–9.
37. Baughman R. Infliximab in cutaneous sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis.
2001;18:70–4.
38. Meyerle J. The use of infliximab in cutaneous sarcoidosis. J Drugs Dermatol. 2003;2:413–4.
39. Heffernan M. Adalimumab for treatment of cutaneous sarcoidosis. Arch Dermatol.
2006;142:17–9.
40. Baughman R. Alternatives to corticosteroids in the treatment of sarcoidosis. Sarcoidosis Vasc
Diffuse Lung Dis. 1997;14:121–30.
41. Rebuck A. Cyclosporin in pulmonary sarcoidosis. Lancet. 1987;1:1486.
42. Kataria Y. Chlorambucil in sarcoidosis. Chest. 1980;78:36–43.
43. Isreal H. Chlorambucil treatment of sarcoidosis. Sarcoidosis. 1991;8:35–41.
44. Baughman R. Leflunomide for chronic sarcoidosis. Sarcoidosis Vasc Diffuse Lung Dis.
2004;21:43–8.
45. Majithia V. Successful treatment of sarcoidosis with leflunomide. Rheumatology.
2003;42:700–2.
46. Cagnoni M. Melatonin for treatment of chronic refractory sarcoidosis. Lancet. 1995;346:
1229–30.
Chapter 46
Seborrheic Dermatitis
46.1 Introduction
46.2 Background
46.2.1 Epidemiology
Seborrheic dermatitis peaks during time periods of increased sebum production ini-
tially during infancy and later during adolescence and adulthood. It is generally
divided into infantile seborrheic dermatitis and adult seborrheic dermatitis which
generally occurs in those aged 30–60 years [1]. Up to 70 % of infants may develop
seborrheic dermatitis within the first 3 months of life which usually resolves by
1 year of age [2]. The prevalence in healthy adults is between 1 % and 3 %, more
commonly seen in males than females, and typically worsens during the winter
months [3]. Many consider dandruff, pityriasis simplex, to be the mildest form of
seborrheic dermatitis which may affect up to 50 % of the population [4].
Seborrheic dermatitis has been noted to be a marker for HIV infection and may
be the presenting sign [5, 6]. The prevalence of seborrheic dermatitis in early HIV
infection is 36 % and 50–83 % in patients with acquired immunodeficiency syn-
drome (AIDS) [7–9]. Patients with Parkinson’s disease have increased sebum pro-
duction as well as an increased prevalence of seborrheic dermatitis. Treatment with
l-dopa results in with both quantitative sebum reduction and clinical improvement
of seborrhea [10]. This increased prevalence also occurs in patients with neuroleptic-
induced Parkinsonism after the use of neuroleptic drugs such as chlorpromazine
hydrochloride and haloperidol and in those with mood disorders such as schizo-
phrenia, depression, and anxiety [11, 12]. Seborrheic dermatitis is seen in other
genetic disorders including familial amyloidosis with polyneuropathy and Down’s
syndrome [13–15].
The role of ultraviolet (UV) light on seborrheic dermatitis is unclear. Patients
frequently report improvement after exposure to sunlight [16]. Also, seborrheic der-
matitis is noted to flare during the winter months [17]. However, mountain guides
who have increased UV exposure were noted to have increased prevalence of sebor-
rheic dermatitis thought to be secondary to UV-induced immunosuppression [18].
Additionally psoralen plus UVA (PUVA) light treatment can induce seborrheic der-
matitis, whereas narrowband UVB (NBUVB) has been used to successfully treat
severe disease [19, 20].
46.2.2 Pathophysiology
twisted red loops, and glomerular vessels in psoriasis in contrast to arborizing vessels
and atypical or absent vascular patterns in seborrheic dermatitis [32].
46.4 Work-Up
46.4.1 Laboratory
46.4.2 Biopsies
46.4.3 Cultures
46.5 Treatment
46.5.1 Antifungals
46.5.2 Antibiotics
46.5.3 Corticosteroids
46.5.4 Immunomodulators
Tacrolimus and pimecrolimus are nonsteroidal anti-inflammatory agents that inhibit cal-
cineurin and decrease subsequent cytokine production and can be used off label as ste-
roid-sparing agents. Tacrolimus 0.1 % ointment showed improvement in seborrheic
dermatitis in open-label pilot studies with side effects of burning and irritation [64, 65].
Tacrolimus was also as effective as topical betamethasone 17-valerate lotion [66].
Pimecrolimus cream 1 % was superior to vehicle and as effective as betamethasone
17-valerate 0.1 % cream in randomized clinical trials [67, 68]. The black-box warning
for rare cases of lymphoma seen in animal models has limited use of these agents.
46.5.8 Phototherapy
References
26. Carr MM, Pryce DM, Ive FA. Treatment of seborrhoeic dermatitis with ketoconazole:
I. Response of seborrhoeic dermatitis of the scalp to topical ketoconazole. Br J Dermatol.
1987;116:213–6.
27. DeAngelis YM, Gemmer CM, Kaczvinsky JR, Kenneally DC, Schwartz JR, Dawson Jr TL.
Three etiologic facets of dandruff and seborrheic dermatitis: Malassezia fungi, sebaceous lip-
ids, and individual sensitivity. J Investig Dermatol Symp Proc. 2005;10:295–7.
28. DeAngelis YM, Saudners CW, Johnstone KR, Reeder NL, Coleman CG, Kaczvinsky JR, et al.
Isolation and expression of a Malassezia globosa lipase gene, LIP1. J Invest Dermatol.
2007;127:2138–46.
29. Xu J, Saunders CW, Hu P, Grant RA, Boekhout T, Kuramae EE, et al. Dandruff-associated
Malassezia genomes reveal convergent and divergent virulence traits shared with plant and
human fungal pathogens. Proc Natl Acad Sci USA. 2007;104:18730–5.
30. Thomas DS, Ingham E, Bojar RA, Holland KT. In vitro modulation of human keratinocyte
pro-and anti-inflammatory cytokine production by the capsule of Malassezia species. FEMS
Immunol Med Microbiol. 2008;54:203–14.
31. Faergemann J, Bergbrant IM, Dohse M, Scott A, Westgate G. Seborrhoeic dermatitis and
Pityrosporum (Malassezia) folliculitis: characterization of inflammatory cells and mediators in
the skin by immunohistochemistry. Br J Dermatol. 2001;144:549–56.
32. Kim GW, Jung HJ, Ko HC, Kim MB, Lee WJ, Lee SJ, et al. Dermoscopy can be useful in dif-
ferentiating scalp psoriasis from seborrheic dermatitis. Br J Dermatol. 2011;164:652–6.
33. Pinkus H, Mehregan AH. The primary histologic lesion of seborrheic dermatitis and psoriasis.
J Invest Dermatol. 1965;46:109–16.
34. Skinner Jr RB, Noah PW, Taylor RM, Zanolli MD, West S, Guin JD, et al. Double-blind treat-
ment of Seborrheic dermatitis with 2% ketoconazole cream. J Am Acad Dermatol.
1985;12:852–6.
35. Elewski B, Ling MR, Phillips TJ. Efficacy and safety of a new once-daily topical ketoconazole
2% gel in the treatment of seborrheic dermatitis: a phase III trial. J Drugs Dermatol.
2006;5:646–50.
36. Elewski BE, Abramovits W, Kempers S, Schlessinger J, Rosen T, Gupta AK, et al. A novel
foam formulation of ketoconazole 2% for the treatment of seborrheic dermatitis on multiple
body regions. J Drugs Dermatol. 2007;6:1001–8.
37. Faergemann J. Treatment of seborrhoeic dermatitis of the scalp with ketoconazole shampoo.
A double-blind study. Acta Derm Venereol. 1990;70:171–2.
38. Apasrawirote W, Udompataikul M, Rattanamongkolgul S. Topical antifungal agents for sebor-
rheic dermatitis: systematic review and meta-analysis. J Med Assoc Thai. 2011;94:756–60.
39. Segal R, David M, Ingber A, Lurie R, Sandbank M. Treatment with bifonazole shampoo for
seborrhea and seborrheic dermatitis: a randomized, double-blind study. Acta Derm Venereol.
1992;72:454–5.
40. Zienicke H, Korting HC, Braun-Falco O, Effendy I, Hagedorn M, Küchmeister B, et al.
Comparative efficacy and safety of bifonazole 1% cream and the corresponding base prepara-
tion in the treatment of seborrhoeic dermatitis. Mycoses. 1993;36:325–31.
41. Shemer A, Nathansohn N, Kaplan B, Weiss G, Newman N, Trau H. Treatment of scalp sebor-
rheic dermatitis and psoriasis with an ointment of 40% urea and 1% bifonazole. Int J Dermatol.
2000;39:532–4.
42. Abrams BB, Hänel H, Hoehler T. Ciclopirox olamine: a hydroxypyridone antifungal agent.
Clin Dermatol. 1991;9:471–7.
43. Lebwohl M, Plott T. Safety and efficacy of ciclopirox 1% shampoo for the treatment of sebor-
rheic dermatitis of the scalp in the US population: results of a double-blind, vehicle-controlled
trial. Int J Dermatol. 2004;43:17–20.
44. Dupuy P, Maurette C, Amoric JC, Chosidow O, Study Investigator Group. Randomized,
placebo-controlled, double-blind study on clinical efficacy of ciclopiroxolamine 1% cream in
facial seborrhoeic dermatitis. Br J Dermatol. 2001;144:1033–7.
45. Shuster S, Meynadier J, Kerl H, Nolting S. Treatment and prophylaxis of seborrheic dermatitis
of the scalp with antipityrosporal 1% ciclopirox shampoo. Arch Dermatol. 2005;141:47–52.
340 E. Farhat and L.S. Gold
67. Warshaw EM, Wohlhuter RJ, Liu A, Zeller SA, Wenner RA, Bowers S, et al. Results of a
randomized, double-blind, vehicle-controlled efficacy trial of pimecrolimus cream 1% for the
treatment of moderate to severe facial seborrheic dermatitis. J Am Acad Dermatol.
2007;57:257–64.
68. Rigopoulos D, Ioannides D, Kalogeromitros D, Gregoriou S, Katsambas A. Pimecrolimus
cream 1% vs. betamethasone 17-valerate 0.1% cream in the treatment of seborrhoeic dermati-
tis. A randomized open-label clinical trial. Br J Dermatol. 2004;151:1071–5.
69. Ballanger F, Tenaud I, Volteau C, Khammari A, Dréno B. Anti-inflammatory effects of lithium
gluconate on keratinocytes: a possible explanation for efficiency in seborrhoeic dermatitis.
Arch Dermatol Res. 2008;300:215–23.
70. Efalith Multicenter Trial Group. A double-blind, placebo-controlled, multicenter trial of lith-
ium succinate ointment in the treatment of seborrheic dermatitis. J Am Acad Dermatol.
1992;26:452–7.
71. Dreno B, Moyse D. Lithium gluconate in the treatment of seborrhoeic dermatitis: a multi-
center, randomised, double-blind study versus placebo. Eur J Dermatol. 2002;12:549–52.
72. Dreno B, Chosidow O, Revuz J, Moyse D, Study Investigator Group. Lithium gluconate 8% vs
ketoconazole 2% in the treatment of seborrhoeic dermatitis: a multicentre, randomized study.
Br J Dermatol. 2003;148:1230–6.
73. Langtry JA, Rowland Payne CM, Staughton RC, Stewart JC, Horrobin DF. Topical lithium
succinate ointment (Efalith) in the treatment of AIDS-related seborrhoeic dermatitis. Clin Exp
Dermatol. 1997;22:216–9.
74. Davies DB, Boorman GC, Shuttleworth D. Comparative efficacy of shampoos containing coal
tar (4.0% w/w; Tarmed), coal tar (4.0% w/w) plus ciclopirox olamine (1.0% w/w/; Tarmed)
and ketoconazole 92.0% w/w/; Nizoral) for the treatment of dandruff/seborrheic dermatitis.
J Deramtol Treat. 1999;10:177–83.
75. Seite S, Rougier A, Talarico S. Randomized study comparing the efficacy and tolerance of a
lipohydroxy acid shampoo to a ciclopiroxolamine shampoo in the treatment of scalp sebor-
rheic dermatitis. J Cosmet Dermatol. 2009;8:249–53.
76. Veraldi S, Menter A, Innocenti M. Treatment of mild to moderate seborrhoeic dermatitis with
MAS064D (Sebclair), a novel topical medical device: results of a pilot, randomized, double-
blind, controlled trial. J Eur Acad Dermatol Venereol. 2008;22:290–6.
77. Ionescu MA, Baroni A, Brambilla L, Cannavò SP, Cristaudo A, Vedove CD, et al. Double
blind clinical trial in a series of 115 patients with seborrheic dermatitis: prevention of
relapses using a topical modulator of Toll like receptor 2. G Ital Dermatol Venereol.
2011;146:185–9.
78. Ryu S, Choi SY, Acharya S, Chun YJ, Gurley C, Park Y, et al. Antimicrobial and anti-
inflammatory effects of cecropin A(1–8)-magainin2(1–12) hybrid peptide analog P5 against
Malassezia furfur infection in human keratinocytes. J Invest Dermatol. 2011;131:1677–83.
79. Shemer A, Kaplan B, Nathansohn N, Grunwald MH, Amichai B, Trau H. Treatment of moder-
ate to severe facial seborrheic dermatitis with itraconazole: an open non-comparative study. Isr
Med Assoc J. 2008;10:417–8.
80. Kose O, Erbil H, Gur AR. Oral itraconazole for the treatment of seborrhoeic dermatitis: an
open, noncomparative trial. J Eur Acad Dermatol Venereol. 2005;19:172–5.
81. Baysal V, Yildirim M, Ozcanli C, Ceyhan AM. Itraconazole in the treatment of seborrheic
dermatitis: a new treatment modality. Int J Dermatol. 2004;43:63–6.
82. Cömert A, Bekiroglu N, Gürbüz O, Ergun T. Efficacy of oral fluconazole in the treatment of
seborrheic dermatitis: a placebo-controlled study. Am J Clin Dermatol. 2007;8:235–8.
83. Vena GA, Micali G, Santoianni P, Cassano N, Peruzzi E. Oral terbinafine in the treatment of
multi-site seborrhoic dermatitis: a multicenter, double-blind placebo-controlled study. Int J
Immunopathol Pharmacol. 2005;18:745–53.
84. Pirkhammer D, Seeber A, Hönigsmann H, Tanew A. Narrow-band ultraviolet B (ATL-01)
phototherapy is an effective and safe treatment option for patients with severe seborrhoeic
dermatitis. Br J Dermatol. 2000;143:964–8.
Chapter 47
Steatocystoma Multiplex
47.1 Introduction
47.2 Background
The exact etiopathogenesis has not been completely elucidated, but keratin 17 (K17)
has been proposed as a central element in the formation of hereditary steatocystomas.
This protein of keratin-containing intermediate filaments is found in several epithelial
structures such as the ungual bed, hair follicles, and sebaceous glands. The area where
the mutations of the gene K17 occur in SM is identical to mutations found in patients
with pachyonychia congenita type 2 (PC-2) [1]. A variety of mutations have been
described in patients with either steatocystoma multiplex or PC-2, and all of them are
localized to the helix initiation 1A domain of the K17 gene [2]. Therefore these two
conditions are thought to be connected and can present simultaneously with the addi-
tional typical features of PC-2, i.e., hypertrophic nail dystrophy and focal hyperkera-
tosis of the palms, soles, knees, and elbows [3]. Hybrid lesions with histological
features of both conditions have been described [4]. SM has also been reported to be
related to acrokeratosis verruciformis, hypertrophic lichen planus [5], and eruptive
vellus hair cysts (EVHCs) [6]. Some authors suggest that SM and EVHCs belong to
one spectrum of the same disease that should be referred as to “multiple piloseba-
ceous cysts” which would be a more appropriate diagnosis than the terms of EVHCs
and steatocystoma multiplex [7]. On the other hand, some state that they are two sepa-
rate entities based on the expression of different keratins. Lesions of SM express kera-
tins 10 and 17, in contrast to EVHCs which express only keratin 17 [8].
Recently two missense mutations (R94H and N92S) of the K17 gene were iden-
tified in two Chinese pedigrees. N92S is a novel mutation for SM, whereas R94H is
a recurrent mutation. Interestingly R94H was also previously found in a sporadic
case of PC-2 [2].
Other hypotheses have been formulated to explain this rare condition including
hamartomatous malformation of the pilosebaceous duct junction [9, 10] sebaceous
retention cysts, a variant of a dermoid cyst or a nevoid formation of abortive hair
follicles where sebaceous glands attach [11]. Also the occurrence of trauma, infec-
tion, or immunological conditions might be implicated [12].
SM usually begins in adolescence or early adult life and seems to be equally
distributed among both genders although it has been proposed that it is more fre-
quently found in males [13]. Although rarely, this condition has been described in
newborns [14] as well as in elderly [15]. The association of SM to the development
of sebaceous glands and common presentation during puberty suggest a hormonal
trigger for lesion growth.
scalp, and proximal extremities (Fig. 47.1). Less common locations include face,
scrotum [16], acral distribution in which involvement of distal extremities is more
prominent [17], and breasts [18] among others (Fig. 47.2). In more severe cases the
lesions can be generalized, polymorphous, and present with joint symptoms and
deteriorated general status, as described in one of the variants referred as to SM
generalisata [19].
Lesions are variable in size, tend to grow slowly, and have a sebum content that
can appear as a clear oily liquid or as a yellowish creamy material [11]. Although
usually asymptomatic, complication with secondary inflammatory changes can
occur. One of the distinctive characteristics that may differentiate this condition
from others, specifically from acne, is the absence of surface punctum.
346 A. Vivas and J. Keri
47.4 Workup
The diagnosis of SM is primarily clinical. However for some cases a biopsy may be
needed. The typical histopathological finding of these lesions is seen in the mid-
dermis with the presence of an encapsulated dermal cyst with infolded walls
composed of stratified, squamous epithelium without a granular layer. In addition
there are flattened sebaceous gland lobules arising from the wall and cellular hyaline
eosinophilic cuticles on the luminal side of the cyst wall similar to that of the
sebaceous duct [25]. Occasionally keratin, vellus hairs, hair follicles, and smooth
muscle components are noted in the lumen [23, 26] adding further evidence to the
hypothesis that SM is a hamartomatous condition [27].
Electron microscopy shows cyst wall cells undergoing trichilemmal keratiniza-
tion similar to that of the isthmus portion of the outer hair sheath [24].
Immunohistochemical analysis has demonstrated that the inner epithelial layer of
the cysts is positive for the specific marker calretinin which could be useful in iden-
tifying these lesions when equivocal [28].
The combined mammographic and sonographic findings can aid in confirming
the diagnosis of the subtype of SM involving the breast [18].
The clinical differential diagnosis of SM includes different forms of acne, erup-
tive vellus hair cysts, epidermoid or dermoid cysts, hidradenitis suppurativa, milia,
pseudofolliculitis barbae, neurofibromatosis, and lipomas [11].
47.5 Treatment
Treatment options are limited and only few present satisfactory results; therefore
this condition carries therapeutic challenge. As the lesions are characteristically
asymptomatic, treatment is usually not necessary. However as previously mentioned
some lesions can grow larger, present with inflammation, and develop symptoms
such as discomfort or pain and even can rupture with concomitant drainage [29].
In addition this condition can be cosmetically undesirable representing a psychologi-
cal burden for the patient, especially when lesions are located in visible areas.
In these cases surgical excision or incision and drainage are the standard treatments.
However conventional excision techniques have shown to be impractical when
treating multiple lesions; therefore new surgical procedures with the use of different
instruments have emerged [30, 31] to increase effectiveness of treatment and patient
47 Steatocystoma Multiplex 347
References
1. Oh SW, Kim MY, Lee JS, Kim SC. Keratin 17 mutation in pachyonychia congenita type 2
patient with early onset steatocystoma multiplex and Hutchinson-like tooth deformity.
J Dermatol. 2006;33:161–4.
2. Wang JF, Lu WS, Sun LD, et al. Novel missense mutation of keratin in Chinese family with
steatocystoma multiplex. Eur Acad Dermatol Venereol. 2009;23:723–4.
3. Kanda M, Natsuga K, Nishie W, et al. Morphological and genetic analysis of steatocystoma
multiplex in an Asian family with pachyonychia congenita type 2 harbouring a KRT17 mis-
sense mutation. Br J Dermatol. 2009;160:465–8.
4. Hurlimann AF, Panizzon RG, Burg G. Eruptive vellus hair cyst and steatocystoma multiplex:
hybrid cysts. Dermatology. 1996;192:64–6.
5. Moritz DL, Silverman RA. Steatocystoma multiplex treated with isotretinoin: a delayed
response. Cutis. 1988;42:437–9.
6. Ahn SK, Chung J, Lee WS, Lee SH, Choi EH. Hybrid cysts showing alternate combination of
eruptive vellus hair cyst, steatocystoma multiplex, and epidermoid cyst, and an association
among the three conditions. Am J Dermatopathol. 1996;18:645–9.
7. Yoshida M, Oiso N, Kurokawa I, Tsubura A, Kimura M, Kawada A. A case of multiple pilo-
sebaceous cysts. Case Rep Dermatol. 2010;2:116–9.
8. Tomkova H, Fujimoto W, Arata J. Expression of keratins (K 10 and K 17) in steatocystoma
multiplex, eruptive vellus hair cysts, and epidermoid and trichilemmal cysts. Am J
Dermatopathol. 1997;19:250–3.
9. Sabater-Marco V, Perez-Ferriols A. Steatocystoma multiplex with smooth muscle. A hamar-
toma of the pilosebaceous apparatus. Am J Dermatopathol. 1996;18:548–50.
10. Plewig G, Wolff HH, Braun-Falco O. Steatocystoma multiplex: anatomic reevaluation, elec-
tron microscopy, and autoradiography. Arch Dermatol Res. 1982;272:363–80.
11. Lima AM, Rocha SP, Batista CM, Reis CM, Leal II, Azevedo LE. Case for diagnosis.
Steatocystoma multiplex. An Bras Dermatol. 2011;86:165–6.
12. Setoyama M, Mizoguchi S, Usuki K, Kanzaki T. Steatocystoma multiplex: a case with unusual
clinical and histological manifestation. Am J Dermatopathol. 1997;19:89–92.
13. Magid ML, Wentzell JM, Roenick Jr HH. Multiple cystic lesions: steatocystoma multiplex.
Arch Dermatol. 1990;126:101. 104.
14. Park YM, Cho SH, Kang H. Congenital linear steatocystoma multiplex of the nose. Pediatr
Dermatol. 2000;17:136–8.
15. Rongioletti F, Cattarini G, Romanelli P. Late onset vulvar steatocystoma multiplex. Clin Exp
Dermatol. 2002;27:445–7.
348 A. Vivas and J. Keri
16. Stollery N. Photo quiz. Case 2. Steatocystoma multiplex. Practitioner. 2007;251(1701):31. 34.
17. Rollins T, Levin RM, Heymann WR. Acral steatocystoma multiplex. J Am AcadDermatol.
2000;43:396–9.
18. Mester J, Darwish M, Deshmukh SM. Steatocystoma multiplex of the breast: mammographic
and sonographic findings. AJR Am J Roentgenol. 1998;170:115–6.
19. Fekete GL, Fekete JE. Steatocystoma multiplex generalisata partially suppurativa-case report.
Acta Dermatovenerol Croat. 2010;18:114–9.
20. Kumakiri M, Yajima C. Eruptive steatocystoma multiplex on the scalp. J Dermatol.
1991;18:537–9.
21. Nishimura M, Kohda H, Urabe A. Steatocystoma multiplex. A facial papular variant. Arch
Dermatol. 1986;122:205–7.
22. Requena L, Martin L, Renedo G, et al. A facial variant of steatocystoma multiplex. Cutis.
1993;51:449–52.
23. Kim SJ, Park HJ, Oh ST, Lee JY, Cho BK. A case of steatocystoma multiplex limited to the
scalp. Ann Dermatol. 2009;21:106–9.
24. Davey MA. Steatocystoma multiplex treatment & management. In: Elston DM; editor.
Medscape reference. http://emedicine.medscape.com/article/1059725-treatment. Accessed 11
Dec 2011.
25. Yamada A, Saga K, Jimbow K. Acquired multiple pilosebaceous cysts on the face having the
histopathological features of steatocystoma multiplex and eruptive vellus hair cysts. Int J
Dermatol. 2005;44:861–3.
26. Thomas VD, Swanson NA, Lee KK. Benign epithelial tumors, hamartomas, and hyperplasias.
In: Wolff K, Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell DJ, editors. Fitzpatrick’s
dermatology in general medicine. 7th ed. New York, NY: McGraw-Hill; 2008. p. 1065–6.
27. Cho S, Chang SE, Choi JH, Sung KJ, Moon KC, Koh JK. Clinical and histologic features of
64 cases of steatocystoma multiplex. J Dermatol. 2002;29:152–6.
28. Riedel C, Brinkmeier T, Kutzne H, Plewig G, Frosch PJ. Late onset of a facial variant of ste-
atocystoma multiplex—calretinin as a specific marker of the follicular companion cell layer.
J Dtsch Dermatol Ges. 2008;6:480–2.
29. Cuperus E, Leguit RJ, Sigurdsson V. Steatocystoma multiplex, a rare distribution of a rare
disease. Eur J Dermatol. 2010;20:402–3.
30. Choudhary S, Koley S, Salodkar A. A modified surgical technique for steatocystoma multi-
plex. J Cutan Aesthet Surg. 2010;3:25–8.
31. Lee SJ, Choe YS, Park BC, Lee WJ, Kim do W. The vein hook successfully used for eradica-
tion of steatocystoma multiplex. Dermatol Surg. 2007;33:82–4.
32. Düzova AN, Sentürk GB. Suggestion for the treatment of steatocystoma multiplex located
exclusively on the face. Int J Dermatol. 2004;43:60–2.
33. Apaydin R, Bilen N, Bayramgürler D, Başdaş F, Harova G, Dökmeci S. Steatocystoma multi-
plex suppurativum: oral isotretinoin treatment combined with cryotherapy. Australas J
Dermatol. 2000;41:98–100.
34. Madan V, August PJ. Perforation and extirpation of steatocystoma multiplex. Int J Dermatol.
2009;48:329–30.
35. Mumcuoğlu CT, Gurel MS, Kiremitci U, Erdemir AV, Karakoca Y, Huten O. Er: yag laser
therapy for steatocystoma multiplex. Indian J Dermatol. 2010;55:300–1.
Chapter 48
Xanthomas
48.1 Introduction
Cutaneous xanthomas result from the localized accumulation of lipid within the
dermis or connective tissue. They can present anywhere on the body with a variety
of morphologies ranging from discrete macules and papules to nodules and diffuse
plaques, typically with a yellow-to-orange color due to the lipid deposition. In
darker skin types, however, the lesions may appear more red-to-brown in color.
Xanthomas are most commonly associated with hyperlipemic states, which may
be due to primary genetic causes or secondary to other metabolic derangements.
Less frequently, normolipemic xanthomas have been reported in association with
monoclonal gammopathies or without any associated underlying disease.
The major subtypes of xanthomas associated with disorders of lipid metabolism
include tuberous, tendinous, eruptive, planar, and palmar. Xanthelasmas, a type of
planar xanthoma, may or may not be associated with a hyperlipidemia. Prompt rec-
ognition and early diagnosis of the lesions can significantly aid in the management
and prognosis of any potential underlying disease as cutaneous lesions may some-
times precede any systemic signs of symptoms. Moreover, the morphology and dis-
tribution of the xanthomas can suggest a particular lipoproteinemia or another
systemic disease.
48.2 Background
The endogenous pathway of lipoprotein synthesis begins in the liver with the
formation of VLDL molecules. The central core of the VLDL, like the chylomicron,
is composed of triglycerides, which are derived from hepatic stores as well as circu-
lating free fatty acids. As with chylomicrons, VLDLs are also cleaved by LPL until
most of the triglycerides are removed and only a similar “remnant” remains. The
VLDL remnant is called an intermediate density lipoprotein (IDL) and is ready to
be taken up by the liver and degraded. The IDLs that escape hepatic uptake are
stripped of their remaining triglyceride core and enter systemic circulation as LDLs.
LDLs deliver cholesterol esters to peripheral tissues where they can be converted
into free cholesterol. Cholesterol is an essential component of many body tissues,
including the selectively permeable cell membrane bilayer and myelin nerve
sheaths. It also serves important roles in adrenal and gonadal steroidogenesis as
well as the production of bile acids. Any excess cholesterol is re-esterified for
hepatic storage [2, 3].
HDL also plays a very important and “protective” role in cholesterol metabo-
lism. Its most important function is to “scavenge” free cholesterol from peripheral
tissues so it can be esterified and transferred to other lipoproteins such as LDLs or
remnants of chylomicrons and VLDLs for reverse transport or transport back to the
liver. The esterification of free cholesterol is mediated by the enzyme lecithin cho-
lesterol acyl transferase (LCAT).
Cholesterol homeostasis involves a complex interweaving of mechanisms.
As such, disorders in lipid metabolism can occur in numerous ways. Inherited
disorders resulting from genetic mutations (primary hyperlipoproteinemia) can
yield absent or defective enzymes, receptors, or receptor ligands, with resultant
overproduction or decreased clearance of lipoproteins. Several mutations are known
to exist and are associated with a unique lipid profile. Additionally, secondary
hyperlipoproteinemias due to an underlying disease that may disrupt lipid metabo-
lism including diabetes mellitus, hypothyroidism, and nephrotic syndrome are not
uncommon. Medications, such as oral retinoids, oral contraceptive pills, antiretrovi-
ral drugs to name a few can also lead to a hyperlipemic state [2, 3].
Cutaneous xanthomas associated with hyperlipidemia are typically divided into five
main groups depending on morphology and/or location: tuberous, tendinous, erup-
tive, planar, and palmar. Xanthelasma palpebrarum is the most common type of
xanthoma presenting around the eyes. It is considered a subtype of planar xan-
thoma, although it is not necessarily associated with aberrant lipid metabolism
(Fig. 48.1). Xanthoma disseminatum and verruciform xanthoma are not usually
associated with any lipid abnormalities.
Tuberous xanthomas present as firm, painless, pink-to-yellow papules or nod-
ules most commonly on extensor surfaces, especially the elbows and knees, as
well as on pressure-prone areas such as the buttocks (Fig. 48.2). Tuberous xantho-
mas are particularly associated with hypercholesterolemic states such as familial
352 L. Rhee and M. Kaufmann
48.4 Workup
Xanthomas may be idiopathic or indicate more serious systemic disease. Key to the
workup is diagnosing and treating any underlying hyperlipidemia in order to mini-
mize the progression atherosclerotic disease. Any potential secondary causes of dis-
ease need to be excluded as well.
Primary, or inherited forms, of hyperlipidemia are generally a diagnosis of exclu-
sion. Laboratory studies used to either primary or secondary hyperlipidemia should
start with a serum lipid panel, including fasting plasma levels of triglycerides, cho-
lesterol, and HDL cholesterol. LDL and VLDL concentrations can be calculated
using the above information. Chylomicrons can be separated via ultracentrifugation
and electrophoresis, then quantified using immunologic methods.
Certain xanthomas are highly suggestive of specific familial hyperlipoprotein-
emias and can guide the workup and diagnosis: xanthoma striatum palmare are seen
in familial dysbetalipoproteinemia and intertriginous xanthomas in homozygous
familial hypercholesterolemia. Rarely, plane xanthomas may be associated with an
354 L. Rhee and M. Kaufmann
48.5 Treatment
Xanthomas are not always associated with hyperlipidemia, but when they are, a
multidisciplinary approach to lowering the lipid levels is essential. First-line thera-
pies include dietary modifications and lipid-lowering agents such as HMG-CoA
reductase inhibitors (“statins”), bile acid-binding resins, fibric acid derivatives, and/
or nicotinic acid. Cutaneous xanthomas generally resolve with correction of the
underlying lipid disorder. Slower growing xanthomas such as tendinous or tuberous
xanthomas may persist for years or never fully resolve in contrast to eruptive xan-
thomas, for example, which often disappear quickly with aggressive lipid-lowering
therapy [1–3].
The cutaneous lesions, particularly xanthelasmas, can be treated by surgical
excision or through locally destructive methods if they are a cosmetic or functional
concern. Laser therapy (CO2; pulsed-dye or erbium:YAG lasers; argon; Q-switched
Nd:YAG; KTP), di- or trichloroacetic acid, electrodesiccation, cryotherapy, intra-
lesional bleomycin, and intralesional corticosteroid injections have all been
reported to be helpful [6]. Unfortunately, the xanthomas often recur despite initial
success, and scarring, pigmentary changes, and koebnerization are potential
adverse effects.
48.6 Conclusion
References
1. Ford ES, Li C, Pearson WS, Zhao G, Mokdad AH. Trends in hypercholesterolemia, treatment
and control among United States adults. Int J Cardiol. 2010 ;140(2):226–35.
2. Parker F. Xanthomas and hyperlipidemias. J Am Acad Dermatol. 1985;13(1):1–30.
3. Bolognia J, Jorizzo J, Rapini R. Dermatology. 2nd ed. New York: Elselvier Limited; 2008. Print.
4. Tsouli SG, Kiortsis DN, Argyropoulou MI, Mikhailidis DP, Elisaf MS. Pathogenesis, detection
and treatment of Achilles tendon xanthomas. Eur J Clin Invest. 2005;35(4):236–44.
5. Streit E, Helmbold P. 65-year-old man with yellow-orange papules on both forearms. Eruptive
xanthomas. Hautarzt. 2009;60(10):834–7.
6. Rohrich RJ, Janis JE, Pownell PH. Xanthelasma palpebrarum: a review and current manage-
ment principles. Plast Reconstr Surg. 2002;110(5):1310–4.
Part VI
Pediatric Dermatoses Mimicking Acne
Chapter 49
Periorificial Granulomatous Dermatitis
49.1 Introduction
histopathologic characteristics of this skin disease because PGD may not be limited
to a perioral distribution and is histopathologically characterized by the presence of
a granulomatous inflammatory reaction [12].
49.2 Background
The etiology of PGD is unknown, although several theories exist. Many believe that
PGD is the less common granulomatous variant of perioral/periorificial dermatitis
(PD) and that PD and PGD share the same etiologic factors [5, 13–17].
The main etiologic factors in PD are thought to be a decreased stratum corneum
(SC) permeability barrier function [14] from intrinsic factors or deficiencies yet to
be identified or from extrinsic causes such as topical products [15, 16]. The topical
products implicated in PD include contact irritants or allergens in cosmetics and
topical corticosteroids (TCs) [4, 11, 14, 17–20]. While there are no reports in the
literature demonstrating SC permeability barrier dysfunction in PGD, there are con-
flicting reports regarding the pathogenic relationship of PGD to topical products.
Some authors report a pathogenic relationship between PGD and TC use, or to topi-
cal allergens and irritants such as formaldehyde and antiseptic solutions [3, 6,
13, 21], while others specifically state that no etiologic relationship is present
between TC and other products in PGD [22].
Whether the inciting factors are intrinsic or extrinsic, the initiating event for PD as
well as PGD is believed to be the same; damage to the hair follicle which results in
damage to the follicular wall, release of its contents into the surrounding dermis, and
subsequent inflammation [14, 22]. The antigenic component of the ruptured follicle
is unknown; however, it is thought in PGD to provoke a granulomatous response
[22]. There are limited data available to support this theory of pathogenesis.
PGD is a rare skin disease that occurs mostly in children with skin of color
[21, 23]. PGD has been seen healthy children ranging in age from 6 months to 18
years [17, 24, 25]. Afro-Caribbean, African American, and Asian children dominate
the reports, but Caucasian patients are also susceptible [21, 23]. There have been
reports of PGD being more common in boys versus girls [26, 27]; however, the
majority of reports state that both sexes are affected equally [21].
involves the vermillion border of the lip which helps distinguishes this eruption
from other diseases such as PD [23]. While the majority of cases reported in the
literature are confined to the face, eight reported cases of PGD also had generalized
skin lesions [21]. The patients with extrafacial involvement had lesions on the neck,
upper trunk, extensor wrists, and vaginal area [28, 29]. Importantly, extensive skin
involvement in PGD does not appear to change the duration of the eruption, the
response to treatment, or have any association with any specific underlying internal
diseases [15].
Scarring is variable in PGD. The initial cases described by Gianotti et al. [1] and
several subsequent authors [6, 28, 30] reported the occurrence of small pitted scars
after resolution of the papules. However, the majority of cases since then have
reported no scarring or other sequelae after the resolution of PGD [4, 13, 23].
In the majority of cases where skin biopsies were performed, a dermal granulo-
matous infiltrate was seen concentrated around the upper half of the hair follicles
[12, 13, 15]. In some biopsy specimens, the granulomatous infiltrate was more dif-
fuse, and in some rare cases no granulomas were detected on histologic examination
[2, 4]. Focal epidermal spongiosis is occasionally described; however, there is never
caseation necrosis, and the results of special stains and cultures for acid-fast bacilli
and fungi are always negative [15].
This granulomatous histologic appearance of PGD is not diagnostic. The dif-
ferential diagnosis of small papules with granulomatous histologic features in chil-
dren includes sarcoidosis, fungal or mycobacterial infection, familial juvenile
systemic granulomatosis (Blau syndrome), and granulomatous rosacea in addi-
tion to PGD [15]. In typical cases of PGD, these entities can be differentiated
362 J. Levin et al.
49.4 Workup
There is some disagreement concerning the recommended workup when the diag-
nosis of PGD is suspected. Because PGD occurs in healthy children and spontane-
ously resolves within a few months to years without scarring or sequelae, it is often
difficult to justify a biopsy or a thorough systemic evaluation [4]. However, it can be
difficult at times to differentiate PGD from other more serious conditions with
extracutaneous manifestations and potential long-term detrimental sequelae [23].
For this reason, when clinical presentation is not sufficient to diagnose PGD [4, 31],
histopathologic evaluation is recommended, with additional workup directed by the
clinician based on the needs of the individual case. Examples of tests that may be
included in the workup are chest radiograph, ophthalmologic examination, tubercu-
lin skin test, serum calcium, serum angiotensin-converting enzyme level, and the
use of special stains and tissue cultures for fungal or mycobacterial organisms on
histologic sections (Table 49.1) [15, 22, 31, 32].
In the literature, PGD has been likened to many dermatologic disorders with both
cutaneous involvement and cutaneous plus systemic involvement. The main differ-
ential diagnoses of PGD include acne vulgaris, periorificial dermatitis (PD), granu-
lomatous rosacea, sarcoidosis, lupus miliaris disseminatus faciei (LMDF), and
seborrheic dermatitis. Many of the facial dermatoses listed the differential of PGD
are discussed in detail elsewhere in the book as they are also difficult at times to
differentiate from acne vulgaris and other facial dermatoses. Table 49.2 summarizes
the defining characteristics of PGD while Table 49.3 lists the characteristics that
help differentiate the skin diseases listed above from PGD [4, 12, 18].
Table 49.1 Summary of possible workup for selected cases of periorificial granulomatous
dermatitisa,b
• Skin biopsy
Confirm consistency with diagnosis and evaluate for other potential diagnoses such as
sarcoidosis, infectious etiologies, others (see text)
Specimen must contain adequate depth to fully evaluate follicular structures
Special stains and cultures to detect fungal or mycobacterial organism if present
• Chest radiograph
• Ophthalmologic examination
• Tuberculin skin test
• Serum calcium
• Serum angiotensin-converting enzyme level
a
Workup to be determined based on judgement of clinician in cases where clinical diagnosis alone
is questionable
b
References [15, 22, 31, 32]
49 Periorificial Granulomatous Dermatitis 363
Less commonly reported skin diseases which should be included in the differential
diagnosis of PGD include allergic/irritant contact dermatitis, impetigo, acrodermati-
tis enteropathica, tinea faciei, lip-lickers dermatitis, atopic dermatitis, benign cephalic
histiocytosis, granulosis rubra nasi, glucagonoma syndrome, Blau syndrome, Haber
syndrome, and facial demodicosis.
49.5 Treatment
Patients and parents should be reassured that PGD is a benign, self-limited condi-
tion. Since the disease is benign and self-limited, treatment may be unnecessary.
However, the average reported time for spontaneous resolution is between 1 and 3
years [1, 6, 10, 30, 33–36], and the resolution may be hastened with the use of
appropriate therapy [15]. The time to resolution with treatment varies greatly in the
literature ranging from 1 week to 6 months [17, 20, 37].
There are no available randomized controlled trials providing guidance for the
best treatment in PGD because of the rare nature of this skin disease. Some reviews
dictate the first step in management to be discontinuation of all TCs or any other
364 J. Levin et al.
Table 49.3 Differentiation of periorificial granulomatous dermatitis from other simulant dermatoses
• Acne vulgaris
Comedones, pustules, nodule, and cysts present
High incidence in puberty
• Perioral dermatitis
Most common in young women in third to fourth decade
Pustules present
Spares the vermillion border
No extrafacial eruptions
• Granulomatous rosacea
Most common in young women in third to fourth decade
Centrofacial distribution
Background erythema, flushing, telangiectasias, pustules may be present
No spontaneous resolution
• Sarcoidosis
Rarely presents in children
Multiorgan involvement
Systemic symptoms present
Granulomas on histology do not have surrounding infiltrate and are not centered around a follicle
• Seborrheic dermatitis
Ears, nasolabial folds, eyebrows, and scalp commonly involved
Scale or hyperkeratosis often present
• Lupus miliaris disseminatus faciei
Scarring is present
Most common in Japanese adults
Caseation necrosis on histology
References [4, 12, 18]
possible inciting chemicals or products [12, 13, 24, 34]. Because there may be an
associated rebound or exacerbation of the skin disease after stopping TC use, many
physicians recommend the use of a mild topical corticosteroids daily for a few
weeks to effectively wean patients off more potent TCs; however, there is no scien-
tific evidence that this approach is beneficial [25, 37, 38]. The authors suggest dis-
continuation of TCs without a weaning approach [39].
In the literature there are several case reports demonstrating the effectiveness of
treatment with topical metronidazole, sulfacetamide-sulfur, topical erythromycin, oral
tetracyclines, or oral erythromycin for PGD [4, 15, 21]. Nguyen and Eichenfeld [17]
recommend a 1–2-month trial of topical metronidazole with the addition of oral eryth-
romycin. Urbatsch states that the administration of oral macrolides or tetracyclines,
alone or in combination with topical erythromycin, metronidazole, or sulfur-based
lotions, hastens resolution in most patients [15, 21]. Although oral tetracyclines can be
effective in treating PGD, they should not be used in PGD patients less than 8 years
old as tetracyclines have been known to cause dental enamel discoloration [24, 39].
The availability of subantimicrobial dosing of doxycycline offers an option that may
be effective without exposing the patient to antibiotic selection pressure [39].
49 Periorificial Granulomatous Dermatitis 365
49.6 Conclusion
PGD is a chronic and potentially disfiguring dermatosis, often affecting the face.
It may commonly be mistaken for acne vulgaris, but clinically appears as monomor-
phic papules rather than the comedones, papules, and pustules observed in patients
with acne vulgaris. Treatment can be challenging and requires often long-term com-
bination therapy with both topical and oral medications.
References
21. James W, Berger T, Elston D. Andrews’ diseases of the skin: clinical dermatology. 11th ed.
USA: Elsevier Health; 2011.
22. Makkar R, Ramesh V. On the diagnosis of facial granulomatous dermatoses of obscure origin.
Int J Dermatol. 2005;44(7):606–9.
23. Choi YL, Lee KJ, Cho HJ, Kim WS, Lee JH, Yang JM, et al. Case of childhood granulomatous
periorificial dermatitis in a Korean boy treated by oral erythromycin. J Dermatol.
2006;33(11):806–8.
24. Kuflik JH, Janniger CK, Piela Z. Perioral dermatitis: an acneiform eruption. Cutis. 2001;67:
21–2.
25. Coskey RJ. Perioral dermatitis. Cutis. 1984;34:8.
26. Kim MS, Kim BS, Koh WS, Cho JJ, Chun DK. A case of childhood granulomatous perioral
dermatitis. Korean J Dermatol. 2000;38:526–9.
27. Lee WJ, Yang JH, Lee MW, Choi JH, Moon KC, Koh JK. A case of childhood granulomatous
periorificial dermatitis. Korean J Dermatol. 2008;46:1570–2.
28. Andry P, Bodemer C, Teillac-Hamel D, Fraitag S, DeProst Y. Granulomatous perioral derma-
titis in childhood: eight cases [abstract]. Pediatr Dermatol. 1995;12:76.
29. Hansen KK, McTigue K, Esterly NB. Multiple facial, neck, and upper trunk papules in a black
child: childhood granulomatous perioral dermatitis with involvement of the neck and upper
trunk. Arch Dermatol. 1992;128:1396–7.
30. Husz S, Korom I. Periocular dermatitis: a micropapular sarcoid-like granulomatous dermatitis
in a woman. Dermatologica. 1981;162:424–8.
31. Frieden IJ, Prose NS, Fletcher V, Turner ML. Granulomatous perioral dermatitis or sarcoid?
Arch Dermatol. 1990;126(9):1237–8.
32. Gupta AK, Goldfarb MT, Rasmussen JE. Papular midfacial eruption in a child. Cutaneous
Sarcoidosis. Arch Dermatol. 1989;125(12):1704–5. 1707–8.
33. Gianotti F. Cutaneous benign histiocytosis of childhood. Mod Probl Paediatr. 1975;17:
193–203.
34. El-Saad E-RM. Perioral dermatitis with epithelioid cell granulomas in a woman: a possible
new etiology. Acta Derm Venereol. 1979;60:359–60.
35. Hansen KK, McTigue K, Esterly NB. Multiple facial, neck, and upper trunk papules in a black
child: childhood granulomatous perioral dermatitis with involvement of the neck and upper
trunk. Arch Dermatol. 1992;128:1396–7.
36. Antony FC, Buckley DA, Russell-Jones R. Childhood granulomatous periorificial dermatitis
in an Asian girl—a variant of sarcoid? Clin Exp Dermatol. 2002;27(4):275–6.
37. Manders SM, Lucky AW. Perioral dermatitis in childhood. J Am Acad Dermatol.
1992;27:688–92.
38. Wilkinson DS, Kirton V, Wilkinson JD. Perioral dermatitis: a 12-year review. Br J Dermatol.
1979;101:245–57.
39. Del Rosso JQ. Management of papulopustular rosacea and perioral dermatitis with emphasis
on iatrogenic causation or exacerbation of inflammatory facial dermatoses: use of doxycycline
modified-release 40 mg capsule once daily in combination with properly selected skin care as
an effective therapeutic approach. J Clin Aesthet Dermatol. 2011;4:20–30.
Chapter 50
Keratosis Pilaris Atrophicans
50.1 Introduction
50.2 Background
Onset of the disease is noted a few months after birth, with the usual phenotype
being Fitzpatrick skin type II, blond-haired boys, with the inherited findings occurring
as an autosomal dominant disease with variable penetrance [7]. Pathogenesis is felt to
occur as the result of a plug that forms in the follicular ostium that causes keratotic
follicular papules and hair shaft deformation. Histologically, evidence of hyperkerato-
sis and hypergranulosis of the isthmus and infundibulum of the follicle lead to sur-
rounding inflammation in the dermis. Subsequently, mononuclear cells appear around
the superior follicle with surrounding mucin and destructive changes to connective
tissue. After the disintegration of the follicle, reactive changes surrounding a naked hair
shaft result in further inflammation and finally scarring in the final chronic phase [1].
KPAF begins shortly after birth and is generally clinically evident before 5 years of
age with erythema and small horny follicular papules emerging laterally from the
eyebrows, often extending to the cheeks and forehead (Fig. 50.1). This condition is
often associated with atopic dermatitis, asthma, and seasonal allergies. On exam many
patients also often have keratosis pilaris on extremity extensor surfaces [8]. Eventually
the affected areas become atrophic with residual scarring alopecia. The cycle contin-
ues through adolescence but generally halts with the beginning of puberty.
50.4 Workup
Diagnosis is based on clinical findings and may be made soon after birth. Since
KPAF is believed to be inherited and is considered a marker of certain genetic
syndromes such as Noonan syndrome [9–11], Rubinstein-Taybi syndrome [12],
50 Keratosis Pilaris Atrophicans 369
50.5 Treatment
50.6 Conclusion
KPAF is a progressive scarring condition affecting the face and the eyebrows. While
treatment is challenging, aggressive therapy early after diagnosis may halt progression
of the disease and minimize the onset of permanent scarring and alopecia later in life.
References
1. Baden HP, Byers HR. Clinical findings, cutaneous pathology, and response to therapy in 21
patients with keratosis pilaris atrophicans. Arch Dermatol. 1994;130:469–75.
2. Wilson, Erasmus (1878) Lectures on Dermatology. London. p 217
3. Paul Taenzer (1889) “Ueber das Ulerythema ophryogenes,” Monats. f. prakt. Derwnat.
Hamburg. pp 197–208.viii
4. Nazarenko SA, Ostroverkhova NV, Vasiljeva EO, et al. Keratosis pilaris and ulerythema
ophryogenes associated with an 18 p deletion caused by a Y/18 translocation. Am J Med
Genet. 1999;85:179–82.
370 O. Pacha and A. Hebert
5. Fiorentini C, Bardazzi F, Bianchi T, Patrizi A. Keratosis pilaris in a girl with monosomy 18p.
J Eur Acad Dermatol Venereol. 1999;12 suppl 2:S221.
6. Stratakis, Zouboulis, Gollnick HPM, Orfanos. Keratosis pilaris/ulerythema ophryogenes and
18p deletion: is it possible that theLAMA1 gene is involved? J Med Genet. 2001;38:127.
7. Oranje AP. Others. Keratosis pilaris atrophicans: one heterogeneous disease or a symptom in
different clinical entities? Arch Dermatol. 1994;130:500.
8. Davenport DD. Ulerythema ophryogenes. Arch Dermatol. 1964;89:134–40.
9. Pierini DO, Pierini AM. Keratosis pilaris atrophicans faciei (Ulerythema ophryogenes): a cuta-
neous marker in the Noonan syndrome. Br J Dermatol. 1979;100:409–16.
10. Snell JA, Mallory SB. Ulerythema ophryogenes in Noonan syndrome. Pediatr Dermatol.
1990;7:77–8.
11. Turner AM. Noonan syndrome. J Paediatr Child Health. 2011. doi:10.1111/j.1440-1754.
2010.01970.x.
12. Centeno PG, Roson E, Peteiro C, Pereiro M, Toribio J. Rubinstein-Taybi syndrome and
Ulerythema ophryogenes in a 9-year-old boy. Pediatr Dermatol. 1999;16:134–6.
13. Florez A, Fernandez-Redondo V, Toribio J. Ulerythema ophryogenes in Cornelia de Lange
syndrome. Pediatr Dermatol. 2002;19:42–5.
14. Manci EA, Martinez JE, Horenstein MG, Gardner TM, Ahmed A, Mancao MC, et al.
Cardiofaciocutaneous syndrome (CFC) with congenital peripheral neuropathy and nonorganic
malnutrition: An autopsy study. Am J Med Genet A. 2005;137A:1–8. doi:10.1002/ajmg.a.30834.
15. Borradori L, Blanchet-Bardon C. Skin manifestations of cardio-facio-cutaneous syndrome.
J Am Acad Dermatol. 1993;28:815–9.
16. Layton AM, Cunliffe WJ. A case of ulerythema ophryogenes responding to isotretinoin. Br J
Dermatol. 1993;129:645–6.
17. Burnett JW, Schwartz MF, Berberian BJ. Ulerythema ophryogenes with multiple congenital
anomalies. J Am Acad Dermatol. 1988;18:437–40.
18. Callaway SR, Lesher JL. Keratosis pilaris atrophicans: case series and review. Pediatr
Dermatol. 2004;21:14–7.
19. Ross EK, Tan E, Shapiro J. Update on primary cicatricial alopecias. J Am Acad Dermatol.
2005;53(1):1–37. doi:10.1016/j.jaad.2004.06.015. ISSN 0190–9622.
20. Di Lernia V, Bisighini G. Discoid lupus erythematosus during treatment with cyclosporine.
Acta Derm Venereol. 1996;76:87–8.
21. Mirmirani P, Willey A, Price VH. Short course of oral cyclosporine in lichen planopilaris.
J Am Acad Dermatol. 2003;49:667–71.
22. Goldman GD. Eyebrow Transplantation. Dermatol Surg. 2001;27:352–4. doi:10.1046/j.1524-4725.
2001.00209.x.
23. Barankin B, Taher M, Wasel N. Successful hair transplant of eyebrow alopecia areata. J Cutan
Med Surg. 2005;9:162–4.
Chapter 51
Neonatal and Infantile Acne
Hilary Baldwin
51.1 Introduction
Acne vulgaris is a disease that typically affects teenagers but may continue into
adulthood in some cases. However, acneiform eruptions can occur in the pediatric
population from birth onward. Neonatal acne refers to lesions that develop in the
neonatal period, from birth through 4 weeks old. Some consider neonatal acne to be
synonymous with neonatal cephalic pustulosis. Infantile acne refers to the acneiform
eruption that typically presents in children between 1 month and 1 year old [1].
Making the correct diagnosis can be a challenge, and in some cases the diagnosis of
infantile acne is given whenever comedones are clinically present even if the patient
is a neonate.
51.2 Background
In one study, pustules of the necks of 8 of 13 neonates with acne revealed M. furfur
[5]. However, not all patients with the eruption have positive cultures, suggesting
hypersensitivity to yeast as the causative factor rather than level of colonization
itself [6–8].
Little epidemiological data is available on infantile acne, as it so uncommon.
Expert opinions suggest that it occurs more commonly in boys than girls. Infantile
acne usually presents between 3 and 6 months of age, but it may develop as early as
immediately after birth or as late as 1 year old. While pustular lesions may be pres-
ent in both NCP and infantile acne, the presence of comedonal lesions distinguishes
infantile acne from the pustular lesions of neonatal acne/ NCP [6].
Neonatal acne typically presents with pustules and erythematous papules on the
cheeks, chin, and forehead (see Fig. 51.1). Lesions less commonly affect the neck,
chest, or scalp. Comedones may occasionally be present as well. Most cases are
mild in severity and self-limited, resolving spontaneously within 3 months [2]. NCP
patients may present predominantly with pustules and inflammatory papules, but
characteristically lack comedones.
Infantile acne may begin in the neonatal period with the presence of comedones
that persist as the child ages. Unlike in neonatal acne, patients with infantile acne
tend to develop inflammatory lesions, including papules, pustules, and in some
cases nodules or cysts [9]. While infantile acne typically resolves by the age of 1
year, it may rarely persist for several years [10]. Inflammatory disease can result in
scarring if not appropriately treated. It also may be associated with the development
of severe acne later in life during adolescence [6].
51.4 Workup
51.5 Treatment
51.6 Conclusion
While most common in adolescence, acne can occur as early as the neonatal period.
The lesions observed in newborns is most commonly associated with Malassezia
yeast on the skin surface. However, true comedonal disease does occur. Therapy
should be initiated based on clinical appearance and the parents’ preferences.
374 H. Baldwin
References
1. Friedlander SF, Eichenfield LF, Fowler Jr JF, et al. Acne epidemiology and pathophysiology.
Semin Cutan Med Surg. 2010;29:2–4.
2. Cantatore-Francis JL, Glick SA. Childhood acne: evaluation and management. Dermatol Ther.
2006;19:202–9.
3. Antoniou C, Dessinoti C, Stratigos A, Katsambas AD. Clinical and therapeutic approaches to
childhood acne: an update. Pediatr Dermatol. 2009;26:373–80.
4. Bergman J, Eichenfield LF. Neonatal acne and cephalic pustulosis: is Malassezia the whole
story? Arch Dermatol. 2002;138:255–7.
5. Rapelanoro R, Mortureux P, Couprie B, Malleville J, Taieb A. Neonatal Malassezia furfur
pustulosis. Arch Dermatol. 1996;132:190–3.
6. Eichenfield LF, Baldwin HE, Friedlander SF, Mancini AJ, Yan AC. Pediatric acne manage-
ment: optimizing outcomes. Semin Cutaneous Med Surg. 2011;30 Suppl 1:3S.
7. Bernier V, Weill FX, Hirigoyen V, et al. Skin colonization by Malassezia special in neonates:
a prospective study and relationship with neonatal cephalic pustulosis. Arch Dermatol.
2002;138:215–8.
8. Ayhan M, Sancak B, Karaduman A, Arikan S, Sarhin S. Colonization of neonate skin by
Malassezia species: relationship with neonatal cephalic pustulosis. J Am Acad Dermatol.
2007;57:1012–10128.
9. Tom WL, Friedlander SF. Acne through the ages: case-based observations through childhood
and adolescence. Clin Pediatr (Phila). 2008;47:639–51.
10. Chew EW, Bingham A, Burrows D. Incidence of acne vulgaris in patients with infantile acne.
Clin Exp Dermatol. 1990;15:376–7.
11. Krakowski AC, Eichenfield LF. Pediatric acne: clinical presentations, evaluation, and manage-
ment. J Drugs of Dermatol. 2007;6(6):589–93.
12. Sancak B, Ayhan M, Karaduman A, Arikan S. In vitro activity of ketoconazole, itraconazole
and terbinafine against Malassezia strains isolated from neonates. Microbiyol Bul. 2005;39:
301–8.
13. Gollnick H, Cunliffe W, Berson D, et al. Management of acne: a report from a Global Alliance
to Improve Outcomes in Acne. J Am Acad Dermatol. 2003;49(1 Suppl):S1–37.
Chapter 52
Papular Granuloma Annulare
Rebecca Smith
52.1 Introduction
52.2 Background
The cause of GA is unknown although it has been reported to follow trauma [3],
viral infections [4], tuberculin skin tests, malignancies [5], solar radiation, and
insect bites [6]. It is hypothesized that a cell-mediated delayed-type hypersensitivity
reaction to an unknown antigen is the inciting event. It has been postulated that the
characteristic rings may form in response to a centralized insult that sets off an
inflammatory chain reaction [7].
Granuloma annulare can occur at any age, but is most commonly diagnosed in
children and young adults. Females are affected twice as commonly as males [8].
The disseminated form is more common in adults [9]. The condition is usually seen
in otherwise healthy patients but has occasionally been associated with diabetes or
thyroid disease [10]. While the disease is usually sporadic, inherited cases have
been described with an increased incidence of HLA-Bw35 in Israel [11], HLA-B8
in Denmark [12], and HLA-A29, B14, and B15 in Belfast [13]. The association with
these particular phenotypes, however, may simply be population specific.
GA presents with skin findings only. The most common form of GA is the localized
type which presents as ringed, firm, and flesh-colored to slightly erythematous pap-
ules without scale or epidermal change (insert Fig. 52.1); the ringed papules can
affect any skin surface, but is most commonly found on the extremities. The face and
scalp are only rarely affected. The distribution of GA is approximately 60 % isolated
to hands and arms, 20 % to legs and feet, and 7 % on both the upper and lower
extremities. Fewer than 5 % of patients present with lesions on the trunk [14]. The
rings vary in size and slowly expand reaching up to several centimeters in diameter
with central clearing. Many patients have only a single ring, but multiple lesions are
common. Over half of the patients with localized disease experience spontaneous
resolution within 2 years. A papular umbilicated form of GA without perforation has
been described in school age children on the dorsal hands and fingers [15].
Only rarely would papular GA be confused clinically with acne due to the dif-
ference in location and the lack of epidermal change. A localized papular eruption
of GA on the chest or back could mimic an acneiform eruption, but the lack of
comedones or pustules and the more monomorphous nature of GA would argue
against acne. Papular GA would more often be in the differential diagnosis of
arthropod assault reaction, secondary syphilis, xanthomas, or non-X-histiocytosis.
With papular GA, the diagnosis is often difficult due to the lack of the more typical
annular plaques.
The differential diagnosis for classic GA includes annular lichen planus and
tinea corporis. Often children have been unsuccessfully treated for tinea before
presenting to a dermatologist for definitive diagnosis. A potassium hydroxide prep-
aration, the lack of scale, and/or a confirmatory biopsy can differentiate these
entities.
Generalized or disseminated GA consists of hundreds to thousands of individual,
small papules that may form small annular plaques, are usually symmetrical, and
can coalesce into reticulated, circinate, or linear patterns [10]. Unlike other forms of
GA, the trunk is usually involved with a propensity for the neck, forearms, legs, and
extensor elbows. This form may exhibit a slightly more violaceous color. It is
unusual to see involvement of the face, palms, soles, or mucous membranes [9].
Subcutaneous GA is a variant generally seen in young children with large,
asymptomatic, skin colored, rapidly enlarging deep dermal or subcutaneous nod-
ules [16]. They may be solitary or multiple and have a predilection for pretibial skin,
palms, soles, buttocks, fingers, toes, and periorbital areas. The skin overlying the
lesions can ulcerate and often these nodules spontaneously regress. Deep GA can
clinically resemble rheumatoid nodules, deep granulomatous infections, and subcu-
taneous sarcoid.
Perforating GA is a more rare form consisting of asymptomatic, superficial
small-grouped papules with central umbilicated ulcerations and scale crust. These
occur most frequently on the dorsal hands and fingers [17]. Differential diagnosis of
perforating GA includes perforating collagenosis, Kyrle disease, and elastosis per-
forans serpiginosa.
52.4 Workup
Patients with GA are generally healthy and laboratory tests are usually normal and
not recommended. The disseminated form may be seen more commonly in immu-
nocompromised patients such as those with human immunodeficiency virus and
lymphomas. Such patients may also have GA in atypical locations.
Often, the diagnosis can be made clinically, but a confirmatory punch biopsy can
be helpful especially with the less common variants. Histologically, GA is a granu-
lomatous dermatitis exhibiting focal degeneration of collagen and elastin fibers,
mucin deposition, and a perivascular and interstitial lymphohistiocytic infiltrate in
the upper and mid-dermis [18]. It is the increased mucin that is the hallmark of GA
[19]. If not readily apparent on routine stains, using colloidal iron or Alcian blue to
highlight mucin can be helpful. Palisaded granuloma with central connective tissue
degeneration surrounded by histiocytes and lymphocytes is a common pattern seen
microscopically. Often, the pattern is more infiltrative or interstitial with scattered
histiocytes infiltrating between collagen fibers. In perforating GA there is transepi-
dermal elimination of the degenerating collagen bundles.
378 R. Smith
52.5 Treatment
GA is rarely symptomatic, and due to the benign and often self-limited nature,
clinical observation and reassurance are often the treatment of choice. While lesions
can have a chronic relapsing course, they usually persist for only 1–4 years, and
73 % of all lesions disappear within 2 years. Cosmetic disfigurement can prompt the
need for intervention and many modalities have been utilized. First-line therapies
include topical steroids with or without occlusion or intralesional injections of tri-
amcinolone. Complete involution of the entire lesion has been reported to follow
biopsy. Other treatment modalities include cryosurgery [20], topical calcineurin
inhibitors, PUVA [21], and CO2 laser [22].
In general, systemic agents are reserved for more severe generalized cases. GA
has been treated with short-term oral corticosteroids, chlorpropamide, pentoxifyl-
line [23], nicotinamide, niacinamide [24], isotretinoin [25], etretinate [26], salicy-
lates, potassium iodide, vitamin E, fumaric acid esters, antimalarials [27], dapsone
[28], cyclosporine [29], and infliximab. Spontaneous resolution of the disease
makes evaluation of the efficacy of such treatments more difficult. No large, ran-
domized placebo-controlled double-blind studies have been performed.
52.6 Conclusion
The papular form of GA may mimic acne vulgaris in some cases. The lack of
comedones and pustules and the monomorphic papular appearance of this rash clin-
ically distinguish it from acne. While self-limited in most cases, lesions can persist
for months to years, so therapies are targeted towards symptomatic relief and the
unsightly appearance.
References
10. Dabski K, Winkelmann RK. Generalized granuloma annulare: clinical and laboratory findings
in 100 patients. J Am Acad Dermatol. 1989;20:39–47.
11. Friedman-Birnbaum R, Gideoni O, Bergman R, Pollack S. A study of HLA antigen association
in localized and generalized granuloma annulare. Br J Dermatol. 1986;115:329–33.
12. Andersen BL, Verdich J. Granuloma annulare and diabetes mellitus. Clin Exp Dermatol.
1979;4:31–7.
13. Middleton D, Allen GE. HLA antigen frequency in granuloma annulare. Br J Dermatol.
1984;110:57–9.
14. Cronquist SD, Stashower ME, Benson PM. Deep dermal granuloma annulare presenting as an
eyelid tumor in a child, with review of pediatric eyelid lesions. Pediatr Dermatol.
1999;16:377–80.
15. Lucky AW, Prose NS, Bove K, White WL, Jorizzo JL. Papular umbilicated granuloma annu-
lare. Arch Dermatol. 1992;128:1375–8.
16. Felner EI, Steinberg JB, Weinberg AG. Subcutaneous granuloma annulare: a review of 47
cases. Pediatrics. 1997;100:965–7.
17. Shimizu H, Harada T, Baba E, Kuramochi M. Perforating granuloma annulare. Int J Dermatol.
1985;24:581–3.
18. Elder D, Elenitsas R, Jaworsky C, Johnson B. Lever’s Histopathology of the skin. 8th ed.
Philadelphia, PA: Lippincott-Raven Publishers; 1997.
19. Dabski K, Winkelmann RK. Generalized granuloma annulare: histopathology and immunopa-
thology. Systematic review of 100 cases and comparison with localized granuloma annulare.
J Am Acad Dermatol. 1989;20:28–39.
20. Blume-Peytavi U, Zouboulis CC, Jacobi H, Scholz A, Bisson S, Orfanos CE. Successful
outcome of cryosurgery in patients with granuloma annulare. Br J Dermatol. 1994;130:
494–7.
21. Hindson TC, Spiro JG, Cochrane H. PUVA therapy of diffuse granuloma annulare. Clin Exp
Dermatol. 1988;13:26–7.
22. Rouilleault PH. CO2 laser and granuloma annulare. J Dermatol Surg Oncol. 1988;14:120.
23. Rubel DM, Wood G, Rosen R, Jopp-McKay A. Generalised granuloma annulare successfully
treated with pentoxifylline. Australas J Dermatol. 1993;34:103–8.
24. Ma A, Medenica M. Response of generalized granuloma annulare to high-dose niacinamide.
Arch Dermatol. 1983;119:836–9.
25. Ratnavel RC, Norris PG. Perforating granuloma annulare: response to treatment with isotreti-
noin. J Am Acad Dermatol. 1995;32:126–7.
26. Botella-Estrada R, Guillen C, Sanmartin O, Aliaga A. Disseminated granuloma annulare: reso-
lution with etretinate therapy. J Am Acad Dermatol. 1992;26:777–8.
27. Simon Jr M, von den Driesch P. Antimalarials for control of disseminated granuloma annulare
in children. J Am Acad Dermatol. 1994;31:1064–5.
28. Steiner A, Pehamberger H, Wolff K. Sulfone treatment of granuloma annulare. J Am Acad
Dermatol. 1985;13:1004–8.
29. Fiallo P. Cyclosporin for the treatment of granuloma annulare. Br J Dermatol. 1998;138:
369–70.
Chapter 53
Precocious Puberty and Acne
53.1 Introduction
M. Miyar, M.D.
Department of Dermatology, UT Southwestern, Austin, TX, USA
M.L. Levy, M.D. (*)
Department of Pediatric Dermatology, Dell Children’s Medical Center,
4900 Mueller Blvd., Austin, TX 78723, USA
e-mail: [email protected]
53.2 Background
The presence of acne in a child between 1 and 7 years old is unusual and raises a
concern for precocious puberty or hyperandrogenemia [16, 17]. Severe or recalci-
trant acne are especially suspicious [17, 18]. These patients usually present with
comedones on the central face, including the mid-forehead, nose, and chin
(Fig. 53.1). Comedones in the concha of the ear are also suspicious (Fig. 53.2).
53 Precocious Puberty and Acne 383
Inflammatory lesions may be present as well [9]. The child should be examined for
signs of advanced pubertal development, such as axillary or pubic hair development
and breast development in girls or testicular development in boys. Other signs of
precocious puberty include advanced osseous maturation and a high height and
weight for age [8]. The increased rate of bone maturation can result in premature
epiphyseal closure translating to a shorter adult stature. Approximately 30 % of
girls and an even higher percentage of boys with precocious puberty grow to less
than the 5th percentile of height as adults [2, 8]. While cognitive development is
normal, these children may have emotional and behavioral issues [8].
384 M. Miyar and M.L. Levy
53.4 Workup
Initial evaluation for precocious puberty should include a review of the growth
chart, examination of bone age, and laboratory evaluations. Bone age measurement
can help determine if there is advanced bone age and is considered by some to be
the best screening evaluation for precocious puberty [16–19]. Children with preco-
cious puberty often have a bone age 2–3 standard deviations above their chronologi-
cal age [8]. Moreover, children with high androgen levels have accelerated growth
across standardized growth percentiles [19]. Children with suspected precocious
puberty or hyperandrogenism require a hormonal evaluation, which can be per-
formed by the dermatologist, pediatrician, or more commonly, referral to a pediatric
endocrinologist [20, 21]. Initial screening tests include serum free and total testos-
terone, DHEA-S, estradiol, and ratio of luteinizing hormone (LH) to follicle-
stimulating hormone (FSH) [19]. Moderate elevation of DHEA-S is suggestive of
an adrenal pathology (e.g., CAH), while extremely high levels of DHEA-S and/or
testosterone are more suggestive of an adrenal tumor [22].
Concentrations of sex hormones may be used to help stage puberty. In both early
normal puberty and the early phase of sexual precocity, serum estradiol is low or
undetectable in girls. Serum testosterone levels are detectable or slightly elevated at
the time of diagnosis in boys. LH levels are also elevated in children with central
sexual precocity. The gonadotropin-releasing hormone (GnRH) stimulation test is
also helpful in diagnosing precocious puberty. The test shows whether there is a
predominance of LH over FSH [23]. Females with poor LH response in the GnRH
stimulation test may also have a leuprolide stimulation test to evaluate for estradiol
levels [8]. Other laboratory tests to be evaluated in working up hyperandrogenism
include prolactin, cortisol, 17α-hydroxyprogesterone, androstenedione, and an
adrenocorticotropic hormone (ACTH) stimulation test [16–18, 22]. Imaging tech-
nology is also helpful as pelvic ultrasonography can detect ovarian and uterine
enlargement in girls. Magnetic resonance imaging (MRI) of the brain is indicated if
true central precocious puberty is suspected [2].
53.5 Treatment
Acne treatment in a child with precocious puberty depends on severity of the acne.
In general, mild cases of comedonal acne can be treated with topical retinoids like
tretinoin or adapalene. Combination therapy is usually most successful using com-
edolytic agent like a retinoid along with topical benzoyl peroxide or salicylic acid
[17]. Mild inflammatory lesions can be treated with topical benzoyl peroxide and/or
topical antibiotics like clindamycin or erythromycin [19]. Fixed-dose combination
products containing more than one active ingredient in one product are good options
for children, as only one product needs to be used. For more severe or recalcitrant
disease, oral antibiotics or oral retinoids may be needed. However, tetracyclines
53 Precocious Puberty and Acne 385
should not be used in children younger than 8 years old due to the potential damage
to the developing bones and teeth [24]. If antibiotics are to be used in children less
than 8 years old, erythromycin and trimethoprim/sulfamethoxazole are treatment
options [18].
If a patient is suspected of precocious puberty, early involvement of a pediatric
endocrinologist is advised. While acne may be managed by dermatologists, cur-
rent hormonal therapies that may be prescribed by endocrinologists include GnRH
agonists and growth hormone (GH). Treatment with GnRH agonists such as leup-
rolide helps prevent early fusion of the epiphyseal plates to avoid unnecessary
short stature [25].
References
1. Marshall WA, Tanner JM. Variations in pattern of pubertal changes in girls. Arch Dis Child.
1969;44(235):291–303.
2. Sørensen K, Mouritsen A, Aksglaede L, Hagen CP, Mogensen SS, Juul A. Recent secular
trends in pubertal timing: implications for evaluation and diagnosis of precocious puberty.
Horm Res Paediatr. 2012;77(3):137–45.
3. Biro FM, Galvez MP, Greenspan LC, Succop PA, Vangeepuram N, Pinney SM, et al. Pubertal
assessment method and baseline characteristics in a mixed longitudinal study of girls.
Pediatrics. 2010;126(3):e583–90.
4. Herman-Giddens ME, Steffes J, Harris D, Slora E, Hussey M, Dowshen SA, et al. Secondary
sexual characteristics in boys: data from the Pediatric Research in Office Settings Network.
Pediatrics. 2012;130(5):e1058–68.
5. Kaplowitz PB, Oberfield SE. Reexamination of the age limit for defining when puberty is
precocious in girls in the United States: implications for evaluation and treatment. Drug and
Therapeutics and Executive Committees of the Lawson Wilkins Pediatric Endocrine Society.
Pediatrics. 1999;104(4 Pt 1):936–41.
6. Midyett LK, Moore WV, Jacobson JD. Are pubertal changes in girls before age 8 benign?
Pediatrics. 2003;111(1):47–51.
7. Cisternino M, Arrigo T, Pasquino AM, Tinelli C, Antoniazzi F, Beduschi L, et al. Etiology and
age incidence of precocious puberty in girls: a multicentric study. J Pediatr Endocrinol Metab.
2000;13 Suppl 1:695–701.
8. Kliegman RM. Nelson textbook of pediatrics. 19th ed. Philadelphia, PA: Elsevier/Saunders;
2011.
9. Lucky AW, Biro FM, Huster GA, Leach AD, Morrison JA, Ratterman J. Acne vulgaris in
premenarchal girls. An early sign of puberty associated with rising levels of dehydroepian-
drosterone. Arch Dermatol. 1994;130(3):308–14.
10. Pochi PE, Strauss JS, Downing DT. Skin surface lipid composition, acne, pubertal develop-
ment, and urinary excretion of testosterone and 17-ketosteroids in children. J Invest Dermatol.
1977;69(5):485–9.
11. Lucky AW, Biro FM, Simbartl LA, Morrison JA, Sorg NW. Predictors of severity of acne
vulgaris in young adolescent girls: results of a five-year longitudinal study. J Pediatr.
1997;130(1):30–9.
12. Mancini AJ, Baldwin HE, Eichenfield LF, Friedlander SF, Yan AC. Acne life cycle: the spec-
trum of pediatric disease. Semin Cutan Med Surg. 2011;30(3 Suppl):S2–5.
13. Lucky AW, Biro FM, Huster GA, Morrison JA, Elder N. Acne vulgaris in early adolescent
boys. Correlations with pubertal maturation and age. Arch Dermatol. 1991;127(2):210–6.
386 M. Miyar and M.L. Levy
14. Reiter EO, Fuldauer VG, Root AW. Secretion of the adrenal androgen, dehydroepiandrosterone
sulfate, during normal infancy, childhood, and adolescence, in sick infants, and in children
with endocrinologic abnormalities. J Pediatr. 1977;90(5):766–70.
15. Goldberg JL, Dabade TS, Davis SA, Feldman SR, Krowchuk DP, Fleischer AB. Changing age
of acne vulgaris visits: another sign of earlier puberty? Pediatr Dermatol. 2011;28(6):645–8.
16. Paller AS, Mancini AJ. Hurwitz clinical pediatric dermatology. 4th ed. Edinburgh, NY:
Elsevier Saunders; 2011.
17. Cantatore-Francis JL, Glick SA. Childhood acne: evaluation and management. Dermatol Ther.
2006;19(4):202–9.
18. Antoniou C, Dessinioti C, Stratigos AJ, Katsambas AD. Clinical and therapeutic approach to
childhood acne: an update. Pediatr Dermatol. 2009;26(4):373–80.
19. Lucky AW. A review of infantile and pediatric acne. Dermatology (Basel). 1998;
196(1):95–7.
20. De Raeve L, De Schepper J, Smitz J. Prepubertal acne: a cutaneous marker of androgen
excess? J Am Acad Dermatol. 1995;32(2 Pt 1):181–4.
21. Eichenfield LF, Fowler Jr JF, Friedlander SF, Levy ML, Webster GF. Diagnosis and evaluation
of acne. Semin Cutan Med Surg. 2010;29(2 Suppl 1):5–8.
22. Lucky AW. Hormonal correlates of acne and hirsutism. Am J Med. 1995;98(1A):89S–94S.
23. Iughetti L, Predieri B, Ferrari M, Gallo C, Livio L, Milioli S, et al. Diagnosis of central preco-
cious puberty: endocrine assessment. J Pediatr Endocrinol Metab. 2000;13 Suppl 1:709–15.
24. Ray WA, Federspiel CF, Schaffner W. Prescribing of tetracycline to children less than 8 years
old. A two-year epidemiologic study among ambulatory Tennessee medicaid recipients.
JAMA. 1977;237(19):2069–74.
25. Appelbaum H, Malhotra S. A comprehensive approach to the spectrum of abnormal pubertal
development. Adolesc Med State Art Rev. 2012;23(1):1–14.
Part VII
Drug-Induced Acneiform Eruptions
Chapter 54
Drug-Induced Acneiform Eruptions
54.1 Introduction
54.2.1 Hormones
54.2.1.1 Corticosteroids
Systemic corticosteroids causing acneiform eruption were first reported in 1950s [1].
Exposure to high levels of systemic (oral [2] or intravenous [3]), topical [4, 5], and
inhaled [6–8] corticosteroids can induce or exacerbate acne. Perioral (periorificial)
with injectable testosterone for premature closure of epiphyseal growth zones had
an increased acne incidence [18]. There is also an increase in acne incidence in
young athletes who take anabolic-androgenic steroid to increase muscle mass
(“body builder acne” or “doping acne”) [19–21].
54.2.1.3 Danazol
54.2.2.2 Lithium
The first few cases linking lithium to causing acneiform eruption appeared in the
early 1970s [29–31]. Lithium triggers neutrophilic cutaneous conditions such as
neutrophilic folliculitis, acneiform, and psoriasiform eruptions [32–34]. Acne
induced by lithium occurs more frequently in males and patients with an allergic
(atopic) history. Inflammatory papules and pustules are distributed on face, axillae,
54 Drug-Induced Acneiform Eruptions 393
groin, arms, and buttocks. There is no direct correlation between lithium dose and
acne appearance or severity. However, high level of lithium can be detected in the
affected areas.
54.2.2.3 Antiepileptics
Acneiform eruptions are becoming a hallmark side effect for several therapies that
target a specific key molecule involved in the pathophysiology of the disease. This
therapeutic group of targeted therapies includes epidermal growth factor receptor
(EGFR) inhibitors (gefitinib [42], erlotinib [43, 44], and imatinib [45]), EGFR
monoclonal antibodies (i.e., cetuximab [46, 47] and panitumumab [48]), TNF
receptor inhibitors [49] (i.e., infliximab and lenalidomide), and BRAF-V600E
inhibitors.
EGFRs belong to the tyrosine kinase family and are thought to play a crucial role in
the development and progression of cancer. Several solid tumors of the head and
neck, breast, lungs, ovary, prostate, and colon overexpress EGFRs. Consequently,
EGFR inhibitors are used to treat these cancers.
394 H.K. Do et al.
Tumor necrosis factor (TNF) inhibitors are used to treat a wide range of autoim-
mune diseases such as inflammatory bowel disease, rheumatoid arthritis, psoriasis
and psoriatic arthritis, and ankylosing spondylitis. Of the TNF inhibitors, infliximab
is the most reported agent associated with an acneiform eruption [56]. Likewise,
lenalidomide, a second generation of thalidomide, has major TNF inhibitory activ-
ity. Lenalidomide-induced acute acneiform eruption in a multiple myeloma patient
has been reported [57]. The acneiform eruption was cleared with oral doxycycline
100 mg daily after 3 months without stopping lenalidomide therapy.
54 Drug-Induced Acneiform Eruptions 395
54.2.3.3 G-CSF
54.2.3.4 Vemurafenib
Vemurafenib, a BRAF (V600E) inhibitor, was FDA approved in January 2012 for
the treatment of advanced metastatic melanoma. Among the most common cutane-
ous adverse effects associated with vemurafenib are verrucous papillomas and
hand-foot syndrome, along with keratoacanthomas and squamous cell carcinomas.
More recently, a case report of acneiform eruption associated with vemurafenib has
been published [59].
54.2.4 Retinoids
In the late 1980s, several case reports appeared in the medical literature linking
acneiform eruption with oral etretinate [60, 61]. These patients developed acute
onset of severe cystic acne within the first few weeks after the initiation of oral
etretinate for psoriasis treatment. The acneiform eruption gradually improved when
etretinate was discontinued and with using topical erythromycin or oral tetracy-
clines. Etretinate is no longer available in the United States.
An acneiform eruption has been reported in a young adult who started propranolol
for migraine prophylaxis [62]. The lesions completely resolved after propranolol
was discontinued. These lesions recurred within 3 weeks when the patient was
started on nadolol for migraine prophylaxis. Facial acne resolved soon after nadolol
was discontinued. In general, cutaneous adverse effects from beta-blockers class are
rare. The exact pathogenesis linking acneiform eruptions with beta-blockers is still
unclear.
In 1981, the case of a 57-year-old man who develop papulopustules on the chest
and back soon after initiating quinidine for treatment of premature ventricular con-
tractions was reported [63]. The acneiform eruption responded well to topical eryth-
romycin lotion and topical benzoyl peroxide.
396 H.K. Do et al.
54.2.6.1 Sirolimus
54.2.6.2 Tacrolimus
54.2.6.3 Cyclosporine
54.2.6.4 Azathioprine
54.2.7.1 Gold
Gold has been reported to be associated with acne. In one case report, a patient with
rheumatoid arthritis first developed an eruption consistent with lichen planus and
subsequently developed acneiform lesions on the face and trunk after gold sodium
thiomalate treatment [80]. Gold is retained in the body for a prolonged duration and
is especially bound in the kidneys, liver, and skin [80].
54.2.7.2 Dactinomycin
Dactinomycin, used in the treatment of solid tumors, has been associated with occa-
sional development of an acneiform eruption. A case was reported of a prepubertal
girl who received dactinomycin in combination with other chemotherapeutic agents
(vincristine and cyclophosphamide), none of which have been implicated in the
development of acne lesions [81]. A rise and fall of serum androstenedione, dehy-
droepiandrosterone, and testosterone levels over a period of time, defined by two
courses of therapy inclusive of dactinomycin, was documented. Gradual improve-
ment of the acneiform eruption was also noted as hormone levels diminished, which
supported a relationship between drug exposure, the presence of the eruption, and
the tested hormone levels [82].
54.2.7.3 Isoniazid
54.2.7.4 Rifampin
54.2.7.5 Dapsone
Dapsone inhibits bacterial synthesis of dihydrofolic acid via competition with para-
aminobenzoate for the active site of dihydropteroate synthetase [86]. A young
female with preexisting mild facial acne vulgaris was treated with oral dapsone fol-
lowing poor response to oral tetracyclines [87]. She soon developed acne fulminans
and hemolysis which resolved with prompt administration of high doses of vitamin
C, oral prednisone, and intravenous methylene blue [87].
54.2.7.6 Halogens
Acneiform eruptions may originate from skin exposure to various industrial chemi-
cals, such as fumes generated in the manufacture of chlorine and its byproducts.
These chlorinated hydrocarbons may cause chloracne, consisting of cysts, pustules,
folliculitis, and comedones. The most potent acneiform-inducing agents are the
polyhalogenated hydrocarbons, notably dioxin (2,3,7,8 tetrachlorodibenzodioxin)
[88]. Notoriously difficult to treat, chloracne can persist for extended time periods
without known additional exposure to chloracnegens [89]. The most acne-prone
locations to “chloracnegens” are the malar crescent (inferior and lateral to the eye)
and the postauricular region. The genitalia, both penis and scrotum, are also vulner-
able anatomic regions. If sufficient exposure has occurred, lesions may involve the
shoulders, chest, back, and, eventually, the buttocks and abdomen. The axillary
regions have been commonly involved only in those patients who have ingested or
inhaled the chloracnegens as the sole or major route of exposure. Cutting and lubri-
cating oils, crude coal tar applied to the skin for medicinal purposes, heavy tar distil-
lates, coal tar pitch, and asbestos are known to cause acneiform eruptions. Acne
venenata is another term applied to this process [90].
Inflammatory acneiform flares have been reported in association with ingestion
of iodides and bromides [91]. Common sources of halogens include some thyroid
medications, expectorants containing potassium iodide, radiographic contrast
media, iodized salt, vitamin and mineral preparations, and some sedatives of histori-
cal interest only. Stimulation of neutrophil function has been suggested, although
the pathogenesis of this reaction is unknown. Initial lesions are often follicular pus-
tules, and later, comedonal lesions can emerge, possibly related to a hyperkeratotic
reaction to chronic inflammation [92].
54 Drug-Induced Acneiform Eruptions 399
Megadoses of vitamins B6 and B12 have been reported to induce a facial acneiform
eruption [93]. The histology shows parakeratosis overlying a focally spongiotic epi-
dermis with a mononuclear, perivascular inflammatory infiltrate in the papillary der-
mis. There was dramatic improvement in the acneiform lesions upon discontinue of
the nutritional supplement [93].
Exacerbation or onset of inflammatory acne lesions correlated with vitamins B2
(riboflavin), B6 (pyridoxine), and B12 (cyanocobalamin) intake has been reported
in the European literature [93]. This manifests as an exacerbation of preexisting
acne vulgaris with onset of multiple papules and papulopustules or as an explosive
facial pustular eruption. The pathogenesis is unknown, although it has been postu-
lated that the origin of B6/B12-induced acne may be similar to that of INH-induced
acne [93, 94].
Most uses for petroleum jelly exploit its lubricating, coating, and moisturizing
potentials. White petrolatum-associated unilateral acne developed in a young
woman attempting to relieve the effects of unilateral facial paralysis (Bell’s palsy)
by massaging the substance onto her face nightly [95]. While generally thought to
be noncomedogenic and safe to use in acne-prone skin, its occlusive properties
could induce a pustular reaction in selected individuals [95].
Application of cow udder ointment used for treatment of atopic eczema and psoria-
sis was associated with development of widespread and atypical acneiform erup-
tions in two patients [96]. Cow udder ointment contains boric acid and starch in a
wax and oil base.
54.2.7.11 Dantrolene
54.3 Conclusion
Acne vulgaris and acneiform drug eruptions can easily be overlooked in daily clini-
cal practice. There are no specific diagnostic criteria to define drug-induced acne.
However, several key clinical characteristics (Table 54.2) may help clinicians to
recognize and diagnose acneiform drug eruptions. Two useful clinical keys to help
diagnose acneiform drug eruptions are the temporal relationship (acne-like eruption
after introduction of a new medication) and the monomorphic lesions (all lesions
are in the same stage and lacking comedones).
References
1. Brunner MJ, Riddell Jr JM, Best WR. Cutaneous side effects of ACTH cortisone and preg-
nenolone therapy. J invest Dermatol. 1951;16:205–10.
2. Clementson B, Smidt AC. Periorificial dermatitis due to systemic corticosteroids in children:
report of two cases. Pediatric Dermatol. 2012;29:331–2.
3. Fung MA, Berger TG. A prospective study of acute-onset steroid acne associated with
administration of intravenous corticosteroids. Dermatology. 2000;200:43–4.
4. Plewig G, Kligman AM. Induction of acne by topical steroids. Arch Dermatol Forsch.
1973;247:29–52.
5. Hengge UR, Ruzicka T, Schwartz RA, Cork MJ. Adverse effects of topical glucocorticoste-
roids. J Am Acad Dermatol. 2006;54:1–15. quiz 6–8.
6. Monk B, Cunliffe WJ, Layton AM, Rhodes DJ. Acne induced by inhaled corticosteroids.
Clin Exp Dermatol. 1993;18:148–50.
7. Hughes JR, Higgins EM, du Vivier AW. Acne associated with inhaled glucocorticosteroids.
BMJ. 1992;305:1000.
8. Guillot B. Adverse skin reactions to inhaled corticosteroids. Exp Opin Drug Saf. 2002;
1:325–9.
9. Cohen HJ. Perioral dermatitis. J Am Acad Dermatol. 1981;4:739–40.
10. Wells K, Brodell RT. Topical corticosteroid “addiction”. A cause of perioral dermatitis.
Postgrad Med. 1993;93:225–30.
11. Hurwitz RM. Steroid acne. J Am Acad Dermatol. 1989;21:1179–81.
12. Bedane C, Souyri N. Induced acne. Ann Dermatol Venereol. 1990;117:53–8.
13. Gloor M, Funder H, Franke M. Effect of topical application of dexamethasone on
Propionibacteria in the pilosebaceous duct. Eur J Clin Pharmacol. 1978;14:53–6.
14. Gloor M, Mildenberger KH. On the influence of an external therapy with dexamethasone-21-
sodium-m-sulfobenzoate on the amount of free fatty acids in the skin surface lipids. Arch
Dermatol Res. 1978;261:33–8.
15. Shibata M, Katsuyama M, Onodera T, Ehama R, Hosoi J, Tagami H. Glucocorticoids enhance
Toll-like receptor 2 expression in human keratinocytes stimulated with Propionibacterium
acnes or proinflammatory cytokines. J Invest Dermatol. 2009;129:375–82.
16. Scott III MJ, Scott AM. Effects of anabolic-androgenic steroids on the pilosebaceous unit.
Cutis. 1992;50:113–6.
17. Scott Jr MJ, Scott 3rd MJ. Dermatologists and anabolic-androgenic drug abuse. Cutis. 1989;
44:30–5.
18. Fyrand O, Fiskaadal HJ, Trygstad O. Acne in pubertal boys undergoing treatment with andro-
gens. Acta Derm Venereol. 1992;72:148–9.
19. Heydenreich G. Testosterone and anabolic steroids and acne fulminans. Arch Dermatol.
1989;125:571–2.
20. Melnik B, Jansen T, Grabbe S. Abuse of anabolic-androgenic steroids and bodybuilding
acne: an underestimated health problem. J Dtsch Dermatol Ges. 2007;5:110–7.
21. Gerber PA, Kukova G, Meller S, Neumann NJ, Homey B. The dire consequences of doping.
Lancet. 2008;372:656.
22. Greenberg RD. Acne vulgaris associated with antigonadotropic (Danazol) therapy. Cutis.
1979;24:431–3.
402 H.K. Do et al.
23. Ilse JR, Greenberg HL, Bennett DD. Levonorgestrel-releasing intrauterine system and
new-onset acne. Cutis. 2008;82:158.
24. Olsson SE, Odlind V, Johansson E. Androgen levels in women using Norplant implants.
Contraception. 1986;34:157–67.
25. Grupper C. New iatrogenic acne: acne caused by amineptin (Survector). Ann Dermatol
Venereol. 1988;115:1174–6.
26. Thioly-Bensoussan D, Charpentier A, Triller R, et al. Iatrogenic acne caused by amineptin
(Survector). Apropos of 8 cases. Ann Dermatol Venereol. 1988;115:1177–80.
27. Vexiau P, Gourmel B, Husson C, et al. Severe lesions of acne type induced by chronic
amineptin poisoning: apropos of 6 cases. Ann Dermatol Venereol. 1988;115:1180–2.
28. Ponte CD. Maprotiline-induced acne. Am J Psychiatry. 1982;139:141.
29. Kusumi Y. A cutaneous side effect of lithium: report of two cases. Dis Nerv Syst.
1971;32:853–4.
30. Ruiz-Maldonado R, Perez de Francisco C, Tamayo L. Lithium dermatitis. JAMA. 1973;224:
1534.
31. Yoder FW. Letter: acneiform eruption due to lithium carbonate. Arch Dermatol. 1975;111:
396–7.
32. Rifkin A, Kurtin SB, Quitkin F, Klein DF. Lithium-induced folliculitis. Am J Psychiatry.
1973;130:1018–9.
33. Heng MC. Lithium carbonate toxicity. Acneiform eruption and other manifestation. Arch
Dermatol. 1982;118:246–8.
34. Heng MC. Cutaneous manifestations of lithium toxicity. Br J Dermatol. 1982;106:107–9.
35. Hesse S, Berbis P, Lafforgue P, Acquaviva PC, Pastor MJ, Privat Y. Acne and enthesiopathy
during anti-epileptic treatment. Ann Dermatol Venereol. 1992;119:655–8.
36. Jenkins RB, Ratner AC. Diphenylhydantoin and acne. New Engl J Med. 1972;287:148.
37. Nielsen JN, Licht RW, Fogh K. Two cases of acneiform eruption associated with lamotrigine.
J Clin Psychiatry. 2004;65:1720–2.
38. Joffe H, Cohen LS, Suppes T, et al. Valproate is associated with new-onset oligoamenorrhea
with hyperandrogenism in women with bipolar disorder. Biol Psychiatry. 2006;59:1078–86.
39. Stankler L, Campbell AG. Neonatal acne vulgaris: a possible feature of the fetal hydantoin
syndrome. Br J Dermatol. 1980;103:453–5.
40. Norris JF, Cunliffe WJ. Phenytoin-induced gum hypertrophy improved by isotretinoin. Int J
Dermatol. 1987;26:602–3.
41. Mishra B, Praharaj SK, Prakash R, Sinha VK. Aripiprazole-induced acneiform eruption. Gen
Hosp Psychiatry. 2008;30:479–81.
42. Warthan MM, Jumper CA, Smith JL. Acneiform eruption induced by Iressa (gefitinib) tablets
used to treat non-small cell lung cancer. J Drugs Dermatol. 2004;3:569–70.
43. Schalock PC, Zug KA. Acneiform reaction to erlotinib. Dermatitis. 2007;18:230–1.
44. Kardaun SH, van Duinen KF. Erlotinib-induced florid acneiform rash complicated by exten-
sive impetiginization. Clin Exp Dermatol. 2008;33:46–9.
45. Martin JM, Jorda E, Monteagudo C, Alonso V, Garcia L, Villalon G. Follicular acneiform
eruption induced by imatinib. J Eur Acad Dermatol Venereol. 2006;20:1368–70.
46. Lee JE, Lee SJ, Lee HJ, Lee JH, Lee KH. Severe acneiform eruption induced by cetuximab
(Erbitux). Yonsei Med J. 2008;49:851–2.
47. Molinari E, De Quatrebarbes J, Andre T, Aractingi S. Cetuximab-induced acne. Dermatology.
2005;211:330–3.
48. Rowinsky EK, Schwartz GH, Gollob JA, et al. Safety, pharmacokinetics, and activity of
ABX-EGF, a fully human anti-epidermal growth factor receptor monoclonal antibody in
patients with metastatic renal cell cancer. J Clin Oncol. 2004;22:3003–15.
49. Sun G, Wasko CA, Hsu S. Acneiform eruption following anti-TNF-alpha treatment: a report
of three cases. J Drugs Dermatol. 2008;7:69–71.
50. Jacot W, Bessis D, Jorda E, et al. Acneiform eruption induced by epidermal growth factor
receptor inhibitors in patients with solid tumours. Br J Dermatol. 2004;151:238–41.
54 Drug-Induced Acneiform Eruptions 403
51. DeWitt CA, Siroy AE, Stone SP. Acneiform eruptions associated with epidermal growth
factor receptor-targeted chemotherapy. J Am Acad Dermatol. 2007;56:500–5.
52. Eames T, Landthaler M, Karrer S. Severe acneiform skin reaction during therapy with
erlotinib (Tarceva), an epidermal growth factor receptor (EGFR) inhibitor. Eur J Dermatol.
2007;17:552–3.
53. Journagan S, Obadiah J. An acneiform eruption due to erlotinib: prognostic implications and
management. J Am Acad Dermatol. 2006;54:358–60.
54. Perez-Soler R, Saltz L. Cutaneous adverse effects with HER1/EGFR-targeted agents: is there
a silver lining? J Clin Oncol. 2005;23:5235–46.
55. Segaert S, Van Cutsem E. Clinical signs, pathophysiology and management of skin toxicity
during therapy with epidermal growth factor receptor inhibitors. Ann Oncol. 2005;16:
1425–33.
56. Sladden MJ, Clarke PJ, Mitchell B. Infliximab-induced acne: report of a third case. Br J
Dermatol. 2008;158:172.
57. Michot C, Guillot B, Dereure O. Lenalidomide-induced acute acneiform folliculitis of the
head and neck: not only the anti-EGF receptor agents. Dermatology. 2010;220:49–50.
58. Lee PK, Dover JS. Recurrent exacerbation of acne by granulocyte colony-stimulating factor
administration. J Am Acad Dermatol. 1996;34:855–6.
59. Petukhova TA, Novoa RA, Honda K, Koon HB, Gerstenblith MR. Acneiform eruptions asso-
ciated with vemurafenib. J Am Acad Dermatol. 2013;68:e97–9.
60. Lowe L, Herbert AA. Cystic and comedonal acne: a side effect of etretinate therapy. Int J
Dermatol. 1989;28:482.
61. Menter A, Boyd A. Cystic acne in psoriatic patients undergoing etretinate therapy. J Am
Acad Dermatol. 1988;18:751–2.
62. Bajwa ZH, Sami N, Flory C. Severe acne as a side effect of propranolol and nadolol in a
migraineur. Headache. 1999;39:758–60.
63. Burkhart CG. Quinidine-induced acne. Arch Dermatol. 1981;117:603–4.
64. Fidan K, Kandur Y, Sozen H, Gonul II, Dalgic A, Soylemezoglu O. How often do we face
side effects of sirolimus in pediatric renal transplantation? Transplant Proc. 2013;45:185–9.
65. Kunzle N, Venetz JP, Pascual M, Panizzon RG, Laffitte E. Sirolimus-induced acneiform erup-
tion. Dermatology. 2005;211:366–9.
66. Lubbe J, Sorg O, Male PJ, Saurat JH, Masouye I. Sirolimus-induced inflammatory papules
with acquired reactive perforating collagenosis. Dermatology. 2008;216:239–42.
67. Momin SB, Peterson A, Del Rosso JQ. A status report on drug-associated acne and acneiform
eruptions. J Drugs Dermatol. 2010;9:627–36.
68. Mahe E, Morelon E, Lechaton S, et al. Acne in recipients of renal transplantation treated with
sirolimus: clinical, microbiologic, histologic, therapeutic, and pathogenic aspects. J Am
Acad Dermatol. 2006;55:139–42.
69. Mahe E, Morelon E, Lechaton S, et al. Cutaneous adverse events in renal transplant recipients
receiving sirolimus-based therapy. Transplantation. 2005;79:476–82.
70. Bakos L, Bakos RM. Focal acne during topical tacrolimus therapy for vitiligo. Arch Dermatol.
2007;143:1223–4.
71. Antille C, Saurat JH, Lubbe J. Induction of rosaceiform dermatitis during treatment of facial
inflammatory dermatoses with tacrolimus ointment. Arch Dermatol. 2004;140:457–60.
72. El Sayed F, Ammoury A, Dhaybi R, Bazex J. Rosaceiform eruption to pimecrolimus. J Am
Acad Dermatol. 2006;54:548–50.
73. el-Shahawy MA, Gadallah MF, Massry SG. Acne: a potential side effect of cyclosporine A
therapy. Nephron. 1996;72:679–82.
74. Azurdia RM, Graham RM, Weismann K, Guerin DM, Parslew R. Acne keloidalis in cauca-
sian patients on cyclosporin following organ transplantation. Br J Dermatol. 2000;143:
465–7.
75. Bencini PL, Crosti C, Sala F, et al. Gingival hyperplasia by nifedipine. Report of a case. Acta
Derm Venereol. 1985;65:362–5.
404 H.K. Do et al.
A etiology, 117
Abnormal keratinization, 8, 9 etiopathogenesis, 118
Acne conglobata immune complexes, 118
atrophic and hypertrophic scars, 108 immunofluorescence testing, 121
chromosomal defects, 107–108 laboratory findings, 119
cryotherapy, nodules, 109 Propionibacterium acnes, 118
environmental factors, 107 rheumatoid factor and ANA tests, 121
fulminans, 107 SAPHO syndrome, 117
inflammatory acne, 107 siblings, 118
isotretinoin, 109 treatment, 121–122
musculoskeletal syndrome, 108 Acneiform eruptions
renal amyloidosis, 108 drug-induced (see Drugs)
work-up, 109 PAPA syndrome (see Pyogenic sterile
Acne excoriée arthritis, pyoderma gangrenosum
characterization, 111 and acne (PAPA) syndrome)
clinical presentation, 113 Acne mechanica
facial acne excoriation, 111, 112 clinical presentation, 126
OCD patients, 114 description, 125
patients, 111–112 diagnosis, 126
psychiatric problems, 111 NIOSH workshop, 125
psychogenic excoriation, 112 rubbing, 125, 126
psychopathology, 113 sealed acne-bearing skin, 126
scar formation, 112 surface skin architecture, 125–126
subjective and physiological treatment, 127
symptoms, 114 Acne scarring
treatment, 114–115 atrophic scars, 238–239
Acne fulminans chemical peels, 241
acne vulgaris, 117 dermal fillers, 241
adolescent boys, 118 fibroblasts, 241
androgenic steroids, 118 FP, 242
antistaphylococcal and antistreptococcal hypertrophic scars, 240
antibody, 121 inflammatory lesions, 237
bacterial cultures, 121 keloids scars, 240
bone, 121 laser resurfacing, 242
Caucasian males, 118 MMP inhibitors, 237
clinical presentation, 118–120 morphologies, 237, 238
description, 117 and pigmentation, 240