Download July/August 2010 • Volume 8 • Issue 4

Document related concepts

Adherence (medicine) wikipedia , lookup

Sjögren syndrome wikipedia , lookup

Management of multiple sclerosis wikipedia , lookup

Multiple sclerosis research wikipedia , lookup

Transcript
July/August 2010 • Volume 8 • Issue 4
EDITORIAL
Updating the Dermatologic Nomenclature:
Names That Are Good or Bad
LETTER TO THE EDITOR
Prolactinoma Can Be Associated With Gynecomastia
Cohen, Robinson, and Gray
Parish and Witkowski
ORIGINAL CONTRIBUTIONS
Serum Lipid Level in Iraqi Patients With Psoriasis
Aldhalimi, Almuhanna, and Alrikabi
Clinical Features and Treatment of
Dermatosis Papulosa Nigra in Migrants to Italy
Calcaterra, Franco, Valenzano, Fazio, and Morrone
DEPARTMENTS
New Therapy Update
Zyclara (Imiquimod) Cream, 3.75%
Gupta, Cooper, and Abramovits
Perils of Dermatopathology
Newtonian Dermatopathology:
When the Reaction to a Lesion Obfuscates the Diagnosis
Rojas, Sarkissian, Heller, and Lambert
REVIEWS
Acute Generalized Exanthematous Pustulosis
Pecina and Cappel
Sporotrichosis: Part I
Schechtman
COMMENTARIES
Sentinel Lymph Node Biopsy in Melanoma:
The Gulf Between Presentation and Reality
Thomas
Sentinel Lymph Node Biopsy in Melanoma:
Coping With Incomplete Information;
Call for Further Investigations
Lambert
Congress Report
Highlights From the 7th EADV Spring Symposium,
Cavtat, Croatia, May 13–16, 2010
Lipozenčić
CASE STUDIES
Segmental Cutaneous Piloleiomyomata
Cook-Norris, Rodriguez, Kovach, Boyd, and Zic
Vulvoperineal Crohn’s Disease:
Response to Metronidazole
Khaled, Ezzine-Sebai, Fazaa, Zeglaoui, Zermani, and Kamoun
Folliculocentric Lichen Sclerosus et Atrophicus
Mann, Vergilis-Kalner, Wasserman, and Petronic-Rosic
Poorly Growing Hair in a Child
Bolotin, Ortel, and Stein
When she asks about her scar,
recommend Mederma® with confidence.
In a recent clinical study,1 77% of subjects noted moderate
to significant improvement in scars treated with
Mederma® in terms of redness, texture and
overall appearance. 98% of patients completed
the study without any adverse events.
1 Draelos, Z., The ability of
onion extract gel to improve
the cosmetic appearance of
postsurgical scars, Cosmetic
Dermatology, June 2008.
2 IMS Health, NDTI, December
2008. 3 Pharmacy Times, OTC
Supplement, June 2009.
The #1 doctor-2 and pharmacist-recommended3 brand for
scars helps empower your patients to take charge of their
appearance so they can get back their confidence.
And you get a very satisfied patient.
Available for office dispensing.
mederma.com
888.925.8989
TABLE OF CONTENTS
July/August 2010 • Volume 8 • Issue 4
EDITORIAL
Updating the Dermatologic Nomenclature: Names That Are Good or Bad....................................................... 199
Lawrence Charles Parish, MD, MD (Hon); Joseph A. Witkowski, MD
LETTER TO THE EDITOR
Prolactinoma Can Be Associated With Gynecomastia.................................................................................... 201
Philip R. Cohen, MD; Floyd W. Robinson, BS; James M. Gray, MD
ORIGINAL CONTRIBUTIONS
Serum Lipid Level in Iraqi Patients With Psoriasis.......................................................................................... 204
Muhsin A. Aldhalimi, CABDV; Sadiq J. Almuhanna, FICMS; Samir H. Alrikabi, MSc
Clinical Features and Treatment of Dermatosis Papulosa Nigra in Migrants to Italy........................................ 207
Roberta Calcaterra, MD; Gennaro Franco, MD; Mariacarla Valenzano, MD; Raffaella Fazio, MD; Aldo Morrone, MD
REVIEWS
Acute Generalized Exanthematous Pustulosis............................................................................................... 210
Jennifer L. Pecina, MD; Mark A. Cappel, MD
Sporotrichosis: Part I................................................................................................................................... 216
Regina Casz Schechtman, MD, PhD
Self-Test Review Questions (p. 221)
COMMENTARIES
Sentinel Lymph Node Biopsy in Melanoma: The Gulf Between Presentation and Reality.................................. 222
J. Meirion Thomas, MS, FRCP, FRCS
Sentinel Lymph Node Biopsy in Melanoma:
Coping With Incomplete Information; Call for Further Investigations............................................................... 226
W. Clark Lambert, MD, PhD
Departments
New Therapy Update
William Abramovits, MD; Aditya K. Gupta, MD, Section Editors
Zyclara (Imiquimod) Cream, 3.75%.............................................................................................................. 227
Aditya K. Gupta, MD; Elizabeth A. Cooper, HBSc; William Abramovits, MD
Perils of Dermatopathology
W. Clark Lambert, MD, PhD, Editor
Newtonian Dermatopathology: When the Reaction to a Lesion Obfuscates the Diagnosis................................ 231
Javier Rojas, MD; Navér Sarkissian, MD, PhD; Debra S. Heller, MD; W. Clark Lambert, MD, PhD
193
July/August 2010
TABLE OF CONTENTS
Departments (continued)
Congress Report
Marcia Ramos-e-Silva, MD, PhD, Section Editor
Highlights From the 7th EADV Spring Symposium, Cavtat, Croatia, May 13–16, 2010.................................... 235
Jasna Lipozenčić, MD, PhD, Chairperson
CASE STUDIES
Vesna Petronic-Rosic, MD, MSc, Section Editor
Segmental Cutaneous Piloleiomyomata........................................................................................................ 238
Robert H. Cook-Norris, MD; Adrian O. Rodriguez, MD; Bradley T. Kovach, MD; Alan S. Boyd, MD; John A. Zic, MD
Vulvoperineal Crohn’s Disease: Response to Metronidazole........................................................................... 240
Aida Khaled, MD; Nadia Ezzine-Sebai, MD; Becima Fazaa, MD; Faten Zeglaoui, MD; Rachida Zermani, MD; Mohamed Ridha Kamoun, MD
Folliculocentric Lichen Sclerosus et Atrophicus............................................................................................. 242
David J. Mann, MD; Irene J. Vergilis-Kalner, MD; Justin R. Wasserman, MD; Vesna Petronic-Rosic, MD, MSc
Poorly Growing Hair in a Child...................................................................................................................... 246
Diana Bolotin, MD, PhD; Bernhard Ortel, MD; Sarah L. Stein, MD
VINTAGE LABEL
Courtesy of BuyEnlarge, Philadelphia, PA
194
For your patients with tinea pedis, tinea cruris, and tinea corporis
For the fungicidal power you trust, turn to Naftin® (naftifine HCl 1%).
Designed to suit a range of patients and preferences, Naftin® delivers
power against tinea pedis, cruris and corporis. Choose Naftin® Gel,
Naftin® Cream, or the innovative Naftin® Cream Pump and give your
patients the customized care they deserve.
Naftin® (naftifine HCl 1%) Cream and Gel are indicated for the topical treatment of tinea pedis,
tinea cruris and tinea corporis caused by Trichophyton rubrum, Trichophyton mentagrophytes,
Epidermophyton floccosum and Trichophyton tonsurans (Gel only).
Naftin® Cream and Gel are contraindicated in individuals who have shown hypersensitivity
to any of their components and are for topical use only.
During clinical trials with Naftin® Cream and Gel, the following side effects were most commonly
reported: burning/stinging, dryness, erythema, itching, local irritation, skin tenderness and rash.
Please see brief summary on the following page.
©2010 Merz Pharmaceuticals
All rights reserved.
5/10
Power made personal
July/August 2010
Volume 8 • Issue 4
ABOUT OUR JOURNAL
SKINmed: Dermatology for the Clinician®, print ISSN 1540-9740, online ISSN
1751-7125, is published bimonthly by Pulse Marketing & Communications,
LLC, located at 4 Peninsula Avenue, Sea Bright, NJ 07760.
BRIEF SUMMARY
Rx ONLY
INDICATIONS AND USAGE: Naftin® Cream, 1% is
indicated for the topical treatment of tinea pedis, tinea
cruris, and tinea corporis caused by the organisms
Trichophyton rubrum, Trichophyton mentagrophytes,
and Epidermophyton floccosum. Naftin® Gel, 1% is
indicated for the topical treatment of tinea pedis, tinea
cruris, and tinea corporis caused by the organisms
Trichophyton rubrum, Trichophyton mentagrophytes,
Trichophyton tonsurans*, Epidermophyton floccosum*.
*Efficacy for this organism in this organ system was
studied in fewer than 10 infections.
CONTRAINDICATIONS: Naftin® Cream and Gel, 1%
are contraindicated in individuals who have shown
hypersensitivity to any of their components.
WARNINGS: Naftin® Cream and Gel, 1% are for
topical use only and not for ophthalmic use.
PRECAUTIONS: General: Naftin® Cream and
Gel, 1%, are for external use only. If irritation or sensitivity develops with the use of Naftin® Cream or Gel,
1%, treatment should be discontinued and appropriate
therapy instituted. Diagnosis of the disease should be
confirmed either by direct microscopic examination of
a mounting of infected tissue in a solution of potassium hydroxide or by culture on an appropriate medium.
Information for patients: The patient should be told to:
1. Avoid the use of occlusive dressings or wrappings
unless otherwise directed by the physician.
2. Keep Naftin® Cream and Gel, 1% away from the
eyes, nose, mouth and other mucous membranes.
Carcinogenesis, mutagenesis, impairment of fertility:
Long-term studies to evaluate the carcinogenic
potential of Naftin® Cream and Gel, 1% have not been
performed. In vitro and animal studies have not
demonstrated any mutagenic effect or effect on fertility.
Printed in the USA.
Disclaimer: The Publisher, Editors, and Editorial Board cannot be held responsible for errors or any consequences arising from the use of information contained
in this journal; the views and opinions expressed herein do not necessarily reflect
those of the Publisher, Editors, and Editorial Board, neither does the publication
of advertisements constitute any endorsement by the Publisher, Editors, and Editorial Board of the products or services advertised. The Publisher, Editors, Editorial
Board, Reviewers, Authors, and Affiliated Agents shall not be held responsible or
in any way liable for the continued accuracy of the information or for any errors,
inaccuracies, or omissions of any kind in this publication, whether arising from
negligence or otherwise, or for any consequences arising thereafter.
Copyright: ©2010 Pulse Marketing & Communications, LLC. All rights reserved.
No part of this publication may be reproduced, stored, or transmitted in any form or
by any means without the prior permission in writing from the Publisher. Requests
should be addressed to the Permissions Editor at: Pulse Marketing & Communications, LLC, 4 Peninsula Avenue, Sea Bright, NJ 07760.
Abstracting & Indexing: The journal is indexed in Index Medicus/MEDLINE.
Editorial
MANAGING EDITOR
Sarah D. Staats
[email protected]
PRODUCTION DIRECTOR
Scott C. Bouchard
Distinct Layouts, LLC
www.distinctlayouts.com
EDITORIAL DIRECTOR
Gila Berkowitz
[email protected]
ASSOCIATE MANAGING EDITOR
Elizabeth Holcomb
[email protected]
Pregnancy: Teratogenic Effects: Pregnancy
Category B: Reproduction studies have been performed
in rats and rabbits (via oral administration) at doses
150 times or more than the topical human dose and
have revealed no evidence of impaired fertility or harm
to the fetus due to naftifine. There are, however, no
adequate and well-controlled studies in pregnant
women. Because animal reproduction studies are not
always predictive of human response, this drug should
be used during pregnancy only if clearly needed.
Nursing mothers: It is not known whether this drug
is excreted in human milk. Because many drugs are
excreted in human milk, caution should be exercised
when Naftin® Cream or Gel,1% are administered to a
nursing woman.
Pediatric use: Safety and effectiveness in pediatric
patients have not been established.
Publishing
PUBLISHER
Art Kalaka
Corporate
President
Arthur Kalaka
[email protected]
skinmedbriefPI.indd 1
Chief Executive Officer
Jo-Ann Kalaka-Adams
[email protected]
General Counsel
Marianne Mckenzie
[email protected]
ADVERSE REACTIONS: During clinical trials with
Naftin® Cream, 1%, the incidence of adverse reactions
was as follows: burning/stinging (6%), dryness (3%)
erythema (2%), itching (2%), local irritation (2%).
During clinical trials with Naftin® Gel, 1%, the
incidence of adverse reactions was as follows:
burning/stinging (5.0%), itching (1.0%), erythema
(0.5%), rash (0.5%), skin tenderness (0.5%).
Manufactured for Merz Pharmaceuticals, Greensboro, NC 27410
© 2010 Merz Pharmaceuticals Rev 3/10
Associate Publisher
James R. Adams
[email protected]
Pulse Marketing & Communications, LLC
4 Peninsula Avenue • Suite 401 • Sea Bright, NJ 07760 • Tel (732) 747-6525 • Fax (732) 747-7010
6/11/10 12:05 PM
196
July/August 2010
EDITORIAL BOARD
EDITOR IN CHIEF
Lawrence Charles Parish, MD, MD (Hon)
Philadelphia, PA
DEPUTY EDITORS
William Abramovits, MD
Dallas, TX
Larry E. Millikan, MD
Meridian, MS
Jennifer L. Parish, MD
Philadelphia, PA
Marcia Ramos-e-Silva, MD, PhD
Rio de Janeiro, Brazil
EDITORIAL BOARD
Mohamed Amer, MD
Cairo, Egypt
Ibrahim Hassan Galadari, MD, PhD, FRCP
Dubai, United Arab Emirates
Jasna Lipozencic, MD, PhD
Zagreb, Croatia
Noah S. Scheinfeld, MD, JD
New York, NY
Robert L. Baran, MD
Cannes, France
Anthony A. Gaspari, MD
Baltimore, MD
George M. Martin, MD
Kihei, HI
Virendra N. Sehgal, MD
Delhi, India
Anthony V. Benedetto, DO
Philadelphia, PA
Michael Geiges, MD
Zurich, Switzerland
David I. McLean, MD
Vancouver, British Columbia
Charles Steffen, MD
Oceanside, CA
Brian Berman, MD, PhD
Miami, FL
Michael H. Gold, MD
Nashville, TN
Marc S. Micozzi, MD, PhD
Bethesda, MD
Alexander J. Stratigos, MD
Athens, Greece
Jack M. Bernstein, MD
Dayton, OH
Orin M. Goldblum, MD
Abbott Park, IL
George F. Murphy, MD
Boston, MA
James S. Studdiford III, MD
Philadelphia, PA
Sarah Brenner, MD
Tel Aviv, Israel
Lowell A. Goldsmith, MD, MPH
Chapel Hill, NC
Oumeish Youssef Oumeish, MD, FRCP
Amman, Jordan
Robert J. Thomsen, MD
Los Alamos, NM
Joaquin Calap Calatayud, MD
Cadiz, Spain
Aditya K. Gupta, MD, PhD, FRCP(C)
London, Ontario
Joseph L. Pace, MD, FRCP
Naxxar, Malta
Julian Trevino, MD
Dayton, OH
Henry H.L. Chan, MB, MD, PhD, FRCP
Hong Kong, China
Seung-Kyung Hann, MD, PhD
Seoul, Korea
Art Papier, MD
Rochester, NY
Snejina Vassileva, MD, PhD
Sofia, Bulgaria
Ncoza C. Dlova, MBChB, FCDerm
Durban, South Africa
Roderick J. Hay, BCh, DM, FRCP, FRCPath
London, UK
Vesna Petronic-Rosic, MD, MSc
Chicago, IL
Daniel Wallach, MD
Paris, France
Richard L. Dobson, MD
Mt Pleasant, SC
Tanya R. Humphreys, MD
Philadelphia, PA
Johannes Ring, MD, DPhil
Munich, Germany
Michael A. Waugh, MB, FRCP
Leeds, UK
William H. Eaglstein, MD
Palo Alto, CA
Camila K. Janniger, MD
Englewood, NJ
Roy S. Rogers III, MD
Rochester, MN
Wm. Philip Werschler, MD
Spokane, WA
Boni E. Elewski, MD
Birmingham, AL
Abdul-Ghani Kibbi, MD
Beirut, Lebanon
Donald Rudikoff, MD
New York, NY
Joseph A. Witkowski, MD
Philadelphia, PA
Charles N. Ellis, MD
Ann Arbor, MI
W. Clark Lambert, MD, PhD
Newark, NJ
Robert I. Rudolph, MD
Wyomissing, PA
Ronni Wolf, MD
Rechovot, Israel
Howard A. Epstein, PhD
Gibbstown, NJ
Andrew P. Lazar, MD
Highland Park, IL
Vincenzo Ruocco, MD
Naples, Italy
197
July/August 2010
Volume 8 • Issue 4
EDITORIAL
Updating the Dermatologic Nomenclature:
Names That Are Good or Bad
Lawrence Charles Parish, MD, MD (Hon);1 Joseph A. Witkowski, MD2
D
ermatology probably has the largest vocabulary of any
of the medical specialties,1 a statement to which we
can attest when we reviewed aspects of the Dermatology Lexicon project. Some terms are derived from Latin or Greek
and present spelling problems to all but a select few. Examples
include acrokeratosis verruciformis and pterigium. Other diseases
carry names that are such a mouthful that few can call them out
without the interruption of breathing. These might include erosio
interdigitalis blastomycetica and exudative discoid and lichenoid
chronic dermatosis of Sulzberger and Garbe. No wonder the former is now referred to simply as candidosis, or is it, candidiasis,
and the latter as oid-oid disease or Sulzberger-Garbe disease.
Mediocre Names
•Neurodermatitis denotes red scaling patches on the body, often with lichenification. Should there be history of allergic rhinitis or asthma, a new diagnosis might be used: atopic dermatitis. This has its problems, however, as the causative allergens
are rarely found, contrary to the case with hayfever or asthma.
Neurodermatitis is better than the synonym of lichen simplex
of Brocq, but, once again, the person is led astray into believing
that “nerves” or the proverbial stress is the etiologic agent.
•Lichen sclerosus atrophicus represents an atrophic process
that is often found on the female genitalia. Its male counterpart is called balnaitis xerotica. Do the red, scaling, and
atrophic lesions really need 9-syllable terminology, let alone
challenges to the spelling capabilities of the clinician?
Bad Names
Be that as it may, we would like to focus on disease names that
we should consider avoiding, because they unnecessarily connote ideas that are not germane to patient welfare. These diseases
can be easily called by other terms.
•Paget’s disease would seem to be a harmless eponym, honoring Sir William Paget who described:
oBone disease
oSquamous cell carcinoma in situ of the areola, being
more common in women
•Chondrodermatitis nodularis chronica helicis: This may sound
more important in its Latinate form; simplifying the diagnosis
to chondrodermatitis of the ear is much more understandable.
The cartilage can still be inflamed, and the annoying pain and
tenderness will exist with the simplified terminology.
•Senile keratoses: These represent the overproduction of
keratin due to excessive exposure to sunlight. While it
may take many years of such exposure, not every patient
with this possible premalignant condition is approaching
Alzheimer’s disease. Even knowing that the histopathology
demonstrates squamous cell carcinoma, grade 1 or 2, use of
the term actinic keratosis would be preferred.
oSquamous cell carcinoma in situ in areas other than the
breast (extramammary)
Using the word Paget then denotes 3 different diseases.
Were not the terms so well established, it might be more
appropriate to select nomenclature that was less confusing
and possibly more explicit.
Good Names
•Senile purpura: While nonpalpable purpura is often seen
in older patients, there is no reason to cast aspersions on
the chronologically challenged. Possibly, just calling the red
collections, often found on the arms, nonpalpable purpura
would suffice. Chronic purpura is just not helpful.
•Basal cell carcinoma is a very descriptive name, an improvement over basal cell epithelioma, and a significant change for
the better over rodent ulcer. The patient is anxious enough
over the possible scarring to be caused by the surgical removal
of this malignancy. There is no good reason to add to the
worry by suggesting that a rat created the problem.
•Acne represents a sebaceous gland disorder where many of
From the Departments of Dermatology and Cutaneous Biology, Jefferson Medical College of Thomas Jefferson University, Philadelphia, PA;1
and the Department of Dermatology, University of Pennsylvania School of Medicine, Philadelphia, PA2
Address for Correspondence: Lawrence Charles Parish, MD, MD (Hon), 1760 Market Street, Suite 301, Philadelphia, PA 19103 •
E-mail: [email protected]
SKINmed. 2010;8:199–200
199
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
EDITORIAL
the lesions can be pointed and so, the misconstruing of acne
to acme is not so terrible. Acne vulgaris is another mouthful
that is unnecessary to perpetuate, while acne rosacea does
not provide more information over the one-word, rosacea.
•Contact dermatitis says just what it means: a reaction due
to contact with a substance. Adding irritant or allergic to
the terminology may be informative, but equally so, causes
problems for the simplicity of the term contact dermatitis.
Conclusions
Perhaps, we have just scratched the surface. While we may not
have reformed dermatologic nomenclature, we have presented
some of our thoughts.
Reference
1 Leider M, Rosenblum M. A dictionary of dermatological words,
terms and phrases. New York, NY: Blakiston; 1968:440.
Wax Moulage
Kerion celsi, Trichophytia profunda capillitii. Moulage Nr. 218, made by Otto Vogelbacher ca. 1920 at the Dermatology
Clinic in Freiburg i.Br. (Germany). Museum of Wax Moulages Zurich, www.moulagen.ch
Courtesy of Michael Geiges, MD
SKINmed. 2010;8:199–200
200
Updating the Dermatologic Nomenclature
July/August 2010
Volume 8 • Issue 4
LETTER TO THE EDITOR
Prolactinoma Can Be Associated With Gynecomastia
Philip R. Cohen, MD;1,2,3 Floyd W. Robinson, BS;1 James M. Gray, MD1
To the Editor:
Dr Kapoor’s review provides an excellent summary of the cutaneous manifestations that characterize the systemic conditions
associated with gynecomastia. In addition, the pathogenesis of
gynecomastia in the included chronic metabolic disorders (cirrhosis and chronic renal failure), endocrine disorders (Graves’ disease
and hypopituitarism), systemic infections (leprosy and human
immunodeficiency virus), gastrointestinal malignancies, genetic
syndromes (lentiginosis syndrome and Klinefelter syndrome), and
alcohol abuse is discussed.1 We respectfully add prolactinoma to
the conditions that can be associated with gynecomastia.
Prolactinoma, the most common secretory pituitary tumor, can
be classified by tumor size: microadenoma (<10 mm), macroadenoma (>10 mm), and giant (a macroadenoma >40 mm).
Clinical symptoms are secondary to either tumor-associated hy-
perprolactinemia (gonadal and sexual dysfunction) or adenomarelated growth (visual field defects, cranial nerve palsies, and
headaches). Prolactinoma typically presents as a microadenoma
with amenorrhea, infertility, and galactorrhea in women.2,3
In men, since the prolactinoma is more often diagnosed as a macroadenoma, initial symptoms may be associated with the tumor’s
larger size: neurologic and vision dysfunction. Endocrine-related
symptoms from hyperprolactinemia (such as hypogonadism, infertility, and impotence with decreased libido and erectile dysfunction), however, can be the presenting finding or a subsequent manifestation in men. In addition, albeit less common, gynecomastia,
galactorrhea, and abnormally sparse body hair can occur.2–5
Prolactinoma-associated gynecomastia results from tumor-associated hyperprolactinemia. Prolactin inhibits the pulsatile secretion
Figure. (A) Distant and (B) closer views of the left chest of a 36-year-old Ukranian man who presented with unilateral galactorrhea
and fatigue. At 13 years of age, he had been exposed to radioactivity for 2 weeks following the Chernobyl nuclear power plant
reactor 4 explosion. White liquid, elicited by palpation of the breast, is visible on the left nipple and areola. A large intrasellar macroadenoma (2.6 × 2.7 × 2.5 cm), which neither compressed the optic apparatus nor invaded the cavernous sinuses, was detected
on the brain magnetic resonance imaging scan. His serum testosterone level was decreased (147 ng/dL, normal = 214–827 ng/
dL) and his serum prolactin level was markedly increased (3440.2 ng/mL, normal = 3.0–30.0 ng/mL); the latter observation confirming the suspected diagnosis of prolactinoma. Follow-up evaluation after 6 weeks of cabergoline treatment showed diminished
left nipple lactation and decreasing serum prolactin level (200 ng/mL).7,8
From the University of Houston Health Center, University of Houston;1 the Department of Dermatology, The University of Texas M.D. Anderson Cancer Center;2 and the Department of Dermatology, University of Texas-Houston Medical School,3 Houston, TX
Address for Correspondence: Philip R. Cohen, MD, 805 Anderson Street, Bellaire, TX 77401-2806 • E-mail: [email protected]
SKINmed. 2010;8:201–202
201
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
LETTER TO THE EDITOR
of gonadotropin-releasing hormone from the hypothalamus, resulting in suppression of the hypothalamic-pituitary-gonadal axis.
In addition, prolactin directly impairs gonadal steroidogenesis by
inhibiting the pituitary release of follicle-stimulating hormone
and luteinizing hormone.3,4
A prior history of therapeutic or accidental exposure to radioactivity
should be sought in all patients with sporadic prolactinoma.
References
Prolactinoma can be associated with the coexistence of parathyroid and pancreatic neoplasms in patients with multiple endocrine neoplasia type 1 syndrome.2,3,6; however, the adenoma
usually presents as a sporadic tumor.2 For example, we recently
observed a man with previous radiation exposure who subsequently developed a prolactinoma that presented with unilateral
galactorrhea (Figure) and fatigue.7,8
Currently, “no risk factors have been identified for sporadic prolactinomas.”2 We hypothesize, however, that a previously unrecognized risk factor for prolactinoma is exposure to accidental or
therapeutic radiation.7–9 We therefore suggest that patients who
present with sporadic prolactinoma be asked whether they have
a prior history of radiation exposure.
In summary, gynecomastia can either be the presenting symptom or
a subsequent development in men with a prolactinoma. Sparse body
hair, secondary to hyperprolactinemia-associated hypogonadism, is
a cutaneous manifestation that can be observed in these individuals.
1 Kapoor S. Cutaneous manifestations of systemic conditions associated with gynecomastia. Skinmed. 2010;8:87–92.
2 Klibanski A. Prolactinomas. N Engl J Med. 2010;362:1219–1226.
3 Mancini T, Casanueva FF, Giustina A. Hyperprolactinemia and prolactinomas. Endocrinol Metab Clin North Am. 2008;37:67–99.
4 Creyghton WM, Custers M. Gynaecomastia: is one cause
enough? Neth J Med. 2004;62:257–259.
5 Walsh JP, Pullan PT. Hyperprolactinaemia in males: a heterogeneous disorder. Aust N Z J Med. 1997;27:385–390.
6 Piecha G, Chudek J, Wiecek A. Multiple endocrine neoplasia
type 1. Eur J Intern Med. 2008;19:99–103.
7 Cohen PR, Robinson FW, Gray JM. Pituitary tumor following radiation exposure: prolactinoma in a man exposed to radioactivity
from the Chernobyl accident. [Poster Abstract] The University of
Texas MD Anderson Cancer Center Division of Internal Medicine
2010 Research Retreat. Houston, TX, May 14, 2010.
8 Cohen PR, Robinson FW, Gray JM. Prolactinoma presenting as
unilateral galactorrhea in a man exposed to radioactivity from
the Chernobyl accident: radiation-induced pituitary tumor? Int J
Dermatol. In press.
9 Cohen PR, Robinson FW, Gray JM. Radiation-associated prolactinoma [letter]. South Med J. In press.
VINTAGE LABEL
Courtesy of BuyEnlarge, Philadelphia, PA
SKINmed. 2010;8:201–202
202
Prolactinoma Can Be Associated With Gynecomastia
SAVE THE DATE FOR THE
7th Annual Maui Derm
A Pan-Pacific Thought Leaders Meeting
FEBRUARY 23-27, 2011 GRAND WAILEA, MAUI
Save the Date & Register Early! $395 before June 30th!
Plan to attend the 7th Annual Maui Derm Conference! “Cutting edge, a great blend of science and clinical medicine and
a world class faculty” describe Maui Derm 2011. Our faculty will share with you the most important developments in
medical and cosmetic dermatology vital to your practice. Our live patient demonstration workshops in lasers, Botulinum
Toxin A and fillers have set the standard for “hands on” workshops. Our format has been specifically designed to allow
optimal time for discussion with our faculty. We are certain that you will find this to be an outstanding and memorable
educational event that you will not want to miss. Put Maui Derm on your calendar for 2011!
FEATURED LECTURES
MAUI DERM 2011 FACULTY
Skin Cancer/Chemoprevention • Acne/Rosacea • Pediatric Dermatology •
Infectious Diseases • Psoriasis • Pigmentary Disorders • Contact Dermatitis • Cosmeceuticals • Collagen Vascular Diseases • Office Surgery Tips
Practice Management • Skin of Color
Dr. Rox Anderson
Dr. Andrew Blauvelt
Dr. Joel Cohen
Dr. Lawrence Eichenfield
Dr. Ilona Frieden
Dr. Sheila Fallon-Friedlander
Dr. Michael Gold
Dr. Mitchel Goldman
Dr. Pearl Grimes
Dr. Arthur Kavanaugh
Dr. Suzanne Kilmer
Dr. David Laub
Dr. Craig Leonardi
Medical Dermatology
Cosmetic Dermatology
Botulinum Toxin A and Fillers • Lasers • Skin of Color
“Lunch with the Faculty” –
Controversial Topics, Case-based Discussions
Interactive Workshops
Small Group, Live Patient, Hands-On Demonstrations:
Botulinum Toxins/Fillers
Chairman: Dr. George Martin
Dr. Stuart Maddin
Dr. Sam Moschella
Dr. Stuart Nelson
Dr. Kevin Pinski
Dr. Ted Rosen
Dr. Vic Ross
Dr. Alan Shalita
Dr. Bruce Strober
Dr. Hensin Tsao
Dr. Sandy Tsao
Dr. Guy Webster
Dr. Philip Werscheler
* Subject to Change
Official Meeting Publication Sponsor
Maui Derm 2011 has special/reduced rates at the
Grand Wailea and the Wailea Marriott
For more details, visit our website at
www.acmd-derm-hawaii.com
July/August 2010
Volume 8 • Issue 4
Original Contribution
Serum Lipid Level in Iraqi Patients With Psoriasis
Muhsin A. Aldhalimi, CABDV;1 Sadiq J. Almuhanna, FICMS;2 Samir H. Alrikabi, MSc3
Abstract
Psoriasis is a chronic inflammatory and proliferative dermatosis. Previous studies have demonstrated that patients with psoriasis may have an
increased risk of occlusive vascular disease. High serum lipid level has been suggested in the pathogenesis of this phenomenon. In this study,
the authors assess the lipid profile in Iraqi patients with psoriasis and compare it with that of nonaffected persons. This study was designed and
conducted as a cross-sectional study with 50 cases in the patient group and 50 patients in the control group. It was performed in the department
of dermatology at Al-Sadr Teaching Hospital in Najaf, Iraq. The lipid profile, including serum levels of triglycerides, total cholesterol, low-density
lipoprotein (LDL), and high-density lipoprotein, were assessed in both groups. The patient and control groups each consisted of 39 men and 11
women. The serum triglyceride, cholesterol, LDL, and very LDL levels were significantly higher in psoriatic patients (P<.05) but not for highdensity lipoprotein (P>.05). Serum lipid level was found to be significantly higher in Iraqi patients with psoriasis. It may be useful to do early
screening and treatment of hyperlipidemia in psoriasis to prevent atherosclerosis and its complications. (SKINmed. 2010;8:204–206)
P
soriasis is a chronic inflammatory skin disorder that affects
more than 2% of the population worldwide.1 It is characterized by an increase in keratinocyte proliferation and alterations in dermal and epidermal T cells, monocytes/macrophages,
and neutrophils.2 Although several previous studies suggest that
patients with psoriasis have abnormalities of plasma lipids and lipoproteins as an important risk factor for cardiovascular disease,
many of the results remain controversial.3–7 Many of these reports
suggest that persons with psoriasis have a proatherogenic lipoprotein profile that includes hypertriglyceridemia, raised plasma concentrations of low-density lipoprotein cholesterol (LDL-C), very
low-density lipoprotein cholesterol (VLDL-C), and lipoprotein
(a) and a lowered high-density lipoprotein cholesterol (HDL-C)
concentration,3,4,7–19 High HDL-C concentrations are well established as a major protective factor against coronary heart disease
because of their ability to remove unesterified cholesterol from
cells and other lipoproteins, where it may have accumulated, and
return it to the liver for excretion in the bile.10
The aim of this study was to investigate the plasma lipid profile
in psoriasis in the Middle Euphrates region in Iraq in comparison with nonaffected persons.
Patients and Methods
The study was done in the dermatology outpatient department at
Al-Sadr Teaching Hospital, Najaf, Iraq, during the period from
January to August 2007. It is a cross-sectional study that included
50 adult patients with psoriasis and 50 sex-, age-, and body mass
index (BMI)–matched healthy individuals as the control group.
The diagnosis of psoriasis was mainly clinical and all doubtful
cases were excluded. Smokers, alcoholics, pregnant women, and
those with a BMI >30 kg/m2 were not included in the study. BMI
was calculated as weight (in kilograms) divided by the square of
height (in meters). Weight and height were recorded as the persons were wearing light clothes and not wearing shoes.16
Psoriasis area severity index (PASI) was evaluated using the standard formula.17 Mild PASI score was defined as 0.1 to 10.9 and
moderate PASI score was defined as 11 to 49.9.14 Patients with
severe (PASI >50), erythrodermic, and pustular types of disease
were excluded from the study. Other exclusion criteria included
patients with diseases that may cause secondary hyperlipidemia
such as diabetes mellitus, hypothyroidism, nephritic syndrome,
chronic renal insufficiency, obstructive liver disease, and connective tissue diseases, as well as patients taking medications such as
b-blockers, thiazide diuretics, corticosteroids, cyclosporine, retinoids, and lipid-lowering agents in the past 6 months.
Blood samples were taken after 12 hours of overnight fasting. Serum levels of total cholesterol, triglycerides (TGs) and HDL-C
were measured by enzymatic colorimetric methods using a Hitachi 704 auto-analyser (Tokyo, Japan) and commercially available
kits (Diasys Diagnostic System, GmbH and Co, Holzheim, Germany). LDL-C was estimated by the Friedewald formula (LDL-C
= total cholesterol – HDL-C – VLDL-C). VLDL-C was calculated as 0.456 × total TG concentration expressed in mmol/L.15
From the Departments of Dermatology,1 Medicine,2 and Pharmacology,3 University of Kufa College of Medicine, Najaf, Iraq
Address for Correspondence: Muhsin A. Aldhalimi, CABDV, PO Box 18, Kufa Post Office, Najaf, Iraq • E-mail: [email protected]
SKINmed. 2010;8:204–206
204
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
ORIGINAL CONTRIBUTION
Table. Comparison of Lipid Profile Between Patients With Psoriasis and Control Patients
Patients With Psoriasis (N=50)
Control Group (N=50)
Mean
SD
Mean
SD
P Value
Total cholesterol, mg/dL
216.4
20.16
208.8
22.09
<.05
Triglycerides, mg/dL
172.6
14.34
139.7
13.93
<.05
HDL-C, mg/dL
39.62
3.76
43.88
3.52
>.05
LDL-C, mg/dL
146.22
25.62
134.56
23.73
<.05
34.52
12.35
27.9
11.67
<.05
VLDL-C, mg/dL
Abbreviations: HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol; SD, standard deviation; VLDL-C,
very low-density lipoprotein cholesterol.
This study was approved by the ethics committee of our university, and all patients gave their informed consent.
Statistical Analyses
Values are presented as mean ± standard deviation. Comparison
between patients with and without psoriasis was performed by
Student t test and Pearson correlation coefficients. P value <.05
was considered statistically significant.
Results
The age of the patients ranged from 20 to 67 years, with a mean of
37.6 years. There were 39 men and 11 women. The age of the control group ranged from 21 to 65 years, with a mean of 37.4 years.
The sex distribution was the same as that of the patients. PASI score
ranged from 2.3 to 24.7, with a mean of 19.6. The mean duration
of disease was 5.45 years (range 0.8–19 years). There was no difference between patients and the control group with regard to BMI,
systolic and diastolic blood pressures, and physical activity. Serum
lipid levels in patients with psoriasis and in controls are shown in
the Table. In the patient group, serum total cholesterol, LDL-C,
VLDL-C, and TG levels were significantly higher than those of controls (P<.05). HDL-C was slightly higher in the control group, but
the difference was not statistically significant (P>.05). There was no
significant association between PASI and serum lipid profile in the
present work (Pearson correlation coefficients P>.05).
Discussion
In spite of an increasing number of studies dealing with plasma
lipid levels in patients with psoriasis, the results are still conflicting.3–13,15–18 It is still controversial whether changes in lipid composition are primary events or secondary to psoriasis or perhaps
due to medications such as cyclosporine and retinoids.5,7,19
In this study we try to assess this relation in a group of Iraqi patients. The measured lipid parameters in this work were significantSKINmed. 2010;8:204–206
ly higher in patients with psoriasis than the control group, apart
from HDL-C, which was slightly higher in the control group. The
results of previous studies were greatly variable. High,3,11,17,19 low,20
or even normal4,5,7,18 values of serum cholesterol levels in patients
with psoriasis have been reported. In this study we found a significantly higher level of total cholesterol values in the psoriatic
group (P<.05). Similar conflicting results of plasma TG level were
observed in the published literature. The previous results varied
from high3,4,7,17 to low20 or even normal5,6,8,18,19 values. This work
showed a significant higher value of serum TG levels in the psoriatic group. A similar controversy was found regarding HDL-C and
VLDL-C values in patients with psoriasis. Normal4,5,7,8,17–19 and
low3,10,12 levels of serum HDL-C and high3,6,11,12 levels of VLDL-C
have been reported. Although the values of HDL-C in the present study were lower in the psoriasis group than in the control
group, the difference was not statistically significant (P>.05). The
recorded values of VLDL-C level in this work were significantly
higher in the psoriatic group (P<.05).
The discrepancy in data from different studies may be due to the
fact that patients included in statistical analysis have different
phases and forms of psoriasis and undergo various treatments. The
other cause may be a result of differences in lipid referential values,
such as sex and age, which considerably change the results when
young and old patients and men and women are collectively investigated.5 Different methods used in disease scoring and laboratory
assessment may be another cause of the discrepancy.
The values of serum lipid in psoriatic patients have been studied
since almost half a century ago. Lea and colleagues21 reported increased serum lipid concentrations in patients with psoriasis.
Since then, much research has been performed in this area, most
of which consistently points to a raised prevalence of lipid abnormalities in individuals diagnosed with psoriasis. The exact cause of
these changes in lipid metabolism in patients with psoriasis is not
yet confirmed. The activation of the immune system in psoriasis
205
Serum Lipid Level in Iraqi Patients With Psoriasis
July/August 2010
ORIGINAL CONTRIBUTION
may cause some changes in patients’ lipid profiles. They may be
related, however, to some structural and functional abnormalities in
the digestive system, which are shown to be found in nearly all the
segments of the digestive system.22 These abnormalities may cause
alteration in decomposition, modification, and synthesis of many
organic compounds, including lipids. Knowing that psoriasis is a
genetic disorder, however, there is a possibility that genetic alterations in the HDL-C and/or apolipoprotein A-I genes may be linked
with this disease. This genetic hypothesis is supported by a study on
lipid and lipoprotein profile among Swedish patients with psoriasis
at the initial stages of the disease.11 It demonstrated that patients
with psoriasis manifest significant lipid abnormalities and support
the notion that the abnormal lipid profile seen in such patients
might be genetically determined rather than acquired.
Previous studies have shown a high prevalence of atherosclerosis
in psoriatic patients.3,19,23,24 High serum lipid level has been suggested in the pathogenesis of this phenomenon. Early assessment
and treatment of abnormal lipid profile may reduce atherosclerosis and cardiovascular accidents in these patients.
References
1 Christophers E. Psoriasis—epidemiology and clinical spectrum.
Clin Exp Dermatol. 2001;26:314–320.
2 Ortonne JP. Recent developments in the understanding of the
pathogenesis of psoriasis. Br J Dermatol. 1999;140:1–7.
3 Rocha-Pereira P, Santos-Silva A, Rebelo I, et al. Dislipidemia and
oxidative stress in mild and in severe psoriasis as a risk for
cardiovascular disease. Clin Chim Acta. 2001;303:33–39.
4 Uyanik BS, Ari Z, Onur E, et al. Serum lipids and apolipoproteins
in patients with psoriasis. Clin Chem Lab Med. 2002;40:65–68.
5 Seckin D, Tokgozoglu L, Akkaya S. Are lipoprotein profile and
lipoprotein (a) levels altered in men with psoriasis? J Am Acad
Dermatol. 1994;31:445–449.
6 Simonetti O, Ferretti G, Salvi A, et al. Plasma lipid changes in
psoriatic children. Dermatology. 1992;185:96–100.
7 Seishima M, Seishima M, Mori S, et al. Serum lipid and apolipoprotein levels in patients with psoriasis. Br J Dermatol.
1994;130:738–742.
8 Piskin S, Gurkok F, Ekuklu G, et al. Serum lipid levels in psoriasis. Yonsei Med J. 2003;44:24–26.
9 Pietrzak A, Lecewicz-Torun B. Activity of serum lipase [EC
SKINmed. 2010;8:204–206
206
3.1.1.3] and the diversity of serum lipid profile in psoriasis. Med
Sci Monit. 2002;8:CR9–CR13.
10 Reynoso-von Drateln C, Martinez-Abundis E, Balcazar-Munoz BR,
et al. Lipid profile, insulin secretion and insulin sensitivity in psoriasis. J Am Acad Dermatol. 2003;48:882–885.
11 Mallbris L, Granath F, Hamsten A, et al. Psoriasis is associated
with lipid abnormalities at the onset of skin disease. J Am Acad
Dermatol. 2006;54:614–621.
12 Vahlquist C, Michaelsson G, Vessby B. Serum lipoproteins in middleaged men with psoriasis. Acta Derm Venereol. 1987;67:12–15.
13 Cimsit G, Orem A, Deger O, et al. The variation of serum lipoprotein (a) level with disease activity in psoriasis. Br J Dermatol.
1998;138:917–919.
14 Vanizor Kural B, Orem A, Cimsit G, et al. Evaluation of the atherogenic tendency of lipids and lipoprotein content and their
relationships with oxidant-antioxidant system in patients with
psoriasis. Clin Chim Acta. 2003;328:71–82.
15 Friedewald WT, Levy RI, Fredrickson DS. Estimation of the
concentration of low-density lipoprotein cholesterol in plasma,
without use of the preparative ultracentrifuge. Clin Chem.
1972;18:499–502.
16 Mallbris L, Akre O, Granath F, et al. Increased risk for cardiovascular mortality in psoriasis inpatients but not in outpatients. Eur
J Epidemiol. 2004;19:225–230.
17 Akhyani M, Ehsani AH, Robati RM, et al. The lipid profile in
psoriasis: a controlled study. J Eur Acad Dermatol Venereol.
2007;21:1330–1332.
18 Farshchian M, Zamanian A, Farshchian M, et al. Serum lipid level
in Iranian patients with psoriasis. J Eur Acad Dermatol Venereol.
2007;21:802–805.
19 Gurkok F, Piskin S, Ekuklu G. Serum lipid and lipoprotein levels in
psoriasis. Bull Leprosy. 1999;30:105–111.
20 Fortinskaia ES, Torkhavskia TI, Sharapova GI, et al. Features of
distribution of free and esterified cholesterol in the epidermis,
biological membrane and plasma lipoproteins in psoriasis. Klin
Lab Diagn. 1996;4:38–43.
21 Lea WA Jr, Cornish HH, Block WD. Studies on serum lipids,
proteins, and lipoproteins in psoriasis. J Invest Dermatol.
1958;30:181–185.
22 Pietrzak A, Lecewicz-Torun B, Kadziela-Wypyska G. Changes in
the digestive system in patients suffering from psoriasis. Ann
Univ Mariae Cruie Sklodowska. 1998;53:187–194.
23 McDonald CJ, Calabresi P. Psoriasis and occlusive vascular disease. Br J Dermatol. 1978;99:469–475.
24 Gelfand JM, Neimann AL, Shin DB, et al. Risk of myocardial infarction in patients with psoriasis. JAMA. 2006;296:1735–1741.
Serum Lipid Level in Iraqi Patients With Psoriasis
July/August 2010
Volume 8 • Issue 4
Original Contribution
Clinical Features and Treatment of
Dermatosis Papulosa Nigra in Migrants to Italy
Roberta Calcaterra, MD;1 Gennaro Franco, MD;2 Mariacarla Valenzano, MD;2
Raffaella Fazio, MD;2 Aldo Morrone, MD1
Abstract
Dermatosis papulosa nigra (DPN) is a benign epithelial tumor that is common in dark-skinned people. Although the diagnosis is easily made
on medical examination, DPN is characterized by a chronic and worsening course. Therefore, even if DPN is a benign disease, the lesions
are unaesthetic and the therapeutic options are quite inefficient. A prospective study was carried out during a period of 24 months (January
2006 to December 2007) at the Department for Preventive Medicine for Migration, Tourism and Tropical Dermatology of San Gallicano
Dermatological Institute in Rome. Among 58 patients, 41 (71%) were women and 17 (29%) were men. The mean age was 33.5 years (range,
8–45 years). One pediatric patient was observed. This study is the first in Italy that, in recent years, has observed an important growth of the
migration. The classic female predominance, family predisposition, and photodistribution of the lesion were found. DPN is frequently associated with patient discomfort, therefore the education of patients to reduce self-treatment is important. (SKINmed. 2010;8:207–209)
D
ermatosis papulosa nigra (DPN) is a nevoid condition
of the pilosebaceous apparatus that predominantly affects adult black persons.1,2 It is characterized by black
or dark brown flattened or cupuliform papules, with a diameter
ranging from 1 mm to 5 mm. The lesions do not itch or hurt (are
asymptomatic) and occur on the face, neck, and trunk. DPN is
a frequent disease among black persons, above all Africans and
African-Americans, but is also common in Native Americans
and persons from Malaysia.
sites involved at onset, morphology of lesions, clinical course,
and psychological impact of the disease.
Despite the diagnosis being easily made at medical examination,
DPN is characterized by a chronic and worsening course. Therefore, even though DPN is a benign disease, the lesions are unaesthetic and the therapeutic options are quite inefficient.
Results
Materials And Methods
A prospective study was performed during a 24-month period
(January 2006 to December 2007) at the Department for Preventive Medicine for Migration, Tourism and Tropical Dermatology of San Gallicano Dermatological Institute in Rome, Italy.
All patients attending our department for this or any other dermatologic reason were assessed.
For each patient, the following data were obtained: sex, age,
country of origin, family history of DPN, age at onset of disease,
A careful clinical examination was performed. Histologic examination was carried out when necessary to confirm the clinical diagnosis. If required by the patient, the lesions were excised using
fine-needle or fine-scissor electrosurgery after the application of
lidocaine-prilocaine cream. The follow-up of the aesthetic results
were evaluated during a 3-month period.
Fifty-eight patients were evaluated, 41 (71%) were women and
17 (29%) were men. The mean age was 33.5 years (range, 8–45
years). One pediatric patient was observed. All patients had dark
skin, in particular, 35 (61%) were Africans and 23 (39%) were
Indomalesian. More than half the patients (42 [73%]) had a
family member with the same disease.
After analysis of the anamnestic data, results showed that lesions
developed gradually during the years. The age of onset of the lesions ranged from 8 to 45 years, with a mean of 25 years. All the
patients presented with lesions on the face (100%), mostly on the
temples, malar regions, and on the forehead (Figure 1 and Figure
2). They were uncommon on the lower parts of the face and chin,
but they were also found on the neck (8%) and on the trunk (4%).
From the National Institute for Health, Migration and Poverty,1 and San Gallicano Dermatological Institute,2 Rome, Italy
Address for Correspondence: Roberta Calcaterra, MD, National Institute for Health, Migration and Poverty, Via di San Gallicano 25/a Roma, Italy •
E-mail: [email protected]
SKINmed. 2010;8:207–209
207
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
ORIGINAL CONTRIBUTION
Clinically, the lesions that we found were round, smooth, brown or
black papules, ranging in size from 1 mm to 5 mm. Rarely we observed, only on the trunk, nodules or pedunculated papules (14%).
These lesions were commonly asymptomatic, although 5 patients (8.4%) reported transitory itch.
Twenty-three women (39.6%) reported the application of
bleaching cream (most containing high-strength corticosteroids)
for a period ranging from 4 months to 5 years, without any improvement. The histopathologic examination was performed in
3 cases and confirmed the diagnosis of DPN (Figure 3).
Regarding psychological impact, discomfort described by these
patients, above all among young adults and women, was remarkable. In fact, aesthetic considerations were the reason for
consultation in 43 patients (74%), 35 of whom were women.
Moreover, in 37 patients (63.8%), DPN caused anxiety and fear
of professional concerns.
Figure 1. Multiple pigmented papules of the face.
All patients were informed that the treatment may be temporary
and could result, above all in black skin, in important dyschromia.
Electrosurgery was performed in 20 patients after the application
of 30 minutes of lidocaine-prilocaine cream. At 1-month followup examination, hypochromia in 15 patients and hyperchromia
in 5 patients were detected. At 3-month follow-up, dyschromia
persisted in only 2 cases; 3 patients were lost to follow-up.
Discussion
DPN was first described by Castellani in 1925 while visiting Central America and Jamaica. He noted that this condition was common in adult black persons and was usually localized on the face.1
Different from that previously reported, we found that DPN occurs
in all people with high Fitzpatrick skin phototype classification (skin
phototype IV, V, or VI), not only in African descendants.1
The incidence of DPN among black persons rises from about
5% to 40%, and women, especially adolescents, are more commonly affected than men.1,3–5
The lesions usually develop during puberty and become more pronounced by the sixth decade; they are rare during childhood. Some
studies have demonstrated the family predisposition of DPN.
DPN is likely to be genetically determined, with 40% to 54% of
patients having a family history of involvement. DPN is believed
to be caused by a nevoid developmental defect of the pilosebaceous follicle. Some researchers have suggested that DPN should
be classified within the group of epithelial nevi.6,7
Figure 2. Multiple pigmented papules of the face.
SKINmed. 2010;8:207–209
Clinically, the single lesions are 1-mm to 5-mm black or dark
brown cupuliform and usually asymptomatic. DPN is localized
on the malar regions and on the forehead, is rare on the lower
208
Dermatosis Papulosa Nigra in Migrants to Italy
July/August 2010
ORIGINAL CONTRIBUTION
parts of the face and the chin, and can sometimes be found on
the neck, chest, and back.
The differential diagnosis of DPN include melanocytic nevi,
warts, acrochordons, adenoma sebaceum, follicular hamartoma,
and seborrheic keratosis.
The diagnosis may be confirmed by biopsy, with DPN resembling an acanthotic seborrheic keratosis.
Lesions of dermatosis papulosa nigra have the histologic appearance of seborrheic keratoses; they display hyperkeratosis, irregular acanthosis, keratin-filled invaginations of the epidermis (horn
cysts), and marked hyperpigmentation of the basal layer. Although
most lesions are of the acanthotic type and show thick interwoven
tracts of epidermal cells, they may have a reticulated pattern in
which the tracts consist of a double row of basaloid cells.8,9
Although DPN could exhibit thick interwoven tracts of epithelial cells, the proliferation of small basal cells is not common in
DPN, such as in seborrheic keratosis.
Treatment is seldom requested because of DPN’s benign nature,
but patients often request some treatment to improve their aspect and reduce discomfort.
Figure 3. Epidermal acantholysis and mild papillomatosis.
2 Micheal JC, Seale ER. Dermatosis papulosa nigra. Arch Dermatol Syph. 1929;20:629.
3 Grimes PE, Arora S, Minus HR, et al. Dermatosis papulosa nigra.
Cutis. 1983;32:385.
4 Hairston MA Jr, Reed RJ, Derbes VJ. Dermatosis papulosa nigra.
Arch Dermatol. 1964;89:655–658.
5 Dunwell P, Rose A. Study of the skin disease spectrum occurring in an Afro-Caribbean. Int J Dermatol. 2003;42:287–289.
6 Hairston MA Jr, Reed RJ, Derbes VJ. Dermatosis papulosa nigra.
Arch Dermatol. 1964;89:655–658.
The papules may be removed with diathermy or cautery; cryotherapy has also be used, but it is important to inform the patient that all this treatment may be temporary and could result
in, above all in black skin, important hypopigmentation or hyperpigmentation and sometimes in keloids.10–13
8 Niang SO, Kane A, Diallo M, et al. Dermatosis papulosa nigra in
Dakar, Senegal. Int J Dermatol. 2007;46:45–47.
Other therapies include keratolytic creams associated with electrodessication, electrodessication, fractional photothermolysis,
and long-pulsed 1064 nm Nd:YAG laser.14,15
10 Kaun YC. A surgical approach for dermatosis papulosa nigra. Int
J Dermatol. 1983;22:1013–1015.
We performed excision using fine-needle electrosurgery to minimize the risk of unaesthetic scarring, and the use of anesthetic
cream was permitted to treat more lesions at the same time.
Conclusions
Although DPN is benign and the treatment, however easy to
perform, is not always required, it is important to promote the
knowledge of this condition common among black persons to
provide the right assistance and treatment.
References
1 Castellani A. Observations on some diseases of Central America. J Trop Med Hyg. 1925;28:1.
SKINmed. 2010;8:207–209
209
7 Hafner C, Landthaler M, Mentzel T, et al. FGFR3 and PIK3CA
mutations in stucco keratosis and dermatosis papulosa nigra.
Br J Dermatol. 2010;162:508–512.
9 Binazzi M, Simonetti S. Dermatosis papulosa nigra in a white
man. Ann Derm Venereol. 1984;111:1013–1015.
11 Schweiger ES, Kwasniak L, Aires DJ. Treatment of dermatosis
papulosa nigra with a 106 nm Nd:YAG laser: report of two cases. J Cosmet Laser Ther. 2008;10:120–122.
12 Schwartzberg JB, Ricotti CA Jr, Ballard CJ, et al. Eruptive dermatosis papulosa nigra as a possible sign of internal malignancy.
Int J Dermatol. 2007;46:186–187.
13 Carter EL, Coppola CA, Barsanti FA. A randomized, double-blind
comparison of two topical anesthetic formulations prior to electrodesiccation of dermatosis papulosa nigra. Dermatol Surg.
2006;32:1–6.
14 Kundu RV, Joshi SS, Suh KY, et al. Comparison of electrodesiccation and potassium-titanyl-phosphate laser for treatment of dermatosis papulosa nigra. Dermatol Surg. 2009;35:1079–1083.
15 Katz TM, Goldberg LH, Friedman PM. Dermatosis papulosa nigra treatment with fractional photothermolysis. Dermatol Surg.
2009;35:1840–1843.
Dermatosis Papulosa Nigra in Migrants to Italy
July/August 2010
Volume 8 • Issue 4
REVIEW
Acute Generalized Exanthematous Pustulosis
Jennifer L. Pecina, MD;1 Mark A. Cappel, MD2
Abstract
Acute generalized exanthematous pustulosis is a cutaneous reaction pattern typically precipitated by medication exposure. It has an estimated incidence of 1 to 5 cases per million per year and can occur in all age groups. Diagnosis is based on typical rash morphology,
histopathology, and clinical course. Treatment consists of withdrawing the causative agent and providing supportive care. Prognosis for
full recovery is excellent. Patients should be counseled to avoid any future use of the causative medication. (SKINmed. 2010;8:210–214)
F
irst described as a distinct entity in 1968,1 acute generalized
exanthematous pustulosis (AGEP) is a relatively uncommon cutaneous reaction pattern typically precipitated by
medication exposure. The disease has been variably termed pustular drug rash, generalized pustular dermatosis, and toxic pustuloderma. The current nomenclature was proposed in the French
literature in 1980.2
Clinical Presentation
AGEP is characterized by acute onset of an edematous erythematous
dermatitis with overlying small pustules, frequently accompanied by
a temperature >38°C. Leukocytosis is common, predominantly as
neutrophilia, although eosinophilia may also occur. Serum chemistry values may show a slight elevation in hepatic transaminases,
hypocalcemia, and a transient decrease in creatinine clearance.3
The typical dermatologic appearance is that of an edematous
erythematous dermatitis that frequently begins on the face or
in intertriginous regions. Patients often describe the dermatitis
as burning and pruritic. This dermatitis is followed by the appearance of numerous small (<5 mm) nonfollicular sterile pustules in the involved areas (Figure 1). By definition, the pustules in AGEP are sterile, but, as with any diffuse dermatitis,
staphylococcal and other bacterial superinfections can occur.
Mucous membrane involvement occurs in 20% of the patients.
Occasionally, blisters, target lesions, and purpura are seen.3 Individual pustules can expand and coalesce, thereby forming
larger confluent pustules. Biopsy may be required to confirm
the diagnosis, and histopathology typically shows spongiform
subcorneal pustules or intraepidermal pustules with papillary
dermal edema and perivascular infiltrates of neutrophils and
sometimes eosinophils.4
An AGEP validation score proposed for diagnosis is based on a
combination of features of the morphology, histopathology, and
clinical course (Table I).5 Often, however, patients with cutaneous drug reactions have been simultaneously exposed to multiple
new medications, making determination of the particular causative agent challenging. Inadvertent reexposure has been reported
to provoke recurrent episodes of AGEP; thus, rechallenge could
be used to confirm clinical suspicions of a causative agent.3,6 Reexposure is potentially unsafe, however, and may raise ethical concerns. Since patch testing results are positive in 50% of patients,
it is a reasonable method for attempting to elucidate the inciting
medication.7 Patch tests are generally considered safe, although an
AGEP reaction provoked by patch testing has been reported.8
Incidence
The incidence of AGEP is estimated to be approximately 1 in 5
cases per million per year, and all age groups may be affected.5
Two studies suggested a female predilection, but this has not
been a consistent finding.3,9,10 One study found a disproportionate association with the HLA B51, DR11, and DQ3 genotypes
compared with the general population. A personal or family
history of psoriasis is slightly more common than would be expected in the general population, but most cases of AGEP occur
in patients without any psoriatic history.
Etiology
The underlying pathogenic mechanism for AGEP remains unclear; however, on the basis of several studies, a T-cell–mediated
process has been hypothesized. In vitro tests have demonstrated
that drug-specific T lymphocytes from AGEP patients produce
chemokines that attract neutrophils as well as prolong their
From the Department of Family Medicine,1 and the Division of Laboratory Dermatology,2 Mayo Clinic, Rochester, MN
Address for Correspondence: Jennifer L. Pecina, MD, Department of Family Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 •
E-mail: [email protected]
SKINmed. 2010;8:210–214
210
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
REVIEW
Figure 1. Acute generalized exanthematous pustulosis. Anterior surfaces of legs (A); back (B).
survival, thus leading to the sterile inflammatory pustules that
are clinically observed.11,12 AGEP is precipitated by systemic
drugs in approximately 90% of patients. Historically, b-lactam
and macrolide antibiotics are the most frequent causative agents,
although multiple drugs have been implicated (Table II). AGEP
has been reported to occur from multiple antibiotics within the
same class: in 1 patient, AGEP developed from 2 different macrolides,13 and in another patient, AGEP developed from 3 different b-lactam antibiotics (piperacillin, ceftazidime, and meropenem).15 A bimodal pattern has been seen for the interval to onset
after starting administration of an inciting drug. With antibiotics, the onset of dermatitis usually occurs within 2 days (mean,
2.5 days), whereas with other medications there is often a delay
of 1 to 3 weeks (mean, 18 days).3 AGEP has been associated with
viral infections (including enteroviruses, cytomegalovirus, and
human parvovirus B19),3 spider bites,2,3 and mercury exposure.3
Differential Diagnosis
The differential diagnosis of AGEP includes various dermatoses
that have a pustular component with a background of diffuse
erythema. Folliculitis, impetigo, and Sweet syndrome can appear
pustular but are typically not associated with a background of
diffuse erythema. Blistering diseases such as Sneddon-Wilkinson
disease, immunoglobulin A pemphigus, and pemphigus foliaceus
can form pustules but often have more chronic and discrete lesions. Impetigo typically presents with larger, flaccid, and nonsterile pustules that remain relatively localized. Sweet syndrome
can occur suddenly and the lesions may appear pustular, but these
SKINmed. 2010;8:210–214
lesions are also relatively localized and discrete. Pustular contact
dermatitis, superinfected eczematous dermatitis, pustular psoriasis, staphylococcal scalded skin syndrome, and drug rash with
eosinophilia and systemic symptoms can present with diffuse
erythematous dermatitis associated with pustules. Staphylococcal
scalded skin syndrome is associated with staphylococcal infection,
and drug rash with eosinophilia and systemic symptoms is associated with significant systemic symptoms. Both tend to be characterized by exfoliation rather than pustule formation. Most of the
above entities are easily distinguished from AGEP on the basis of
clinical findings and history. The primary disease process to be excluded is generalized pustular psoriasis (von Zumbusch psoriasis)
(Figure 2), which has a longer clinical course than AGEP and is
not associated with drug administration.
Treatment
Treatment consists of discontinuing use of the suspected offending agent and providing supportive care. If they are not
thought to be causative agents, antipyretics and antihistamines
can be used for symptomatic relief. Various topical treatments,
including topical corticosteroids and cool compresses, can also
be considered, but they may provide only mild symptomatic relief. Systemic corticosteroids may be empirically used, but supportive evidence is lacking. In at least two published reports,
pretreatment with corticosteroids did not prevent AGEP from
occurring.6,18 Paradoxically, there are also reports of corticosteroids provoking an AGEP reaction.19,20 Patients should be counseled to avoid use of the offending drug in the future, and their
211
Acute Generalized Exanthematous Pustulosis
July/August 2010
REVIEW
Table I. AGEP: Validation Scorea of the EuroSCAR Study
Groupb (1 of 2)
Table I. AGEP: Validation Scorea of the EuroSCAR Study
Groupb (2 of 2)
Feature
Feature
Points
Morphology
Points
Histology
Pustules
Other disease
Typical
-10
+2
Not representative/no histology
Compatible
+1
Exocytosis of PNN
+1
Insufficient
0
Subcorneal and/or intraepidermal nonspongiform or NOS pustule(s) with papillary edema or
subcorneal and/or intraepidermal spongiform or
NOS pustule(s) without papillary edema
+2
Spongiform subcorneal and/or intraepidermal
pustule(s) with papillary edema
+3
c
d
e
Erythema
Typical
+2
Compatible
+1
Insufficient
0
Abbreviations: AGEP, acute generalized exanthematous pustulosis;
EuroSCAR, European Study of Severe Cutaneous Adverse Drug Reactions; NOS, not otherwise specified; PNN, polynuclear neutrophil
count. aInterpretation: ≤0, no AGEP; 1–4, possible; 5–7, probable;
8–12, definite. bPatients were not included in the study if only localized pustules were reported, the pustular dermatitis already lasted
longer than 3 weeks, or a clear alternative diagnosis was made by a
dermatologist. cTypical: typical morphology. dCompatible: not typical, but not strongly suggestive of other disease. eInsufficient: lesions
cannot be judged (mostly because of late stage of the disease or poor
quality of pictures). Adapted with permission from Sidoroff et al.5
Distribution/pattern
Typical
+2
Compatible
+1
Insufficient
0
Postpustular desquamation
Yes
+1
No/insufficient
+0
0
Course
Outcomes
Mucosal involvement
Yes
-2
No
0
Outcomes in AGEP are uniformly excellent. The dermatitis is
topographically self-limited in distribution and usually spontaneously resolves in less than 15 days. Resolution is followed
by pinpoint cutaneous desquamation and eventual healing.5 No
data exist on cross-sensitization between similar classes of medications (eg, penicillins and cephalosporins). Serious consequences are extremely rare: an early article reported a 5% mortality
rate,24 but this has not been substantiated in later reports. Two
cases of AGEP-related death have been described; however, 1
patient died of hepatic coma and had other comorbidities15 and
the other died of acute renal failure and sepsis.9
Acute onset (≤10 d)
Yes
0
No
-2
Resolution ≤15 d
Yes
0
No
-4
Temperature ≥38°C
Yes
+1
No
0
Conclusions
AGEP is a relatively rare dermatologic disease typically provoked
by medication exposure. Specific criteria exist for establishing
the diagnosis. Symptoms usually spontaneously resolve after
withdrawal of the precipitating medication.
PNN ≥7000/μL
Yes
+1
No
0
adverse reaction should be documented in their medical record.
Although no specific guidelines are available, it may be prudent
to recommend that patients avoid future exposure to medications in the same class.
SKINmed. 2010;8:210–214
References
212
1 Baker H, Ryan TJ. Generalized pustular psoriasis: a clinical and epidemiological study of 104 cases. Br J Dermatol.
1968;80:771–793.
2 Beylot C, Bioulac P, Doutre MS. Acute generalized exanthematic
Acute Generalized Exanthematous Pustulosis
July/August 2010
REVIEW
Table II. Drugs and Miscellaneous Causes Reported to Be
Implicated in Acute Generalized Exanthematous Pustulosis
Antibiotics
Antihypertensives
Amoxicillin13
Diltiazem7
Ampicillin13
Enalapril5
Azithromycin14
Nifedipine3
Cefaclor13
Corticosteroids
Cefazolin
Dexamethosone19
Ceftazidime15
Methylprednisolone20
14
Cefuroxime13
Antiepileptics
Cephalexin13
Carbamazepine3,7
Chloramphenicol13
Phenobarbital7
Ciprofloxacin9
Other medications
Clindamycin
Allopurinol5
Doxycycline17
Chromium picolinate5
Erythromycin13
Cimetidine5
Gentamicin5
Disulfiram5
Imipenem13
Ferrous fumarate5
Isoniazid13
Furosemide5
Meropenem15
Ginkgo biloba21
Metronidazole5
Intravenous contrast media22
Penicillin13
Lansoprazole5
Piperacillin15
Quinidine3
Trimethoprim-sulfamethoxazole5
Sulfasalazine5
16
Vancomycin3
Figure 2. Pustular psoriasis on posterior surface of leg.
Miscellaneous
Other antimicrobials
pustuloses (four cases) (author’s transl) [in French]. Ann Dermatol Venereol. 1980;107:37–48.
Enterovirus3
Acyclovir9
Human parvovirus B1923
Griseofulvin5
Influenza vaccine9
Hydroxychloroquine5
Mercury3
Itraconazole5
Pneumococcal vaccine17
Mefloquine14
Psoralen–UV-A5
Nystatin5
Spider bites23
Terbinafine18
Tonsillitis9
3 Roujeau JC, Bioulac-Sage P, Bourseau C, et al. Acute generalized exanthematous pustulosis: analysis of 63 cases. Arch Dermatol. 1991;127:1333–1338.
4 James WD, Berger TG, Elston DM. Andrews’ Diseases of the
Skin: Clinical Dermatology. 10th ed. Philadelphia, PA: Saunders
Elsevier; 2006:125.
5 Sidoroff A, Halevy S, Bavinck JN, et al. Acute generalized exanthematous pustulosis (AGEP): a clinical reaction pattern. J Cutan
Pathol. 2001;28:113–119.
6 Bracke A, Van Marck E, Lambert J. Acute generalized exanthematous pustulosis after pemetrexed, and recurrence after re-introduction. Clin Exp Dermatol. 2009;34:337–339. Epub 2008 Aug 9.
Analgesics
7 Wolkenstein P, Chosidow O, Fléchet ML, et al. Patch testing in
severe cutaneous adverse drug reactions, including StevensJohnson syndrome and toxic epidermal necrolysis. Contact Dermatitis. 1996;35:234–236.
Acetaminophen8
Diclofenac14
Paracetamol17
8 Mashiah J, Brenner S. A systemic reaction to patch testing for
the evaluation of acute generalized exanthematous pustulosis.
SKINmed. 2010;8:210–214
213
Acute Generalized Exanthematous Pustulosis
July/August 2010
REVIEW
Arch Dermatol. 2003;139:1181–1183.
9 Tamir E, Wohl Y, Mashiah J, et al. Acute generalized exanthematous pustulosis: a retrospective analysis showing a clear predilection for women. Skinmed. 2006;5:186–188.
10 Davidovici B, Dodiuk-Gad R, Rozenman D, et al. Israeli RegiSCAR
Network. Profile of acute generalized exanthematous pustulosis
in Israel during 2002–2005: results of the RegiSCAR Study. Isr
Med Assoc J. 2008;10:410–412.
11 Schmid S, Kuechler PC, Britschgi M, et al. Acute generalized
exanthematous pustulosis: role of cytotoxic T cells in pustule
formation. Am J Pathol. 2002;161:2079–2086.
12 Britschgi M, Steiner UC, Schmid S, et al. T-cell involvement in
drug-induced acute generalized exanthematous pustulosis. J
Clin Invest. 2001;107:1433–1441.
13 Moreau A, Dompmartin A, Castel B, et al. Drug-induced acute
generalized exanthematous pustulosis with positive patch tests.
Int J Dermatol. 1995;34:263–266.
14 Beylot C, Doutre MS, Beylot-Barry M. Acute generalized exanthematous pustulosis. Semin Cutan Med Surg. 1996;15:244–249.
15 Mysore V, Ghuloom A. A case of recurrent acute generalized
exanthematous pustulosis due to beta-lactam antibiotics: a case
report. J Dermatolog Treat. 2003;14:54–56.
16 Sulewski RJ Jr, Blyumin M, Kerdel FA. Acute generalized exanthematous pustulosis due to clindamycin. Dermatol Online J.
2008;14:14.
17 De Coninck AL, Van Strubarq AS, Pipeleers-Marichal MA, et
al. Acute generalized exanthematous pustulosis induced by
paracetamol: a case with severe hemodynamic disturbances.
Dermatology. 1996;193:338–341.
18 Bajaj V, Simpson N. Oral corticosteroids did not prevent AGEP
due to terbinafine. Acta Derm Venereol. 2006;86:448–449.
19 Demitsu T, Kosuge A, Yamada T, et al. Acute generalized exanthematous pustulosis induced by dexamethasone injection.
Dermatology. 1996;193:56–58.
20 Mussot-Chia C, Flechet ML, Napolitano M, et al. Methylprednisolone-induced acute generalized exanthematous pustulosis [in
French]. Ann Dermatol Venereol. 2001;128(3 pt 1):241–243.
21 Pennisi RS. Acute generalised exanthematous pustulosis
induced by the herbal remedy Ginkgo biloba. Med J Aust.
2006;184:583–584.
22 Peterson A, Katzberg RW, Fung MA, et al. Acute generalized exanthematous pustulosis as a delayed dermatotoxic reaction to
IV-administered nonionic contrast media. AJR Am J Roentgenol.
2006;187:W198–W201.
23 Davidovici BB, Pavel D, Cagnano E, et al. EuroSCAR; RegiSCAR
study group. Acute generalized exanthematous pustulosis following a spider bite: report of 3 cases. J Am Acad Dermatol.
2006;55:525–529.
24 Roujeau JC. Clinical heterogeneity of drug hypersensitivity. Toxicology. 2005;209:123–129.
VINTAGE LABEL
Courtesy of BuyEnlarge, Philadelphia, PA
SKINmed. 2010;8:210–214
214
Acute Generalized Exanthematous Pustulosis
Available soon...
%
A New Tretinoin Therapy
From Triax Pharmaceuticals
©2010 Triax Pharmaceuticals, LLC.
All Rights Reserved.
Printed in USA
TX-0610-02
July/August 2010
Volume 8 • Issue 4
REVIEW
Sporotrichosis: Part I
Regina Casz Schechtman, MD, PhD
Abstract
Sporotrichosis is the most common subcutaneous mycosis in South America. Classic infection is associated with traumatic inoculation of soil,
vegetables, and organic material contaminated with Sporothix schenckii. Animals of various species, including humans, are affected by this disease.
This subcutaneous mycosis is an infection of implantation. The most frequent clinical form is the lymphocutaneous form. The fixed cutaneous
form is characterized by infiltrated nodular, ulcerated, or erythematosquamous lesions located on exposed areas where fungal inoculation occurred. The disseminated cutaneous forms have mainly been observed among immunosuppressed patients, especially human immunodeficiency
virus–positive individuals. Sporotrichosis is the only subcutaneous mycosis for which direct examination or histology is of little or no value for
diagnosis. The diagnosis rests solely on the isolation of S schenckii in culture. Since 1998, researchers from Brazil suggested that feline transmission
of sporotrichosis was associated with a large and long-lasting outbreak of the disease in the city of Rio de Janeiro. (SKINmed. 2010;8:216–220)
S
porotrichosis is the most common subcutaneous mycosis
in South America. Classic infection is associated with traumatic inoculation of soil, vegetables, and organic material
contaminated with Sporothix schenckii. Zoonotic transmit ion
has been described in isolated cases or in small outbreaks. Since
1998, researchers from Fiocruz Institute have been observing an
increasing number of cases of sporotrichosis in the population of
the city of Rio de Janeiro, Brazil. According to researchers,1 their
study suggests that feline transmission of sporotrichosis was associated with a large and long-lasting outbreak of the disease in
Rio de Janeiro city and surroundings.
Sporotrichosis is a subcutaneous subacute or chronic mycosis
caused by the dimorphic fungus S schenckii, which is found in
the soil, on plant debris, and on decaying vegetable material. Animals of various species, including humans, are affected by this
disease. This subcutaneous mycosis is an infection of implantation. In most cases it develops following traumatic injury and the
inoculation of environmental organisms into the host where they
affect the subcutaneous tissue, skin, and other adjacent structures.
It usually presents as a skin infection, cutaneous sporotrichosis,
when the organism gains entry by traumatic injury. Persons in certain occupational groups, such as those who handle plant materials or soil, are frequently exposed to the organism and have been
known to become infected.2,3
In 1998, an outbreak of in-house sporotrichosis transmitted by domestic cats was reported in the city of Rio de Janeiro. Since then,
human sporotrichosis with feline transmission took place as a public health disease in Brazil, and domestic cats are considered an important vector of sporotrichosis.1 In July 2006, Brazilian researchers
demonstrated 44 cases of street dogs with sporotrichosis in the city of
Rio de Janeiro. In the near future, dogs may play an important role in
the transmission of Brazilian zoonotic sporotrichosis as well as cats.4
History
In 1898, Bernard Schenck first isolated and described S schenckii.
He was attending medical school at Johns Hopkins University at
the time, and he had the assistance of a US Department of Agriculture mycologist, Erwin Smith, who identified the organism and
classified it as “sporotricha.”5 In 1900, the disease was reported for
the second time by Hektoen and Perkins, who classified the etiologic agent as S schenckii, with the pathogen being isolated from
a specimen aspirated from the cutaneous lesions of a patient.6 In
Europe, the first case was described in 1903, and more than 200
cases were reported during the following 10 years.7
The first sporotrichosis in Brazil was reported in 1907 by Lutz
and Splendore, who also found that it was possible to culture the
yeast form in vitro.8 The dimorphic transition of this fungus was
described by Howard in 1961.9
Taxonomy
S schenckii is, therefore, a dimorphic pathogenic fungus and the
etiologic agent of human and animal sporotrichosis. The fungus
From the Instituto de Dermatologia Prof Rubem David Azulay (IDPRDA), Santa Casa da Misericórdia do Rio de Janeiro, Rio de Janeiro, Brazil
Address for Correspondence: Regina Casz Schechtman, MD, PhD, rua Ribeiro de Almeida 44 Apt 102, Rio de Janeiro, Brazil, CEP 22240-060 •
E mail: [email protected]
SKINmed. 2010;8:216–220
216
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
REVIEW
belongs to the subdivision Deuteromycotina, class Hyphomycetes.3 According to Mariat and Taylor, the fungus Ceratocystis stenoceras (Ophiostoma stenoceras) presents a conidial stage similar to that
of S schenckii; however, no conclusions exist regarding the description of the teleomorphic phase of this species.10 Other investigators have recently postulated that stenoceras may correspond with
the teleomorph of S schenckii.2 Previous studies based on DNA
hybridization experiments, however, provided strong evidence
that stenoceras is not the teleomorph of S schenckii. The degree of
hybridization observed among S scheckii and O stenoceras DNAs
was as low as 30% while a high degree of cross-hybridization was
observed among 4 S schenckii strains studied.10
Epidemiology
Figure 1. Initial presentation of sporotrichosis. Single eroded
papule on the hand.
S schenckii is widely distributed in nature and can be found in
soil associated with plant organic matter (eg, thorns, dry leaves,
and wood), water, and decomposing organic matter, among others. S schenckii is present in the soil throughout the world. Sporotrichosis is more frequent in tropical and subtropical countries,
however, and is endemic in many parts of Latin America. Sporotrichosis has been mainly reported in tropical and temperate
zones.11,12 It is endemic in Mexico, Central America, and South
America, as well as other areas such as South Africa. It can also
occur in temperate climates such as in southern United States,
Japan, and Australia. In the south of the American continent,
the disease more frequently occurs in humid autumn or summer
seasons,13,14 whereas in Mexico, the highest incidence is observed
in cold and dry seasons.15 No seasonal difference, however, has
been reported by other authors.11,16
Figure 2. Dermal and subcutaneous eroded nodules and
ulcers up the arm along the path of lymphatic drainage.
Sporotrichosis can affect persons of all ages,17 and the number of
cases involving men and women varies from region to region.11,17
In some regions, the difference in the distribution of cases according to age and sex might be explained by the type of fungal
exposure, but no association has been found in other regions.
Generally, infection results from inoculation of the fungus
through thorns, splinters, scratches, and small traumas during
leisure and occupational activities such as floriculture, horticulture, gardening, fishing, hunting, farming and cattle raising,
mining, and wood exploration.13,14,17–22 Laboratory professionals
can be accidentally infected while manipulating S schenckii cultures.23,24 Sporotrichin skin test is mainly applied for epidemiologic studies and is not commercially available in Brazil.
Figure 3. Sporotrichosis. Initial ulcerated lesion with additional subcutaneous nodules up the arm along the path of lymphatic drainage.
Epidemic Outbreaks
Sporotrichosis usually occurs in isolated cases or in small family
or professional outbreaks. Epidemics are rare and, if present, have
been related to a single source of infection.25 The largest reported
epidemic occurred in South Africa, with about 3000 gold miners
SKINmed. 2010;8:216–220
infected with the fungus, which occurred in the wood girders of
the mine structure.26 Another epidemic burst affected 84 workers who participated in reforestation programs in 15 states of the
217
Sporotrichosis: Part I
July/August 2010
REVIEW
in various species such as horses, rats, dolphins, cows, and fish.2
Animal to human transmission is rare, however, and there are
few reports on animal transmission of sporotrichosis to humans,
with the exception of some cases of armadillo transmission in
Uruguay35 and feline transmission in Brazil. Other risk groups
include animal health personnel, such as veterinary doctors and
technicians, and animal owners as a result of the increasing number of sporotrichosis case reports transmitted by domestic cats.
Domestic cats can acquire the disease following traumatic injury
from infected street cats during fights or from direct inoculation
of environmental organisms that inhabit soil and vegetation.
They transmit the infection to humans in the domestic environment through biting or scratching human skin and consequently
inoculating the organisms that are present under the nails. Another way of transmission is through direct contact of human
skin with the infected cat skin, mucous membranes, or exudates
from open lesions. Domestic dogs have been identified with sporotrichosis, probably transmitted by domestic cats.1,4
Figure 4. Sporotrichosis. Fixed cutaneous form. Infiltrated
nodular eroded papule on the leg.
Pathogenesis
This subcutaneous mycosis is an infection of implantation. In
most cases it develops following traumatic injury, especially by
vegetation such as thorns and wood, and the inoculation of environmental organisms into the host where they affect the subcutaneous tissue, skin, and other adjacent structures. Certain
occupational groups such as those who handle plant materials
or soil are frequently exposed to the organism and have been
known to become infected.
Multiple inoculations may occur simultaneously, not to be confused with spread of a single primary lesion. The presentation and
course of sporotrichosis depends on the host immune response as
well as the size and virulence of the inoculums. In hosts not previously inoculated, involvement of regional lymphatic ensues. In
those with a prior history of exposure to S schenckii, no lymphatic
spread occurs and a “fixed ulcer” develops at the site of inoculums
or granulomatous plaque (particularly on the face).36
Figure 5. Sporotrichosis. Fixed cutaneous form. Infiltrated
nodular eroded papule on the nose.
United States and was associated with the sphagnum moss used to
store the seedlings that originated from Pennsylvania.27
Zoonotic Sporotrichosis
Human sporotrichosis has been sporadically related to the scratch
or bite of animals.21,28 The presence of the fungus in the mouth or
nails of the animals, however, was not demonstrated in any of the
cases described.28,29 Since the 1980s, domestic cats have gained
importance in the transmission of the mycosis to humans.20,30–34
Between animals, the acquired infection is reported in various
species. There are few reports, however, on animal transmission
of sporotrichosis to humans. The acquired infection is reported
SKINmed. 2010;8:216–220
Extensive cutaneous disease with or without systemic involvement is also possible, especially in an immunocompromised
host. Cases of inhaled sporotrichosis have been reported with
systemic and cutaneous dissemination, similar to disseminated
histoplasmosis and other dimorphic fungal infections.
Clinical Features
Sporotrichosis has diverse clinical manifestations, and investigators
disagree regarding the clinical classification of the disease.2 The most
frequent clinical form (about 80%) is the lymphocutaneous form.
The initial presentation of sporotrichosis is that of a single papule
at the precise location of injury, most commonly on the hand, that
218
Sporotrichosis: Part I
July/August 2010
REVIEW
appears several weeks after inoculation. The lesion then becomes
eroded or ulcerated with purulent drainage, and is generally not
painful (Figure 1). Weeks after development of the initial lesion, additional lesions appear, typically as dermal and subcutaneous nodules and ulcers along the path of lymphatic drainage (often up the
arm) (Figure 2). This clinical description led to naming the disease
as “ascending nodular lymphangitis.”37 This is the well-known “sporotrichoid” pattern (Figure 3). Usually, lesions located in the deep
dermis or subcutaneous tissue result in skin scars.
In general, the fixed cutaneous form is characterized by infiltrated
nodular, ulcerated, or erythematosquamous lesions located on
exposed areas where fungal inoculation occurred (Figure 4 and
Figure 5) The lesions of fixed cutaneous sporotrichosis can appear
granulomatous (Figure 6), and disseminated lesions typically present as subcutaneous nodules. The disseminated cutaneous forms
have mainly been observed among immunosuppressed patients,
especially human immunodeficiency virus–positive individuals.38,39 Mucosal involvement is not common but may occur and
preferentially affects the ocular mucosa (Figure 7).40 Among the
extracutaneous forms, osteoarticular and pulmonary involvement
are the most common, but there are reports of cases of severe hematogenic dissemination with involvement of multiple organs.
Figure 6. Sporotrichosis. Fixed cutaneous form. Granulomatous lesion typically on the face.
Differential Diagnosis
Due to the diversity of the clinical forms of sporotrichosis, there
is also a vast set of differential diagnoses with other pathologic
conditions. Examples include leishmaniasis, nocardiosis, chromomycosis, tuberculosis, rosacea, noninfectious granulomatous
diseases, and psoriasis, among others.
When there is a sporotrichoid pattern, the major entity to be excluded in endemic areas from Brazil is leishmaniasis. In Europe, Canada,
and the United States, the major differential diagnosis is an atypical mycobacterial infection, in particular Mycobacterium marinum.
Other less common causes are rhinoscleroma, granuloma inguinale,
histoplasmosis, leishmaniosis, and Penicillium marneffei infection.
The differential diagnosis of fixed plaque sporotrichosis and disseminated lesions is broad and includes other granulomatous
disorders, both infectious and inflammatory.
References
1 de Lima Barros MB, Schubach TM, Gutierrez-Galhardo MC, et al.
Sporotrichosis: an emergent zoonosis in Rio de Janeiro. Mem
Inst Oswaldo Cruz. 2001;96:777–779.
2 da Silva Lacaz CS, Porto E, Martins JE, Heins-Vaccari EM, Melo
NT, eds. Tratado De Micologia Médica. Sao Paulo, Brazil: Sarvier; 2002:479–497.
3 Bennett JE. Sporothricosis. In: Kwon-Chung K, Bennett J, eds.
Medical Mycology. 2nd ed. Philadelphia, PA: Lea & Febiger;
1992:707–729.
SKINmed. 2010;8:216–220
219
Figure 7. Sporotrichosis with ocular mucosal involvement.
4 Schubach TMP, Schubach A, Okamoto T, et al. Canine sporotrichosis in Rio de Janeiro, Brazil: clinical presentation, laboratory
diagnosis and therapeutic response in 44 cases (1998–2003).
Med Mycol. 2006;44:87–92.
5 Davis BA. Sporotrichosis. Dermatol Clin. 1996;14:69–76.
6 Hektoen L, Perkins CF. Refractory subcutaneous abcesses
caused by sporothrix schenkii. a new pathogenic fungus. J Exp
Med. 1900;5:77–89.
7 Mariat F. The epidemiology of sporotrichosis. In: Wolstenholme
GE, Porter R, eds. Systemic Mycoses. London, England: Little,
Brown Book Group; 1968:144–159.
8 Lutz A, Splendore A. Ueber eine bei menschen und ratten
beobachtete mykose: ein beitrag zur kenntnis der sogenannten
Sporotrichosis: Part I
July/August 2010
REVIEW
sporotrichosen. Centralblatt Bakteriologie, Parasitenkunde und
Infektionskrankheiten. 1907;45:631–637.
24 Cooper CR, Dixon DM, Salkin IF. Laboratory-acquired sporotrichosis. J Med Vet Mycol. 1992;30:169–171.
9 Howard DH. Dimorphism of sporotrichum schenckii. J Bacteriol.
1961;81:464–469.
25 Bustamante B, Campos PE. Endemic sporotrichosis. Curr Opin
Infect Dis. 2001;14:145–149.
10 Travassos LR, Lloyd KO. Sporothrix schenckii and related species of ceratocystis. Microbiol Rev. 1980;44:683–721.
26 Quintal D. Sporotrichosis infection on mines of the Witwatersrand. J Cutan Med Surg. 2000;4:51–54.
11 de Lima Barros MB. Estudo de uma série de casos de esporotricose atendidos no instituto de pesquisa clínica evandro chagas,
Rio de Janeiro. Universidade Federal do Rio de Janeiro, Faculdade de Medicina. 2004.
27 Centers for Disease Control (CDC). Multistate outbreak of
sporotrichosis in seedling handlers. Morb Mortal Wkly Rep.
1988;37:652–653.
12 Schubach TM, Schubach A, Okamoto T, et al. Evaluation of an
epidemic of sporotrichosis in cats: 347 cases (1998–2001).
J Am Vet Med Assoc. 2004;224:1623–1629.
13 Mackinnon JE, Conti-Diaz IA, Gezuele E, et al. Isolation of sporothrix schenckii from nature and considerations on its pathogenicity and ecology. Sabouraudia. 1969;7:38–45.
14 Lopes JO, Alves SH, Mari CR, et al. Epidemiology of sporotrichosis in the central region of Rio Grande do Sul [in Portuguese].
Rev Soc Bras Med Trop. 1999;32:541–545.
15 Gonzalez-Ochoa A. Contribuciones recientes al conociemiento
de la esporotrichosis. Gac Med Mex. 1965;95:463–474.
16 de Lima Barros MB, Schubach ADO, do Valle ACF, et al. Cat-transmitted sporotrichosis epidemic in Rio de Janeiro, Brazil: description of a series of cases. Clin Infect Dis. 2004;38:529–535.
17 Rippon JW. Medical Mycology: The Pathogenic Fungi and the
Pathogenic Actinomycetes. Philadelphia, PA: WB Saunders;
1988:325–352.
18 Mayorga R, Caceres A, Toriello C, et al. An endemic area
of sporotrichosis in Guatemala [in French]. Sabouraudia.
1978;16:185–198.
19 Mayorga R, Caceres A, Toriello C, et al. Study of an endemic
sporotrichosis zone in the Ayarza lake region of Guatemala [in
Spanish]. Bol Oficina Sanit Panam. 1979;87:20–34.
20 Read SI, Sperling LC. Feline sporotrichosis. Transmission to
man. Arch Dermatol. 1982;118:429–431.
21 Kauffman CA. Sporotrichosis. Clin Infect Dis. 1999;29:231–
236; quiz 237.
22 de Lima Barros MB, de Oliveira Schubach A, Galhardo MC, et
al. Sporotrichosis with widespread cutaneous lesions: report of
24 cases related to transmission by domestic cats in Rio de
Janeiro, Brazil. Int J Dermatol. 2003;42:677–681.
23 Thompson DW, Kaplan W. Laboratory-acquired sporotrichosis.
Sabouraudia. 1977;15:167–170.
SKINmed. 2010;8:216–220
220
28 Moore JJ, Davis DJ. Sporotrichosis following mouse bite, with
certain immunologic data. J Infect Dis. 1918;23:252–266.
29 Fischman O, Alchorne MM, Portugal MA. Human sporotricosis following rat bite. Rev Inst Med Trop Sao Paulo. 1973;15:99–102.
30 Dunstan RW, Reimann KA, Langham RF. Feline sporotrichosis.
J Am Vet Med Assoc. 1986;189:880–883.
31 Schubach TMP, Schubach A. Sporotrichosis of cat and dog: review. Clin Vet (Milano). 2000;29:21–24.
32 Werner AH, Werner BE. Sporotrichosis in man and animal. Int J
Dermatol. 1994;33:692–700.
33 Reed KD, Moore FM, Geiger GE, et al. Zoonotic transmission
of sporotrichosis: case report and review. Clin Infect Dis.
1993;16:384–387.
34 Larsson CE, Goncalves MD, Araujo VC, et al. Feline sporotrichosis: clinical and zoonotic aspects. Rev Inst Med Trop Sao Paulo.
1989;31:351–358.
35 Conti-Diaz IA. Sporotrichosis in Uruguay: epidemiologic and clinical aspects. Pan American Health Organization Scientific Publication. Washington, DC: PAHO; 1980:312–321.
36 Elgart ML. Subcutaneous and miscellaneous mycoses. In:
Elewski BE, ed. Cutaneous Fungal Infections. New York, NY:
Igaku-Shoin Medical Publishers; 1998:155–197.
37 Hoeprich PD. Sporotrichosis. In: Hoeprich PD, Jordan MC, Ronald AR, eds. Infectious Diseases. Philadelphia, PA: Lippincott Williams and Wilkins; 1994:1050–1054.
38 Shaw JC, Levinson W, Montanaro A. Sporotrichosis in the acquired immunodeficiency syndrome. J Am Acad Dermatol.
1989;21:1145–1147.
39 Donabedian H, O’Donnell E, Olszewski C, et al. Disseminated cutaneous and meningeal sporotrichosis in an AIDS patient. Diagn
Microbiol Infect Dis. 1994;18:111–115.
40 Vieira-Dias D, Sena CM, Orefice F, et al. Ocular and concomitant
cutaneous sporotrichosis. Mycoses. 1997;40:197–201.
Sporotrichosis: Part I
July/August 2010
Volume 8 • Issue 4
SELF-TEST REVIEW QUESTIONS
W. Clark Lambert, MD, PhD, Editor
Instructions: For each of the following numbered questions, choose the single most appropriate lettered response.
1) Sporotrichosis is most commonly transmitted to humans by
contact with:
4) Of the following, the organ most likely to be involved by extracutaneous sporotrichosis is the:
a. cats
a. colon
b. dogs
b. kidney
c. hamsters
c. liver
d. lice
d. lung
e. snakes
e. stomach
2) The “sporotrichoid pattern” is best described as a/an:
5) Of the following, the tissue most likely to be involved by extracutaneous sporotrichosis is:
a. acral ulcerating dermatosis
a. central nervous system tissue
b. ascending nodular lymphangitis
b. endometrial tissue
c. dependent edematous erythema
c. osteoarticular tissue
d. nodular intertrigo
d. peritoneal tissue
e. reticulated mucinosis
e. retroperitoneal tissue
3) Of the following, the most common mucosal site to be involved
with sporotrichosis is the:
a. conjunctiva
b. oral cavity
c. urethra
d. vagina
e. vulva
ANSWERS TO SELF-TEST REVIEW QUESTIONS:
1) a, 2) b, 3) a, 4) d, 5) c
From the Departments of Pathology and Dermatology, UMDNJ-New Jersey Medical School, Newark, NJ
Address for Correspondence: W. Clark Lambert, MD, PhD, Room C520 MSB, UMDNJ-NJMS, 185 South Orange Avenue, Newark, NJ 07101 •
E-mail: [email protected]
221
July/August 2010
Volume 8 • Issue 4
COMMENTARY
Sentinel Lymph Node Biopsy in Melanoma:
The Gulf Between Presentation and Reality
J. Meirion Thomas, MS, FRCP, FRCS
P
atients with newly diagnosed melanoma are frightened
and vulnerable. At times of crisis, pessimism prevails and
patients inevitably consider the worst possible outcome as
one of the range of possibilities. That fear is compounded when
they realize that, unlike most other malignancies, there is no systemic therapy that can prolong overall survival. In this frame of
mind, patients are offered sentinel lymph node biopsy (SLNB),
which, like ultrasound, in the best of hands,1 is an effective staging and prognostic tool.2 Inevitably, many patients accept SLNB
in the hope that their disease proves to be sentinel node (SN)–
negative; however, during the consent process, 3 other possible
benefits of SLNB in melanoma may be mentioned that are either
speculative or unproven.
First, it is suggested that even if the disease is SN-positive, then
immediate (early) lymphadenectomy vs delayed lymphadenectomy (when the nodal disease becomes palpable) may provide
a survival advantage. This was a conclusion of the first Multicenter Selective Lymphadenectomy Trial (MSLT-1),2 but the observation is better explained by an incidence of false-positivity
within the SN.3 Second, it is suggested that immediate vs delayed lymphadenectomy provides a benefit in disease-free survival (DFS), but this outcome is an inevitable consequence of
the MSLT-1 trial design and does not amount to a therapeutic advantage for patients.4 Third, it is suggested that immediate lymphadenectomy is associated with fewer complications
than delayed lymphadenectomy. This contribution will address
these 3 issues and will argue that the benefits of SLNB in melanoma have been overstated. It will conclude that the procedure
is merely a staging and prognostic tool and, for most patients,
represents excessive and unnecessary treatment.
diate lymphadenectomy comparing 2 computer-selected groups
of patients matched for all other prognostic factors.5 Sadly, this
survival advantage was not confirmed by the results of MSLT-1,
which showed no difference in overall survival from the point of
randomization, when immediate and delayed lymphadenectomy
were compared in the context of a randomized controlled trial.2
The authors of these papers2,5 have declined to explain the large
difference in survival between the 2 studies, but the most likely
explanation is that some of the minimally involved SNs in the
matched-pair analysis were prognostically false-positive, which
means that they were destined for dormancy or destruction and
not for progression to palpable nodal recurrence.
Breast cancer is another disease where SN status directs treatment options, but microscopic tumor deposits in the SN are
believed to be of no adverse prognostic significance and, according to size, are not an indication for completion lymphadenectomy nor necessarily for adjuvant systemic therapy.6 In biological
terms, why should it be assumed that tiny subcapsular deposits
of melanoma in the SN will behave so differently and require
more radical treatment?
False-Positivity in the SN
The authors of MSLT-1 and others refuse to consider the possibility of false-positivity in the SN in melanoma, possibly because
the statistical analysis of MSLT-1 assumes, without proof, that
all positive SNs will inevitably progress to palpable nodal recurrence if not removed. The incidence of false-positivity in the SN
in MSLT-1 at the third interim analysis has been estimated to be
24% for patients with intermediate thickness tumors and 34%
for all patients entered into the trial.3 These figures suggest that a
large number of patients may have been wrongly up-staged and
may have had unnecessary completion lymphadenectomy and
possibly unnecessary adjuvant therapy.3,4,7
It was the matched-pair analysis that first concluded that immediate vs delayed lymphadenectomy provided a survival advantage.5 In that study, a survival advantage of 22%, 32%, and
37% at 5, 10, and 15 years, respectively, was claimed for imme-
In a remarkable paper, investigators8 describe performing delayed SLNB in 6 patients at a median of 3.9 years after wide
excision of the primary melanoma. Two patients were found to
have tiny subcapsular deposits of melanoma in the SN 4 years
From the Department of Surgery, Royal Marsden Hospital and Imperial College, London, England
Address for Correspondence: J. Meirion Thomas, MS, FRCP, FRCS, Department of Surgery, Royal Marsden Hospital, Fulham Road, London
SW3 6JJ, UK • E-mail: [email protected]
SKINmed. 2010;8:222–224
222
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
COMMENTARY
and 4.9 years after the removal of their primary tumor (Figure).
Were these patients destined for late nodal recurrence, or is this a
perfect, if unintended, illustration of prognostic false-positivity?
Disease-Free Survival
Neither is it correct to claim, as was published,2 that immediate
vs delayed lymphadenectomy provides an advantage in DFS. A
benefit in DFS in the biopsy arm of MSLT-1 was inevitable due
to trial design.4 The most likely site of first recurrence in patients with intermediate thickness primary melanoma (1.2 mm
to 3.5 mm) is the regional lymph nodes, and, in the biopsy arm
of MSLT-1, the patients most likely to develop nodal metastases were identified as being SN-positive at the outset and were
treated with prophylactic lymphadenectomy. Therefore, it is inevitable that there are more regional node recurrences as site of
first recurrence in the observation arm. When the incidence of
first recurrence was broken down by site,2 nodal recurrence was
more than 3 times more common in the observation arm than
in the biopsy arm. Consequently, the advantage in DFS simply
amounts to lead-time bias and is of no therapeutic advantage.
This error was successfully challenged with the National Cancer
Institute (NCI), who funded the MSLT trials. They issued guidance in June 20074 that stated the following:
We agree that in a trial where one arm has nodal disease
removed at the outset, and the other arm does not, calculation of either distant DFS or exclusion of nodal recurrences from the DFS calculation is appropriate. We have
indicated to Dr Morton our belief that he should include
a DFS analysis in the next update of study, that excludes
nodal recurrences.
Patients who die of melanoma succumb to distant metastasis;
therefore, the most important end point for the fourth interim
analysis and (fifth) final analysis of MSLT-1 is distant DFS. Despite this guidance from NCI, the authors of MSLT-1 and others
have continued to stress the importance of DFS as previously
calculated.9–13 It is surprising that this crucial error was not identified before publication.2
Comparing Morbidity
Following Lymphadenectomy
Patients are also encouraged to undergo SLNB for the following reason:
We must also understand the safety and efficacy of salvage
node dissection at the time of nodal relapse. Available data
suggests that lymphadenectomy performed for clinically evident nodal disease is associated with more complications and
poorer regional control rates than when the same procedure
is done for microscopic disease.13
SKINmed. 2010;8:222–224
Figure. Delayed sentinel lymph node biopsy: subcapsular
deposits of melanoma in the sentinel node 4 years (A) and 4.9
years (B) after wide excision of the primary tumor.8
Statements similar to this are used frequently by advocates of
SLNB but are not evidence-based. The only randomized controlled trial of immediate vs delayed lymphadenectomy is MSLT1, which does not describe any greater morbidity following delayed lymphadenectomy in patients with intermediate thickness
tumors.2 It is conceded that retrospective studies of therapeutic
lymph node dissection do show a high incidence of complications, but this is due to the inclusion of patients who, for a variety of reasons, present with advanced, neglected nodal disease
and who, unlike patients in the observation arm of MSLT-1,
did not undergo regular follow-up, with or without ultrasound
surveillance, after wide excision of their primary tumor. In other
words, there is no evidence that patients who are closely observed
by clinical or ultrasound surveillance following wide excision of
their primary melanoma and who then develop palpable nodal
recurrence, have more complications or more nodal basin recurrences following delayed lymphadenectomy.
Excessive and Unnecessary Surgery
Patients must understand that the SLNB procedure involves excessive and unnecessary surgery for the vast majority, and that
SLNB alone carries a risk of immediate and late complications.
We know that the incidence of SN-positivity is about 20% and
that only 20% of SN-positive patients will have positive nonSNs on completion lymphadenectomy. Of 100 patients undergoing SLNB, at least 80 will be SN-negative and will have had
an unnecessary operation. The prevailing opinion among SLNB
experts is that all patients with positive SNs should have completion lymphadenectomy, irrespective of the tumor burden12,13;
therefore, 16 patients will have unnecessary completion lymphadenectomy and only 4 of the original 100 patients will have the
exact amount of surgery required. In terms of complications of
SLNB alone, 13.8% of patients will have minor complications14
and 12% will have permanent lymphedema at 6 years.15 Patients
should also understand that although SN status is an important
223
Sentinel Lymph Node Biopsy in Melanoma
July/August 2010
COMMENTARY
prognostic factor, it is no guarantee of remaining disease-free. In
MSLT-1, approximately 17% of SN-negative patients had recurrence by 5 years and 9.7% had died of metastatic melanoma.
Conclusions
When considering complex management issues, physicians and
patients have the right to expect accurate information from opinion leaders, untainted by personal preference, creative statistical
interpretation, or unjustified extrapolation. Patients who consider
the SLNB procedure should understand that it is a staging and
prognostic tool and nothing more. There is no evidence at present that the procedure offers an advantage in DFS or that any
subgroup of patients benefits from a survival advantage. There is
also no evidence that delayed lymphadenectomy results in more
complications than immediate lymphadenectomy, providing that
patients are effectively followed-up after wide excision of their primary tumor by regular clinical examination, preferably with ultrasound surveillance. There has been a reluctance to disclose and
discuss the arguments against SLNB in melanoma, and it has even
been suggested that “… to deny the patient the opportunity of a
successful SLNB may have medicolegal consequences.”16
Prognostic false-positivity in the SN in melanoma is potentially
the most serious error in the SLNB hypothesis, a suggestion that
advocates of the procedure refuse to consider at present. Some
tiny deposits of melanoma in the subcapsular sinus of SNs are
of no adverse prognostic significance and are destined for dormancy or destruction. Such patients are at risk for being wrongly
up-staged with the inevitable consequences described. This risk
has been compounded by the 2009 American Joint Committee on Cancer melanoma staging classification, which classifies
all micrometastasis in the SN, including solitary tumor cells, as
node-positive.17 Inevitably, this change, which is not evidencebased, will result in more unnecessary completion lymphadenectomies and will not enrich the pool of patients most suitable for
studies of adjuvant therapy.
This commentary highlights clinical, biological, and ethical issues relating to the initial treatment of melanoma. The term
standard of care has no authenticity in medicine.18 Physicians and
patients should focus on reality rather than presentation.
References
1 Voit C, van Akkooi AC, Schafer-Hesterberg G, et al. Rotterdam
criteria for sentinel node (SN) tumour burden and the accuracy of ultrasound (US)-guided fine-needle aspiration cytology
SKINmed. 2010;8:222–224
224
(FNAC): can US-guided FNAC replace SN staging in patients with
melanoma? J Clin Oncol. 2009;27:4994–5000.
2 Morton DL, Thompson JF, Cochran AJ, et al. Sentinel-node
biopsy or nodal observation in melanoma. N Engl J Med.
2006;355:1307–1317.
3 Thomas JM. Prognostic false-positivity of the sentinel node in
melanoma. Nat Clin Pract Oncol. 2008;5:18–23.
4 Thomas JM. Concerns relating to the conduct and statistical
analysis of the Multicenter Selective Lymphadenopathy Trial
(MSLT-1) in patients with melanoma. J Plast Reconstr Aesthet
Surg. 2009;62:442–446.
5 Morton DL, Hoon DSB, Cochran AJ, et al. Lymphatic mapping
and sentinel lymphadenectomy for early-stage melanoma. Ann
Surg. 2003;238:538–549.
6 Langer I, Guller U, Viehl CT. Axillary lymph node dissection for sentinel lymph node metastases may be safely omitted in early-stage
breast cancer patients. Ann Surg Oncol. 2009;16:3366–3374.
7 van Akkooi AC, de Wilt JH, Verhoef C, et al. Clinical relevance
of melanoma micrometastases (<0.1 mm) in sentinel nodes:
are these nodes to be considered negative? Ann Oncol.
2006;17:1578–1585.
8 Murughar P, Azawi K, Humzah D. The sleeper cells: delayed
sentinel lymph node biopsy. J Plast Reconstr Aesthet Surg.
2007;60:1357–1360.
9 Cochran AJ, Thompson JF. Lymphatic mapping in sentinel node
biopsy: the data unchanged by speculation. Arch Dermatol.
2008;144:687–689.
10 Morton DL, Elashoff R. Sentinel node biopsy: facts to clear the
alleged clouds. Arch Dermatol. 2008;144:685–687.
11 Cochran AJ, Ohsie SJ, Binder SW. Pathology of the sentinel
node. Curr Opin Oncol. 2008;20:190–195.
12 Balch CM, Morton DL, Gershenwald JE, et al. Sentinel node biopsy and standard of care for melanoma. J Am Acad Dermatol.
2009;60:872–875.
13 Sondak VK. Nonsentinel node metatases in melanoma: do they
reflect the biology of the tumour, the lymph node or the surgeon? Ann Surg Oncol. 2009;16:2965–2967.
14 Kretschmer L, Thomas KM, Peeters S, et al. Postoperative morbidity of lymph node excision for cutaneous melanoma-sentinel
lymphonodectomy versus complete regional lymph node dissection. Melanoma Res. 2008;18:16–21.
15 Estourgie SH, Nieweg OE, Valdes Olmos RA, et al. Review
and evaluation of sentinel node procedures in 250 melanoma
patients with a median follow-up of 6 years. Ann Surg Oncol.
2003;10:681–688.
16 Thompson JF, Scolyer RA, Kefford RF, et al. Melanoma management in 2007. Aust Fam Physician. 2007;36:487–488.
17 Balch CM, Gershenwald JE, Soong SJ, et al. Final version of
2009 AJCC melanoma staging and classification. J Clin Oncol.
2009;27:6199–6206.
18 Strauss DC, Thomas JM. What does the medical profession
mean by “standard of care”? J Clin Oncol. 2009;27:e192–e193.
Sentinel Lymph Node Biopsy in Melanoma
July/August 2010
Volume 8 • Issue 4
COMMENTARY
Sentinel Lymph Node Biopsy in Melanoma:
Coping With Incomplete Information;
Call for Further Investigations
W. Clark Lambert, MD, PhD
T
he commentary in this issue of SKINmed, “Sentinel
Lymph Node Biopsy in Melanoma: The Gulf Between
Presentation and Reality,”1 makes a very valid point:
that in evaluating sentinel lymph node biopsies (SLNBs), statistical analyses performed to date have lumped together all positive biopsies, regardless of whether only a few cells (ie, a “micrometastasis”) or larger masses of tumor cells have been detected.
Some or all of those biopsies read as positive on the basis of only
small tumor volumes may not be prognostically significant, and
this may, in turn, lead to unnecessary surgery to remove whole
fields of lymph nodes with attendant morbidity.
Limitations
This may also limit the value of SNLBs as a starter. The author correctly points out that breast surgeons have recommended that small
metastases in lymph node biopsies may not be considered significant, and may not recommend additional surgery in some instances.
It may also be self-serving for the physician to recommend
SLNB, as well as additional surgery, if the biopsy is positive, even
with a micrometastasis. He or she becomes more important, and
he or a colleague gets paid for the surgery.
The problem is that no one knows whether such micrometastases in SNLBs of melanomas can or should be ignored. The
studies simply have not been performed as of this date. Thus, the
physicians recommending further surgery after detecting such
metastases may in fact be benefitting the patient, even if they are
not without bias in recommending this course of action.
Recommendations
What is needed are additional studies to examine the point raised
by the author. Until these are done, it may indeed be proper to
advise patients differently who face the prospect of a possible
SLNB. They are indeed “frightened and vulnerable,” but with
good reason. Thus, a recommendation for more restraint in advising the patient needs to be carefully considered.
It may be appropriate for the patient to be advised differently before
giving consent for an SNLB. It may also be advisable for the patient
to be advised differently if a micrometastasis is found. The patient
should be told objectively of the potential risks and benefits at each
step, allowing them to make decisions based on the best evidence
available. They may well still opt for the procedure, and then may or
may not opt for further surgery if a micrometastasis is found.
One of the major problems with SNLB is that it is still relatively
new, and much relevant data have not yet been compiled and
analyzed. We need to give patients our best information and advice in the meantime.
Conclusions
Knowing what I know, I would probably accept SNLB, even though
I may well have less faith in it than the physician who recommended
it to me. If a metastasis is found it may be larger than a micrometastasis, making the decision for further surgery less challenging. If I
were a patient in this situation, even if informed that SNLB has value
only as a staging procedure, I might opt for it simply so that I could
have better knowledge of my prognosis. This may not only affect my
anxiety level, but also influence decisions such as whether to have
more children or to hurry up in writing that “great American novel.”
We need to give patients the best advice we can and allow them
to make such decisions. Most importantly, as much as possible,
we need to remove our own self interests from all of this, as the
accompanying commentary states very clearly. I know a name
for this, it is called “honesty.”
Reference
1 Thomas JM. Sentinel lymph node biopsy in melanoma: the gulf
between presentation and reality. SKINmed. 2010;8:222–224.
From the Departments of Pathology and Dermatology, UMDNJ-New Jersey Medical School, Newark, NJ
Address for Correspondence: W. Clark Lambert, MD, PhD, Room C520 MSB, UMDNJ-NJMS, 185 South Orange Avenue, Newark, NJ 07103 •
E-mail: [email protected]
SKINmed. 2010;8:226
226
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
Volume 8 • Issue 4
New Therapy Update
William Abramovits, MD; Aditya K. Gupta, MD, Section Editors
Zyclara (Imiquimod) Cream, 3.75%
Aditya K. Gupta, MD;1,2 Elizabeth A. Cooper, HBSc;2 William Abramovits, MD3
Description
Zyclara has recently been approved by both the US Food and
Drug Administration (FDA) and Health Canada as a new topical therapy for multiple actinic keratoses (AKs) of the face and
balding scalp. Topical imiquimod 5% cream (Aldara cream, 5%)
has been available for many years as a 25 cm2 field treatment for
AKs. Dosage uses application 2 times per week for periods of 8
to 10 hours for 16 weeks.1,2 The lower concentration of imiquimod in Zyclara allows for a daily-use schedule and application
over larger areas than the 5% cream, with a much shorter application period of 6 weeks.
It may also increase antigen-presenting cell antigen presentation
to T cells, leading to a strong T-helper type I immune response.3
Pharmacokinetics
Mean peak serum drug concentration for patients with AK
(n=17) was approximately 0.323 ng/mL at the end of a 3-week
treatment period, using once-daily applications of Zyclara (2
packets, 18.75 mg) to the entire face and/or balding scalp.4,5
Steady state levels were achieved in 2 weeks, and the Tmax ranged
from 6 to 9 hours.4
Clinical Studies
AK lesions appear in areas of sun-damaged skin and have some
potential to transform into squamous cell carcinoma, although
there are currently no markers to indicate which lesions may become malignant.3 Treatment of all AKs is therefore prudent in
prevention of more serious outcomes. The mode of action of
imiquimod is stimulation of immune response to destroy abnormal cells within the field of drug application. The resultant immune response to imiquimod can also reveal subclinical lesions
not previously detected in the sun-damaged areas, providing
effective early destruction that may help sustain clearance rates
post-treatment.3 Overall, imiquimod cream provides good field
destruction of multiple AKs.
Clinical efficacy was studied for both a 2.5% and a 3.75% formulation of imiquimod (Zyclara), compared with placebo (identical
excipients to the active formulations). The treatment regimen for
all arms was once-daily application for two 2-week cycles, with
a 2-week no-treatment interval between cycles.6 Eligible patients
had 5 to 20 visible or palpable AK lesions in an area >25 cm2 on
either the face or the balding scalp, but not both. Patients applied cream to either the total face or the total balding scalp, but
not both, as selected by the investigator. A total of 479 patients
were enrolled in the trial (3.75% cream = 160 patients; 2.5%
cream = 160 patients; and placebo = 159 patients).
Clinical Pharmacology
Efficacy
Mechanism of Action
Although the mechanism of action of imiquimod has not been
studied or conclusively defined in AK, there are many known actions produced by imiquimod that may be involved. Imiquimod
is a toll-like receptor-7 agonist and has been shown to induce the
release of interferon-a and other cytokines from human monophages/macrophages and keratinocytes in vitro.3,4 The upregulation of cytokines may enhance apoptosis of neoplastic epidermal
cells and upregulation of DNA repair enzymes after UV damage.3
Final efficacy was evaluated 8 weeks after the end of treatment.6 Primary efficacy measured was complete clearance, defined as proportion of patients with zero lesions in the treatment area. Complete
clearance was found in 35.6% of patients in the Zyclara group, vs
30.6% in the 2.5% imiquimod group and 6.3% in the placebo
group. Both active formulations showed significantly higher complete clearance than placebo (P<.001 for pairwise comparison vs
placebo). Considering partial clearance (≥75% reduction in AK
lesions compared with baseline), the clearance rates were 59.4%
for Zyclara, 48.1% for 2.5% imiquimod, and 22.6% for placebo.
From the Department of Medicine, University of Toronto, Toronto, Canada;1 Mediprobe Research Inc, London, Ontario, Canada;2 and the
Department of Medicine, Baylor University Medical Center, and Department of Dermatology & Family Practice, The University of Texas
Southwestern Medical School, Dallas, Dermatology Treatment and Research Center, Dallas, TX3
Address for Correspondence: Elizabeth A. Cooper, HBSc, Mediprobe Research Inc, 645 Windermere Road, London, Ontario, N5X 2P1 Canada •
E-mail: [email protected]
SKINmed. 2010;8:227–229
227
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
NEW THERAPY UPDATE
Table. Local Skin Reactions in the Treatment Area as Assessed by the Investigator
Zyclara
(n=160)
Placebo
(n=159)
All Grades
Severe
All Grades
Severe
Erythema
154 (96%)
40 (25%)
124 (78%)
0 (0%)
Edema
120 (75%)
9 (6%)
31 (19%)
0 (0%)
Weeping/exudate
81 (51%)
9 (6%)
6 (4%)
0 (0%)
Flaking/scaling/dryness
147 (92%)
13 (8%)
123 (77%)
2 (1%)
Scabbing/crusting
149 (93%)
22 (14%)
72 (45%)
0 (0%)
Erosion/ulceration
99 (62%)
17 (11%)
14 (9%)
0 (0%)
Again, both active treatment groups showed significantly higher
partial clearance rates than placebo (P<.001), and Zyclara showed
higher partial clearance than the 2.5% imiquimod (P=.047).
Patients receiving Zyclara showed a higher frequency of need for
a rest period during treatment, with 10.6% (17 of 160) requiring at least 1 treatment rest period, compared with 6.9% in the
2.5% cream group and 0% in placebo.6
Safety/Adverse Events
In postmarketing surveillance of imiquimod 5% cream, rare reports have been received of either onset or exacerbation of autoimmune conditions (including thyroiditis, multiple sclerosis,
spondyloarthropathy, psoriasis, and ulcerative colitis).4
No unexpected safety issues were found in any group, and vital
sign measure/physical examination did not show any significant
safety issues during the trial.6 The most frequently reported adverse events (AEs) on nonapplication sites with use of 3.75% imiquimod were headache (6.3%), fatigue (4.4%), nausea (3.8%),
and pyrexia (3.1%). Treatment-related AEs were reported in
19.4% of the 3.75% cream group (31 of 160), compared with
11.9% in the 2.5% cream group and 2.5% in the placebo group.
One case of severe diarrhea was reported as probably related to
use of Zyclara.
Local skin reactions (LSRs) were independently collected of
other AEs/application site reactions (ASRs) and were reported
in almost all patients using Zyclara4–6 (Table). Severe erythema
was found in 25.2% of Zyclara patients, with severe scabbing/
crusting in 13.8%. The other LSR groups had severe grades in
approximately 5% to 10% of patients (Table). The mean erythema score increased during the treatment periods but was reduced below baseline level during the follow-up period for both
imiquimod groups.6
ASRs were reported in 10.6% (17 of 160) of the 3.75% cream
group, compared with 6.3% in the 2.5% cream group and 1.3%
in the placebo group.6 Pruritus was the most frequently reported
ASR in the Zyclara group (4.4%), followed by irritation (3.1%),
pain (3.1%), and swelling (1.3%).
Of the 6 patients who discontinued the study due to AEs, 2
were in the Zyclara group.6 Only 2 AEs leading to discontinuation were considered related to study treatment: 1 Zyclara patient had headache, fatigue, pain, and application site pain, and
1 placebo patient had headache.
SKINmed. 2010;8:227–229
A second Zyclara study used a similar design to investigate two
3-week application cycles, with a 3-week no-treatment interval between cycles, vs 2.5% imiquimod cream and placebo.7
Although the safety profiles were similar between 2-week and
3-week cycles, the increased duration of use did not appear to
increase efficacy compared with the 2-week cycle.
Use of imiquimod may reveal the presence of subclinical AKs
during therapy. This was noted as a transient increase in AK
counts during trial therapy and did not reduce treatment efficacy.6,7 This effect has been noted in previous imiquimod studies.
The clearance of subclinical lesions may result in sustained lesion
clearance and could be an advantage compared with other treatment methods that do not reveal subclinical lesions, reducing
the need for future treatment.
Almost half of patients treated with Zyclara (78 of 160) were 65
years and older. No overall differences in safety or effectiveness
were detected in these patients vs younger patients, although
greater sensitivity of some older individuals cannot be ruled out.
Indication, Dosage, and Administration
Zyclara is indicated for the topical field (area) treatment of multiple clinically typical, visible, or palpable actinic keratoses, presented at start of therapy or revealed during therapy, of the face
or balding scalp in adults.4,5 The cream is provided in 250-mg
single-dose packets containing 9.4 mg of imiquimod (3.75%
w/w). Up to 2 packets are applied once daily before bedtime
228
Zyclara (Imiquimod) Cream, 3.75%
July/August 2010
NEW THERAPY UPDATE
to the affected treatment field for 2 treatment cycles of 2 weeks
each, separated by a 2-week no-treatment period. Prior to application, patients should wash the treatment area with mild
soap and water and allow the area to dry thoroughly. A thin
film of imiquimod should be applied to the entire treatment area
and rubbed in until the cream is no longer visible, and hands
should be washed after application. Use near eyes, lips, and nostrils should be avoided, and the application site should not be
occluded. Zyclara should be left on the skin for approximately
8 hours overnight and then removed by washing the area and
hands with mild soap and water.
Most patients will experience some LSRs, which may include erythema, scabbing/crusting, flaking/scaling/dryness, and edema.
These LSRs may require an interruption of dosing for several
days if the reaction is severe or produces intolerable patient discomfort. Treatment may be continued after consulting with a
physician. The dosage period should not be prolonged to make
up for missed doses and rest periods.
Safety has not been established in pregnant women, and use
should be restricted to cases where the potential benefit outweighs the potential risk to the fetus. It is not known whether
topical imiquimod is excreted in human breast milk, therefore
caution should be used in nursing women.
The maximum treated area tested is approximately 200 cm2.
Safety and efficacy of application over larger areas has not been
established and is not recommended.
Discussion
The new imiquimod formulation provides a lower potency preparation and a less taxing protocol of therapy for the patient (2 weeks on,
2 weeks off, 2 weeks on, and stop). This results in a shorter course of
therapy, with no loss of efficacy in eradicating actinic keratosis. This
formulation should be a welcome addition to the dermatologist’s
armamentarium for the management of actinic damage.
References
1 Aldara (imiquimod) cream, 5% [product monograph]. London,
ON, Canada: Graceway Pharmaceuticals; March 04, 2009.
Safety and Precautions
Some patients may experience flu-like signs and symptoms including fatigue, nausea, fever, myalgias, arthralgias, and chills preceding or accompanying LSRs. A patient assessment should be
considered, and dosing may need to be interrupted or adjusted.
Patients should minimize or avoid natural or artificial sunlight
exposure during treatment, due to concerns of heightened sunburn susceptibility and reports of shortened tumor formation
time in an animal photocarcinogenicity study. Protective clothing (eg, hat) should be used during treatment.
Safety in immunosuppressed patients has not been established,
and imiquimod should be used with caution in patients with preexisting autoimmune conditions including thyroiditis, multiple
sclerosis, spondyloarthropathy, psoriasis, and ulcerative colitis.
SKINmed. 2010;8:227–229
229
2 Aldara cream, 5% (imiquimod) [prescribing information]. Bristol,
TN: Graceway Pharmaceuticals, LLC; April 2009.
3 Gaspari AA, Tyring SK, Rosen T. Beyond a decade of 5% imiquimod topical therapy. J Drugs Dermatol. 2009;8:467–474.
4 Zyclara (imiquimod) cream, 3.75% [product monograph]. London,
ON, Canada: Graceway Pharmaceuticals; December 31, 2009.
5 Zyclara (imiquimod) cream, 3.75% [product monograph]. Bristol, TN: Graceway Pharmaceuticals, LLC; March 2010.
6 Swanson N, Abramovits W, Berman B, et al. Imiquimod 2.5%
and 3.75% for the treatment of actinic keratoses: results of
two placebo-controlled studies of daily application to the face
and balding scalp for two 2-week cycles. J Am Acad Dermatol.
2010;62:582–590.
7 Hanke CW, Beer KR, Stockfleth E, et al. Imiquimod 2.5% and 3.75%
for the treatment of actinic keratoses: results of two placebo-controlled studies of daily application to the face and balding scalp for
two 3-week cycles. J Am Acad Dermatol. 2010;62:573–581.
Zyclara (Imiquimod) Cream, 3.75%
July/August 2010
Volume 8 • Issue 4
Perils of Dermatopathology
W. Clark Lambert, MD, PhD, Editor
Newtonian Dermatopathology:
When the Reaction to a Lesion Obfuscates the Diagnosis
Javier Rojas, MD;1 Navér Sarkissian, MD, PhD;1,2,3 Debra S. Heller, MD;1 W. Clark Lambert, MD, PhD1,2,3
S
ir Isaac Newton’s third law states that for every action there is
an equal and opposite reaction.1 The same may be said of many
biological phenomena, except that the reaction is rarely equal
and may, in fact, vastly exceed the impetus. Causes include trauma,
stasis, and masses, including tumors. Many of the latter, such as desmoplastic basal cell carcinomas and melanomas, induce marked connective tissue reactions. Other such reactive phenomena include vascular reactions, several types of inflammation, and reactions including
granulation tissue. In the preceding column in this series we examined
ways in which the latter, on the surface, may obfuscate diagnosis of a
skin or mucus membrane biopsy.2 We now consider how such reactions in deeper tissues may produce similar diagnostic misdirection.
A Masson tumor may occur anywhere a venous thrombus may arise.
It commonly occurs in the hemorrhoidal veins. We have previously
reported its occurrence on the vulva.3
Case 2
A painless, nontender deep nodular lesion on the thigh of a 38-yearold man was biopsied near its edge. Histopathologic examination
showed a somewhat storiform spindle cell lesion (Figure 2), which
was initially suspected to be a possible dermatofibrosarcoma protuberans. A CD34 immunohistochemical stain, which is characteristically positive in dermatofibrosarcoma protuberans, was reactive in
lesional cells (Figure 3), consistent with this diagnosis. This stain also
Case 1
A painless, slightly tender bluish nodule on the upper lip of a 48-yearold human immunodeficiency virus–positive woman was biopsied
and read by the referring pathologist as an angiosarcoma, possibly
Kaposi sarcoma. Review of slides received in consultation showed
a poorly defined nodular lesion composed of numerous dissecting
vascular channels lined by a single layer of prominent endothelial
cells (Figure 1). Further evaluation revealed overlying lichen simplex
chronicus, indicative of chronic trauma or manipulation, and areas
in which the lesion was bounded by a thin remnant of a venous wall.
The final diagnosis was Masson tumor (Masson pseudotumor; intravascular papillary endothelial hyperplasia).
A Masson tumor is formed when a venous thrombus is invaded by
normal reactive endothelial cells attempting to recanalize the lesion. Diagnosis in early lesions is facilitated by the presence of fibrin within the
connective tissue. Diagnosis in later lesions is made by identification of
the remnant of a venous wall at the border of the lesion, but chronic
trauma may cause the thrombus and, therefore, the reactive lesion to
extend beyond the vessel wall in places. Thus the lesion is not actually a
tumor (neoplasm) and is not papillary, and the terms Masson tumor and
intravascular papillary endothelial hyperplasia are not really appropriate.
Figure 1. Case 1: Dissecting endovascular channels (Hematoxylin and eosin stain, magnification ×380).
From the Department of Pathology, Divisions of Anatomic Pathology1 and Dermatopathology2 and the Department of Dermatology,3 University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, NJ
Address for Correspondence: W. Clark Lambert, MD, PhD, Room C520 MSB, UMDNJ-NJMS, 185 South Orange Avenue, Newark, NJ 07103 •
E-mail: [email protected]
SKINmed. 2010;8:231–233
231
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
Perils of Dermatopathology
Figure 2. Case 2: Spindle cells forming a vaguely storiform
pattern (Hematoxylin and eosin stain, magnification ×380).
Figure 3. Case 2: CD34 immunohistochemical staining of
spindle cell lesion (magnification ×380).
histochemical staining for smooth muscle actin within this portion
of the lesion (Figure 4). The final diagnosis was leiomyoma with a
peripheral vascular reaction.
Although leiomyomas may be tender, many lack this characteristic.
Notifying the pathologist that the lesion was biopsied at its periphery may have helped in its evaluation. Cutaneous leiomyomas may
arise from multiple sites within the skin and subcutis, including
blood vessels and pilar erectus muscles, and may occur anywhere.
Discussion
The issue of whether a lesion or tissue reacting to it is biopsied is very
much a matter of discussion between the dermatopathologist and
the clinician, because, unless an excisional biopsy is performed, the
clinician chooses which portion of the lesion to biopsy. In addition
to obtaining diagnostic tissue, this decision is influenced by factors
such as cosmetic result and other disabilities resulting from the biopsy. If a lesion is biopsied at its edge, or at another special site, this
information needs to be communicated to the pathologist.
Figure 4. Case 2: Smooth muscle actin immunohistochemical
staining of different area of spindle cell lesion (magnification ×380).
decorates endothelial cells, however, and immunohistochemical staining with CD31, which stains only the latter, was also positive. Further
examination of deeper levels revealed a part of the lesion consistent
with a leiomyoma, and this diagnosis was confirmed by immunoSKINmed. 2010;8:231–233
As the dermatopathology laboratory continues to become more remote from the clinical setting, the lines of communication between
clinician and dermatopathologist may become more tenuous. Use
of electronic medical records, whatever their other advantages and
limitations, may facilitate this process, provided that a complete and
accurate record of the biopsy procedure is entered into the patient’s
record and this portion of the chart is transmitted to the pathologist.
232
Newtonian Dermatopathology
July/August 2010
Perils of Dermatopathology
References
1 Guicciardini N. Isaac Newton on Mathematical Certainty and
Method. Cambridge, MA: The MIT Press; 2009:422.
2 Sarkissian N, Patel PP, Fleeger EJ, et al. Anatomically correct,
histopathologically correct, diagnostically disastrous biopsies.
Skinmed. 2010;8:98–99.
3 Maghari A, Lambert WC, Sherman N, et al. Intravascular papillary endothelial hyperplasia (Masson’s tumor) manifesting as a
cystic vulvar lesion. J Lower Gen Tract D. In press.
HISTORICAL DIAGNOSIS & Treatment
Diagnosis and treatments have advanced over the past century. This feature depicts conditions from a collection of steroptic cards published
in 1910 by The Stereoscopic Skin Clinic, by Dr. S. I. Rainforth.
Synonyms: Pediculosis vestimenti seu vestimentorum; Phthiriasis capitis; Vagabond’s disease; Body-lousiness.
DIAGNOSIS: The discovery of a louse or the nits in the seams of the underclothes is sufficient, but even in their absence,—as
when the patient has recently changed his clothing,—the distribution of the lesions, the punctiform hemorrhages, parallel excoriations and pigmentation make the diagnosis comparatively easy.
TREATMENT: Attention should be directed mainly toward the clothing and bed linen, which are to be immersed in boiling water
or subjected to a temperature of 165° or 170° F. in an oven. To destroy any nits that may be present on the body, the latter
should be anointed with a 20 per cent ointment of staphisagria, made by adding two drachms of the freshly powdered seed to
the ounce of hot lard, which is then to be strained and cooled.
SKINmed. 2010;8:231–233
233
Newtonian Dermatopathology
Aklarus Multi-Wavelength
Phototherapy System
The Aklarus is a multi-wavelength phototherapy
system FDA cleared and medically proven
to provide a drug-free and pain-free treatment
for acne, photodamaged skin and wrinkles.
HA90D Chair
Shown with
optional contour
seat and back
and removable
fold-down
armrests.
See Before and After
Photos and Video at
www.HillTherapy.com
The Versatile, Affordable
Chair for Dermatology!
$9650
$8150
Includes cart,
treatment arm
and accessories
Working closely with
a leading dermatologist,
Hill Laboratories has
designed the ideal chair
for dermatology...
• Electrically controlled height - 22”-35”
• Standard width - 26” or 24”
• Length - 70"
• 2.5” Foam top
• Electric Lift back
• Full-width foot section-locks at any angle
• Adjustable headrest
• Standard Flat Cushions
• Paper retainer on headpiece
• Paper roll bracket on foot cushion
• One-year warranty
Starts at
$3495
HA90D fully reclined with
matching stool
Since 1945
Optional
Tapered-Top
& headpiece
90ALB for Aesthetics
starts
$3175
Call Today!
1-877-445-5020
www.HillLabs.com
Optional
Removable, FoldDown Armrests
July/August 2010
Volume 8 • Issue 4
Congress Report
Marcia Ramos-e-Silva, MD, PhD, Section Editor
Highlights From the 7th EADV Spring Symposium,
Cavtat, Croatia, May 13–16, 2010
Jasna Lipozenčić, MD, PhD, Chairperson
T
he 7th Spring Symposium of the European Academy of
Dermatology and Venereology (EADV) took place with
more than 2000 participants, gathering at the Congress
Centre of the Hotel Croatia in Cavtat (antique Epidaurus) near
Dubrovnik (antique Ragusa and called “South Pearl of the Adriatic Sea”). The locale is located on the beautiful Croatian coast,
which boasts of 1244 islands and peninsula sand overwhelmed
with rich and unique vegetation.
matology and Venereology” and by Professor Stefan Beissert entitled
“Nowdays and Future of Dermatovenereological Research.”
Background
This 7th EADV Spring Symposium was organized under the high
auspice of the president of the Republic of Croatia, Professor Ivo
Josipović. Participants came from 78 countries worldwide.
We wanted to achieve harmony in dermatology and venereology,
and we succeeded with a successful blend of the scientific program
and the social events. The president of EADV, Professor Andreas
Katsambas, expressed his sincere thanks to each and every one of the
organizers and board members for their support and valuable input
for Cavtat (Figure 1). He congratulated them for “their amazing
job in organizing such a terrific meeting.” There were positive comments from the many who attended the meeting, giving proof of its
success. “Everything was more than perfect” (Figure 2).
Figure 1. Jasna Lipozenčić (center, left) and Andreas Katsambas (center, right) during the welcome reception with Penny
Emmanouil (far left) and Electra Nikolaidau (far right).
In the opening ceremony, the traditional good spirit of Dubrovnik
was demonstrated by the folk dance Ensemble Linđo with two
performances and by the renaissance Ragvseum Ensemble Plazzarius with three presentations: La Magdalena, Branle, and Troto.
The biggest confirmation for the organizers of the 7th EADV
Spring Symposium was the compliment paid that we have done
for Croatian dermatology today just as the famous Academician
Franjo Kogoj (1894–1983) did in his time.
Scientific Program
The highlights of the Symposium were the opening lectures by Professor Amir Muzur entitled “Milestones in the History of Croatian Der-
Figure 2. From left to right: Bruce Thiers, Stefan Beissert, Angela
Robinson, and Steingrimur Davidsson at the opening reception.
From the University Department of Dermatology and Venereology, Zagreb University Hospital Center and School of Medicine, Zagreb, Croatia
Address for Correspondence: Jasna Lipozenčić, MD, PhD, Professor, University Department of Dermatology and Venereology, Zagreb University
Hospital Center and School of Medicine, Salata 4, 10000 Zagreb, Croatia • E-mail: [email protected]
SKINmed. 2010;8:235–237
235
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
CONGRESS REPORT
Friday, May 14, 2010, had a packed program with:
oPractical Considerations in Cutaneous Lymphomas
oHair and Nail Diseases
•Free Communications
•Symposia
•Case Report Sessions
•Scholarship Ceremony
oAcne and Rosacea
•Continuing Medical Education/Continuing Professional
Development
oBullous Diseases Revisited
oSkin Diseases in the Mediterranean
•Media Committee
•Workshops
•Scientific Programming Committee
oApproaches to Wound Healing and Leg Vein Treatment
•Fostering Trainee Committee
oManagement of Chronic Urticaria
•Euromelanoma Meeting
oVitiligo and Pigmentary Disorders
•Fostering Skill Specialists Committee
On Saturday, May 15, 2010, the program was just as exciting:
•Statutes Committee
•Forum Session
•Free Communications
•Update on Melanoma Management
•Poster Forum (awards given to presenters from Denmark,
Iran, and Moldavia)
•Psoriasis
•Case Report Sessions
•Skin Conditions and Endocrinology
•President’s Symposium
•Workshops
oMetabolic Comorbidities and Psoriasis
oSyphilis: The Resurgence of an Old Problem
oPearls in Dermatological Surgery
oNovel Drugs and Adverse Reactions in Dermatovenereology
•Plenary Sessions
oCumulative Experience From the Sexually Transmitted
Disease Clinic
oMelanocytes and Melanogenesis: News to Tell
•Symposia
oAllergy, Environment, and the Skin
oSkin Clues of Systemic and Connective Tissue Diseases
oBrain-Skin-Axis
oImmunotherapy for Skin Diseases in the Molecular Era
•Satellite Symposia
oPhotodermatology
oAtopic Dermatitis
oNonsurgical Treatment of Non-Melanoma Skin Cancer
oAzithromycin in Dermatovenereology
o Optimizing Psoriasis Treatment Outcomes With Adalimumab
oDilemmas in Prevention of Superinfections in Dermatology
oGeneral Topics in Sirolimus
oNew Approach in Sun Protection for Allergy-Prone Skin
oTopical Retinoids in Treatment of Acne
oHuman Immunodeficiency Virus Infection in Dermatology
oDermatomycoses
•Workshops
oLaser Therapy
oSexually Transmitted Infections (STIs)
oPediatric Dermatology
oWhat’s New in Photoprotection in 2010?
oPsyche and the Skin
oThe Human Papillomavirus (HPV) Vaccine—Impossible Without Dermatology and Venereology
oHPV in Dermatovenereology
oFacial Rejuvenation
•Plenary Sessions
•Focus Sessions
oBotulinum Toxin: Use and Misuse
oNovel Imaging Techniques for Diagnosis of Skin Disorders
oThe Skin and Internal Malignancy
oExogenous Acne Revisited
oTest Yourself in Dermoscopy
oSTIs: Progress, Challenges, and Opportunities
•Focus Sessions
•Forum Sessions
SKINmed. 2010;8:235–237
236
Highlights From the 7th EADV Spring Symposium
July/August 2010
CONGRESS REPORT
oManaging Adult-Onset Acne
oSTIs and Adolescence
oOffice-Based Cryotherapy
oPractical Consideration in Treating Cutaneous Lupus
Erythematosus
•Forum Sessions
oBiologicals in Psoriasis: When and Which to Use?
oDermatoses in the Genital Area
Closing Ceremony
•What’s New Sessions
Figure 3. From left to right: Jasna Lipozenčić and Andreas
Katsambas hand over the EADV flag to Peter Arenberger, the
next EADV Spring Symposium chairperson. The next EADV
Spring Symposium will be held April 14–17, 2011, at Karlovy
Vary, Czech Republic.
oWhat’s New in Clinical Dermatology
oWhat’s New in Venereology
oWhat’s New in Pediatric Dermatology
Conclusions
As one participant remarked: “It was a wonderful meeting! Perfectly
organized, good science, nice atmosphere. I not only enjoyed the
EADV Spring Symposium but I also found the organization to be
perfect” (Figure 3).
VINTAGE LABEL
Courtesy of BuyEnlarge, Philadelphia, PA
SKINmed. 2010;8:235–237
237
Highlights From the 7th EADV Spring Symposium
July/August 2010
Volume 8 • Issue 4
CASE STUDY
Vesna Petronic-Rosic, MD, MSc, Section Editor
Segmental Cutaneous Piloleiomyomata
Robert H. Cook-Norris, MD;1,2 Adrian O. Rodriguez, MD;2 Bradley T. Kovach, MD;2
Alan S. Boyd, MD;2 John A. Zic, MD2
A 60-year-old white man seen in consultation for “chronic zoster” presented with a 30-year history of persistent, painful cutaneous lesions
on his right flank. His pain was episodic and exacerbated by physical touch and emotional stress. He denied lesion sensitivity to cold or
heat. Application of capsaicin cream had been without benefit. There was no family history of similar lesions. Physical examination revealed
multiple, pink to violaceous, firm, tender nodules (3–7 mm) arising on the right flank in a segmental distribution (Figure 1). No other
lesions were noted. A biopsy specimen showed findings of dermal proliferation of spindle-shaped cells with eosinophilic cytoplasm and
blunt-ended nuclei without cytologic atypia (Figure 2A and Figure 2B). All of these findings were compatible with a diagnosis of segmental
cutaneous piloleiomyomata. He declined surgical or medical intervention.
C
utaneous piloleiomyomata (CPL) are benign smooth
muscle tumors most commonly presenting as multiple,
grouped, red to dark brown, firm, dermal papules and nod-
Figure 1. Multiple pink nodules in a segmental distribution on
the right flank.
ules, typically no larger than 1.5 cm.1,2 An unusual and suggestive
clinical finding is sharp, shooting pain, resulting from cold contact,
emotional stress, or physical trauma.3 CPL tends to occur in the
second and third decades without sex predilection.3 Common lesion locations include the extensor surfaces of the extremities, trunk,
and face. Linear or segmental distribution is rare. Some authors
have described the segmental pattern as zosteriform leiomyomatosis but, as stated by some experts,4 the term segmental is preferred
since the lesions do not follow a true dermatomal distribution. The
pathophysiology of segmental cutaneous leiomyomatosis (SCL) is
unknown but may represent an autosomal dominant genetic mosaicism.5 SCL has since been divided into two distinct segmental
patterns, type 1 and 2, based on a loss of wild-type allele(s) following
a mutational event.4 Type 1 SCL manifests as purely segmental lesions reflecting heterozygosity for the underlying mutation. In type
2 SCL, a postzygotic mutation in a heterozygous embryo results in a
loss of heterozygosity, leading to a segmental pattern superimposed
on disseminated areas of involvement. Our patient, with only segmental lesions, would best fit type 1 SCL. The etiology of the pain
associated with CPL is unclear but thought to be either the result of
smooth muscle contraction, an increased number of nerve bundles,
or pressure on surrounding cutaneous nerves.3,6
Treatment depends on the severity of symptoms and extent of skin
involvement. Surgical excision is possible in patients with limited
skin involvement. Recurrence is common, however, particularly in
patients with multiple lesions.1 If extensive involvement is present,
From the Department of Dermatology, Mayo Clinic College of Medicine, Rochester, MN;1 and the Department of Dermatology, Vanderbilt
University Medical Center, Nashville, TN2
Address for Correspondence: Alan S. Boyd, MD, Vanderbilt Health - One Hundred Oaks, 719 Thompson Lane, Suite 26300, Nashville, TN 37204 •
E-mail: [email protected]
SKINmed. 2010;8:238–239
238
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
CASE STUDY
medications including nifedipine, nitroglycerin, and phenoxybenzamine have been used with limited success.7
This rare neoplasm is likely clinically underrecognized,7 particularly when presenting in a segmental distribution. Increased awareness of this unusual presentation is important due to the associated risks of uterine fibroids and aggressive renal malignancies in
these patients and their offspring if germline mosaicism is present.
References
1 Smith CG, Glaser DA, Leonardi C. Zosteriform multiple leiomyomas. J Am Acad Dermatol. 1998;38:272–273.
2 Sahoo B, Radotra BD, Kaur I, et al. Zosteriform pilar leiomyoma.
J Dermatol. 2001;28:759–761.
3 Agarwalla A, Thakur A, Jacob M, et al. Zosteriform and disseminated lesions in a cutaneous leiomyoma. Acta Derm Venereol.
2000;80:446–458.
4 Lang K, Reifenberger J, Ruzicka T, et al. Type 1 segmental cutaneous leiomyomatosis. Clin Exp Dermatol. 2002;27:649–650.
5 Happle R. Loss of heterozygosity in human skin. J Am Acad
Dermatol. 1999;41:143–161.
6 Raj S, Calonje E, Kraus M, et al. Cutaneous pilar leiomyoma:
clinicopathologic analysis of 53 lesions in 45 patients. Am J
Dermatopathol. 1997;19:2–9.
7 Holst VA, Junkins-Hopkins JM, Elenitsas R. Cutaneous smooth
muscle neoplasms: clinical features, histologic findings, and
treatment options. J Am Acad Dermatol. 2002;46:477–490.
Figure 2. Photomicrograph showing abundant spindle-shaped
smooth muscle cells (hematoxylin and eosin original magnification [A] ×4 and [B] ×40).
VINTAGE LABEL
Courtesy of BuyEnlarge, Philadelphia, PA
SKINmed. 2010;8:238–239
239
Segmental Cutaneous Piloleiomyomata
July/August 2010
Volume 8 • Issue 4
CASE STUDY
Vulvoperineal Crohn’s Disease:
Response to Metronidazole
Aida Khaled, MD;1 Nadia Ezzine-Sebai, MD;1 Becima Fazaa, MD;1 Faten Zeglaoui, MD;1 Rachida Zermani, MD;2
Mohamed Ridha Kamoun, MD1
A 46-year-old woman with a medical history of chronic juvenile arthritis with bilateral prosthetic hips presented with vulvoperineal ulcerations of
3 years’ duration. There was no diarrhea or recent weight loss. Cutaneous examination showed asymmetrical vulvar edema of the labia minora and
labia majora with deep and linear ulcerations having verrucous borders located on the inguinocrural regions and the buttocks fold (Figure 1). On
physical examination there was bilateral limited mobilization of the hips. A biopsy specimen was taken from the border of the vulvar ulceration
and histologic examination showed under a hyperplasic epidermis an epithelioid granuloma with multinucleated giant cells of the dermis without
caseification. Laboratory analyses and results from chest x-ray were normal. Results for Koch bacilla in the spittle, microbiologic studies (staining
for microorganisms and cultures), and tuberculin intradermoreaction were negative. There was no Crohn’s disease aspect on colonoscopy, and
there was normal small bowel enterography. Systematic intestinal biopsies were also with normal aspect. Based on the clinical data and granulomatous histologic characteristics, the diagnosis of metastatic Crohn’s disease without digestive involvement was obtained. The patient was started on
metronidazole 1 g/d. After 6 months of treatment, there was an almost-complete healing of ulcerations (Figure 2). Treatment was well-tolerated.
V
ulvoperineal involvement in Crohn’s disease is rare
and may constitute the sole manifestation of the disease since it may precede bowel symptoms by several
months to years in 20% of cases. In this localization, treatment
of Crohn’s disease is difficult and not standardized.1 We report a
case of vulvoperineal Crohn’s disease that responded to metronidazole in a 46-year-old woman.
Figure 1. Asymmetric vulvar edema with deep and linear
ulcerations of the inguinocrural regions.
Our patient had cutaneous Crohn’s disease without digestive involvement. Diagnosis may not be obvious if vulvoperineal involvement precedes active bowel disease. Abscesses, draining sinuses,
edema, and sharply demarcated ulceration of the perineum and/
or vulva with usually verrucous borders (as demonstrated by our
case) are the characteristic manifestations of the disease. They are
caused by direct extension from the involved bowel or by granulomas separated from the bowel by normal tissue. The latter form is
usually named metastatic Crohn’s disease. In the absence of digestive involvement, the diagnosis is based on clinical and histologic
aspects and on the absence of other clinical and paraclinical signs
of a specific infection. Because of the scarcity of this topographic
form of Crohn’s disease, and in the absence of randomized essay,
treatment remains not standardized.1 Multiple treatments have
been tried. They include corticosteroids, which can be injected either topically or locally or administered orally. They are often efficient with a frequent corticodependence and multiple side effects,
From the Departments of Dermatology1 and Pathology,2 Charles Nicolle Hospital, Tunis, Tunisia
Address for Correspondence: Aida Khaled, MD, Boulevard 9 Avril, 1006 Tunis, Tunisia • E-mail: [email protected]
SKINmed. 2010;8:240–241
240
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
CASE STUDY
especially for oral steroids. Sulfasalazine may lead to a partial healing of cutaneous lesions but is more efficient in digestive forms.
Immunosuppressive agents such as azathioprine, methotrexate,
and cyclosporine are also efficient, but a long-term treatment may
expose the patient to severe side effects. Immunosuppressive treatments should be indicated for digestive involvement and may be
an ultimate option in resistant forms of vulvoperineal Crohn’s
disease.2 Thalidomide also leads to a satisfactory result in refractory vulvar ulcerations associated with Crohn’s disease.3 Recently,
investigators stated that infliximab is a good therapeutic option for
recalcitrant vulvar Crohn’s disease, but no optimum regimen was
provided.4 Only a few small studies have evaluated metronidazole
for the treatment of Crohn’s disease with low convincing results.
A better activity has been demonstrated in colonic disease and for
perianal fistulas.5 Metronidazole seems to be a good therapeutic
option since it has led to complete healing of perineal lesions with
a persistent result in our patient and in several uncontrolled reports.2,6–9 Its efficiency is due to antibacterial, antiinflammatory,
and immunosuppressive action. As demonstrated by researchers,
the efficacy of metronidazole is dependent on the dose and duration of the treatment. According to the largest follow-up study, the
optimal dose is 20 mg/kg/d and the duration is at least 12 months,
with a maximum duration of 36 months.7 The most commonly
reported side effects are digestive and hematologic, but also reversible sensory peripheral neuropathy.10
References
1 Moyes LH, Glen P, Pickford IR. Perineal metastatic Crohn’s disease: a case report and review of the literature. Ann R Coll Surg
Engl. 2007;89:W1–3.
2 Conscience I, Perceau G, Durlach A, et al. Vulvar involvement in
Crohn’s disease: efficacy of metronidazole. Ann Dermatol Venereol. 2006;133:585–587.
3 Kolivras A, De Maubeuge J, André J, et al. Thalidomide in refractory vulvar ulcerations associated with Crohn’s disease. Dermatology. 2003;206:381–383.
4 Preston PW, Hudson N, Lewis FM. Treatment of vulval Crohn’s
disease with infliximab. Clin Exp Dermatol. 2006;31:378–380.
5 Rubin DT, Kornblunth A. Role of antibiotics in the management
SKINmed. 2010;8:240–241
241
Figure 2. Almost-complete healing of the ulcerations after 6
months of metronidazole treatment.
of inflammatory bowel disease: a review. Rev Gastroenterol Disord. 2005;5(suppl 3):S10–S15.
6 Bernstein LH, Frank MS, Brandt LJ, et al. Healing of perineal Crohn’s disease with metronidazole. Gastroenterology.
1980;79:599.
7 Brandt LJ, Bernstein LH, Boley SJ, et al. Metronidazole therapy
for perineal Crohn’s disease: a follow-up study. Gastroenterology. 1982;83:383–387.
8 Bourrat E, Faure C, Vignon-Pennamen MD, et al. Anitis, vulvar edema and macrocheilitis disclosing Crohn disease in a child: value
of métronidazole. Ann Dermatol Venereol. 1997;124:626–628.
9 Andreani SM, Ratnasingham K, Dang HH, et al. Crohn’s disease
of the vulva. Int J Surg. 2010;8:2–5. Epub 2009 Oct 1.
10 Bernstein LH, Frank MS, Brandt LJ, et al. Healing of perineal Crohn’s disease with metronidazole. Gastroenterology.
1980;79:357–365.
Vulvoperineal Crohn’s Disease
July/August 2010
Volume 8 • Issue 4
CASE STUDY
Folliculocentric Lichen Sclerosus et Atrophicus
David J. Mann, MD;1 Irene J. Vergilis-Kalner, MD;2 Justin R. Wasserman, MD;3 Vesna Petronic-Rosic, MD, MSc4
A 71-year-old black woman presented to the dermatology clinic with a several-year history of increasing numbers of itchy white spots
spreading over the chest, back, elbows, and legs. In 2004, the patient developed clusters of mildly pruritic hypopigmented and depigmented macules on her back and left shoulder along with depigmented flat papules on the lower extremities. The patient’s condition was unresponsive to topical triamcinolone and tacrolimus, after which she was lost to follow-up for 3 years until her entire chest became involved.
Her medical history was significant for hypertension and diabetes mellitus and her medications included amlodipine, aspirin, atorvastatin,
hydrochlorothiazide, irbesartan, and metformin. Family history, social history, and review of systems were unremarkable. Physical examination revealed multiple shiny white perifollicular macules, some with slight scale, on the trunk (Figure 1) and extremities (Figure 2). On the
left shoulder and back were 2 shiny plaques with hypopigmented macules and follicular scale within. Punch biopsies from the abdomen and
back showed atrophy of the epidermis, edema and sclerosis of the papillary dermis, and a patchy lichenoid lymphocytic infiltrate (Figure 3).
Despite inspecting multiple step sections, we were unable to histopathologically confirm the clinically apparent folliculocentric distribution.
Based on the clinical and histological findings, extragenital lichen sclerosus et atrophicus (LSA) was diagnosed and the patient was treated
with clobetasol proprionate 0.05% ointment. While being prepared for narrowband UV-B therapy, a serologic screen revealed antinuclear
antibodies at 1:640 with a homogeneous pattern. A subsequent rheumatologic evaluation was negative for autoimmune disorders; pertinent negative serologies included anti–double-stranded DNA, smooth muscle, mitochondrial, thyroid, and parietal cell antibodies.
L
SA is a chronic inflammatory scarring disease, described
clinically by Hallopeau in 1887 and histologically by
Darier in 1892. The etiology of the disease still remains
unknown. While it can be seen in all age groups, LSA affects
women more commonly than men, with a bimodal peak of incidence: the first occurs between 8 and 13 years of age and the
second between the fifth and sixth decade, both in women with
physiologically low estrogen states.
LSA has a predilection for the anogenital region, seen in approximately 85% of cases. While 15% to 20% of those have extragenital lesions,1 the folliculocentric distribution of the lesions in our
patient is unique and the generalized distribution itself is uncommonly reported and thus less well understood. We report a case of
extragenital LSA that had a folliculocentric and generalized clinical presentation in a patient with antinuclear antibodies.
Discussion
Extragenital LSA without accompanying genital involvement is
uncommon and has been reported in 2.5% to 15% of patients.2,3
Extragenital LSA is histologically identical to the more common
genital presentation of LSA and is characterized by homogenization of papillary dermal collagen, extensive edema, a band-like
lymphocytic infiltrate, and thinning of the overlying epidermis.
Clinically, this results in atrophic or sclerotic ivory white papules, which often coalesce into large plaques that, aside from dryness and itching, are generally asymptomatic.
The lesions typically occur on the trunk and proximal extremities, often favoring the neck, shoulders, and wrists,2 although the
tongue and oral mucosa may also be involved.1,2 Occasionally, microguttate lesions have been reported. Early on, the white sclerotic
or atrophic papules are found between hair follicles and then often
coalesce into larger plaques. This case is unusual given its folliculocentric clinical appearance and generalized distribution.
The cause of LSA remains undetermined but it is believed to
be multifactorial, with genetic, hormonal, and infectious influences, including Borrelia burgdorferi infection.4,5 Patients of both
sexes with LSA have a high incidence of systemic autoimmune
disease, most commonly autoimmune gastritis and Hashimoto’s
thyroiditis.6–10 An exuberant immunologic reaction to an as-yet
unidentified antigen has been proposed to be responsible for the
characteristic disease manifestations. Supportive evidence for this
hypothesis includes the occurrence of a lymphocytic vasculitis and
a T-lymphocyte–dominant inflammatory infiltrate with monoclonally rearranged g-chain gene of the T-cell receptor detected in
skin lesions of lichen sclerosus and occasionally in blood.11–14
From the Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL
Address for Correspondence: Vesna Petronic-Rosic, MD, MSc, Department of Medicine, Section of Dermatology, University of Chicago,
5841 South Maryland Avenue, MC 5067, Chicago, IL 60637 • E-mail: [email protected]
SKINmed. 2010;8:242–244
242
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
CASE STUDY
A 1981 study of 50 women with histologically confirmed LSA
showed that 37 (74%) had tissue autoantibodies and 17 (34%)
had at least 1 autoimmune disease.15 A second study in 1988
confirmed this finding, as 147 of 350 (42%) women with LSA
had tissue autoantibodies, 74 (21.1%) had ≥1 first-degree relatives with an autoimmune disease, and 75 (21.4%) had ≥1 autoimmune diseases (by history).10 No identifiable difference in
clinical features was found among women who developed autoimmune related phenomena, nor was it clear whether screening for such diseases was justifiable, as only a small percentage
developed autoimmune diseases after the diagnosis of LSA.
First-line treatment for LSA includes ultrapotent topical steroids and intralesional triamcinolone injections. Alternative
therapies that have been reported include photodynamic therapy
with aminolevulinic acid (effective in relieving pruritus) in one
study16 and phototherapy for extragenital LSA, with a marked
reduction or clearing after treatment with 40 sessions of UV-A1 at 20 J/cm2 in 10 patients with extragenital lichen sclerosus
without adverse effects.17 There are other recent case reports of
successful therapy using thrice-weekly narrowband UV-B.18 It
is believed that both UV-A-1 and narrowband UV-B work by
increasing matrix-metalloproteinase levels.
Vitamin D and vitamin A derivatives have also been reported
in the treatment of LSA. One study demonstrated a marked
reduction in the number of hypertrophic plaques in a patient within 3 weeks of beginning treatment with calcipotriol
0.005% ointment applied twice daily under occlusion.19 Systemic vitamin A has shown some success with both oral and
parenteral administration.2 This is thought to work by inhibiting transforming growth factor b, which decreases collagen
synthesis by fibroblasts.
Figure 1. Myriad folliculocentric white macules on the patient’s chest.
Figure 2. Multiple folliculocentric lesions of lichen sclerosus
et atrophicus on the patient’s proximal legs.
Topical testosterone or progesterone is no longer recommended
given the conflicting data and possible adverse effects.16 Likewise,
earlier studies showed no benefit of using penicillin, other antibiotics, or penicillamine. Additional treatment options reported
to be effective include adrenocorticotropic hormone,20 hydroxychloroquine,21 sulfasalazine,22 oral calcitriol,23 and topical retinoids.24 Surgery is typically reserved for symptomatic patients
who have failed multiple medical modalities. Recent research has
focused on the use of topical tacrolimus for LSA, with one successful report of treating extragenital LSA with 0.1% tacrolimus
ointment and psoralen plus UV-A.25
We find this presentation to be of interest due to the unique
folliculocentric and generalized clinical presentation. A diagnosis of LSA should be considered in the differential of perifollicular hypopigmentation.
SKINmed. 2010;8:242–244
243
Figures 3. Histological findings characteristic of lichen sclerosus
et atrophicus.
Folliculocentric Lichen Sclerosus et Atrophicus
July/August 2010
CASE STUDY
References
1 Powell JJ, Wojnarowska
1999;353:1777–1783.
F.
Lichen
sclerosus.
Lancet.
cytic infiltrate in penile lichen sclerosus: an immunohistochemical
and molecular investigation. Histopathology. 2006;48:730–735.
2 Meffert JJ, Davis BM, Grimwood RE. Lichen sclerosus. J Am
Acad Dermatol. 1995;32:393–416; quiz 417–398.
15 Harrington CI, Dunsmore IR. An investigation into the incidence
of auto-immune disorders in patients with lichen sclerosus and
atrophicus. Br J Dermatol. 1981;104:563–566.
3 Corbalan-Velez R, Perez-Ferriols A. Lichen sclerosus et atrophicus affecting the wrists and left ankle and clinically simulating
lichen planus. Cutis. 2001;67:417–419.
16 Smith YR, Haefner HK. Vulvar lichen sclerosus: pathophysiology
and treatment. Am J Clin Dermatol. 2004;5:105–125.
4 Eberz B, Berghold A, Regauer S. High prevalence of concomitant anogenital lichen sclerosus and extragenital psoriasis in
adult women. Obstet Gynecol. 2008;111:1143–1147.
17 Kreuter A, Gambichler T, Avermaete A, et al. Low-dose ultraviolet A1 phototherapy for extragenital lichen sclerosus: results of
a preliminary study. J Am Acad Dermatol. 2002;46:251–255.
5 Smith YR, Haefner HK. Vulvar lichen sclerosus: pathophysiology
and treatment. Am J Clin Dermatol. 2004;5:105–125.
18 Colbert RL, Chiang MP, Carlin CS, et al. Progressive extragenital lichen sclerosus successfully treated with narrowband UV-B
phototherapy. Arch Dermatol. 2007;143:19–20.
6 Goolamali SK, Barnes EW, Irvine WJ, et al. Organ-specific antibodies in patients with lichen sclerosus. BMJ. 1974;4:78–79.
7 Harrington CI, Dunsmore IR. An investigation into the incidence
of auto-immune disorders in patients with lichen sclerosus and
atrophicus. Br J Dermatol. 1981;104:563–566.
19 Kreuter A, Gambichler T, Sauermann K, et al. Extragenital lichen
sclerosus successfully treated with topical calcipotriol: evaluation by in vivo confocal laser scanning microscopy. Br J Dermatol. 2002;146:332–333.
8 Meyrick Thomas RH, Ridley CM, Black MM. The association of
lichen sclerosus et atrophicus and autoimmune-related disease
in males. Br J Dermatol. 1983;109:661–664.
20 Di Silverio A, Serri F. Generalized bullous and haemorrhagic lichen sclerosus et atrophicus. Marked improvement with ACTH.
Br J Dermatol. 1975;93:215–217.
9 Thomas RH, Ridley CM, Black MM. Lichen sclerosus et atrophicus associated with systemic lupus erythematosus. J Am Acad
Dermatol. 1985;13(5 pt 1):832–833.
21 Wakelin SH, James MP. Extensive lichen sclerosus et atrophicus with bullae and ulceration–improvement with hydroxychloroquine. Clin Exp Dermatol. 1994;19:332–334.
10 Meyrick Thomas RH, Ridley CM, McGibbon DH, et al. Lichen
sclerosus et atrophicus and autoimmunity–a study of 350 women. Br J Dermatol. 1988;118:41–46.
22 Taveira M, Selores M, Costa V, et al. Generalized morphea and
lichen sclerosus et atrophicus successfully treated with sulphasalazine. J Eur Acad Dermatol Venereol. 1999;12:283–284.
11 Lukowsky A, Muche JM, Sterry W, et al. Detection of expanded
T cell clones in skin biopsy samples of patients with lichen sclerosus et atrophicus by T cell receptor-gamma polymerase chain
reaction assays. J Invest Dermatol. 2000;115:254–259.
23 Ronger S, Viallard AM, Meunier-Mure F, et al. Oral calcitriol: a
new therapeutic agent in cutaneous lichen sclerosis. J Drugs
Dermatol. 2003;2:23–28.
12 Regauer S, Reich O, Beham-Schmid C. Monoclonal gamma-Tcell receptor rearrangement in vulvar lichen sclerosus and squamous cell carcinomas. Am J Pathol. 2002;160:1035–1045.
13 Regauer S. Immune dysregulation in lichen sclerosus. Eur J Cell
Biol. 2005;84:273–277.
14 Regauer S, Beham-Schmid C. Detailed analysis of the T-cell lympho-
24 Guerriero C, Manco S, Paradisi A, et al. Extragenital lichen
sclerosus and atrophicus treated with topical steroids and
retinoids in a child with vitiligo. Int J Immunopathol Pharmacol.
2008;21:757–759.
25 Valdivielso-Ramos M, Bueno C, Hernanz JM. Significant improvement in extensive lichen sclerosus with tacrolimus ointment and
PUVA. Am J Clin Dermatol. 2008;9:175–179.
HYPERTRICHOSIS
Medications reported to cause excessive hair growth
Clomid
Erbitux
Rogaine
Soriatane
Corticosteroids
Loniten
Sandimmune
Timoptic
Dilantin
Neoral
Simulect
Topamax
Zyrtec
Adapted from Litt, JZ. Curious, Odd, Rare, and Abnormal Reactions to Medications. Fort Lee, NJ: Barricade Books; 2009:162.
SKINmed. 2010;8:242–244
244
Folliculocentric Lichen Sclerosus et Atrophicus
July/August 2010
Volume 8 • Issue 4
CASE STUDY
Poorly Growing Hair in a Child
Diana Bolotin, MD, PhD; Bernhard Ortel, MD; Sarah L. Stein, MD
A healthy 5-year-old Caucasian girl presented to the pediatric dermatology clinic for poor hair growth. The patient’s father described slow hair growth
and finely textured hair since birth. The patient had her first haircut 1 month before presentation. The family denied bald areas on the scalp, significant
hair shedding, and sores on the scalp. The child did not scratch at her scalp or pull her hair. She ate a normal diet and had normal growth and development otherwise. She was not taking any medications or over-the-counter supplements. There was no family history of hair disorders. On physical
examination, the patient had short, fine, reddish blond hair that was of different lengths (Figure 1). There were no papules, pustules, scale, or crust on her
scalp. Lymphadenopathy was absent. She had normal eyelashes and eyebrows. There were no lesions on her oral mucosa. No tooth or nail abnormalities
were present. The rest of the physical examination was normal. The hair pull test resulted in 4 easily pulled hairs (3 anagen and 1 telogen). A hair mount
was performed (Figure 2). The hair mount analysis revealed anagen hairs with distorted bulbs and ruffled cuticles extending a short distance distally from
the bulb, consistent with loose anagen hair (Figure 2). All of the anagen hairs on the pull test demonstrated the above findings. Based on the patient’s
clinical presentation and the findings seen on light microscopy of the hair mount preparation, the patient was diagnosed with loose anagen syndrome.
L
oose anagen syndrome (LAS) is a hair disorder that is primarily diagnosed in childhood. The most frequent complaint on presentation is poor hair growth and thin hair,
although other symptoms include unruly lusterless hair.1,2 In most
cases, patients tend to be blonde, although a few dark-haired patients have been described. Three different patterns of LAS have
been reported, all of which show loose anagen hair (LAH) phenotype on hair pull test.3 Patients with type A phenotype have sparse
Figure 1. Reddish brown fine hair of variable lengths was appreciated on the scalp.
From the Section of Dermatology, University of Chicago Medical Center, Chicago IL
Address for Correspondence: Diana Bolotin, MD, PhD, University of Chicago Medical Center, Section of Dermatology, MC5067 5841, South
Maryland Avenue, Chicago IL 60637 • E-mail: [email protected]
SKINmed. 2010;8:246–247
246
© 2010 Pulse Marketing & Communications, LLC
July/August 2010
CASE STUDY
hair with poor growth; patients with type B phenotype present
with sparse, dull, and unruly hair; and patients with type C phenotype present with increased hair shedding.3
The pathogenesis of LAS remains largely unclear. One study has
found a mutation in a hair keratin K6hf, present in the companion cell layer of the outer root sheath, detected in 3 of 9 families
with LAS.4 It is unclear, however, whether other keratins or adhesion molecules in the inner root sheath may also be responsible
for this hair disorder. In addition, it remains to be seen whether
genetic differences account for the various subtypes of LAS.
Diagnosis of LAS is based on the combination of characteristic
clinical findings as well as the presence of microscopically defined
LAH on hair pull test.1 On light microscopy, LAH hairs show
misshapen hair bulbs, absence of root sheaths, and the characteristically ruffled cuticle.1,2,5,6 It is thought that anywhere from
70% to 100% of hairs in patients with LAS demonstrate LAH
features.3 It should be noted, however, that up to 10% of cases
of LAH have been reported in normal individuals, although it is
unclear whether these individuals may have a milder expression
of LAS. A biopsy is usually nondiagnostic for this condition,
although one characteristic feature is premature keratinization of
the Henle and Huxley layers of the inner root sheath.1,5
The clinical severity of LAS tends to improve with age, with
some cases leading to complete resolution.1,2 Generally, aggressive treatment is not warranted, and gentle hair care is recommended for these patients.
References
1 Tosti A, Piraccini BM. Loose anagen hair syndrome and loose
anagen hair. Arch Dermatol. 2002;138:521–522.
Figure 2. Trichogram showing anagen hairs with ruffled cuticles.
2 Cheng AS, Bayliss SJ. The genetics of hair shaft disorders. J Am
Acad Dermatol. 2008;59:1–22.
3 Olsen EA, Bettencourt MS, Cote NL. The presence of loose
anagen hairs obtained by hair pull in the normal population. J
Investig Dermatol Symp Proc. 1999;4:258–260.
4 Chapalain V, Winter H, Langbein L, et al. Is the loose anagen
hair syndrome a keratin disorder? A clinical and molecular study.
Arch Dermatol. 2002;138:501–506.
5 Smith VV, Anderson G, Malone M, et al. Light microscopic examination of scalp hair samples as an aid in the diagnosis of
paediatric disorders: retrospective review of more than 300
cases from a single centre. J Clin Pathol. 2005;58:1294–1298.
6 Cantatore-Francis JL, Orlow SJ. Practical guidelines for
evaluation of loose anagen hair syndrome. Arch Dermatol.
2009;145:1123–1128.
Formulary of Dr George C. Andrews
Bleach
• #1 Powder
o
Bismuth oxychloride – 1 part
o
Sodium perborate – 2 parts
o
Talc – 7 parts
• #2 Solution
o
Hydrogen peroxide
Sig. Use enough #2 to make a thick paste.
Submitted by Douglas D. Altchek, MD, New York, NY
SKINmed. 2010;8:246–247
247
Poorly Growing Hair in a Child
Locoid Lipocream® Cream, 0.1%
(hydrocortisone butyrate 0.1% cream)
For Topical Use Only
Rx Only
BRIEF SUMMARY
INDICATIONS AND USAGE
Locoid Lipocream is a topical corticosteroid indicated for: relief of the inflammatory
and pruritic manifestations of corticosteroid-responsive dermatoses in adults and
the treatment of mild to moderate atopic dermatitis in patients 3 months to 18 years
of age.
WARNINGS AND PRECAUTIONS
Reversible hypothalamic-pituitary-adrenal (HPA) axis suppression may occur, with
the potential for glucocorticosteroid insufficiency. Consider periodic evaluations for
HPA axis suppression if Locoid Lipocream is applied to large surface areas or used
under occlusion. If HPA axis suppression is noted, reduce the application frequency,
discontinue use, or switch to a lower potency corticosteroid.
Systemic effects of topical corticosteroids may also include manifestations of
Cushing’s syndrome, hyperglycemia, and glucosuria.
Pediatric patients may be more susceptible to systemic toxicity due to their larger
skin surface-to-body-mass ratios.
Initiate appropriate therapy if concomitant skin infections develop.
Discontinue use if irritation develops.
ADVERSE REACTIONS
The most common adverse reactions (>1%) are HPA axis suppression and
application site reactions.
The following additional local adverse reactions have been reported infrequently
with topical corticosteroids, and they may occur more frequently with the use of
occlusive dressings and higher potency corticosteroids. These reactions included:
irritation, folliculitis, acneiform eruptions, hypopigmentation, perioral dermatitis,
allergic contact dermatitis, secondary infection, skin atrophy, striae, miliaria and
telangiectasia.
USE IN SPECIFIC POPULATIONS
Pregnancy
Pregnancy Category C. Corticosteroids have been shown to be teratogenic in
laboratory animals when administered systemically at relatively low dosage levels.
Some corticosteroids have been shown to be teratogenic after dermal application
in laboratory animals.There are no adequate and well-controlled studies in
pregnant women. Therefore, Locoid Lipocream should be used during pregnancy
only if the potential benefit justifies the potential risk to the fetus.
Please refer to full prescribing information for detailed information regarding
systemic embryofetal development studies.
Nursing Mothers
Systemically administered corticosteroids appear in human milk and could
suppress growth, interfere with endogenous corticosteroid production, or cause
other untoward effects. It is not known whether topical administration of
corticosteroids could result in sufficient systemic absorption to produce detectable
quantities in human milk. Because many drugs are excreted in human milk, caution
should be exercised when Locoid Lipocream is administered to a nursing woman.
Pediatric Use
Safety and efficacy in pediatric patients below 3 months of age have not been
established.
Because of higher skin surface-to-body-mass ratios, pediatric patients are at a
greater risk than adults of HPA axis suppression when they are treated with topical
corticosteroids. They are therefore also at a greater risk of glucocorticosteroid
insufficiency after withdrawal of treatment and of Cushing’s syndrome while on
treatment.
Eighty-six (86) pediatric subjects (5 months to less than 18 years of age) with
moderate to severe atopic dermatitis affecting at least 25% of body surface area
(BSA) treated with Locoid Lipocream three times daily for up to 4 weeks were
assessed for HPA axis suppression. The disease severity (moderate to severe
atopic dermatitis) and the dosing regimen (three times daily) in this HPA axis study
were different from the subject population (mild to moderate atopic dermatitis) and
the dosing regimen (two times daily) for which Locoid Lipocream is indicated.
Five of the 82 evaluable subjects (6.1%) demonstrated laboratory evidence of
suppression, where the sole criterion for defining HPA axis suppression was a
serum cortisol level of less than or equal to 18 micrograms per deciliter after
cosyntropin stimulation. Suppressed subjects ranged in age from 5 months to
16 years and, at the time of enrollment, had 25% to 95% BSA involvement. These
subjects did not develop any other signs or symptoms of HPA axis suppression.
At the first follow up visit, approximately one month after the conclusion of
treatment, cosyntropin stimulation results of all subjects had returned to normal,
with the exception of one subject. This last subject recovered adrenal function by
the second post treatment visit, 65 days post-treatment.
Cushing’s syndrome, linear growth retardation, delayed weight gain, and
intracranial hypertension have also been reported in pediatric patients receiving
topical corticosteroids. Manifestations of adrenal suppression in pediatric patients
include low plasma cortisol levels to an absence of response to ACTH stimulation.
Manifestations of intracranial hypertension include bulging fontanelles, headaches,
and bilateral papilledema.
Geriatric Use
Clinical studies of Locoid Lipocream did not include sufficient numbers of subjects
aged 65 and over to determine whether they respond differently from younger
subjects.
Carcinogenesis, Mutagenesis, Impairment of Fertility
No studies were conducted to determine the photococarcinogenic or dermal
carcinogenic potential of Locoid Lipocream.
Hydrocortisone butyrate revealed no evidence of mutagenic or clastogenic
potential based on the results of two in vitro genotoxicity tests (Ames test and
L5178Y/TK+ mouse lymphoma assay) and one in vivo genotoxicity test (mouse
micronucleus assay).
No evidence of impairment of fertility or effect on mating performance was
observed in a fertility and general reproductive performance study conducted in
male and female rats at subcutaneous doses up to and including 1.8 mg/kg/day
(0.7X maximum topical human dose [MTHD]). Mild effects on maternal animals,
such as reduced food consumption and a subsequent reduction in body weight gain,
were seen at doses ≥0.6 mg/kg/day (0.2X MTHD).
PATIENT COUNSELING INFORMATION
Patients using Locoid Lipocream should receive the following information
and instructions:
Apply a thin layer to the affected skin two or three times daily for corticosteroidresponsive dermatoses in adults. Consult with your physician to determine if
treatment is needed beyond 2 weeks. Apply a thin film to the affected skin areas
two times daily for atopic dermatitis in patients 3 months of age and older.
Safety of Locoid Lipocream in pediatric patients has not been established beyond 4
weeks of use.
Rub in gently.
Avoid contact with the eyes.
Do not bandage, otherwise cover, or wrap the affected skin area so as to be
occlusive unless directed by your physician.
Do not use Locoid Lipocream in the diaper area, as diapers or plastic pants may
constitute occlusive dressings.
Do not use Locoid Lipocream on the face, underarms, or groin areas unless
directed by your physician.
If no improvement is seen within 2 weeks, contact your physician.
Do not use other corticosteroid-containing products while using Locoid Lipocream
without first consulting your physician.
Store at 25°C (77°F); excursions permitted to 15-30°C (59-86°F) [see USP Controlled
Room Temperature]. Protect from freezing. Keep out of the reach of children.
Manufactured for: Triax Pharmaceuticals, LLC
Cranford NJ 07016
By: Ferndale Laboratories, Inc.
Ferndale MI 48220
Locoid Lipocream is a registered trademark of
Astellas Pharma Europe BV licensed to
Triax Pharmaceuticals, LLC.
Marketed and Distributed By:
Triax Pharmaceuticals, LLC
Cranford NJ 07016
www.Locoid.com
131B301
Rev 11/09
Now younger eczema
patients have something
to smile about
Now approved for use in children
down to 3 months of age
The power of an ointment with the elegance of a cream
Locoid Lipocream is indicated for the relief of the inflammatory and pruritic manifestations of corticosteroid-responsive dermatoses,
including the treatment of mild to moderate atopic dermatitis in patients 3 months of age and older.
Safety and effectiveness in pediatric patients below 3 months of age have not been established.
Reversible HPA axis suppression may occur, with the potential for corticosteroid insufficiency. Consider periodic evaluations for HPA
axis suppression if applied to large surface areas or used under occlusion. Systemic effects of topical corticosteroids may also include
manifestations of Cushing’s syndrome, hyperglycemia, and glucosuria. Pediatric patients may be more susceptible to systemic toxicity
due to their large skin surface-to-body-mass ratios. Initiate appropriate therapy if concomitant skin infection develops. Discontinue use
if irritation develops. Please see full Prescribing Information on adjacent page.
Visit us at www.locoid.com
(hydrocortisone butyrate 0.1%) Cream
©2010 Triax Pharmaceuticals, LLC. All rights reserved. Locoid is a registered trademark of Astellas Pharma Europe B.V. licensed to Triax Pharmaceuticals, LLC. LOC-0410-01