Dermatology

Casal's Necklace

Authors
Kiran H.S
Article Citation and PDF Link
BJMP 2021;14(1):a007
Abstract / Summary
Abstract: 

This article presents an interesting case of Casal's necklace in a patient with Pellagra-like dermatitis and the response to treatment.

Abbreviations: 
HIV-Human Immnodeficiency Virus; ELISA - Enzyme linked immunosorbent assay
Keywords: 
Pellagra; Dermatitis; Casal's necklace

A 40 years old non-alcoholic and non-diabetic agricultural laborer presented with skin lesions around his neck, forearms and feet (sun exposed areas) along with glossitis. Pellagra was suspected because of Casal's necklace (i.e., erythematous, hyperpigmented, scaly lesions around his neck- arrow mark in figure 1). However he did not have diarrhea or neurological manifestations. Pellagra is due to Niacin (Vitamin B3) deficiency. Typical cases of pellagra are associated with 3 Ds - Dermatitis, Diarrhea, Dementia, (and if not treated, the 4th D- Death).1,2  Not many will have all the three Ds. Most commonly involved is skin – dermatitis (Pelle-skin; agra -rough). The patient belonged to poor socioeconomic status.2 His vital parameters and basic investigations were all within normal limits and HIV-ELISA was negative.

The diagnosis of a pellagra-like dermatitis was entertained.3 He was treated with multivitamin capsules which included Niacinamide.2  

The skin lesions had disappeared dramatically at the time of follow-up after one month (figure 2).


Figure 1: Casal's necklace before treatment


Figure 2: Improvement after treatment

Acknowledgements / Conflicts / Author Details
Acknowledgement: 
I thank our respected Principal, HOD of Gen.Medicine and department of Dermatology.
Competing Interests: 
None declared
Details of Authors: 
Dr Kiran H.S; MD, PGDTSP, FAIMER fellow, Professor, Department of Internal Medicine, JSS Medical College and Hospital, JSSAHER, Mysore, Karnataka, India.
Corresponding Author Details: 
Dr Kiran H.S; MD, PGDTSP, FAIMER fellow, Professor, Department of Internal Medicine, JSS Medical College and Hospital, JSSAHER, Mysore, Karnataka, India.
Corresponding Author Email: 
drhskiran@gmail.com
References
References: 
  1. Hegyi, J., Schwartz, R.A. and Hegyi, V. (2004), Pellagra: Dermatitis, dementia, and diarrhea. International Journal of Dermatology, 43: 1-5. https://doi.org/10.1111/j.1365-4632.2004.01959.x
  2. Frank G-PGM, Voorend DM, Chamdula A, van Oosterhout JJ, Koop K. Pellagra: a non-communicable disease of poverty. Tropical Doctor. 2012;42(3):182-184. doi:10.1258/td.2012.120155
  3. Hendricks WM. Pellagra and pellagralike dermatoses: etiology, differential diagnosis, dermatopathology, and treatment. Semin Dermatol. 1991 Dec;10(4):282-92. PMID: 1764355. 

The use of dermoscopy amongst dermatology trainees in the United Kingdom

Authors
Claire Reid, Kimberlee Lim & Catriona Henderson
Article Citation and PDF Link
BJMP 2018;11(2):a1110

A dermatoscope is a hand-held device for examining the appearance of the skin. Dermoscopy has become an increasingly used and valued tool in the assessment of various skin lesions, and more recently, inflammatory rashes. It is quick, cheap and when used correctly, dermoscopy is an essential tool in helping clinicians detect early stage skin cancer. Various national and international guidelines recommend routine use of dermoscopy in the assessment of pigmented lesions1,2 because it enhances melanoma detection rates3,4 and can help confirm the diagnosis of benign lesions such as haemangiomas and seborrhoeic keratoses. As with any skill, competency takes time to develop and a combination of various learning and assessment methods is best. The dermatology specialist training curriculum in the United Kingdom (UK) states that trainees should be competent in using a dermatoscope and interpreting findings, while recognizing the limitations of this tool5. Assessment of these clinical skill and behavioural competencies using direct observation of procedural skills (DOPS), case-based discussion (CBD), mini clinical examination (mini-CEX), and/or multisource feedback (MSF) is suggested. There is no specific guidance on what resources a trainee should use to achieve these competencies, nor on what is the minimum expected dermoscopy skillset at completion of specialist training.

The aim of this survey was to explore dermoscopy use amongst dermatology specialist trainee registrars in the UK including frequency of use, how it is being taught and whether trainees feel their dermoscopy training has been adequate.

An online survey was designed and distributed to dermatology trainees in the United Kingdom using an email link and hard copies were distributed at a national dermoscopy course. Respondents who did not identify themselves as dermatology trainees were removed from the analysis. Responses were collected anonymously, then collated and analysed using SurveyMonkey® computer software.

Twenty-five percent (59/238) of dermatology trainees completed the survey. On average, 92% (54/59) use dermoscopy more than once daily. Eighty-five percent (50/59) always use dermoscopy when assessing pigmented lesions while 34% (20/59) always and 59% (35/59) sometimes use it to assess non-pigmented lesions. When asked about specific tools used to learn dermoscopy, 41% (24/59) have been on a previous course, 42% (25/59) reported attendance at a lecture or seminar, 46% (27/59) have used a dermoscopy text book, 14% (8/59) have attended a conference, 19% (11/59) have used online resources. Seventeen percent (10/59) have never used any of the above learning methods. (Figure 1a). Amongst those who have attended a formal dermoscopy course (n=24), 92% (22/24) of these were ≤1 day in duration. When questioned about informal teaching in clinical practice, 12% (7/59) frequently, 56% (33/59) sometimes, 31% (18/59) rarely and 2% (1/59) never receive teaching from their supervising dermatology consultant. (Figure 1b). Fifty-four percent (32/59) feel they have received adequate training in dermoscopy while the remaining 46% (27/59) feel their dermoscopy training is inadequate for their training stage (Figure 1c). Seventy-three percent (43/59) have access to dermoscopic photography within their local dermatology department.

Fig 1a - Have you undertaken any formal study in dermoscopy? 49% of trainees have attended a lecture, 2% a seminar, 14% a conference, 41% a course, 19% have used an online resource, 46% have used a book, 17% have not used any resource.

Fig 1b- Do you receive dermoscopy training from your supervisor in clinic? 56% of trainees sometimes, 31% rarely, 12% frequently, and 2% have never received training from their seniors in clinic.

Fig 1c- Do you believe that you have received adequate training in the use of a dermoscopy for your training grade?

These results of this survey highlight the need for dermoscopy training to be reviewed within the UK national training curriculum for dermatology. Despite daily use by the vast majority, dermoscopy training is largely self-directed and highly variable amongst individual trainees. Of concern, a significant proportion of those who responded feel their dermoscopy skills are inadequate for their training stage. Of note, the 25% response rate means that the results of this survey may not be representative of dermatology trainees in the United Kingdom as a whole.

This is the first time that dermoscopy use has been explored through a national survey of dermatology trainees in the UK, to the best of our knowledge. A survey on dermoscopy use was carried out by The British Association of Dermatologists (BAD) in 20126 but the majority of responses were from dermatology consultants. This confirmed that 98.5% of respondents regularly used dermoscopy, while 81% had received any training. The most frequent source of training was UK based courses, which 62% of respondents reported attending. Of note, 39% of all respondents lacked confidence when making a diagnosis based on their interpretation of dermoscopy findings. It is not clear how many of those lacking in confidence were consultants, trainees or specialty doctors. Although the situation may have improved since 2012, these results do suggest that dermoscopy training needs have not been met for a proportion of doctors across the dermatology community.

Dermoscopy training is an important issue to address for several reasons. The volume of cutaneous lesions being referred to dermatology is increasing, and skin cancer referrals and treatment now account for 50% of a UK dermatologists’ workload7. For every melanoma diagnosed, a dermatologist may expect to see 20–40 benign lesions referred from general practitioners (GPs)7. These facts highlight the importance of maximising diagnostic skills which frequently include using dermoscopy as part of clinial assessment. Lack of adequate training is a common self-reported reason for dermatologists not using dermoscopy8. Both trainees and their supervising bodies have a responsibility to maximize training opportunities and embed the use of dermoscopy in routine practice.

In conclusion, we feel UK dermatology trainees and indeed any clinician who utilizes this tool, would benefit from a more standardized and integrated approach to dermoscopy teaching to ensure safe practice of this skill and deliver high quality evidence-based patient care.

Acknowledgements / Conflicts / Author Details
Competing Interests: 
Dr Catriona Henderson teaches at a dermoscopy course which is ran by the British Association of Dermatology
Details of Authors: 
CLAIRE REID, Medical Doctor, Dermatology Department, University Hospital Southampton NHS Foundation Trust, SO140YG England. KIMBERLEE LIM, Medical Doctor, Dermatology Department, University Hospital Southampton NHS Foundation Trust, SO140YG England. CATRIONA HENDERSON, Medical Doctor, Dermatology Department, University Hospital Southampton NHS Foundation Trust SO140YG England.
Corresponding Author Details: 
CLAIRE REID, Medical Doctor, Dermatology Department, University Hospital Southampton NHS Foundation Trust, SO140YG England
Corresponding Author Email: 
clairereid85@gmail.com
References
References: 
  1. European Dermatology Forum; update of the guideline on the diagnosis and treatment of melanoma, developed by the guideline sub-committee ‘Melanoma’ of the European Dermatology Forum, 2012. Accessed at http://www.euroderm.org/edf/index.php/edf-guidelines/category/5-guidelines-miscellaneous
  2. Melanoma; assessment and management. National Institute for Health and Care Excellence. NICE guideline [NG-14], July 2015. Accessed at https://www.nice.org.uk/guidance/ng14/chapter/1-recommendations#assessing-melanoma-2
  3. Watts C.G., Dieng M., Morton R.L., Mann G.J., Menzies S.W., Cust A.E. Clinical practice guidelines for the identification, screening and follow-up of individuals at high risk of primary cutaneous melanoma: a systematic review. British Journal of Dermatology 2015;172;1:33-47
  4. Vestergaard M.E., Macaskill P., Holt P.E., Menzies S.W. Dermoscopy compared with naked eye examination for the diagnosis of primary melanoma: a meta-analysis of studies performed in a clinical setting. British Journal of Dermatology 2008:159:3;669-676
  5. Specialty Training Curriculum for Dermatology August 2010 (amended August 2012) Joint Royal Colleges of Physicians Training Board. Accessed at https://www.jrcptb.org.uk/sites/default/files/2010%20Dermatology%20%28amendment%202012%29.pdf
  6. Butler T, Martin R, Affleck A, Fleming C, Bowling J. Trends in dermoscopy use in the UK: results from surveys in 2003 and 2013. Dermatology Practical and Conceptual 2015;5;(2);4:29-38
  7. Eedy D. Dermatology: a specialty in crisis. Clinical Medicine 2015;15;6 509–10
  8. Engasser H.C., Warshaw E.M. Dermatoscopy use by US dermatologists: A cross-sectional survey. Journal of the American Academy of Dermatology. 2010 Sep 1;63(3). Available from, DOI: 10.1016/j.jaad.2009.09.050

Photo Quiz: Localized, reticulated erythema

Authors
Andrew A Lawson & Thomas C Michels
Article Citation and PDF Link
BJMP 2017;10(1):a1004
Abstract / Summary
Abstract: 

A 70-year-old man presented in the winter with a four-week history of redness of the left anterolateral leg. On exam, the patient has an 8 cm, irregular patch of reticulated erythema with both hyperpigmentation and scaling. This manuscript contains the history and physical exam, a photo of the lesion, a differential diagnosis, and a discussion of management.

Abbreviations: 
EAI - Erythema ab igne
Keywords: 
Erythema ab igne, reticulated erythema, livedo reticularis, livedo racemosa, first-degree burn

A 70-year-old man presented in the winter with a four-week history of redness of the left anterolateral leg. He first noticed a slight “tenderness” in the area when showering; the discomfort lasted only a few days. Over the next week, he noticed redness developing. It is now painless and not pruritic, warm, or peeling. He has not applied any topical lotions or creams. He has not had an exposure to new soaps or detergents. He feels well, without fever or weight loss. He has a diagnosis of hypertension and lumbar radiculopathy with an L5 discectomy and resultant leg numbness. He is retired and does not smoke or drink alcohol; his hobby is woodworking in his garage.

Physical examination reveals normal vital signs. On his left anterolateral leg, he has an 8 cm, irregular patch of reticulated erythema with both hyperpigmentation and scaling. The lesion is non-palpable. He has decreased sensation in an L5 distribution on that leg, which was unchanged from prior examinations. These skin findings are shown in Figure 1.

Figure 1

Question: Based on history and physical examination, which of the following is the most likely diagnosis?

  • Livedo reticularis
  • Erythema ab igne
  • Livedo racemosa
  • First-degree burn

Discussion

The answer is erythema ab igne (EAI; literally “redness from fire,”) which results from chronic exposure to moderate-intensity heat. EAI presents as a reticulated erythematous patch over the area of exposed skin. Possible secondary changes include epidermal atrophy and scaling.1,2 With repeated exposure, brown hyperpigmentation may develop.1 Most patients are asymptomatic, although some note a mild burning sensation. A history of repeated exposure to heat is key to the diagnosis. While cases were historically noted on skin exposed to fire, such as the arms of bakers and coal shovellers, EAI can result from our many, modern heat-sources, such as laptop computers, car seat heaters, heating pads, and, in this case, the portable space heater under the patient’s woodworking bench.2-4 With removal of the heat source, hyperpigmentation typically regresses but may take years.1,3 The diagnosis is clinical. A biopsy is not required to make the diagnosis, but is indicated if malignant transformation is suspected. EAI can increase risk of squamous cell carcinoma, Merkel cell carcinoma, and cutaneous marginal zone lymphoma.1,5 Treatment is typically not necessary; topical steroids or retinoids and laser have had variable success.1,3,4 If pre-malignant changes are detected, topical 5-flourouracil is recommended.1,4

See Table 1 for a summary of the key characteristics and distinguishing features of each diagnosis in this selected differential.

Table 1. Selected Differential Diagnosis of Reticulated Skin Lesions in Adults

Condition Characteristics
Livedo reticularis Violaceous mottled or reticulated patches; painless; typically temperature sensitive; may be physiologic or secondary to systemic disease; no hyperpigmentation.
Erythema ab igne Erythematous reticulated patch, with possible secondary changes including epidermal atrophy and scaling; chronic exposure may lead to hyperpigmentation; painless or associated with a mild burning sensation; history of heat exposure.
Livedo racemosa Violaceous reticulated patch with larger branching pattern than livedo reticularis, often with asymmetric or “broken” net appearance; typically involves the trunk and proximal limbs; generally secondary to chronic disease; frequently painful; no hyperpigmentation.
First-degree burn Erythematous, dry, painful lesion which includes the entire area of skin that contacted the high-intensity heat source; not reticulated; no hyperpigmentation.

Livedo reticularis is typically more violaceous in appearance, with net-like, mottled discolouration of the skin due to deoxygenation and dilation of the venous plexus. Primary, physiologic livedo reticularis is often brought on by cold and alleviated by warming. It usually involves a larger area, such as the bilateral thighs, rather than a confined area of skin.1,2

Livedo racemosa is a persistent variant of livedo reticularis with a characteristic, large, broken, branching pattern, often on the trunk and proximal limbs. It is generally secondary to a systemic disease, such as antiphospholipid antibody syndrome or Sneddon syndrome.6

First-degree burns are erythematous, dry, and painful. Instead of a reticulated pattern, as shown here, the erythema of first degree burns covers the entire area of skin that contacted the high-intensity heat source.

Acknowledgements / Conflicts / Author Details
Acknowledgement: 
None
Competing Interests: 
None declared
Details of Authors: 
ANDREW LAWSON, BS, 8253 Bagley Ave N., Seattle, WA 98103, USA. THOMAS MICHELS, MD, MPH, 1037 NE 65th St #80631, Seattle, WA 98115, USA.
Corresponding Author Details: 
ANDREW LAWSON, 8253 Bagley Ave N., Seattle, WA 98103, USA
Corresponding Author Email: 
aalawson@uw.edu
References
References: 
  1. Miller K, Hunt R, Chu J, et al. Erythema ab igne. Dermatology Online Journal. 2011;17(10):28.
  2. Riahi RR, Cohen PR. What Caused This Hyperpigmented Reticulated Rash On This Man’s Back? The Dermatologist.  2013;21(1).  http://www.the-dermatologist.com/content/what-caused-hyperpigmented-reticulated-rash-man%E2%80%99s-back
  3. Salgado F, Handler MZ, Schwartz RA. Erythema ab igne: new technology rebounding upon its users? Int J Dermatol. 2017. doi: 10.1111/ijd.13609. [Epub ahead of print]
  4. Beleznay K, Humphrey S, Au S. Erythema ab igne. CMAJ. 2010;182(5):E228.
  5. Sigmon JR, Cantrell J, Teague D, et al. Poorly differentiated carcinoma arising in the setting of erythema ab igne. Am J Dermatopathol. 2013;35(6):676-8.
  6. Uthman IW, Khamashta MA. Livedo racemosa: a striking dermatological sign for the antiphospholipid syndrome. J Rheumatol. 2006;33:2379-2382. 

Striae distensae: What’s new at the horizon?

Authors
Mohammad Abid Keen
Article Citation and PDF Link
BJMP 2016;9(3):a919
Abstract / Summary
Abstract: 

Striae distensae, commonly known as stretch marks, are benign skin lesions associated with considerable cosmetic morbidity. Despite considerable investigations into their origins, the pathogenesis of striae distensae remains unknown.Currently, there is no treatment which consistently improves the appearance of striae. With a high incidence and unsatisfactory treatments, stretch marks remain an important target of research for an optimum consensus of treatment. The aim of present article is to appraise the readers with various newer treatment options in the management of this difficult condition.

Keywords: 
Striae distensae, stretch marks, cosmetic

Introduction

Striae distensae, or stretch marks, are linear scars in the dermis which arise from rapid stretching of the skin over weakened connective tissue. It is a common skin condition that rarely causes any significant medical problems but is often a significant source of distress to those affected. Striae distensae were described as a clinical entity hundreds of years ago, and the first histological descriptions appeared in the medical literature in 1889.1 With a high incidence and unsatisfactory treatments, stretch marks remain an important target of research for an optimum consensus of treatment. These appear initially as red, and later, as white lines on the skin, representing scars of the dermis, and are characterized by linear bundles of collagen lying parallel to the surface of the skin, as well as eventual loss of collagen and elastin. The estimated prevalence of striae distensae range from 50 to 80%.2,3 The anatomical sites affected vary, with areas commonly affected including the abdomen, breasts, thighs and buttocks.4 The three maturation stages of striae include the acute stage (striae rubra) characterized by raised, erythematous striae, the sub-acute stage characterized by purpuric striae, and the chronic stage (striae alba), characterized by white or hypo-pigmented, atrophied striae.5 Although stretch marks are only harmful in extreme cases, even mild stretch marks can cause distress to the bearer6 (Table 1).

Table 1: Histological comparisons between striae rubrae and striae albae

Epidermis Oedema
Increased melanocytes
Epidermal atrophy
Loss of rete ridges
Decreased melanocytes
Papillary dermis Dilatation of blood vessels No vascular reaction
Reticular dermis Structural alteration of collagen fibres
Reduced and reorganized elastic fibres
Fine elastic fibres in dermis
Densely packed collagen parallel to skin surface.
Thick elastic fibres in dermis
Inflammatory cells Lymphocytes and fibroblasts Eosinophills

Aetiology

Striae may result from a number of causes, including, but not limited to, rapid changes in weight, adolescent growth spurts, corticosteroid use or Cushing Syndrome, and generally appear on the buttocks, thighs, knees, calves, or lumbosacral area.7 In addition, approximately 90% of all pregnant women develop stretch marks either on their breasts and/or abdomen by the third trimester.8 Genetic predisposition is also presumed, since striae distensae have been reported in monozygotic twins.9,10 There is decreased expression of collagen and fibronectin genes in affected tissue.11 The role of genetic factors is further emphasised by the fact that they are common in inherited defects of connective tissue, as in Marfan’s syndrome.12,13 Obesity and rapid increase or decrease in weight have been shown to be associated with the development of SD.14 Young male weight lifters develop striae on their shoulders.15 Striae distensae also occurs in cachetic states, such as tuberculosis, typhoid and after intense slimming diets.16 Rare etiologies include human immunodeficiency virus positive patients receiving the protease inhibitor indinavir and chronic liver disease.13,15 A case of idiopathic striae was also reported.17

Rosenthal18 proposed four aetiological mechanisms of striae formation: insufficient development of tegument, including elastic properties deficiency; rapid stretching of the skin; endocrinal changes; and other causes, possibly toxic.

Pathogenesis

The pathogenesis of striae is unknown but probably relates to changes in the components of extracellular matrix, including fibrillin, elastin and collagen.19 There has been emphasis on the effects of skin stretching in the pathogenesis of striae because the lesions are perpendicular to the direction of skin tension.20 A possible role of glucocorticoids in the pathogenesis of striae has been suggested because of an increase in the levels of steroid hormones and other metabolites found in patients exhibiting striae.21 There are studies suggesting the role of fibroblasts in the pathogenesis of striae. Compared to normal fibroblasts, expression of fibronectin and both type I and type III procollagen were found to be significantly reduced in fibroblasts from striae, suggesting that there exists a fundamental aberration of fibroblast metabolism in striae distensae.22

Pathological aspects

The earliest pathological changes are subclinical to be detected by electron microscopy only. These changes include mast cell degranulation and the presence of activated macrophages in association with mid-dermal elastolysis.23 When the lesions become become clinically visible, collagen bundles start showing structural alterations, fibroblasts become prominent, and mast cells are absent.23 On light microscopic examination, Inflammatory changes are conspicuous in the early stage, with dermal oedema and perivascular lymphocytic cuffing.24 In later stages, there is epidermal atrophy, loss of rete ridges and other appendages including hair follicles are absent.25

Evaluation of striae distensae

Approaches to evaluating SD severity visually include the Davey 26 and Atwal scores,27 although these have not been validated specifically for SD. An objective evaluation of SD may be carried out using skin topography, imaging devices including three-dimensional (3D) cameras, reflectance confocal microscopy and epiluminescence colorimetry.28,29,30

Table 2: Visual scoring systems for the assessment of striae distensae

Davey method Used for evaluating striae rubrae and albae.
Divide the abdomen into quadrants using midline vertical and horizontal lines.
Each quadrant given a score (0 no SD; 1 moderate number of SD; 2 many SD).
Score given out of 8.
Atwal score Used for evaluating striae rubrae and albae.
Six sites chosen (abdomen, hips, breasts, thigh/buttocks).
Each site given a maximum score of six.
Total score out of 24.
Score 0–3 for the presence of striae (0 no SD; 1 < 5 SD; 2 5–10 SD; 3 > 10 SD).
Score 0–3 for the presence of erythema (0 no erythema; 1 light red/pink; 2 dark red; 3 purple).

Management

Striae distensae (striae alba) is a very challenging cosmetic problem for dermatologists to treat. Various modalities of treatment have been tried. Although therapeutic strategies are numerous, there is no treatment which consistently improves the appearance of striae and is safe for all skin types.31 Weight loss by diet alone or a combination of diet and exercise do not change the degree of striae distensae.32

Topical treatments

Topical tretinoin (0.1%) ameliorates striae and the improvement may persist for almost a year after discontinuation of therapy.33 More recently, tretinoin has been shown to improve the clinical appearance of stretch marks during the active stage (striae rubra), although with not much effect during the mature stage (striae alba).34 Some of the studies have proven the inefficacy of the vitamin A derivative in the treatment of SD, but most of the patients included in these early studies presented with old lesions that had evolved into whitish atrophic scars.35 A study comparing topical 20% glycolic acid and 0.05% tretinoin versus 20% glycolic acid and 10% L-ascorbic acid, found that both regimens improved the appearance of striae alba.36

Hydrant Creams: 1) Trofolastin (a cream containing Centella asiatica extract, vitamin E, and collagen-elastin hydrolysates). The exact mechanism of action was identified as the stimulation of fibroblastic activity 37 and an antagonistic effect against glucocorticoids.38 2) Verum (a cream containing vitamin E, panthenol, hyaluronic acid, elastin and menthol). The results suggest that the product may show the benefit of massage alone.39 3) Alphastria (a cream composed of hyaluronic acid, allantoin, vitamin A, vitamin E, and dexpanthenol). Only one study was conducted, which concluded that the product markedly lowered the incidence of stretch mark development after pregnancy.40

Glycolic acid (GA): The exact mechanism of action of GA in the management of striae distensae is still unknown because, although GA is reported to stimulate collagen production by fibroblasts and to increase their proliferation in vivo and in vitro, which may be useful for the treatment of stretch marks.41,42 A study comparing topical 20% glycolic acid and 0.05% tretinoin versus 20% glycolic acid and 10% L-ascorbic acid, found that both regimens improved the appearance of striae alba.43

Trichloroacetic acid (TCA; 10–35%): It has been used for many years as a treatment option for striae distensae and is repeated at monthly intervals with reported improvement in texture and color of marks.44

Other topical products: Several oils have been used in the prevention of SD. A non-randomized, comparative study investigated the effect of almond oil in the prevention of SD in which they noted significant differences in the frequency of SD between the groups (almond oil and massage 20%, almond oil alone 38.8%, control 41.2%).45

Overall, there is limited evidence for the efficacy of topical therapy for the treatment of SD.

Microdermabrasion

Microdermabrasion may improve many skin problems including acne scars, skin texture irregularities, mottled pigmentation and fine wrinkles. Karimipour et al reported that microdermabrasion induces epidermal signal transduction pathways associated with remodelling of the dermal matrix.46 However, studies documenting the efficacy of rnicrodermabrasion in treatment of striae are lacking. Published in 1999, a book on microdermasion written by a French dermatologist, Francois Mahuzier, and translated to English, has a chapter "Microdermabrasion of stretch marks.47 The author states that 10-20 sessions of microdermabrasion at an interval of not less than 1 month, each session resulting in bleeding points, provide satisfactory results. The author concludes that, "microdermabrasion is the only effective treatment of stretch marks today."

Lasers

Lasers have recently become a popular therapeutic alternative to ameliorate and improve the appearance of stretch marks. Most commonly used lasers used include pulsed-dye laser (PDL), short- pulse carbon dioxide and erbium-substituted yttrium aluminium garnet (YAG), neodymium- doped YAG (Nd:YAG), diode, and Fraxel.

Pulsed dye laser: The dilated blood vessels render the striae rubrae a good candidate for PDL.48 The 585- nm pulsed dye laser has a moderate beneficial effect in the treatment of striae rubra.49 To evaluate the effectiveness of the 585-nm flashlamp-pumped pulse dye laser in treating cutaneous striae, 39 striae were treated with four treatment protocols.50 Subjectively, striae appeared to return toward the appearance of normal skin with all protocols. Objectively, shadow profilometry revealed that all treatment protocols reduced skin shadowing in striae. Laser treatment of SD should be avoided or used with great caution in darker skin types (IV–VI), because of the possibility of pigmentary alterations after treatment.51

Excimer laser: Studies have shown temporary repigmentation and improvement of leukoderma in SD with excimer laser, although it failed to show any improvement in skin atrophy.52,53 To evaluate the true efficacy of the 308-nm excimer laser for darkening striae alba, 10 subjects were treated using the excimer laser on the white lines of striae, while the normal skin near to and between the lines was covered with zinc oxide cream. The results of this study showed the weakly positive effect of the 308-nm excimer laser in the repigmentation of striae alba.54

Copper Bromide laser: copper-bromide laser (577-511 nm) has been used for stretch marks. A clinical study was conducted in 15 Italian women with stretch marks, treated with the CuBr laser (577-511 nm) and followed-up for 2 years.55 The results of the study concluded that the copper-bromide laser was effective in decreasing the size of the SD and there were some pathogenic considerations that justified the use of this laser.

1,450-nm Diode Laser: The non-ablative 1,450-nm diode laser has been shown to improve atrophic scars and may be expected to improve striae. To evaluate the efficacy of the 1,450-nm diode laser in the treatment of striae rubra and striae alba in Asian patients with skin types 4-6, striae on one half of the body in 11 patients were treated with the 1,450-nm diode laser with cryogen cooling spray with the other half serving as a control.56 None of the patients showed any noticeable improvement in the striae on the treated side compared to baseline and to the control areas. The study concluded that the non-ablative 1,450-nm diode laser is not useful in the treatment of striae in patients with skin types 4, 5, and 6.

1,064-nm Nd:YAG Laser: A study was aimed to verify the efficacy of this laser in the treatment of immature striae in which 20 patients with striae rubra were treated using the 1,064-nm long-pulsed Nd:YAG laser.57 A higher number of patients (55%) considered the results excellent when compared to the same assessment made by the doctor (40%).

Intense Pulsed Light: In order to assess the efficacy of IPL in the treatment of striae distensae, a prospective study was carried out in 15 women, all of them having late stage striae distensae of the abdomen.58 All the study subjects showed clinical and microscopical improvement after IPL. It seems to be a promising method of treatment for this common problem with minimal side-effects, a wide safety margin and no downtime.

Fractional Photothermolysis: To determine the efficacy of fractional photothermolysis in striae distensae, 22 women with striae distensae were treated with two sessions each of fractional photothermolysis at a pulse energy of 30 mJ, a density level of 6, and eight passes at intervals of 4 weeks and response to treatment was assessed by comparing pre- and post-treatment clinical photography and skin biopsy samples.59 Six of the 22 patients (27%) showed good to excellent clinical improvement from baseline, whereas the other 16 (63%) showed various degrees of improvement. This study concluded that Fractional photothermolysis may be effective in treating striae distensae, without significant side effects.

Ablative 10,600-nm carbon dioxide fractitional laser: Ablative 10,600-nm carbon dioxide fractional laser systems (CO₂ FS) have been used successfully for the treatment of various types of scars. To assess the therapeutic efficacy of CO₂ FS for the treatment of striae distensae, 27 women with striae distensae were treated in a single session with a CO₂ FS and clinical improvement was assessed by comparing pre- and post-treatment clinical photographs and participant satisfaction rates.60 The evaluation of clinical results 3 months after treatment showed that two of the 27 participants (7.4%) had grade clinical 4 improvement, 14 (51.9%) had grade 3 improvement, nine (33.3%) had grade 2 improvement, and two (7.4%) had grade 1 improvement. None of the participants showed worsening of their striae distensae.To assess and compare the efficacy and safety of nonablative fractional photothermolysis and ablative CO(2) fractional laser resurfacing in the treatment of striae distensae, 24 ethnic South Korean patients with varying degrees of atrophic striae alba in the abdomen were enrolled in a randomized blind split study and were treated with 1,550 nm fractional Er:Glass laser and ablative fractional CO(2) laser resurfacing.61 These results of the study support the use of nonablative fractional laser and ablative CO(2) fractional laser as effective and safe treatment modalities for striae distensae of Asian skin with neither treatment showing any greater clinical improvement than the other treatment.

UVB/UVA1 Combined Therapy: Besides lasers, light sources emitting ultraviolet B (UVB) irradiation have been shown to repigment striae distensae. A study was conducted on 9 patients with mature striae alba who received 10 treatment sessions, and biopsies were taken at the baseline and end of the study.62 At the end of the study, all patients reported some form of hyperpigmentation that was transient and did not affect any surrounding tissues. No changes were seen on biopsy to indicate an effective remodelling collagen effect of the device, although it needs further assessment. Another study was conducted to analyse the histologic and ultrastuctural changes seen after UVB laser- or light source-induced repigmentation of striae distensae in which analyses of biopsied skin after treatment with both the UVB laser and light source showed increased melanin content, hypertrophy of melanocytes, and an increase in the number of melanocytes in all patients.63

Radiofrequency devices: RF devices are based on the principle of heat generation that occurs in response to poor electrical conductance according to Ohm’s law (heat generation is directly correlated with tissue resistance). The heat that is generated is sufficient to cause thermal damage to the surrounding connective tissue,64 which is responsible for the partial denaturation of pre-existing elastic fibers and collagen bundles.65 Initial collagen denaturation within thermally modified deep tissue is thought to represent the mechanism for immediate tissue contraction; subsequent neocollagenesis further tightens the dermal tissue and reduces striae.66 The efficacy and safety of combination therapy with fractionated microneedle radiofrequency (RF) and fractional carbon dioxide (CO2) laser in the treatment of striae distensae has been evaluated revealing that this combination therapy is a safe treatment protocol with a positive therapeutic effect on striae distensae.67 A recent study evaluating the effectiveness of a RF device in combination with PDL subjected 37 Asian patients with darker skin tone with SD to a baseline treatment with a RF device and PDL.68 All histological evaluations demonstrated an increase in the amount of collagen fibers, and six of the nine specimens showed an increase in the number of elastic fibers.TriPollar RF device appears to be a promising alternative for the treatment of striae distensae in skin phototypes IV-V.69

Needling therapy:

To evaluate the effectiveness and safety of a disk microneedle therapy system (DTS) in the treatment of striae distensae, 16 Korean volunteers with striae distensae alba or rubra were enrolled which received three treatments using a DTS at 4-week intervals.70 Marked to excellent improvement was noted in seven (43.8%) patients, with minimal to moderate improvement in the remaining nine. This study revealed that Disk microneedle therapy system (DTS) can be effectively and safely used in the treatment of striae distensae without any significant side effects. Another study assessed and compared the efficacy and safety of needling therapy versus CO2 fractional laser in treatment of striae and the results supported the use of microneedle therapy over CO2 lasers for striae treatment.71

Platelet-rich plasma:

Platelet-rich plasma has these wound-healing properties, affecting endothelial cells, erythrocytes, and collagen,72 which potentially aids in the healing of the localized chronic inflammation believed to be a factor in the aetiology of striae distensae. Platelet-rich plasma is well tolerated by the patients and is a safe and cost effective treatment option for striae distensae.

Platelet-rich plasma alone is more effective than microdermabrasion alone in the treatment of striae distensae, but it is better to use the combination of both for more and rapid efficacy.73

The plasma fractional radiofrequency and transepidermal delivery of platelet-rich plasma using ultrasound has also been found to be useful in the treatment of striae distensae.74

Since thermal damage from intradermal RF has characteristics similar to those of many wounds, combination treatment with intradermal RF and autologous PRP would eventuate in enhanced localized collagen neogenesis and redistribution. In one of the studies, three sessions of intradermal RF were used combined with autologous PRP administered once every four weeks.75 All of the participants showed satisfactory changes and no patient was reported to show no improvement.

Transepidermal retinoic acid:

Transepidermal retinoic acid delivery using ablative fractional radiofrequency associated with acoustic pressure ultrasound has also been used for the treatment of stretch marks.76

Conclusion

Striae distensae are an extremely common, therapeutically challenging form of dermal scarring. Adequate scientific knowledge and the evidence behind both preventative and therapeutic agents are vital in order to understand striae and to offer patients the best therapeutic options. The treatment of this cosmetically distressing condition has been disappointing and there is no widely accepted surgical procedure for improving the appearance of stretch marks. Laser therapy has been advocated as a treatment for striae distensae.

Acknowledgements / Conflicts / Author Details
Competing Interests: 
None declared
Details of Authors: 
MOHAMMAD ABID KEEN (MBBS, MD DERMATOLOGY, STD and LEPROSY), Senior Resident, Postgraduate Department of Dermatology, STD and Leprosy, Government Medical College and Associated SMHS Hospital Srinagar.
Corresponding Author Details: 
DR MOHAMMAD ABID KEEN (MBBS, MD DERMATOLOGY, STD and LEPROSY), Senior Resident, Postgraduate Department of Dermatology, STD and Leprosy, Government Medical College and Associated SMHS Hospital Srinagar., Jammu & Kashmir, Pin code: 190010
Corresponding Author Email: 
keenabid31@gmail.com
References
References: 
  1. Garcia Hidalgo L. Dermatological complications of obesity. Am J Clin Dermatol 2002; 3: 497-506.
  2. Atwal GS, Manku LK, Griffiths CE et al. Striae gravidarum in primiparae. Br J Dermatol 2006; 155: 965–969.
  3. Cho S, Park ES, Lee DH, et al. Clinical features and risk factors for striae distensae in Korean adolescents. J Eur Acad Dermatol Venereol 2006; 20: 1108-1113.
  4. Sisson WR. Colored striae in adolescent children. J Pediatr 1954; 45:520–30.
  5. Kim BJ, Lee DH, Kim MN, et al. Fractional photothermolysis for the treatment of striae distensae in asian skin. Am J Clin Dermatol 2008; 9: 33-37.
  6. Dosal J, Handler MZ, Ricotti CA et al. Ulceration of abdominal striae distensae (stretch marks) in a cancer patient—quiz case. Arch Dermatol 2012; 148:385–390.
  7. Nieman LK, Ilias I. Evaluation and treatment of Cushing's syndrome. Am J Med 2005; 118: 1340- 1346.
  8. Tunzi M, Gray GR. Common skin conditions during pregnancy. Am Fam Physician 2007; 75: 211-218.
  9. Novak M. Colored striae in adolescent children. J Pediatr 2004; 145: 645.
  10. DiLernia V, Bonci A, Cattania M, et al. Striae distensae in monozygotic twins. Pediatr Dermatol 2001; 18: 261-2.
  11. Lee KS, Rho YJ, Jang SI, et al. Decreased expression of collagen and fibronectin genes in striae distensae tissue. Clin Exp Dermatol 1994; 19: 285-8.
  12. Viennet C, Bride J, Cohen-Letessier A, et al. Mechanical behavior of fibroblasts included in collagen lattices. J Soc Biol 2001; 195: 427-30.
  13. Burrows NP, Lovell CR. Disorders of connective tissue. In: Burns T, Breathnach S, Cox N, Griffith C, editors. Rook’s Textbook of dermatology, 7th edn. Oxford: Blackwell Science; 2004. p. 46- 7.
  14. Stevanovic DV. Corticosteroid induced atrophy of the skin with telegiectasia: a clinical and experimental study. Br J Dermatol 1972; 87: 548–56.
  15. Sparker MK, Garcia-Gonzalez E, Sanchez LT. Sclerosing and atrophying conditions. In: Schachner LA, Hansen RC, editors. Pediatric dermatology. 2nd edn. New York: Churchill Livingstone; 1996. p. 897.
  16. Johnston GA, Graham-Brown RA. The skin and disorders of the alimentary tract and the hepato biliary system. In: Freedberg IM, Eisen AZ, Wolff K, Austen KF, Goldsmith LA, Katz SI, editors. Fitzpatrick’s Dermatology in general medicine. 6th edn. New York: McGraw-Hill; 2003. p. 16-7.
  17. Basak P, Dhar S, Kanwar AJ. Involvement of the legs in idiopathic striae distensae – a case report. Indian J Dermatol 1989; 34: 21-2.
  18. Rosenthal DB. Striae atrophicae cutis. Lancet 1937; 232: 557-560.
  19. Watson RE, Parry EJ, Humphries JD, Jones CJ, Polson DW, Kielty CM, et al. Fibrillin microfibrils are reduced in skin exhibiting striae distensae. Br J Dermatol 1998; 138: 931-7.
  20. S Shuster. The cause of striae distensae. Acta Derm Venereol 1979; 59: 161.
  21. Y Shirai. Studies on striae cutis of puberty. Hiroshima Med sci 1959; 8: 215.
  22. K S Lee, YJ Rho, S I Jang, et al. Decreased expression of collagen and fibronectin genes in striae distensae tissue. Clin Exp Dermatol 1994; 19: 285.
  23. H M Sheu, H S Yu, C H Chang. Mast cell degranulation and elastolysis in the early stage of striae distensae. J Cut Pathol 1991; 18: 410.
  24. Burrows NP, Lovell CR. Disorders of connective tissue. In: Burns T, Breathnach S, Cox N, Griffith C, editors. Rook’s Textbook of dermatology, 7th edn. Oxford: Blackwell Science; 2004. p. 46- 7.
  25. Watson RE, Parry EJ, Humphries JD, et al. Fibrillin microfibrils are reduced in skin exhibiting striae distensae. Br J Dermatol 1998; 138: 931-7.
  26. Davey CM. Factors associated with the occurrence of striae gravidarum. J Obstet Gynaecol Br Commonw 1972; 79:1113–14.
  27. Atwal GS, Manku LK, Griffiths CE et al. Striae gravidarum in primiparae. Br J Dermatol 2006; 155: 965–9.
  28. Hermanns JF, Pierard GE. High-resolution epiluminescence color- imetry of striae distensae. J Eur Acad Dermatol Venereol 2006; 20: 282–7.
  29. Bleve M, Capra P, Pavanetto F et al. Ultrasound and 3D skin imaging: methods to evaluate efficacy of striae distensae treatment. Dermatol Res Pract 2012; 2012: 1–10.
  30. Rolfe H, Wurm E, Gilmore S. An investigation of striae distensae using reflectance confocal microscopy. Australas J Dermatol 2012; 53: 181–5.
  31. Elsaie ML, Baumann LS, Elsaaiee LT. Striae distensae (stretch marks) and different modalities of therapy: an update. Dermatol Surg 2009; 35: 563-573.
  32. Schwingel AC, Shimura Y, Nataka Y, et al. Exercise and striae distensae in obese women. Med Sci Sports Exerc 2003; 35: 33.
  33. Elson ML. Treatment of striae distensae with topical tretinoin. J Dermatol Surg Oncol 1990; 16: 267-70.
  34. Kang S, Kim KJ, Griffith CE, et al. Topical tretinoin (retinoic acid) improves early stretch marks. Arch Dermatol 1996; 132: 519–26
  35. Pribanich S, Simpson FG, Held B, et al. Low-dose tretinoin does not improve striae distensae: a double-blind, placebo controlled study. Cutis 1994; 54: 121–4.
  36. Ash K, Lord J, Zukowski M, et al. Comparison of topical therapy for striae alba (20% glycolic acid/0.05% tretinoin versus 20% glycolic acid/10% L-ascorbic acid). Dermatol Surg 1998; 24: 849-56.
  37. Velasco M, Romero E. Drug interaction between asiaticoside and some anti inflammatory drugs in wound healing of the rat. Curr Ther Res 1976; 18: 121–5.
  38. Brinkhaus B, Lindner M, Schuppan D, et al. Chemical, pharmacological and clinical profile of the East Asian medical plant Centella asiatica. Phytomedicine 2000; 7: 427–48.
  39. Wierrani F, Kozak W, Schramm W, et al. Attempt of preventive treatment of striae gravidarum using preventive massage ointment administration. Wiener Klinishe Wochenschrift 1992; 104: 42–4.
  40. De-Bauman M, Walther M, de-Weck R. Effectivness of alphastria cream in the prevention of pregnancy stretch marks (striae distensae). Results of a double-blind study. Gynakologische Rundschau 1987; 27: 79–84.
  41. Kim SJ, Park JH, Kim DH, et al. Increased in vivo collagen synthesis and in vitro cell proliferative effect of glycolic acid. Dermatol Surg 1998; 24: 1054–8.
  42. Okano Y, Abe Y, Masake H, et al. Biological effects of glycolic acid on dermal matrix metabolism mediated by dermal fibroblasts and epidermal keratinocytes. Exp Dermatol 2003; 12: 57–63.
  43. Ash K, Lord J, Zukowski M, et al. Comparison of topical therapy for striae alba (20% glycolic acid/0.05% tretinoin versus 20% glycolic acid/10% L-ascorbic acid). Dermatol Surg 1998; 24: 849-56.
  44. Obagi ZE, Obagi S, Alaiti S, et al. TCA-based blue peel: a standardized procedure with depth control. Dermatol Surg 1999; 25: 773–80.
  45. Timur S, Kafkasli A. The effect of bitter almond oil and massaging on striae gravidarum in primiparaous women. J Clin Nurs 2012; 21: 1570– 1576.
  46. Karimipour DJ, Kang S, Johnson TM, et al. Microdermabrasion: a molecular analysis following a single treatment. J Am Acad Dermatol 2005; 52: 215-23.
  47. Mahuzier F. Microdermabrasion of stretch marks in microdermabrasion or Parisian peel in practice. Marseille: Solalιditeurs; 1999.
  48. Karsai S, Roos S, Hammes S, et al. Pulsed dye laser: what’s new in non-vascular lesions. JEADV 2007;21:877–90.
  49. Jimeenez GP, Flores F, Berman B, et al. Treatment of striae rubra and striae alba with the 585 nm pulsed dye laser. Dermatol Surg 2003; 29: 362-5.
  50. McDaniel DH, Ash K, Zukowski M. Treatment of stretch marks with the 585-nm flashlamp-pumped pulsed dye laser. Dermatol Surg 1996; 22: 332-7.
  51. Nouri K, Romagosa R, Chartier T, et al. comparison of the 585 nm pulse dye laser and the short pulsed co2 laser in the treatment of striae distensae in skin types IV and VI. Dermatol Surg 1999; 25: 368–70.
  52. Goldberg DJ, Marmur ES, Hussain M. 308 nm excimer laser treatment of mature hypopigmented striae. Dermatol Surg 2003; 29: 596–9.
  53. Goldberg DJ, Marmur ES, Schmults C, et al. Histologic and ultrastructural analysis of ultraviolet B laser and light source treatment of leukoderma in striae distensae. Dermatol Surg 2005; 31: 385–7.
  54. Ostovari N, Saadat N, Nasiri S, et al. The 308-nm excimer laser in the darkening of the white lines of striae alba.J Dermatolog Treat 2010; 21: 229-31.
  55. Longo M, Postiglione MG, Marangoni O, et al. Two-year follow up results of copper bromide laser treatment of striae. J Clin Laser Med Surg 2003; 21: 157–60.
  56. Tay YK, Kwok C, Tan E. Non-ablative 1,450-nm diode laser treatment of striae distensae. Lasers Surg Med 2006; 38: 196–9.
  57. Goldman A, Rossato F, Pratti C. Stretch marks: treatment using the 1,064 nm Nd:YAG laser. Dermatol Surg 2008; 34: 1–7.
  58. Hernandez-Perez E, Charrier EC, Valencia-Ibiett E. Intense pulsed light in the treatment of striae distensae. Dermatol Surg 2002; 28: 1124–30.
  59. Bak H, Kim BJ, Lee WJ, et al. Treatment of striae distensae with fractional photothermolysis.Dermatol Surg 2009; 35: 1215-20.
  60. Lee SE, Kim JH, Lee SJ, et al. Treatment of striae distensae using an ablative 10,600-nm carbon dioxide fractional laser: a retrospective review of 27 participants.Dermatol Surg 2010; 36: 1683-90.
  61. Yang YJ, Lee GY. Treatment of Striae Distensae with Nonablative Fractional Laser versus Ablative CO2 Fractional Laser: A Randomized Controlled Trial.Ann Dermatol 2011; 23: 481-9.
  62. Sadick N, Magro C, Hoenig A. Prospective clinical and histological study to evaluate the efficacy and safety of a targeted high- intensity narrow band UVB/UVA1 therapy for striae alba. J Cosmet Laser Ther 2007; 9: 79–83.
  63. Goldberg DJ, Marmur ES, Schmults C, et al. Histologic and ultrastructural analysis of ultraviolet B laser and light source treatment of leukoderma in striae distensae.Dermatol Surg 2005; 31: 385-7.
  64. Khan MH, Victor F, Rao B, et al. Treatment of cellulite: Part II. Advances and controversies. J Am Acad Dermatol 2010; 62: 373–384.
  65. Fitzpatrick R, Geronemus R, Goldberg D, et al. Multicenter study of non-invasive radiofrequency for periorbital tissue tightening. Lasers Surg Med 2003; 33: 232–242.
  66. Arnoczky SP, Aksan A. Thermal modification of connective tissues: basic science considerations and clinical implications. J Am Acad Orthop Surg 2000; 8: 305–313.
  67. Ryu HW, Kim SA, Jung HR, et al. Clinical improvement of striae distensae in Korean patients using a combination of fractionated microneedleradiofrequency and fractional carbon dioxide laser.Dermatol Surg 2013; 39: 1452-8.
  68. Suh DH, Chang KY, Son HC, et al. Radiofrequency and 585-nm pulsed dye laser treatment of striae distensae: a report of 37 Asian patients. Dermatol Surg 2007; 33: 29–34.
  69. Manuskiatti W, Boonthaweeyuwat E, Varothai S.Treatment of striae distensae with a TriPollar radiofrequency device: a pilot study.J Dermatolog Treat 2009; 20: 359-64.
  70. Park KY, Kim HK, Kim SE, et al. Treatment of striae distensae using needling therapy: a pilot study. Dermatol Surg 2012; 38: 1823-8.
  71. Khater MH, Khattab FM, Abdelhaleem MR. Treatment of striae distensae with needling therapy versus CO2 fractional laser.J Cosmet Laser Ther2016; 18: 75-9.
  72. Anitua E, Andia I, Ardanza B, et al. Autologous platelets as a source of proteins for healing and tissue regeneration. Thromb Haemost 2004; 91: 4–15.
  73. Ibrahim ZA, El-Tatawy RA, El-Samongy MA, et al. Comparison between the efficacy and safety of platelet-rich plasma vs. microdermabrasion in the treatment of striae distensae: clinical and histopathological study. J Cosmet Dermatol 2015; 14: 336-46.
  74. Suh DH, Lee SJ, Lee JH, et al. Treatment of striae distensae combined enhanced penetration platelet-rich plasma and ultrasound after plasma fractional radiofrequency.J Cosmet Laser Ther2012; 14: 272-6.
  75. Kim IS, Park KY, Kim BJ, et al. Efficacy of intradermal radiofrequency combined with autologous platelet-rich plasma in striae distensae: a pilot study. Int J Dermatol 2012; 51: 1253-8.
  76. Issa MC, de Britto Pereira Kassuga LE, Chevrand NS, et al. Transepidermal retinoic acid delivery using ablative fractional radiofrequency associated with acoustic pressure ultrasound for stretch marks treatment.Lasers Surg Med 2013; 45: 81-8.

A comprehensive review on the pregnancy dermatoses.

Authors
Mohammad Adil,Tasleem Arif and Syed Suhail Amin
Article Citation and PDF Link
BJMP 2016;9(1):a906
Abstract / Summary
Abstract: 

Pregnancy results in cutaneous changes in more than 90% of women. This is the result of the altered endocrine, metabolic and immunological state in the female. Many cutaneous changes are common and benign; these are referred to as the physiological changes of pregnancy. They may be of cosmetic concern to the patient and seldom require intervention. These changes are so well recognized that they act as contributory evidence of pregnancy. Many pre-existing dermatological conditions tend to change in pregnancy; some are aggravated while others may be relieved. Knowledge of these conditions is important to forewarn the patient and to prepare for upcoming complications. There are a group of dermatoses specific to pregnancy and there has been much confusion in the literature about their classification and nomenclature. Atopic Eruption of Pregnancy is the most common pregnancy specific dermatoses followed by Polymorphic Eruption of Pregnancy. These are benign conditions with no risk to the mother or baby. Pemphigoid Gestationis and Intrahepatic Cholestasis of Pregnancy carry fetal risk and require antepartal surveillance. This article discusses the current knowledge of the various cutaneous changes of pregnancy with emphasis on their clinical features, diagnosis, management and prognosis.

Abbreviations: 
Melanocyte Stimulating Hormone (MSH), Systemic Lupus Erythematosus (SLE), Pemphigoid Gestationis (PG), Herpes Gestationis (HG), Polymorphic Eruption of Pregnancy (PEP), Pruritic Urticarial Papules and Plaques of Pregnancy (PUPPP), Prurigo of Pregnancy (PP), Pruritic Folliculitis of Pregnancy (PF), Intrahepatic Cholestasis of Pregnancy (ICP), Atopic Eruption of Pregnancy (AEP), Bullous Pemphigoid Antigen 2 (BPAg2)
Keywords: 
Dermatoses, endocrine, physiological changes, pregnancy.

INTRODUCTION

The state of pregnancy results in a multitude of cutaneous changes in the female. These are a reflection of the profound alterations in the endocrine, metabolic and immunological profiles that occur during this period.1 Skin manifestations occur due to the production of a number of proteins and steroid hormones by the fetoplacental unit and also by the maternal pituitary, thyroid and adrenals.2 The placenta, a new endocrine organ in the woman, produces progesterone. Dehydroepiandrosterone is produced by the fetal adrenals from pregnenolone and this is aromatized to estriol. At term, the level of progesterone is 7 times, estradiol is 130 times and prolactin level is 19 times of that present at 8 weeks of gestation.3 There occurs an overall preference for the Th2 cytokine profile, which helps in fetal protection from the immune system.4 This is due to the high levels of progesterone, which promotes Th2 cytokines like IL-4, IL-5 and IL-10 and has inhibitory effects on TNF alpha production. Oestrogen suppresses IL-2 production. The postpartum period is marked by withdrawal of hormones and consequent elevation of Th1 cytokine levels.4

Cutaneous changes develop in more than 90% of all pregnant females.5 These include common cutaneous changes that occur in most cases to severe diseases, some of which are seen exclusively in the pregnant and postpartum state. Cutaneous manifestations can be grouped into three broad categories: physiological cutaneous changes related to pregnancy; diseases modified by pregnancy and specific dermatoses of pregnancy.6

PHYSIOLOGICAL CHANGES IN PREGNANCY

These changes are so common that they are not considered abnormal. Rather, they provide contributory evidence of a pregnant state. This however, does not mean they are cosmetically acceptable to all patients. The various physiological changes during pregnancy have been summarized in Table 1.

Table 1: Physiological changes in pregnancy

Pigmentation
Generalized hyperpigmentation
Pigmentation of inner thigh, genitalia, axilla
Secondary areola
Linea nigra
Chloasma
Prominence/ appearance of pigmentary demarcation lines
Enlargement and darkening of freckles, naevi and scars
Connective tissue changes
Striae distensae (Striae gravidarum)
Molluscum fibrosum gravidarum
Vascular changes
Oedema of distal extremities and hands
Spider angiomas
Palmar erythema
Leg varicosities
Rectal haemorrhoids
Cutis marmorata
Capillary haemangioma
Glandular changes
Miliaria
Dyshidrotic eczema
Montgomery’s tubercles
Aggravation of acne
Oral mucosal changes
Oedema and hyperaemia of gingivae
Pregnancy epulis
Hair changes
Hirsuitism
Hypertrichosis
Delayed anagen release after delivery
Nail changes
Brittle nail plate
Onycholysis
Beau’s lines after delivery

Pigmentation:

Hyperpigmentation is one of the most common and early signs of pregnancy, seen in more than 90% of patients.7 High levels of Melanocyte Stimulating Hormone (MSH), oestrogen and progesterone are believed to be responsible for hyperpigmentation. Progesterone augments the oestrogen mediated melanin output, the levels of which correlate with pigmentary changes.8

Generalized hyperpigmentation is seen which is more marked in the dark haired skin.6 Pigmented areas of the body, namely the genitalia, perineum, areolae and upper medial thighs, demonstrate more pronounced pigmentation. Linea nigra, a hyperpigmented line extending from the pubic symphysis to umbilicus and further up to the xiphisternum, replaces the linea alba.9 Chloasma, also termed as mask of pregnancy, is the well marginated brownish pigmentation of the face like melasma. It is seen in 45-75% of pregnant women in western literature but in less than 10% cases in women with pigmented skin.5,10,11 Pigmentary demarcation lines appear on the limbs with borders of abrupt transition; freckles, naevi and scars tend to darken and enlarge.12

The pigmentation gradually fades after delivery, though the resolution of skin colour is usually incomplete. Chloasma tends to persist in 30% cases postpartum.13 Sun protection and reassurance is all that is needed. Topical formulations containing hydroquinone and tretinoin are avoided in pregnancy and can be added after delivery.

Physiological connective tissue changes:

Gross distension of abdomen with adrenocortical activities are responsible for the red-blue depressed streaks seen on abdomen and breasts in 70-90% pregnancies, called striae distensae.5,14 These usually develop in the second trimester. Females with pre-existing striae on breasts and thighs are more likely to develop striae gravidarum15, seen in White women more than Asian and African-American.14 Preventive therapies are controversial and postpartum treatment options include topical tretinoin, excimer laser or surgery.10

Soft tissue fibromas of pregnancy are called molluscum fibrosum gravidarum. They appear in the second trimester on the neck, face and beneath the breasts. These disappear after delivery.16

Physiological vascular changes:

Vascular growth factors released during pregnancy by the pituitary, adrenals and placenta are believed to be causative and this has been demonstrated in vitro as well.17 Non-pitting oedema of the face, hands and feet is present in around half of all females in the later part of pregnancy.13 This is probably due to sodium and fluid retention and pressure of the gravid uterus on the inferior vena cava. Spider naevi or spider angiomas are small raised lesions with a central pulsatile punctum and radiating telangiectatic vessels frequently present over the area drained by the superior vena cava. They are present in 67% of White women and 11% Black women during the second trimester.5 Palmar erythema is seen in two-thirds of White and one-third of Black women.8 Other vascular changes include varicosities of legs and anus (40%)13, cutis marmorata (0.7%)18 and capillary haemangioma (5%)9. These changes revert after the postpartum period.

Physiological glandular changes:

Eccrine gland activity is usually increased but the palms show decreased sweating. Thus, the incidence of miliaria and dishidrotic eczema is increased. There is inconclusive evidence to suggest that apocrine gland activity is decreased during pregnancy.19 Sebaceous activity increases in the third trimester leading to acne and enlargement of Montgomery’s tubercles.14 One-third to half of all pregnant women develop these tubercles, which are modified sebaceous glands.5,8 However, sebum excretion has not been found to decrease in lactating females post-delivery.20

Oral mucosal changes:

Oedema and hyperaemia of the gingivae in pregnancy is attributable to local irritation and nutritional deficiencies and is seen in around 80% women.5 Gingivitis not related to poor oral hygiene may occur. Granuloma gravidarum or pregnancy epulis might occur that regresses postpartum.

Hair changes:

Hair changes are seen in 3-12% of pregnant females.21 Hirsuitism and hypertrichosis occurs due to oestrogen. This leads to an increase in the percentage of hair in anagen.2 Approximately 2-3 months after delivery, loss of telogen hair occurs.22 This is termed as late anagen release as the hair follicles are no longer stimulated to stay in anagen phase by the maternal hormones. The hair recovery occurs in 3-12 months. A small number of females may experience episodic shedding of hair for long periods. This has been proposed to be due to the inability of some hair follicles to revert to asynchronous shedding.23 Rarely, male pattern baldness may occur in women.2

Nail changes:

Nail growth increases during pregnancy.6 Brittleness of the nail plate and distal onycholysis may be seen.19 Beau’s lines may develop after delivery.12 Reassurance is all that is needed for these benign nail problems.

DISEASES MODIFIED BY PREGNANCY

Many pre-existing dermatoses may be exacerbated or ameliorated by pregnancy. Certain tumours may also show remission or exacerbation. This is due to the shift in pregnancy to the Th2 state and a return to Th1 state in the postpartum period and also the discontinuation of some drugs due to their teratogenic potential.

Infections:

Depressed cell-mediated immunity makes the pregnant woman susceptible to more severe and frequent infections.24

Candidiasis is quite common and was found to be the commonest cause of white discharge per vagina, being present in 22% pregnant females.5 Half of all neonates born to infected mothers are positive for Candida and some may show signs of infection.25 Pityrosporum folliculitis, caused by Pityrosporum ovale, is more common in pregnancy.25

Genital warts are the commonest sexually transmitted disease seen in 4.7% subjects, these increase in size during pregnancy.9,25 Prophylactic caesarian section to prevent laryngeal papillomas in the neonate is not recommended now.26 Herpes simplex virus infection carries 50% risk of transmission to neonate in the primary episode and 5% risk in recurrent episode, caesarean section might be warranted to prevent such transmission.26 Varicella zoster virus infection has been reported to cause pneumonia in 14% of mothers and death in 3%.27 Bowenoid papulosis, caused by human papilloma virus appears first during pregnancy or may get aggravated.6

Pregnancy prepones the clinical manifestations in HIV infected females, possibly due to additive immune suppression. Pneumocystis pneumonia or listeriosis may prove to be fatal.27 Kaposi’s sarcoma may occur in these females.27 20-30% women present with leprosy for the first time in pregnancy and the postpartum period.28 The disease tends to downgrade towards the lepromatous pole in pregnancy and upgrades during lactation.29 Type 1 lepra reactions are more frequent in the first trimester and after delivery, whereas type 2 lepra reactions peak in third trimester.29 Trichomoniasis is diagnosed in 60% of pregnant women.25

Autoimmune diseases:

Systemic Lupus Erythematosus (SLE) is associated with a better prognosis than previously thought, if the disease is in remission and nephropathy and cardiomyopathy are not present.10 If the disease is active, half of the patients’ disease will get worse and there might be fatalities.14 SLE tends to be more severe if it first presents in pregnancy.14 Babies of such mothers are likely to develop neonatal lupus.

Patients with scleroderma are usually unaffected and some are improved in pregnancy. However, occasional reports of renal crisis, hypertension and pre-eclampsia are reported.30 Course of dermatomyositis is usually unaltered but the disease may worsen in some patients.31

Pemphigus tends to be exacerbated or present for the first time in pregnancy.32 The clinical presentation in pregnancy is similar to that of the regular presentation. Differentiation from herpes gestationis is important.

Metabolic diseases:

Effect of pregnancy on porphyria cutanea tarda is not clear, though some females show biochemical and clinical deterioration.33 Acrodermatitis enteropathica shows clinical worsening.34

Connective tissue diseases:

Pregnancy can lead to bleeding, uterine lacerations and wound dehiscence in patients of Ehlers-Danlos syndrome. Pseudoxanthoma elasticum patients may suffer massive gastrointestinal bleeds.35 Lichen sclerosis et atrophicus of the vulva usually improves in pregnancy and a normal delivery is mostly possible.

Disorders of glands:

Acne can aggravate during pregnancy. Hidradenitis suppurativa and Fox-Fordyce disease become better as a result of decreased apocrine gland activity.27

Keratinization diseases:

The course of psoriasis remains unaltered in 40% females during pregnancy while it improves in a similar percentage of females and worsens in the remaining.36 It is more likely to deteriorate in the postpartum period.37 Psoriatic arthritis has been found to worsen or present for the first time in pregnancy.2

Generalized pustular psoriasis of Von Zambusch may rarely occur. Though most patients have a preceding or family history of psoriasis, some may develop the disease without ever having a preceding episode.38 Peak incidence is seen in the last trimester and the disease tends to recur.38 Multiple, discrete, sterile pustules at the margins of erythematous macules on the umbilicus, medial thigh, axillae, inframammary folds, gluteal creases and sides of neck are seen. These break to form erosions and crusts. Painful, circinate mucosal erosions may form. Prednisolone is used for management.12 Von Zambusch pustular psoriasis of pregnancy was earlier termed ‘Impetigo Herpetiformis’ but the term is best avoided as it is impossible to differentiate it from the former, both clinically and histologically.6 Erythrokeratoderma variabilis is reported to worsen during pregnancy.27

Tumours:

A melanoma that develops during pregnancy carries worse prognosis but if pregnancy occurs after the tumour is resected, the prognosis is unaltered.39 Metastasis in the fetus has been seen and a minimum period of two years following tumour resection is recommended.32 A female with neurofibromatosis may develop neurofibroma for the first time in pregnancy or older neurofibromas may grow in size. Rupture of major vessels may occur.6 Pregnancy may worsen mycosis fungoides and eosinophilic granuloma.6

Miscellaneous diseases:

Prognosis of atopic dermatitis is unpredictable in pregnancy, with reports of both improvement and worsening.27 Predisposed patients may first develop atopic dermatitis during pregnancy.40 Allergic contact dermatitis may improve in pregnancy.12 Hand eczema may worsen in the puerperal period.6 Erythema multiforme may be precipitated by pregnancy.6 Autoimmune progesterone dermatitis has been described in pregnancy.12 This disease is characterized by hypersensitivity to progesterone demonstrated by a positive intradermal skin test and cutaneous lesions resembling urticaria, eczema, erythema multiforme and dermatitis herpetiformis.41 The disease is associated with fetal mortality and recurs in subsequent pregnancies.12

PREGNANCY SPECIFIC DERMATOSES

These are a heterogeneous group of inflammatory skin diseases specific for pregnancy.42 Most of these conditions are benign and resolve spontaneously in the postpartum period but a few of these are associated with fetal complications.42 Almost all of them present with pruritus and a cutaneous eruption of varying severity.5

Classification:

The first attempt to classify these conditions was made by Holmes and Black in 1982-83 who classified them into: a) Pemphigoid Gestationis (PG) or Herpes Gestationis(HG), b) Polymorphic Eruption of Pregnancy (PEP) or Pruritic Urticarial Papules and Plaques of Pregnancy (PUPPP), c) Prurigo of Pregnancy (PP) and d) Pruritic Folliculitis of Pregnancy (PF).43,44 Shornick was of the view that all patients with PF also had papular dermatitis, so he included PF in the PP group. He included Intrahepatic Cholestasis of Pregnancy (ICP) in his classification for dermatoses where secondary skin lesions due to scratching are produced. He proposed that failure to consider ICP in the classification has led to confusion in terminology of pregnancy specific diseases. Thus, his classification included PG, PEP, PP and ICP.45 Ambros-Rudolph et al carried out a retrospective review of 505 pregnant patients over a 10 year period and gave a more rationalised classification system in 2006. They clubbed PP, PF and eczema of pregnancy in one group called Atopic Eruption of Pregnancy (AEP) due to their overlapping features and found this group to be the most common pruritic condition in pregnancy. Thus, they proposed four conditions: a) AEP, b) PEP, c) PG and d) ICP.46 The various specific pregnancy dermatoses have been elaborated in Table 2.

Table 2: Comparison of different pregnancy specific dermatoses in relation to clinical characteristics, prognosis, investigations and treatment.

  AEP PEP PG ICP
Pruritus + + + +
Primary cutaneous involvement + + + -
Skin lesions Eczematous or papular Papules, vesicles and urticarial lesions Vesiculo bullous lesion on urticarial base Excoriations, papules secondary to scratching
Site of lesions Trunk, extensors of limbs, rest of the body also involved Abdominal involvement, in striae distensae, periumbilical sparing Abdominal, particularly periumbilical involvement Palms and soles followed by rest of the body
Time First trimester Third trimester, Post partum Second and third trimester, post partum Second and third trimester
Risk with primigravidae - + - -
Association with multiparity - + - +
Flare at delivery - - + -
Recurrence + - + +
Family history + + - +
Histopathology Non-specific Non-specific Specific, sub epidermal vesicle Non-specific
Immunofluorescence - - Linear deposition of C3 -
Other lab findings Ig E elevated - Indirect IMF + Increased serum bile acids
Maternal risk - - Progression to pemphigoid, thyroid dysfunction Gallstones, Jaundice
Fetal risk - - Prematurity, Small for age baby, neonatal blistering Premature births, fetal distress, stillbirth
Treatment Steroids, antihistaminics Steroids, antihistaminics Oral steroids, antihistaminics Ursodeoxycholic acid

Atopic eruption of Pregnancy (AEP): (Syn: Besnier’s prurigo, prurigo gestationis, Nurse’s early onset prurigo of pregnancy)

It is the most common pregnancy specific dermatoses that includes eczematous or papular lesions in females with personal or family history of atopy and elevated IgE - accounting for nearly half of all patients.46 The disease tends to recur in subsequent pregnancies with 75% of all cases occurring before the start of the third trimester.47 It carries no risk for the mother or baby however, infant may develop atopy later in life.48 Treatment is symptomatic with antihistamines and corticosteroids.

E-type AEP: This group comprises of 67% of AEP patients and includes patients with eczematous features; previously referred to as Eczema of Pregnancy (EP). It was not until 1999 that a high prevalence of atopic eczema was noted in pregnancy.49 80% of pregnant women develop the first episode of atopic dermatitis during pregnancy.46 This is attributed to the Th2 cytokine profile in pregnancy and a dominant humoral immunity.4 It is more common in primigravida, in single gestation, begins in early pregnancy and affects whole body including face, palms and soles.46

P-type AEP: This group includes what was referred to previously as Prurigo of Pregnancy and Pruritic Folliculitis of Pregnancy. Prurigo of Pregnancy (PP) is seen in one out of 300 to 450 pregnancies and occurs predominantly in the second to third trimester.50 Excoriated or crusted papules are seen over the extensors of extremities and abdomen and are associated with some eczematization. The eruption lasts up to 3 months after delivery and recurrences in subsequent pregnancies are common.51 PP is associated with ICP with the differentiating feature being the absence of a primary lesion in the latter.50 Personal and family history of atopic dermatitis or raised IgE may be seen in PP.52 Serology is normal. There are no specific changes on histopathology and immunofluorescence results are found to be negative.50 There appears to be no maternal or fetal risk.45

Pruritic Folliculitis of Pregnancy (PF), first described by Zoberman and Farmer, is now believed to be as common as PG or PP, though only a few cases have been reported.50 It begins in the latter two trimesters and affects roughly one in 3000 pregnancies.51 Pruritus is not a defining feature, despite what the name suggests.2 Multiple, follicular papules and pustules occur on the shoulders, arms, chest, upper back and abdomen and are acneiform in nature.42 The lesions tend to resolve in a couple of months following delivery. Histopathological examination reveals non-specific features with sterile folliculitis and immunofluorescence studies are negative.50 No maternal or fetal risk is described except for low birth weight neonates in a single study.52 Pathogenesis of PF is unknown with no definite role of androgens or immunologic abnormalities.53 There is no evidence to suggest that it is a hormonally aggravated acne as proposed by some workers.54

Polymorphic Eruption of Pregnancy (PEP): (Syn: Pruritic Urticarial Papules and Plaques of Pregnancy or PUPPP, Bourne’s Toxaemic Rash of Pregnancy, Toxic Erythema of Pregnancy, Nurse’s Late Prurigo of Pregnancy)

With a prevalence of a case in every 130-300 pregnancies, this disease is the second most common pregnancy specific dermatoses and was seen in 21.6% pregnancies reviewed by Ambros-Rudolph et al.46 They found it began in late pregnancy in 83% cases and 15% in the postpartum period.46 The disease occurs predominantly in primigravida and a familial predisposition is present.55 Lesions are pleomorphic, usually urticarial but purpuric, vesicular, polycyclic and targetoid lesions may be present. The striae on the abdomen are the first to be involved and there is a characteristic periumbilical sparing.56 The lesions seldom occur on the body above the breast and on hands and feet.12 The lesions resolve with scaling and crusting in six weeks. The disease is more common in excessive weight gain during pregnancy and in multiple gestation.57,58 Histopathology is non-specific and shows spongiosis, occasional subepidermal split and eosinophilic infiltration. Serology and immunofluorescence is negative.50 Treatment is symptomatic, oral steroids are needed in severe cases. There are no associated maternal or fetal complictions,59 although infants may later develop atopic dermatitis.2

The pathogenesis is unknown however, the abdominal distension leading to collagen and elastic fibre damage in striae is hypothesized, leading to formation of antigens and triggering inflammatory cascade.60 The role of progesterone has been suggested by the increased progesterone receptor immunoreactivity in skin lesions of PEP.61 The discovery of fetal DNA in skin lesions of women with PEP has furthered the hypothesis that abdominal distension leads to increased permeability of vessels and permit chimeric cell migration in the maternal skin.62 Linear IgM dermatosis of pregnancy is an entity characterized by pruritic, red, follicular papules and pustules on the abdomen and proximal extremities seen after 36 weeks gestation and a linear band of IgM deposition on basement membrane zone. It has been characterized as a variant of PEP or PP by different authors.12

Pemphigoid Gestationis (PG): (Syn: Herpes Gestationis or HG, Gestational Pemphogoid, Dermatitis Herpetiformis of Pregnancy)

PG is the most clearly characterized pregnancy dermatosis and the one which also affects the fetal skin.63 It is a rare, self-limiting, autoimmune bullous disease with an incidence of 1:1700 to 1:50000 pregnancies.63 Mean onset occurs at 21 weeks gestation, though it occurs in the postpartum period in a fifth of all cases.64 Constitutional symptoms, burning and itching herald the onset of the disease. Half of patients develop urticarial lesions on the abdomen, particularly in the periumbilical region, that change rapidly to a generalized bullous eruption usually sparing the face, palms, soles and mucosae. Vesicles may arise in herpetiform or circinate distribution. Face is involved in 10% cases and oral mucosa in 20%.12 The disease shows spontaneous improvement in late gestation but flares may occur at the time of delivery in 75% of the cases.63 Though the disease may remit after a few weeks after delivery, a protracted course, conversion to bullous pemphigoid or recurrence with menstrual cycle and use of oral contraceptive pills has been reported.50 PG tends to recur in subsequent pregnancies in a more severe form and at an early stage with longer stay in postpartum.50 Skipped pregnancies have been described.63,65 The disease is also linked with hydatiform mole and choriocarcinoma.66

The classical histopathological finding is the presence of a subepidermal vesicle, spongiosis and an infiltrate consisting of lymphocytes, histiocytes and eosinophils.64 An inverted tear drop appearance due to oedema in the dermal papilla is seen in early urticarial lesions.15 Direct immunofluorescence reveals a linear deposition of C3 along the dermo-epidermal junction in 100% cases and is diagnostic of the disease, while a salt split skin shows an epidermal staining.67 Antithyroid antibodies may be present but thyroid dysfunction is not common.63 Systemic corticosteroids are the mainstay of management. About one in ten children born to women with PG develop blisters due to passive transfer of antibodies, this resolves on its own. Severity of the disease has been correlated with the risk of prematurity and small for gestational age babies.68

Pathogenesis of PG involves the production of IgG1 antibodies against NC16A domain of carboxyl terminus of Bullous Pemphigoid Antigen 2 (BPAg2), leading to activation of complement, recruitment of eosinophils to the local site and damage of the basement membrane and consequent blistering.2 The aberrant expression of MHC class II antigens of paternal haplotype is believed to stimulate an allogenic response to placental basement membrane and this is believed to cross react with the skin in PG.63,69

Intrahepatic Cholestasis of pregnancy (ICP): (Syn: Obstetric Cholestasis, Pruritus Gravidarum, Icterus Gravidarum, Recurrent Jaundice of Pregnancy, Idiopathic Jaundice of Pregnancy)

Pruritus in pregnancy is fairly common and can be due to various reasons like pregnancy specific dermatoses and other co-existing dermatoses such as scabies, urticaria, atopic dermatitis, drug reactions etc. It was found to be present in more than half of 170 pregnant women in an Indian study.70 This must be differentiated from ICP where the skin lesions arise secondary to itching.

ICP was first described by Kehr in 1907.63 ICP being referred to Pruritus Gravidarum (for pruritus without skin changes occurring early in pregnancy and related to atopic diathesis and no cholestasis) and Prurigo Gravidarum (for pruritus associated with PP like skin lesions and associated with cholestasis) lead to much confusion regarding nomenclature.63 The disease has an incidence of 10-150 cases per 10,000 pregnancies71, being more common in South America and Scandinavia, probably due to dietary factors.50 Patients complain of sudden onset pruritus beginning from the palms and soles and later generalizing to the whole body. Skin lesions are secondary to itching and range from excoriations to prurigo nodularis, extensors are more severely involved. Jaundice is seen in 20% cases only.72 Clay coloured stools, dark urine and haemorrhage secondary to vitamin K malabsorption can occur. Family history can be elicited in half of the cases and an association with multiple gestation is described.73 Resolution of ICP occurs soon after delivery. Recurrence in subsequent pregnancies is seen in 45-70% cases and routinely with the use of oral contraceptive pills, though no detectable abnormalities are seen in the duration between two pregnancies.63 Histopathology is non-specific and immunofluorescence is negative. Diagnosis is made by increased serum bile acid levels, transaminases are elevated. Prothrombin time may be prolonged. A 2.7 times increased risk of gallstones is reported in primigravida with ICP compared to non-pregnant women.74 ICP is associated with significant fetal morbidity including premature births in 20-60% cases, intrapartum fetal distress including meconium aspiration in 20-30% and fetal mortality in 1-2%.71 Risk is particularly more if serum bile acid levels exceed 40 micromoles per litre.75 Meconium may cause umbilical vein compression and induction of labour at 36 weeks gestation has been recommended in severe cases.50 The goal of treatment is reduction of serum bile acids. Ursodeoxycholic acid, given in the dose of 15mg/kg orally daily is the only proven therapeutic agent that decreases fetal mortality.63,76 Cholestyramine reduces vitamin K absorption and increases the risk of haemorrhage. Other agents like S-adenosylmethionine, dexamethasone, silymarin, phenobarbitone, epomediol and activated charcoal are not that effective and do not affect fetal risk.63 Topical emollients and antipruritic agents offer symptomatic relief but antihistamines are not that effective.50

The key event in the pathogenesis of ICP is elevation of bile acids. Oestrogens are said to have cholestatic properties by reducing hepatocyte bile acid uptake and also by inhibiting basolateral transport proteins.50 Progesterone may additionally saturate the transport capacity of these transport proteins in hepatocyte.71 Genetic predisposition occurs due to mutation in genes encoding bile transport proteins, with cholestasis developing in pregnancy as their capacity to secrete substance is exceeded.63 Bile acids passing through the placenta produce vasoconstriction of placental veins, fetal cardiomyocyte dysfunction and also abnormal uterine contractility, all leading to fetal hypoxia.71

CONCLUSION

Pregnancy is associated with a wide variety of cutaneous changes. These may range from common, benign changes termed physiological or more severe, posing significant risk to the mother as well as the baby. Physiological pregnancy changes may be of cosmetic concern to the patient and seldom need anything more than counselling. Pre-existing dermatoses may aggravate during this period, posing a challenge to the treating physician. Women suffering from such diseases need to be warned of complications and risks before trying to conceive. A strict watch for possible complications and appropriate management at an early stage is warranted. Women should also be looked for pregnancy specific dermatoses and their complaints should not be lightly overlooked as non-specific or physiological. Careful history and examination with a judicious use of investigations will help to arrive at a diagnosis and in prompt institution of treatment.

Acknowledgements / Conflicts / Author Details
Acknowledgement: 
None
Competing Interests: 
None declared
Details of Authors: 
MOHAMMAD ADIL; MBBS, MD (Dermatology, STD’s & Leprosy); Senior Resident, Postgraduate Department of Dermatology, STDs and Leprosy; Jawaharlal Nehru Medical College (JNMC), Aligarh Muslim University (AMU), Aligarh, India. TASLEEM ARIF; MBBS, MD (Dermatology, STD’s & Leprosy); Assistant Professor, Postgraduate Department of Dermatology, STDs and Leprosy; Jawaharlal Nehru Medical College (JNMC), Aligarh Muslim University (AMU), Aligarh, India. SYED SUHAIL AMIN; MBBS, MD (Dermatology, STD’s & Leprosy); Head of the Department, Postgraduate Department of Dermatology, STDs and Leprosy; Jawaharlal Nehru Medical College (JNMC), Aligarh Muslim University (AMU), Aligarh, India.
Corresponding Author Details: 
TASLEEM ARIF; MBBS, MD (Dermatology, STD’s & Leprosy); Assistant Professor, Postgraduate Department of Dermatology, STDs and Leprosy; Jawaharlal Nehru Medical College (JNMC), Aligarh Muslim University (AMU), Aligarh, India.
Corresponding Author Email: 
dr_tasleem_arif@yahoo.com
References
References: 
  1. Karen JK, Pomeranz MK. Skin changes and diseases in pregnancy. In: Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell DJ, Wolff K, editors. Fitzpatrick’s dermatology in general medicine. 8th ed. New York: McGraw Hill; 2012. p 1204-12. 
  2. Vora RV, Gupta R, Mehta MJ, Chaudhari AH, Pilani AP, Patel N. Pregnancy and skin. J Family Med Prim Care 2014;3(4):318-24.
  3. Freinkel N, Metzger BE. Metabolic changes in pregnancy. In: Wilson JD, Foster DW, editors. Williams Textbook of Endocrinology, 8th ed. Philadelphia: Appleton & Lange; 1992. p 993-1005.
  4. Wilder RL. Hormones, pregnancy and autoimmune disease. Ann N Y Acad Sci 1998;840;45-50.
  5. Kumari R, Jaisankar TJ, Thappa DM. A clinical study of skin changes in pregnancy. Indian J Dermatol Venereol Leprol 2007;73(2):141.
  6. Millington GWM, Graham-Brown RAC. Skin and skin disease throughout life. In: Burns T, Brethnach S, Cox N, Griffiths C, editors. Rook’s textbook of dermatology. 8th ed. West Sussex: Blackwell Publishing; 2010. p 8.9-13.
  7. Elling SV, Powell FC. Physiological changes in the skin during pregnancy. Clin Dermatol 1997;15:35-43.
  8. Martin AG, Leal-Khouri S. Physiologic skin changes associated with pregnancy. Int J Dermatol 1992;31:375-8.
  9. Nussbaum R, Benedetto AV. Cosmetic aspects of pregnancy. Clin Dermatol 2006;24:133–41.
  10. Barankin B, Silver SG, Carruthers A. The skin in pregnancy. J Cutan Med Surg 2002;6:236-40.
  11. Raj S, Khopkar U, Kapasi A, Wadhwa SL. Skin in pregnancy. Indian J Dermatol Venereol Leprol 1992;58:84-8.
  12. Kar S, Krishnan A, Shivkumar PV. Pregnancy and skin. J Obstet Gynecol India 2012;62:268-75.
  13. Wilton GB, Lewis CW. Dermatoses of pregnancy. J Am Acad Dermatol 1982;6:977-98.
  14. Kroumpouzos G, Cohen LM. Dermatoses of pregnancy. J Am Acad Dermatol 2001;45:1-19.
  15. Chang AL, Agredano YZ, Kimball AB. Risk factors associated with striae gravidarum. J Am Acad Dermatol 2004;51:881-5.
  16. Tunzi M, Gray GR. Common skin conditions during pregnancy. Am Fam Physician 2007;75:211-8.
  17. Garcia-Gonzalez E, Ahued-Ahued R, Arroyo E, Montes-De-Oca D, Granados J. Immunology of the cutaneous disorders of pregnancy. Int J Dermatol 1999;38:721-9.
  18. Muzaffar F, Hussain I, Haroon TS. Physiologic skin changes during pregnancy: a study of 140 cases. Int J Dermatol 1992;31:375-8.
  19. Muallem MM, Rubeiz NG. Physiological and biological skin changes in pregnancy. Clin Dermatol 2006;24:80-3.
  20. Burton JL, Shuster S, Cartlidge M, Libman LJ, Martell U. Lactation, sebum excretion and melanocyte-stimulating hormone. Nature 1973;243:349–50.
  21. Rathore SP, Gupta S, Gupta V. Pattern and prevalence of physiological cutaneous changes in pregnancy: A study of 2000 antenatal women. Indian J Dermatol Venereol Leprol 2011;77:402.
  22. Dawber RP, Connor BL. Pregnancy, hair loss, and the pill. Br Med J 1971;4:234.
  23. Headington JT. Telogen effluvium. New concepts and review. Arch Dermatol 1993;129:356-63.
  24. Yip L, McCluskey J, Sinclair R. Immunological aspects of pregnancy. Clin Dermatol 2006; 24: 84-7.
  25. Winton GB. Skin diseases aggravated by pregnancy. J Am Acad Dermatol 1989;20:1-13.
  26. Torgerson RR, Marnarch ML, Bruce AJ, Rogers RS 3rd. Oral and vulvar changes in pregnancy. Clin Dermatol 2006;24:122-32.
  27. Sharma R. Skin and pregnancy. In: Valia RG, Valia AR, editors. IADVL textbook of dermatology. 3rd ed. Mumbai(Ind):Bhalani publishing house;2008. p. 1432-8.
  28. Lockwood DNJ, Sinha HH. Pregnancy and leprosy: a comprehensive literature review. Int J Lepr 1999;67:6-12.
  29. Khanna N. Leprosy and pregnancy. In: Kar HK, Kumar B, editors. IAL textbook of leprosy. New Delhi (Ind):Jaypee publishers;2010. p. 313-24.
  30. Steen VD. Pregnancy in scleroderma. Rheum Dis Clin North Am 2007; 33:345-58.
  31. Williams L, Chang PY, Park E, Gorson KC, Bayer-Zwirello L. Successful treatment of dermatomyositis during pregnancy with intravenous immunoglobulin monotherapy. Obstet Gynecol 2007;109:561-3.
  32. Jones SAV, Black MM. Pregnancy dermatoses. J Am Acad Dermatol 1999;40:233-41.
  33. Loret de Mola JR, Muise KL, Duchon MA. Porphyria cutanea tarda and pregnancy. Obstet Gynecol Surv 1996;51:493-7.
  34. Perez-Maldonado A, Kurban AK. Metabolic diseases and pregnancy. Clin Dermatol 2006;24:88-90.
  35. Ramos-E-Silva M, Libia Cardozo Pereira A, Bastos Oliveira G, Coelho da Silva Carneiro S. Connective tissue diseases: pseudoxanthoma elasticum, anetoderma, and Ehlers–Danlos syndrome in pregnancy. Clin Dermatol 2006;24:91-6.
  36. Dunna SF, Finlay AY. Psoriasis: Improvement during and worsening after pregnancy. Br J Dermatol 1989;120:584.
  37. Boyd AS, Morris LF, Phillips CM, Menter MA. Psoriasis and pregnancy: hormone and immune system interaction. Int J Dermatol 1996;35:169-72.
  38. Dacus JV. Pruritus in pregnancy. Clin Obstet Gynecol 1990;33:738-45.
  39. MacKie RM, Bufalino R, Sutherland C. The effect of pregnancy on melanoma prognosis. Br J Dermatol 1990;123(Suppl. 37):40.
  40. Koren G, Pastuszak A, Ito S. Drugs in pregnancy. N Eng J Med 1998;338:1128-37.
  41. Snyder JL, Krishnaswamy G. Autoimmune progesterone dermatitis and its manifestation as anaphylaxis: a case report and literature review. Ann Allergy Asthma Immunol 2003;90:469-77.
  42. Sachdeva S. The dermatoses of pregnancy. Indian J Dermatol 2008;53:103-5.
  43. Holmes RC, Black MM. The specific dermatoses of pregnancy: a reappraisal with special emphasis on a proposed simplified clinical classification. Clin Exp Dermatol 1982;7:65-73.
  44. Holmes RC, Black MM. The specific dermatoses of pregnancy. J Am Acad Dermatol 1983;8:405-12.
  45. Shornick JK. Dermatoses of pregnancy. Semin Cutan Med Surg 1998;17:172-81.
  46. Ambros-Rudolph CM, Mullegger RR, Vaughan-Jones SA, Kerl H, Black MM. The specific dermatoses of pregnancy revisited and reclassified: Results of a retrospective two-center study on 505 pregnant patients. J Am Acad Dermatol 2006;54:395-404.
  47. Paunescu MM, Feier V, Paunescu M, Dorneanu F, Sisak A, Ambros-Rudolph CM. Dermatoses of pregnancy. Acta Dermatovenereol Alp Pannonica Adriat 2008;17:4-11.
  48. Ambros-Rudolph CM. Dermatoses of pregnancy-clues to diagnosis, fetal risk and therapy. Ann Dermatol 2011;23:265-75.
  49. Vaughan-Jones SA, Black MM. Pregnancy dermatoses. J Am Acad Dermatol 1999;40:233-41.
  50. Kroumpouzos G, Cohen LM. Specific dermatoses of pregnancy: An evidence-based systematic review. Am J Obstet Gynecol 2003;188;1083-92.
  51. Roger D, Vaillant L, Fignon A, Pierre F, Bacq Y, Brechot JF, et al. Specific pruritic dermatoses of pregnancy: a prospective study of 3192 women. Arch Dermatol 1994;130:734-9.
  52. Vaughan-Jones SA, Hern S, Nelson-Piercy C, Seed PT, Black MM. A prospective study of 200 women with dermatoses of pregnancy correlating clinical findings with hormonal and immunopathological profiles. Br J Dermatol 1999;141:71-81.
  53. Kroumpouzos G, Cohen LM. Pruritic folliculitis of pregnancy. J Am Acad Dermatol 2000;43:132-4.
  54. Reed J, George S. Pruritic folliculitis of pregnancy treated with narrowband (TL-01) ultraviolet B phototherapy [Letter]. Br J Dermatol 1999;141:177-9.
  55. Weiss R, Hull P. Familial occurrence of pruritic urticarial papules and plaques of pregnancy. J Am Acad Dermatol 1992;26:715-7.
  56. Aronson IK, Bond S, Fiedler VC, Vomvouras S, Gruber D, Ruiz C. Pruritic urticarial papules and plaques of pregnancy: clinical and immunopathologic observation in 57 patients. J Am Acad Dermatol 1998;39:933-9.
  57. Cohen LM, Capeless EL, Krusinski PA, Maloney ME. Pruritic urticarial papules and plaques of pregnancy and its relationship to maternal-fetal weight gain and twin pregnancy. Arch Dermatol 1989;125:1534-6.
  58. Elling SV, McKenna P, Powell FC. Pruritic urticarial papules and plaques of pregnancy in twin and triplet pregnancies. J Eur Acad Dermatol Venereol 2000;14:378-81.
  59. Alves GF, Nogueira LSC, Varella TCN. Dermatology and pregnancy. An Bras Dermatol 2005;80:179-86.
  60. Pawels C, Bucaille-Fleury L, Recanati G. Pruritic urticarial papules and plaques of pregnancy: relationship to maternal weight gain and twin or triplet pregnancies. Arch Dermatol 1994;130:801-2.
  61. Im S, Lee E-S, Kim W, Song J, Kim J, Lee M, et al. Expression of progesterone receptor in human keratinocytes. J Korean Med Sci 2000;15:647-54.
  62. Aractingi S, Bertheau P, Le Goue C, Dausset J, Uzan S, Carosella ED. Fetal DNA in skins of polymorphic eruptions of pregnancy. Lancet 1998;351:559-62.
  63. Ambros Rudolph CM, Shornick JK. Pregnancy Dermatoses. In: Bolognia JL, Jorizzo JL, Schaffer JV, editors. Dermatology. 3rd ed. China: Elsevier;2007. p. 439-448.
  64. Engineer L, Bhol K, Ahmed AR. Pemphigoid gestationis: a review. Am J Obstet Gynecol 2000;183:483-91.
  65. Jenkins RE, Hern S, Black MM. Clinical features and management of 87 patients with pemphigoid gestationis. Clin Exp Dermatol 1999;24:255-9.
  66. Giam YC. Dermatoses of pregnancy. J Pediatr Obstet Gynecol 2003;29:22-7.
  67. Kalaaji AN, Nicolas MEO. Mayo clinic atlas of immunofluorescence in dermatology: patterns and target antigens. Rochester: Mayo clinic scientific press;2006. p.4.
  68. Chi CC, Wang SH, Charles-Holmes R, Ambros-Rudolph C, Powell J, Jenkins R, et al. Pemphigoid gestationis: early onset and blister formation are associated with adverse pregnancy outcomes. Br J Dermatol 2009;160:1222–8.
  69. Shornick JK, Jenkins RE, Briggs DC, Welsh KI, Kelly SE, Garvey MP, et al. Anti-HLA antibodies in pemphigoid gestationis (herpes gestationis). Br J Dermatol 1993;129:257-9.
  70. Shivakumar V, Madhavamurthy P. Skin in pregnancy. Indian J Dermatol Venereol Leprol 1999;65:23-5.
  71. Lammert F, Marschall H-U, Glantz A, Matern S. Intrahepatic cholestasis of pregnancy: molecular pathogenesis, diagnosis and management. J Hepatol 2000;33:1012-21.
  72. Kroumpouzos G. Intrahepatic cholestasis of pregnancy: what’s new. J Eur Acad Dermatol Venereol 2002;16:316-8.
  73. Yakasai IA, Thomson AJ, Fitzsimons C. Specific dermatoses of pregnancy: a review. West Afr J Med 2011;30:239-44.
  74. Geenes V, Williamson C. Intrahepatic cholestasis of pregnancy. World J Gastroenterol 2009;15:2049-66.
  75. Glantz A, Marschall HU, Mattsson LA. Intrahepatic cholestasis of pregnancy: relationships between bile acid levels and fetal complication rates. Hepatology 2004;40:467-74.
  76. Ambros-Randolph CM, Glatz M, Trauner M, Kerl H, Müllegger RR. The importance of serum bile acid level analysis and treatment with ursodeoxycholic acid in intrahepatic cholestasis of pregnancy: a case series from central Europe. Arch Dermatol 2007;143:757-62.

A rarity that can lead to a casualty - A retrospective study of 12 cases of Dermatomyositis

Authors
Matilda Naesström, Monika Kakol, Victoria Kamkar, Wioletta Baranska-Rybak, Malgorzata Sokolowska-Wojdylo, Marta Stawczyk and Roman Nowicki
Article Citation and PDF Link
BJMP 2015;8(3):a822
Abstract / Summary
Abstract: 

Aims: Lesions of the skin are omnipresent in Internal Medicine practice. The varying etiopathology when facing multiple system involvement may pose a challenge when it comes to diagnostics and management, especially when faced with less common skin diseases. Dermatomyositis is a rare skin disorder that manifests on the skin and in muscle; it also comes with a higher risk of comorbid cancers. Therefor we present the cases of dermatomyositis diagnosed at our departmet during the last 17 years, with the specific attention to ocurrance of oncological processes.
Method: A retrospective study was performed on 12 cases hospitalized between 1996 to 2011 due to dermatomyositis. The analysis was based on the course of the disease, clinical picture, treatment and frequency of neoplasms.
Results: Within those 12 patients (in addition to dermatomyositis) five patients had concomitant oncological process. The tumors of these five patients were located in discrete anatomical locations. The oncological process occured before, during, or after the appearance of dermatomyositis.
Conclusions: The combination of hallmark signs and symptoms seen in dermatomyositis are specific for the disease. Physicians need to be better informed about this rare, yet important disease, because it can be considered a paraneoplastic process.

Abbreviations: 
DM - Dermatomyositis, EMG - Electromyography, ANA - Antinuclear antibodies, CK - Creatine kinase, LDH - Lactate dehydrogenase, AST- aspartate transaminase, ALT - Alanine transaminase, CT - Computer tomography
Keywords: 
skin diseases; neoplasms; dermatomyositis

Introduction

Dermatomyositis (DM) is a rare autoimmune process with not yet fully understood aetiology. It is characterised by a combination of striated muscle inflammation and cutaneous changes. The pathogenesis of the cutaneous manifestations of DM is not well understood either. DM occurs in all age groups. Therefore, two clinical subgroups of DM are described: adult and juvenile. The adult form is predominant among female patients with a clinical presentation which includes a Heliotrope rash (Fig. 1), Gottron’s papules (Fig. 2), nail fold telangiectasia and other various cutaneous manifestations in association with inflammatory myopathy.1 In addition to the previous mentioned symptoms, juvenile patients also commonly suffer from ulcerative skin and recurrent abdominal pain due to vasculitis. An increased occurrence of oncological processes in combination with adult DM has been observed with a slight predominance for the female gender.2 These patients carry a higher risk for comorbid cancers. The most common ones include malignant processes of the ovary, lung, pancreas, stomach, urinary bladder and haematopoietic system.3 The significance of these observations is that the development of DM should raise suspicion with regard to a possible parallel oncological process.

Figure 1

Figure 2

Materials and Methods

A retrospective consecutive case series was performed on a group of 12 patients that were hospitalised at the Department of Dermatology, Venereology and Allergology at the Medical University of Gdansk between 1996 and 2013. The diagnostic criteria for DM included: hallmark cutaneous lesions of DM, clinically significant muscle weakness evaluated by electromyography (EMG), indicative laboratory findings - muscle enzymes, muscle biopsy, autoantibodies. All 12 cases had muscle biopsy, serum studies and EMG performed. The retrospective study analysed the age and sex of the patients, course of the disease, accompanying diseases, clinical picture and treatment. The patients with malignancies were analysed by the primary organs of origin, and the period between the diagnosis of DM and that of malignancy (Table 1).

Table 1. Patient characteristics

No. Sex Previous medical history Age of onset of DM Clinical picture Diagnostics Treatment Malignancy and age at diagnosis
1 F Chronic eosinophilic leukaemia 54 Muscle weakness of shoulder and hip area, facial oedema and erythema, palmar erythema CK 2550, ANA Hep-2 1:640, LDH 901, AST 69, ALT 143, X-ray = N, USG = N, EMG = N Azathioprine, Prednisone Stage IIA ovarian cancer at 55
2 F Peptic ulcer disease 66 Facial erythema, Gottron’s papules on the hands, muscular weakness creating difficulty in movement, weight loss, decreased appetite ANA Hep-2 1:1280, CT = N, EMG = N Glucocortico- steroids Small cell carcinoma at 66
3 F None 23 Muscular weakness of shoulder and hip area; difficulty in standing up and walking up stairs, Gottron’s papules, Heliotrope rash, upper chest erythema ANA Hep-2 1: 2580, CPK 12022; AST 595, ALT 210, CK-MB 534; Jo 1 = N, Mi = N Azathioprine, Prednisone Methotrexate None
4 F Chronic obstructive pulmonary disease 42 Muscular weakness of shoulder and hip area, facial oedema and erythema   Cyclo- phosphamide, Methyl- prednisolone Stomach tumour at 43
5 F None 22 Muscle weakness, painful extremities, facial oedema and erythema ANA Hep-2 = N, CT = N Cyclo- phosphamide, Prednisone None
6 F None 42 Muscle weakness, paraesthesia of hands, facial oedema and erythema ANA Hep-2 1:640 Cyclo- phosphamide, Prednisone None
7 F Hypertension, diabetes type II, osteopenia, leiomyoma. 65 Muscle weakness of shoulder and hip area, facial oedema and erythema ANA Hep-2 1:1280, LDH 650 Cyclo- phosphamide, Prednisone None
8 F Hyper-thyroiditis 46 Muscle weakness; difficulty in moving, facial oedema and erythema ANA Hep-2 1:160 Cyclosporine A, Prednisone None
9 F Autoimmune hepatic disease, leiomyoma. 45 Muscular weakness of shoulder and hip area, facial oedema and erythema ANA Hep-2 1:2560, CK 3700, Mi-2 = P Azathioprine, Methyl-prednisolone None
10 F Hypertension, diabetes type 2, hypo-thyroidism, ovarian cysts 57 Muscle weakness of shoulder and hip area, facial oedema and erythema, upper chest erythema, Gottron’s papules, Gottron’s papules, fatigue, dysphagia ANA Hep-2 1: 640, CK 747, LDH 363, AST 78, Ro52 = P, Mi 2 = N, Jo 1 = N, PM/Scl = N, CT= two pulmonary lesions that were biopsied and diagnosed as pneumoconiosis Prednisone, Methotrexate Cervical Carcinoma at 51, Breast Cancer at 57, Pulmonary Metastasis at 58
11 F hypertension 80 Muscle weakness, Heliotrope rash

.

ANA Hep-2 = P; Mi = N, CK 171.5, AST 45.22 Azathioprine, Prednisone None
12 F hypertension, diabetes Type 2, hypo-thyroidism 42 Muscle weakness, Heliotrope rash, upper chest erythema ANA Hep-2 1:320, Jo 1(-), M(-), CT=N Cyclosporin A, Methyl- prednisolone, Methotrexate None

No. = number (patient), DM = dermatomyositis, F = female, M = male, CK = creatine phosphokinase, ANA = antinuclear antibodies, LDH = lactate dehydrogenase, AST = aspartate transaminase, ALT = alanine transaminase, N = negative, P = positive, USG = ultrasonography, EMG = electromyography, CT = computerised tomography

Limitations

The small sample size is a significant limitation in this retrospective analysis. DM is a rare disease with a prevalence of 1:1000. Increasing sample size, by combining cases from multiple institutions, and implementing control would further strengthen the presented material.

Results

The average age of onset of the disease was 48 years. All 12 subjects were female. Previous medical history included chronic eosinophilic leukaemia, diabetes mellitus type II, hypertension, leiomyomas, hypo- and hyper- thyroid disease, chronic obstructive pulmonary disease, peptic ulcer disease, autoimmune hepatitis and osteopenia. The two most common are diabetes mellitus type II and hypertension. The clinical picture of each case was similar in that all of the patients presented with some form of muscle weakness. In addition, typical features of DM with Gottron’s papules, periorbital oedema, facial oedema and erythema were noted in five patients. Antinuclear Antibodies (ANA) Hep-2 of values >1:160 were identified in nine patients. Additional laboratory markers such as creatine kinase (CK), lactate dehydrogenase (LDH), aspartate transaminase (AST) and alanine transaminase (ALT) were elevated in five patients. Two patients had muscle biopsies performed. The immunohistopathology picture consisted of Immunglobulin G (IgG), fibrinogen, C1q, and C3 deposition around the perimysium and granular deposits of Immunoglobulin M (IgM) in the dermal epidermal junction. Of the 12 patients, four had neoplasms in addition to the diagnosed DM. The primary cancers were originating from the cervix, breast, stomach and ovary. Of these four patients, all had the diagnosis of DM prior to the diagnosis of a malignancy.

Discussion

The diagnosis of DM is made by combining the clinical picture with the results of various laboratory findings: skin and muscle biopsies, EMG, serum enzymes and ANAs.

The clinical picture varies. The typical dermatological presentation consists of a erythematous and oedematous periorbital rash - the Heliotrope rash (Fig. 1). Symmetrical redness and flaking can be observed on the elbows and dorsal sides of the phalanges, especially over the distal metacarpal joints - Gottron’s papules (Fig. 2). Erythematous lesions can also be found on other locations such as the face, upper chest and knees.4 The dermatitis heals with atrophy, leaving behind areas that resemble radiation-damaged skin. The striated muscle inflammation most often involves the shoulder and hip area, leading to muscle weakness and atrophy. The intercostal muscles and the diaphragm may be involved causing alarm with regards to respiratory compromise. Dysphagia can be present due to inflammation of the smooth and skeletal muscles of the oesophagus. These inflammatory processes often lead to muscle calcification.5 The sum of all these changes clinically is seen most often as weakness, weight loss and subfebrile temperatures. All patients in our study had co-existing muscle and cutaneous symptoms, with variation in severity and localisation. Five patients had the classical picture of shoulder and hip area weakness. The rest of the patients had a more general muscle weakness. Two patients had atypical complaints of hand paraesthesia and extremity pain respectively.

Subtypes of DM exist for the purpose of epidemiological research and sometimes prognosis. They are categorised by the clinical presentation and presence or absence of specific laboratory findings. These subtypes are as follows: Classic DM, Amyopathic DM, Hypo-amyopathic DM and Clinically Amyopathic DM. These subtypes have little impact on routine diagnosis. Common laboratory findings in DM are enzymatic elevation of CK, AST, ALT and LDH; these mainly reflect the muscle involvement. Amyopathic DM lacks both abnormal muscle enzymes and weakness.6 Enzymatic elevation may sometimes precede the clinical symptoms of muscle involvement. Hence, an enzymatic raise in a patient with a history of DM, should raise suspicion of recurrence. Positive ANA findings are frequent in DM but not necessary for diagnosis. More myositis-specific antibodies include anti-Mi 2 and anti-Jo 1. A typical histopathological examination shows: myofiber necrosis, perifascicular atrophy, patchy endomysial infiltrate of lymphocytes and occasionally the capillaries may contain membrane attack complexes.7

Cutaneous changes and muscular complaints can correspond to: 1. Systemic scleroderma which often has a positive ANA; 2. Trichinosis, in which periorbital swelling and myositis occurs, but there is a prominent eosinophilia and a history of consuming undercooked swine or bear meat; 3. Psoriasis with joint involvement which may give a clinically similar picture to DM. However, the skin changes in psoriasis have a more flaking pattern. In doubtful cases, a skin and muscle biopsy together with an electromyography will set the diagnoses apart. A facial rash may also be observed in systemic lupus erythematosus together with nail fold telangiectasia. They are usually distinguished by a clinical picture with more organ system involvement in systemic lupus and by serological studies. A drug-induced picture of DM exists and is particularly associated with statins and hydroxyurea.8

It is estimated that around 25% of DM cases are associated with a neoplastic process that can occur prior, during or after the episode of DM. The risk of developing a malignancy is highest in the first year of DM and remains elevated for years after diagnosis. 9, 10, 11 This was the case with patient number 1, 2 and 4 in our study, where the malignant process appeared in the first year following onset of DM. Risk factors seen in DM patients include male gender, advanced age and symptoms of dysphagia.12 The age range of the four patients in our study with malignancy was between 43 and 66. Symptoms that clinically raised suspicion of a malignant process included weight loss, lack of appetite and dysphagia. All neoplasms were discovered within one year after the diagnosis of DM was made. One patient had a previous history of cervical cancer, six years prior to the onset of DM.

The most common neoplasms seen in patients with DM vary in the world. In Europe the malignancies are located mainly in the ovaries, lungs, and stomach. The cancer types associated with the DM correlate with common cancers seen in the same area. For instance, in Asia, nasopharyngeal carcinoma (which is a rare malignancy in Europe) is a frequent occurrence in DM.1, 3 The location of neoplasms seen in our study varied from gastric, breast, ovary and pulmonary. The screening in regards to malignancies in patients with DM is individualised and should be based on risk factors such as previous malignancies, alarming symptoms such as weight loss or dysphagia, or abnormal findings on physical exam. This was the case with patient number 10 in our study who had a previous history of cancer, and patient number 2 who had symptoms of weight loss and decreased appetite. Initial screening was negative for patient number 1 and 2, where the malignancy developed first after the onset of DM. Age-appropriate screening with mammography, faecal-occult blood test and Papanicolaou smear should be considered. Additional investigations with chest films, computerised tomography (CT) scanning of chest, abdomen or pelvis; colonoscopy, cancer antigens; and gynaecological ultrasonography should be done when indicated.

The main objective of treatment in DM is to improve muscle strength and obtain remission, or at least clinical stabilisation. No specific protocol exists with regard to treatment of DM. Treatment is individualised and adapted to the specific condition of the patient. High-dose corticosteroids are the basis of treatment. However, randomised placebo clinical trials failed to show their efficacy. Clinical efficacy of corticosteroid therapy demonstrates itself and hence is the initial treatment of choice. Doses start at around 1 mg/kg/day depending on the corticosteroid of preference. This dosing is maintained for approximately two months until clinical regression is achieved, followed by approximately 10 mg decrease in dose for the coming three months. A maintenance dose of approximately 5-10 mg should be achieved. The exact parameters are patient-specific. In the case of a severe flare of dermatomyositis, 1 g per day for three days of methylprednisolone intravenous pulses can be administered. The systemic effects of long term therapy with corticosteroids have to be kept in mind. Hence, yearly dual-energy X-ray absorptiometry bone scans can be administered to monitor the development of osteopenia.

Further treatment options are offered in situations where the initial disease presentation is severe, involves internal organs, if relapse occurs during steroid dose reduction, and steroid side-effects. It has been proposed that combination therapy is a better method of approach due to lower reported relapse rates and lower need to use high-dose corticosteroids. Methotrexate is second-line therapy when steroids fail alone. Methotrexate is used with a maximum dose of 25 mg per week plus folate supplementation. The limitations of Methotrexate are immunosuppression and pulmonary fibrosis. Methotrexate is considered preferable to Azathioprine because the latter has a longer onset of efficacy. Azathioprine is administered at doses ranging from 1.5 - 3 mg/kg/day and has a side-effect profile is similar to that of other immunosuppressants. Cyclosporin A is a T-cell cytokine moderator that has a similar efficacy profile to Methotrexate. Side-effects include renal impairment, gingival hyperplasia, and hypertrichosis. Dosing of Cyclosporin A ranges from 2 - 3 mg/kg/day.

An expensive but effective and rather low side-effect alternative is intravenous immunoglobulins. The dosage of this medication has not been officially established in the treatment of DM, but options are: 2 g/kg given either in 1 g/kg/day for two days every four weeks; or 0.4 mg/kg/day for five days initially, and then for three days monthly for three to six months. Other alternatives include Mycophenolate Mofetil, Cyclophosphamide, Chlorambucil, Fludarabine, Eculizumab, Rituximab.9 Further options might be treatment targeted toward malignancy when associated with DM. This was observed in our patient number 10, where full remission of DM was obtained first after lobectomy and chemotherapy for the mammary carcinoma.

Conclusion

DM mainly affects women and all 12 cases presented in our study were female. One third of our cases had malignancies associated with their course of DM. We conclude that it is reasonable to screen these patients, especially in those with already established cancer risk factor. Age-appropriate screening and beyond is indicated by high risk factors or clinical presentation. High suspicion should be raised in patients with a previous history of oncological treatment since DM can be the first clinical sign of cancer recurrence.

Acknowledgements / Conflicts / Author Details
Competing Interests: 
None declared
Details of Authors: 
MATILDA NAESSTRÖM, M.D., Department of Clinical Dermatology, Venerolgy and Allergology, Medical University of Gdansk, Poland. MONIKA KAKOL, M.D. , Department of Clinical Dermatology, Venerolgy and Allergology, Medical University of Gdansk, Poland. VICTORIA KAMKAR, M.D. , Department of Clinical Dermatology, Venerolgy and Allergology, Medical University of Gdansk, Poland. WIOLETTA BARANSKA-RYBAK, M.D PhD, Department of Clinical Dermatology, Venerolgy and Allergology, Medical University of Gdansk, Poland. MALGORZATA SOKOLOWSKA-WOJDYLO, M.D. , Department of Clinical Dermatology, Venerolgy and Allergology, Medical University of Gdansk, Poland. MARTA STAWCZYK, M.D. , Department of Clinical Dermatology, Venerolgy and Allergology, Medical University of Gdansk, Poland. Prof ROMAN NOWICKI, M.D. , Department of Clinical Dermatology, Venerolgy and Allergology, Medical University of Gdansk, Poland.
Corresponding Author Details: 
MATILDA NAESSTRÖM, Department of clinical Dermatology, Venerology and Allergology Medical University of Gdansk, M. Skłodowskiej-Curie 80-210, Gdansk, Poland.
Corresponding Author Email: 
matilda.naesstrom@hotmail.com
References
References: 
  1. Liu WC, Ho M, Koh WP et al.. An 11-year review of dermatomyositis in Asian patients. Ann Acad Med Singapore 2010;39:843-847.
  2. Ohashi M, Shu E, Tokozumi M Fujioka K et al. Anti-p155/140 antibody-positive dermatomyositis with metastases origins from an unknown site. Acta Derm Venereol 2011;91:84-85.
  3. Chen YJ, Wu CY, Huang YL et al. Cancer risks of dermatomyositis and polymyositis: a nationwide cohort study in Taiwan. Arthritis Res Ther2010;12:R70.
  4. Callen JP. Dermatomyositis. Lancet. Jan 1 2000;355(9197):53-7
  5. Wananukul S, Pongprasit P, Wattanakrai P. Calcinosis cutis presenting years before other clinical manifestations of juvenile dermatomyositis: report of two cases. Australas J Dermatol. 1997 Nov;38(4):202-5.
  6. Sontheimer RD. Would a new name hasten the acceptance of amyopathic dermatomyositis (dermatomyositis siné myositis) as a distinctive subset within the idiopathic inflammatory dermato myopathies spectrum of clinical illness?. J Am Acad Dermatol. Apr 2002;46(4):626-36
  7. Smith ES, Hallman JR, DeLuca AM et al. Dermatomyositis: a clinicopathological study of 40 patients. Am J Dermatopathol. Feb 2009;31(1):61-7.
  8. Seidler AM, Gottlieb AB. Dermatomyositis induced by drug therapy: a review of case reports. J Am Acad Dermatol. 2008 Nov;59(5):872-80.
  9. Callen JP, Hyla JF, Bole GG Jr et al. The relationship of dermatomyositis and polymyositis to internal malignancy. Arch Dermatol. Mar 1980;116(3):295-8.
  10. Buchbinder R, Forbes A, Hall S et al.. Incidence of malignant disease in biopsy-proven inflammatory myopathy. A population-based cohort study. Ann Intern Med. Jun 19 2001;134(12):1087-95.
  11. Chow WH, Gridley G, Mellemkjaer L et al.Cancer risk following polymyositis and dermatomyositis: a nationwide cohort study in Denmark. Cancer Causes Control. Jan 1995;6(1):9-13.
  12. Chen D, Yuan S, Wu X et al. Incidence and predictive factors for malignancies with dermatomyositis: a cohort from southern China. Clin Exp Rheumatol. Jul 28 2014
  13. Cordeiro AC, Isenberg DA. Treatment of inflammatory myopathies. Postgrad Med J 2006;82:417-424

Topical medicament allergy: the importance of patch testing

Authors
Amelia Cussans, Natalia Spierings, Amanda Woods and Lucy Ostlere
Article Citation and PDF Link
BJMP 2015;8(2):a819
Abstract / Summary
Abstract: 

A 41-year-old woman with a 6-year history of mild psoriasis presented with a rash under her breasts. She was prescribed Trimovate cream (GlaxoSmith Kline) and had a florid weeping eczema within 48 hours of application. This settled with the withdrawal of Trimovate. Contact dermatitis is type IV allergy and usually appears within 2-3 days after contact with an external allergen. Detection of the allergen, or allergens, is important, as avoidance results in resolution of the eczema. Our patient was patch tested and showed positives to three components of Trimovate; cetearyl alcohol, sodium metabisulphite, and clobetasone butyrate. These are important allergens to identify, because they are also present in other products. Clobetasone butyrate is often used in facial and flexural psoriasis. Cetearyl alcohol is particularly significant, as it is found in many products including commonly used moisturizers such as Diprobase (MSD), Cetraben (Genus) and Epaderm (Mölnlycke) cream, and most steroid creams. Our patient highlights the fact that is insufficient to simply advise a patient to avoid the topical medicament that has caused a reaction. Patch testing is necessary to identify which components the patient is allergic to, so that they can be avoided in all products. This is of particular significance for our patient given her history of psoriasis, as she will likely require moisturizers and topical steroid preparations in the future. Since she began avoiding these allergens, she has had no recurrence of eczema. To conclude, GPs should consider sending their patients with contact dermatitis for patch testing, as the identification of all allergens is valuable to management.

Keywords: 
patch testing, contact dermatitis, concomitant sensitivity, Trimovate cream, sodium metabisulphite, clobetasone butyrate, cetearyl alcohol.

Case Report

A 41-year-old woman with a 6-year history of mild psoriasis presented with a rash under her breasts. The differential diagnosis included flexural psoriasis, an allergy to the nickel in her under wired bra, and intertriginous dermatitis (moisture-associated skin damage). She was prescribed Trimovate cream (GlaxoSmith Kline) and developed a florid weeping eczema within 48 hours of application (Figure 1). The eczema settled with the withdrawal of Trimovate and application of Betnovate RD cream (GlaxoSmith Kline). The history was very suggestive of a contact dermatitis to Trimovate cream.


Figure 1 showing eczema

She was referred to the Dermatology department and was patch tested to the European standard, medicament and steroid batteries. She had a number of positives including cetearyl alcohol, sodium metabisulphite, and clobetasone butyrate. These are all components of Trimovate.  She was given advice sheets on all her allergens and on avoiding them she has had no recurrence of eczema.

Discussion

Contact dermatitis is a type IV allergy and usually appears within 2 to 3 days after contact with an external allergen. This case is likely to be an example of concomitant sensitisation, where one sensitivity facilitates the acquisition of another sensitivity to a chemically unrelated ingredient within a product.Whilst there has been a previous case report of concomitant sensitivity to sodium metabisulphite and clobetasone butyrate in a patient using Trimovate cream,1 this is the first report of a patient reacting to three of the ingredients found in Trimovate - sodium metabisulphite, clobetasone butyrate, and cetearyl alcohol. Allergy to clobetasone butyrate is rare, with only 5 previously reported cases.1, 2, 3 Allergy to sodium metabisulphite is not uncommon, producing a positive reaction in approximately 4% of patients who are patch tested.4, 5 Allergy to cetearyl alcohol is also rare, with one study estimating the incidence of positive reactions to be 0.8% among 3062 patients that were patch tested.6

Detection of the allergen, or allergens, is important, as avoidance results in resolution of the eczema. Our patient highlights the fact that it is insufficient to simply advise a patient to avoid the topical medicament that has caused a reaction. Ideally, patients with a topical medicament allergy should be patch tested to identify which components the patient is allergic to, so that they can be avoided in all products. In this case, in addition to Trimovate, there are a number of other products that our patient will now avoid. This is of particular significance in view of her history of psoriasis, for which she has used moisturizers and topical steroid preparations in the past, and will likely need again in the future. Clobetasone butyrate is often used in facial and flexural psoriasis. Cetearyl alcohol is a particularly important allergen to identify, as it is found in many products including a number of commonly used moisturizers such as Diprobase (MSD), Cetraben (Genus) and Epaderm (Mölnlycke) cream, and most steroid creams although not steroid ointments. Our patient was therefore advised to use only steroid ointments and has had no recurrence of the contact dermatitis. To conclude, GPs should consider sending their patients with contact dermatitis for patch testing, as the identification of all allergens is valuable to management.  

Acknowledgements / Conflicts / Author Details
Competing Interests: 
None declared
Details of Authors: 
AMELIA CUSSANS, University of Southampton School of Medicine, Southampton, UK. NATALIA SPIERINGS, BSc MBBS MRCP(UK), St George's Hospital, London, UK. AMANDA WOODS, MBBS (Hons) DCH MRCP Derm, Chelsea and Westminster Hospital, London, UK. LUCY OSTLERE, BSc MB BS FRCP MD, St George's Hospital, London, UK.
Corresponding Author Details: 
AMELIA CUSSANS, 11A Routh Road, London, SW18 3SW, UK.
Corresponding Author Email: 
ac43g11@soton.ac.uk
References
References: 
  1. Harrison DA, Smith AG. Concomitant sensitivity to sodium metabisulfite and clobetasone butyrate in Trimovate cream. Contact Dermatitis 2002;46(5):310.
  2. Murata T, Tanaka M, Dekio I, Tanikawa A, Nishikawa T. Allergic contact dermatitis due to clobetasone butyrate. Contact dermatitis 2000;42(5):305-305.
  3. Boyle J, Peachey RD. Allergic contact dermatitis to Dermovate and Eumovate. Contact Dermatitis 1984;11(1):50-1.
  4. Madan V, Walker SL, Beck MH. Sodium metabisulfite allergy is common but is it relevant? Contact Dermatitis 2007;57(3):173-6.
  5. Garcia-Gavin J, Parente J, Goossens A. Allergic contact dermatitis caused by sodium metabisulfite: a challenging allergen: a case series and literature review. Contact Dermatitis 2012;67(5):260-9.
  6. De Groot AC, Weyland JW, Nater JP. Unwanted effects of cosmetics and drugs in Dermatology. 3rd ed. Amsterdam: Elsevier; 1994.

Blue Macular Skin Lesions of Unknown Cause in a Tyre Factory Worker: A Case Report

Authors
Mohammed Al Abadie, Dilhara Karunaratne, Nabeel Salmons, Audrey Fong Juan Chin, Soha Ammar and Nisal Karunaratne
Article Citation and PDF Link
BJMP 2015;8(2):a818
Abstract / Summary
Abstract: 

This report describes the case of a 61 year old gentleman who developed blue macular skin lesions of unknown cause. Biopsy of the lesion showed pigment deposition in the dermis that had the appearance of tattoo pigment, but oddly the patient had never been tattooed in the past. Carbon black is a chemical which can be used to give blue tattoos their colour. The patient was exposed to carbon black in his job as a tyre maker and it may have accumulated in the dermis by an unknown route. An occupational exposure may be the cause of the skin lesions and this case may play a part in the identification of more cases and a confirmation of the true diagnosis.

Keywords: 
Macular lesion, dermis, carbon black, tattoo, tyre worker, occupational exposure

Case Report

Blue discolouration of the skin can have a multitude of causes, including Mongolian spots, blue naevi, the naevi of Ito and Ota and metallic discolouration1 or the use of drugs such as minocycline. Here we report the case of a 61 year old gentleman who developed blue macular skin lesions that were not attributable to any obvious cause and may be the result of an unidentified occupational exposure.

A 61 year old Caucasian gentleman developed blue macular skin lesions over a 14 year period. The very first lesion appeared in the middle phalanx of the right middle finger (figure 1). It was light blue and pinpoint, eventually darkening and increasing in size to approximately 1mm x 1mm, at which point becoming permanent and non-evolving. The lesion had no notable associated features and the patient was in otherwise good health.


Figure 1: The first blue macular lesion on the middle phalanx of the right middle finger.


Figure 2: A blue macular lesion on the terminal phalanx of the left middle finger.


Figure 3: A lesion on the anterior abdomen from which a punch biopsy was taken.


Figure 4: Haematoxylin and Eosin stained slide at 10x magnification. Abdominal skin biopsy showing dermal interstitial and perivascular distribution of black coloured pigment deposits

At present, he has approximately thirteen blue macular lesions in total, all of which have developed in the same manner. They are distributed predominantly on his hands with one on his left forearm and one on the right abdominal flank. New spots still continue to arise on his hands (figure 2).

A punch biopsy of the abdominal lesion (figure 3) was carried out. The histological findings were those of skin with normal intact epidermis and the presence of black coloured pigment granular deposits, located largely within the papillary dermis and occasional smaller deposits in the superficial reticular dermis (figure 4). The deep reticular dermis and subcutaneous fat were normal.  The pigment had a perivascular distribution and in dendritic histiocytic cells, with close association to fibroblasts. Histiocytic cells form part of the mononuclear phagocyte system and these cells are abducted mainly for phagocytosis removal or storing material2. Apart from the pigment, the remaining skin was normal. The use of light microscopy alone does not identify all substances on examination of a Haematoxylin and Eosin (H&E) stained section of tissue. Applying polarisation light microscopy enables the identification of numerous structures, for example crystals, pigments, bone and amyloid3. However, the black coloured material here was non polarisable (no refractile foreign material could be identified).  These appearances as seen on light microscopy alone are most frequently seen where there is a history of tattoo artistry, but tattoo pigment is typically identified as showing reflective properties using polarisation4. Interestingly, the patient had no clinical history of deliberate tattooing and other causes were considered.

Discussion

The discovery of black coloured deposits in the dermis excludes the diagnoses of Mongolian spots or blue naevi and the naevi of Ito and Ota, all of which are disorders of dermal melanocytes. Another important differential is malignant melanoma, but it is not the diagnosis as the histopathological findings did not find any evidence of dysplasia or malignancy.

In a disorder known as anthracosis, similar findings of black coloured deposits can be seen in other organs such as within the lung and draining lymph nodes. It is often found in smokers and urban populations and reflects the deposition of carbon which is the most commonly identified exogenous mineral substance within tissue sections. The skin is not a site where such carbon pigment is typically seen and therefore, this is not a credible diagnosis in this case.

Agyria is a condition that occurs as a result of silver particle impregnation of skin leading to blue-grey skin discolouration. Silver exposure may be due to occupational or surgical exposure (by use of silver sutures) or medication with silver salts. On interview, the patient denied any occupational exposure to silver and the use of silver salts. Although the patient had had previous shoulder surgery, silver sutures are no longer used in modern day surgical practice and therefore this cannot be the cause of his skin discolouration.

Unfortunately, histological examination of paraffin embedded tissue sections can only confirm the presence and distribution of an exogenous substance and it is not possible to precisely differentiate the exact type of material which is present. The use of an electron probe micro analyser may have been useful in identifying the substance, however, such equipment is not currently available and was not used in this case.

Interestingly, in tattoo artistry, carbon black may be used to give blue tattoos their colour5 and this is also a component of tyres and industrial rubber products6. This provided us with a link to occupational exposure, given that this gentleman is a tyre worker and has been involved in both the manufacture and assembly of tyres for 34 years. Carbon can cause discoloration of the skin, depending on the extent of deposition.

It is notable that in his 34 years of working with tyres, this gentleman did not routinely use gloves or protective uniform until only 10 years ago. This was as workplace safety precautions were not as strongly enforced in previous times. He admitted to have been in direct contact with the materials involved in tyre building and also suffered accidental superficial cuts on his hands whilst working, which may be a route by which carbon may have been introduced into the dermis. This is supported by the observation that the majority of the blue macular lesions were on the hands. Adding credibility to this theory is the identification of a colleague of this gentleman’s (who did not wish to be identified), whose job also involved the manufacture and assembly of tyres, who also has a similar single blue macular lesion on his hand.

In addition to this we have identified a forum on the internet7 that reports other similar cases of blue pin-point macular lesions appearing on the skin of tyre factory workers – some of whom worked for the same tyre company that this gentleman did. This may suggest that there is an association between exposure to a chemical, possibly carbon black, involved in the manufacture of tyres, and the appearance of these blue macular lesions.

In this case report, the identity of the material deposited and the route by which it accumulated in the dermis is unclear, but may have been related to an occupational exposure – this was in keeping with the general consensus upon presentation of this case at the West Midlands Dermatology Conference at New Cross Hospital Wolverhampton. We welcome any new case reports or literature that may be able to shed further light on this subject.

Acknowledgements / Conflicts / Author Details
Competing Interests: 
None declared
Details of Authors: 
MOHAMMED AL ABADIE, MBBS, DSBD, FAAD, PhD, FRCP, Consultant Dermatologist, The Royal Wolverhampton NHS Trust, New Cross Hospital, Wednesfield Road, Wolverhampton, WV10 0QP, United Kingdom. DILHARA KARUNARATNE, BSC FIRST CLASS HONOURS, Medical student at The University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom. NABEEL SALMONS, MBChB, FRCPATH, Consultant Histopathologist, The Royal Wolverhampton NHS Trust, New Cross Hospital, Wednesfield Road, Wolverhampton, WV10 0QP, United Kingdom. AUDREY FONG JUAN CHIN, Medical student at The University of Birmingham, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom. SOHA AMMAR, Medical student at The University of Birmingham, The University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom. NISAL KARUNARATNE, Year 13 student at Brentwood School, Middleton Hall Lane, Brentwood, Essex, CM15 8EE, United Kingdom.
Corresponding Author Details: 
DILHARA KARUNARATNE, BSC FIRST CLASS HONOURS, Medical student at The University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
Corresponding Author Email: 
diliekaru@yahoo.co.uk
References
References: 
  1. Park JY, Shin DH, Choi JS, Kim KH. Metallic Discoloration on the Right Shin Caused by Titanium Alloy Prostheses in a Patient with Right Total Knee Replacement. Annals of Dermatology. 2013;25(3):356-9.
  2. Stevens A, Anderson PG. Immune System. In: Stevens A, Anderson PG (eds.) Stevens and Lowe's Human Histology. 4th edition. United Kingdom. Elsevier Mosby; 2015. p. 128-129.
  3. Bancroft JD, Floyd AD. Light Microscopy. In: Bancroft JD, Gamble M (eds.) Theory and Practice of Histological Techniques. 6th edition. United Kingdom. Churchill Livingstone; 2008. p. 45-48.
  4. Churukian CJ. Pigments and Minerals. In: Bancroft JD, Gamble M (eds.) Theory and Practice of Histological Techniques. 6th edition. United Kingdom. Churchill Livingstone; 2008. p. 252-257.
  5. Lehner K, Santarelli F, Vasold R, Koenig B, Landthaler M, Baeumler W. Black tattoo inks are a source of problematic substances such as dibutyl phthalate. Contact Dermatitis. 2011;65(4):231-8.
  6. International Carbon Black Association. Carbon Black Uses. [Online] Available from: http://www.carbon-black.org/index.php/carbon-black-uses [Accessed 19th December 2014]
  7. Topix LLC. Blue dots under skin. [Online] Available from: http://www.topix.com/forum/com/gt/TOIIO5NUTJE2050KS [Accessed 19th December 2014].

Prevalence and implications of genital tattoos: A site not forgotten

Authors
Thomas Neluis, Myrna L. Armstrong, Cathy Young, Alden E. Roberts, LaMicha Hogan, and Katherine Rinard
Article Citation and PDF Link
BJMP 2014;7(4):a732
Abstract / Summary
Abstract: 

Purpose:  To provide information on men who have tattooed one anatomical site, the genital region (pubic and/or glans penis), that is uncommonly noted.
Methods: Two methods were used.  First, the limited cultural and medical literature was reviewed. Secondly, a subsample of 14 men were analyzed, taken from a primary study examining male genital piercings (N = 445), who responded affirmatively to one survey question about penile tattoos.
Findings:  The literature (n = 25) was limited.  Cultural literature revealed a long, rich history of genital markings for esthetics, sexual enhancement, and tribal status, whereas the medical literature reflected limited observational type information, some actual case histories, and few studies.  From the small subsample, qualitative and quantitative data were provided.  Similarities to those who wore general body tattoos were validated such as being single, heterosexual, having some college/vocational education, monthly binge drinking, no skin complications, and a strong propensity for a Need for Uniqueness.  Reportedly, they were major body art wearers and continue to enjoy them.  First age occurrence of sexual intercourse was similar to the national average of 17 years. Challenged assumptions included (a) no consensus regarding being risk takers, (b) significant reported forced sexual activity, and no (c) physical, sexual or mental abuse.
Conclusions:  From our experience, those with genital tattoos are seen primarily for a normal range of developmental and physiological urologic issues, not their decorative markings; these genital tattoos are an integral part of their cultural and personal expression and most likely will increase. Yet, the markings are only skin deep so clinicians should adopt a nonjudgmental approach and employ methods of proactive patient health education.

Keywords: 
Key Words: Penis, tattoos, males, genital tattoos, Need For Uniqueness

INTRODUCTION

Maintaining its longstanding presence as one of the oldest forms of art, body tattooing has increased exponentially within mainstream society, as well as in social acceptance. Generally worn to display individuality and creativity, these distinctive forms of indelible markings are present in every culture, whether on tribal men, or people of status. Procedurally when inserting the decorative markings, the approach in studio tattooing has not changed significantly as artists are still using “an electrically powered, vertically vibrating instrument to inject tattoo pigment 50 to 3,000 times per minute up to or into the dermis at a depth of 1/64th to 1/16th of an inch”1

While no national registry provides prevalence, a 2012 Harris Poll cited one in five United States (U.S.) adults have at least one tattoo (21%), an increase of 16% and 14% from previous surveys taken in 2003 and 2008 respectively.2 Tattoo numbers were even higher in some variables including age between 30-39 years (38%), Hispanics (30%), females (23%), and those living in the Western part of the U.S. (26%). No questions were identified in the 2012 poll that queried tattooed body site locations. Other studies cite almost a 25% presence of tattoos.1,3-7 The amount of tattoo studios also echoes the growing body art phenomenon.

Given the societal blaze of tattooing, the medical literature on body art has also increased. Yet, most of the information still remains focused on small case reports6 about traditional locations (arms, legs, chest, back), their decision-making, various risk-taking behaviors,8 and the small amount of complications.7 Those with various adverse skin reactions or major complications seem to have had tattoos with colored pigment.6

While body art can be found virtually everywhere on the human anatomy, several articles have surfaced concerning genital body piecing.4-5, 9-11 Current studies validate the increasing rate of all types of tattooed 4,8 people, from a variety of occupations and social classes, with markings on visible and non-visible locations.7 This article reports on the limited medical literature found about men with genital tattoos (pubic and/or the glans penis). Also presented is a subsample data analysis of 14 men from a primary study examining male genital piercings,11-12 who responded affirmatively to one survey question about penile tattoos. This synopsis and subsample data analysis are provided for clinicians to have further, recent evidence about men with genital tattoos for decision making during patient encounters in health care settings. The terminology of penile and genital tattoos will be used interchangeably in this article.

METHODS

Literature Synopsis

Historically, the cross-cultural literature is rich in visual genital tattoo descriptions. In South America, the Moche on the North Coast of Peru (A.D. 150-800) produced ceramics illustrating vivid sexual imagery and highly decorated male genitals.13 Phallus decorations with dots, concentric lines, and other tattoo markings on the penile skin and mucosa during the Upper Paleolithic era in Europe 12,700 to 11,000 years ago have been reported.14-15 Likewise, the Samoan Island culture, where the word ”tattoo” is believed to have originated from “tatau,”, has maintained ritualistic16 traditions for over two thousand years; they are initiated at the time of puberty for future leadership roles. These 10+ days of ceremonies include very painful, repeated tattooing of the scrotum (tafumiti) and the penis (tafito). Other nearby primitive Polynesian tribes have believed this tattooing as highly erotic,16 whereas the indigenous Maori (New Zealand) trust that the pigment for these tattoos can trap cosmic energy.14 Circumcision and tattooing were thought to produce the same effect of magic protection and healing powers after scar healing.14 In the Japanese culture, an examination of Yakuza (racketeers or gangsters) also describes the genitalia as a site that is tattooed,17 fulfilling their principles of tattoos always being covered.

Searching for information about genital tattoos was more challenging within the medical literature. A comprehensive longitudinal 40 year search of the national and international electronic medical literature (1973-2013) published in English and their associated reference lists was conducted with MEDLINE, EMBASE, CINAHL, SCOPUS, and OVID. Only 20 articles were located that mentioned genital tattoos. Articles were from international authors (n = 11) and the U.S. (n = 9); they all produced interesting reading. One reference cited women with genital tattoos.7

Genital tattoos in the early literature were labeled as criminal, or personality disorders tattoos;18 one recent article discussed them under the header of genital self-mutilation.16 Others described them as a valuable clue for forensic pathology identification.19-20 World War II articles cited descriptive stories of soldiers with penile tattoos,21-22 with one reporting up to 10 sailors being seen.23 Besides reporting on how the fate of Bulgaria was determined by three tattooed men (Churchill with an anchor on his left arm, Roosevelt with a family coat of arms tattoo, and Stalin with a death’s head on his chest),24 Kazandjieva25 then provides vivid examples of auto-aggression markings that his countrymen self-inflicted after the Communist takeover. This included glans penis tattoos which are described as producing great pain.15,25 One political candidate, while campaigning, is reported as suggesting punitive action for those HIV+ by “putting indelible, glow-in-the-dark tattoos on [their] genitals.”26 Traumatic tattoos associated with gunpowder explosions and blast burns are also mentioned on the glans penis.27

Two studies also described inmates with genital tattoos and discussed how these markings demonstrated aggressive behavior within this type of environment. Here large, colorful tattoo designs and wording on the glans penis tattoos were described28-29 which seemed to satisfy the inmate’s flaunt of personal pain endurance. Additionally, Cuban refugees (Marielitos) fleeing to the U.S. were reported as having genital tattoos; they also were from prison subcultures and their markings had various sexual overtones.29

Four other reports described those with penile tattoos also routinely inserting foreign bodies12,30 and paraffinoma12,31,32 into the penis. In Pehlianov’s study (also in Bulgaria) they included a control group of another 25 men with genital tattoos. Recently, a unique case of non-ischemic priapism for 3 months was reported33 following prolonged bleeding from a manual penile tattoo procedure in Iran. The authors suggested the hand-held tattoo needle had penetrated too deeply producing an arteriovenous fistula and the subsequent persistent half-rigid priapism. The authors also noted that the 21 year old patient expressed no regret, depression, or other complications related to the genital tattoo.

Original Study

The initial study queried males with genital piercings using available internet survey software,12 as it was considered a hidden variable. Anonymity and access to people nationally and internationally were major advantages for using this nontraditional approach. The university institutional review board deemed the study status as Exempt. To obtain quantitative and qualitative data about those men with genital piercings, an 83 item web-based survey was used; overall results, and another subsample of this data, are published elsewhere.11-12

Subsample of those with Penile Tattoos

From the original 445 male genital pierced individuals that responded to the question regarding having tattoos on their penis, 14 replied affirmatively. This subsample had previously been determined not be an outlier of the larger group of genital pierced men.12 While a short general description (age span at the time of tattoo procurement, urethral “play,” design types, motives, and tattooists) about the 14 member genital tattoo subsample was published in 2010,12 further investigation leading to quantitative and qualitative (Figure 1) data is presented here.

Figure 1: Subsample Respondent Qualitative Quotes
*Black tribal flames on the top of the shaft, done at [age] 38
* For erotic reasons, self done with no complications, done at [age] 54
*I got it because I wanted it. After it was finished I realized I needed it, done at [age] 30
* I self tattoo’d my penis on the glans and around the corona ridge in order to make up for its’ lack of size and to enhance its appearance. I used a sailmaker’s needle and Indian ink and there were no complications, done at [age] 43.
*one small cross pigment tattoo!
*I’m a little more than average in size, but I still have issues with my genitals. The way they look and their size. Piercings and tattoos have helped me quite a lot.
*I sketched a rose one day, like[d] the design, decided to get it tattooed on my penis. The stem is green with some yellow highlights, the bud is red, all black outline. The tattoo was applied with a standard machine . . .healing was actually quicker and easier than any of my other tattoos.
*It’s a little heart just next to ‘captain hemingway’ which I hand poked and used india ink for it when I was 17. . . thought our penis deserved a reminder of our affection . . .no complications experienced but since it was hand done with a [sterile] needle it’s kind of blurry

This subsample had significantly more foreskin genital piercings (chi-square = 11.5) = 1; P = .001), whereas the most common genital piercing of the larger group of those without genital tattoos11 had Prince Albert piercings (inserted through external urethra). No question inquired which came first, the genital piercing or genital tattoos.

Data Analysis

For this subsample analysis, (and original study11-12), IBM SPSS 21was used to obtain frequencies and chi-square analysis. Cross tabulations for the subsample were obtained by comparing those with and without penile tattoos.

RESULTS

Demographics

Almost all of the subsample respondents with penile tattoos were reportedly Caucasian (92%) and their ages ranged from 18 to 67 years (average 42.3). Of those that replied, six lived in the U.S. and five cited various international locations. Over half had vocational or college education (64%) and significantly more were likely to be single (25%) or divorced (25%), (chi-square 12.6) = 5; P = .027). Data regarding religious faith was weak to non-existent (75%). Respondents self-reported a good state of health (92%) (chi-square = 8.7) = 3; P = .034), yet 50% cited no annual health check-ups.

Risk Behaviors

Within this subsample, there was no consensus about being a “risk taker”. Recreational drugs were reportedly not used (91%), over half were non-smokers (55%), but monthly alcohol use with binge drinking (5+ or more drinks) was cited (78%). Their “motives for genital tattoos were for esthetics, sexual, and personal pleasure”12; a variety of penile tattoo designs were described (Figure 1), created either “by studio artists (n = 11) or self-inflicted (n = 3)”.12 All of them described having other body art, such as piercings and other general body tattoos. Some reported an average of 4 piercings (81%) and a significant amount of general body tattoos (average 3.5) (chi-square = 11.1) = 5; P = .049), that still interest them (85%) (chi-square = 8.9) = 3) P = .031).

Sexual Activity

This subsample’s average age of first intercourse was 17 years, with most citing women as their sexual partners (92%), most preferred penile/vaginal intercourse (79%), and only one respondent reported a sexually transmitted infection (gonorrhea). When asked about any forced sexual activity (rape), this subsample had a significant amount of those who answered affirmatively (23%) (chi-square = 7.7) = 1; P = .005). Virtually no sexual, physical, or mental abuse was reported.

Need for Uniqueness

A four-item scale called the Self-Attributed Need for Uniqueness (SANU)34 was present in the survey to determine the respondent’s self-view (Cronbach alpha = .86). Using a Likert scale, the subsample’s moderate, strong and very strong perspectives were collectively summarized. These respondents with penile tattoos preferred to be different (79%), distinctive (86%), intended to do things to make themselves different than those around them (72%), and reported a Need For Uniqueness (93%) (Cronbach alpha = .77). To validate this finding, when all 5 responses of SANU were totaled,12 the mean was 12.43 documenting a more positive perspective for intentionally wanting to be different, distinctive, and unique.

DISCUSSION

This article reviewed both the cross cultural and medical literature about those with genital tattoos, as well as included both a quantitative and qualitative subsample data analysis of a small group of men who specifically reported penile tattoos. Yet, with certainty this small sample size produced limitations and reporting/survey bias. Additionally, any generalizability with the findings of this subsample should be noted as the respondents could have self-selected their participation and used their personal judgment to interpret the survey questions in this non-experimental cross-sectional study using internet survey methodology.12

From this review and to our knowledge, few have studied groups of men with genital tattoos, a difficult group of subjects to find with this hidden variable.12,31 Cultural descriptions documented a long, rich history 12, 14-17, 29,31 of genital markings for esthetics, sexual enhancement, and tribal status, whereas the medical literature reflected limited observational type information, and few actual case histories or scientific studies. Although there were no mental health evaluations12 cited in this medical literature, more psychopathic, deviant behavior discussions were made about the individuals with genital tattoos.16-18,26,32,35 In contrast, two authors30,33 comment on the “normalcy” of their patients that presented with genital tattoos.

Genital tattoos may be more common than this very small subsample size suggested as great emphasis has been placed on male penile size in many cultures, for a long time.31,36 The augmentation of these genital markings and decorative designs seemed to have motivated their sexual health, self-enhancement9-10 and well-being.31 Thus, when further studies are considered for this population with a hidden variable, these findings should assist with further ideas of investigation.

Current society has a strong 25 year renaissance of procuring tattoos with at least one in five, and perhaps even four, individuals possessing a tattoo, on virtually every part of their body, without major complications. This small subsample of those who have genital tattoos validates some similarities to those who wear general body tattoos such as a single heterosexual orientation, possessing some college/vocational education, monthly binge drinking,1,3-5,10 and a strong propensity for a Need for Uniqueness.4-5,37 They were major body art wearers and continue to enjoy them, as others have also reported.4-5,10-12

Yet other demographic assumptions were challenged for this subsample of men with genital tattoos. These international respondents tended to be older Caucasians and not as ethnically diverse; there also was not a consensus as to them being risk takers, as has been repeatedly reported by many other body art respondents.1,3-5,11-12

Subsample respondents reported their average first occurance of sexual intercourse at age 17, similar to the national figures.38 Significant experiences of rape were also reported in this subsample, as in women with genital piercings.4-5,9-10 The national rate for forced sexual activity is 10.5%38 and those with genital tattoos reported over twice that amount (23%). No sexual abuse was reported in contrast to a recent German study39 examining general body tattooing.

As with any type of invasive procedure, there can be complications with certain types of body art. When these complications occur, body art wearers typically first seek the internet and/or their studio artist for health advice before presenting to clinicians.1,8,10-12 Yet, overall for the amount of general tattooing done, this type of body art produced limited documented complications and more potential concerns.7-8,11, 30,33 More complications were reported when the tattoos contained colored pigments.6

These tattoos are an integral part of their cultural and personal expression.12,31,33 From our experience many of these male patients with genital tattoos are not seen primarily because of their decorative markings,31 but during clinical evaluations for other issues presented with the normal range of urologic issues involving overall genitourinary and sexual function. Genital tattoos can be an ambivalent findings for many clinicians, but these indelible skin markings (tattoos) are only skin deep,40 and provide valuable cues such as a history of sexual trauma.39 Currently more genital tattoos are seen among our freedom-impaired patients, where the prevalence of general body tattoos among the inmates can be as high as 67%.41

Anecdotally, when healthcare staff discover a patient with genital body art, this discovery can be met with judgmental attitudes and behaviors which could impact care. To adequately assess, evaluate and treat the individuals that have chosen to have genital tattooing, clinicians should strive to provide a thoughtful, nonjudgmental patient-centered approach, along with a generous application of health education, for their present, or even future body art.11

Acknowledgements / Conflicts / Author Details
Acknowledgement: 
The authors acknowledge the grateful assistance of Margaret Vugrin, MSLS, AHIP, Reference Librarian, Texas Tech University Health Sciences Library, Lubbock, TX The authors disclose no conflict of interest or research support for the development of this manuscript. Author Contributions: TN/MLA/AR-research design & analysis; Draft manuscript-TN/MLA/CY/LH/KR/AR
Competing Interests: 
None declared
Details of Authors: 
THOMAS NELIUS MD, Ph.D. Assistant Professor, Texas Tech University Health Sciences Center Department of Urology, School of Medicine, Lubbock, TX 79430 USA. MYRNA L. ARMSTRONG Ed.D., RN, FAAN, Professor Emerita, Texas Tech University Health Sciences Center School of Nursing, TX 78654 USA. CATHY YOUNG, DNSc, FNP-BC, FAANP, FAAN, Associate Professor, Tarleton State University, Texas 76028 USA. ALDEN E. ROBERTS, Ph.D. Professor, Department of Sociology, Anthropology and Social Work, Texas Tech University, Lubbock, TX 79409, USA. LA MICHA HOGAN, MSN, RN, FNP-BC, Clinical Instructor, Texas Tech University Health Sciences Center School of Nursing, Lubbock, TX, USA 79430. KATHERINE RINARD, MD, Tx 79601, USA.
Corresponding Author Details: 
MYRNA L. ARMSTRONG Ed.D., RN, FAAN, Professor Emerita, Texas Tech University Health Sciences Center School of Nursing, 39 Augusta Dr, Marble Falls, TX 78654 USA.
Corresponding Author Email: 
myrna.armstrong@ttuhsc.edu
References
References: 
  1. Armstrong ML, Pace-Murphy K.  Tattooing: Another risk-behavior in adolescents warranting national health teaching.  Applied Nurs Res.  1997;10(4): 181-189.
  2. Braverman S.  The Harris Poll:  One in five US adults now has a tattoo.  Retreived 5/16/13 from www.harrisinteractive.com/NewsRoom/HarrisPolls/tabid/.../Default.aspx
  3. Armstrong ML, Roberts AE, Owen DC, et al.  Toward building a composite of college student influences with body art.  Issues Comprehensive Ped Nurs. 2004;27:277-295. 
  4. Koch JR, Roberts AE, Armstrong ML, et al.  Body art, deviance, and American college students.  Soc Science J.  2010;47(1):151-161.
  5. Owen DC, Armstrong ML, Koch JR, et al.  College students with body art: Well being or high risk behavior.  J Psych Soc & Mental Health Services.  2013;51(10): 20-28.
  6. Wenzel SM, Rittmann I, Landthaler M, et al.  Adverse reactions after tattooing: Review of the literature and comparison to results of a survey.  Dermat 2013;226(2):138-147.  DOI:10.1159/000346943
  7. Mayers L, Chiffriller SH.  Body art (body piercing and tattooing) among undergraduate university students:  Then and now  J Adol Health.  2008;42:201-203. 
  8. O‘Malley PA.  Pharmacology Consult:  Tattoos and piercings: Reasons, risks, and reporting.  Cl Nurs Spec.  2013;27(1):14-16  DOI: 10.1097/NURS.Ob013e31827c28a5.
  9. Hogan L, Rinard K, Young C, et al.   A cross-sectional study of men with genital piercings.  Br J Med Pract.  2010;3(2):315-322.  
  10. Young C, Armstrong ML, Mello I, et al.  A triad of evidence for care of women with genital piercings.  J Am Acad NP.  2010;22:70-80.
  11. Nelius T, Armstrong ML, Rinard K, et al. Genital piercings: Diagnostic and therapeutic implications for Urologists. J Urology.  2011;78;998-1008.
  12. Rinard K, Nelius T, Hogan L, Young C, et al. Cross-sectional study examining four types of male penile and urethral "play". Urology.  2010;76(6):1326-1333.
  13. Weismantel M.  Moche sex pots: Reproduction and temporality in Ancient South America.  Am Anthropologist.  2004;106(3):495-505. 
  14. Angulo JC, Garcia-Diez M, Martinez M.  Phallic decoration in paleolithic art:  Genital scarification, piercing and tattoos.  J Urology.  2011;186:2498-2503. 
  15. Rowanchilde T. Male genital modification: A sexual selection interpretation.  Human Nature.  1996;7(2):189-215.
  16. Van Der Horst C, Martinez P, Seif C, et al.  Male genital injury:  Diagnostics and treatment.  BJU Int.  2002;93:927-930.
  17. Tsunenari S, Yonemitsu K, Kanbe T, et al.  How to identify the Yakuza, Japanese racketeers—their sociology, criminology and physical characteristics.  Ann Acad Med. 1984;13(1);25-31. 
  18. Post RS.  The relationship of tattoos to personality disorders.  J Criminal Law, Criminology and Police Science.  1968;59(4): 516-520.
  19. Fatteh A.  Handbook of Forensic Pathology.  1973  Philadelphia:  J.B. Lippincott.
  20. Burton JL.  The external examination:  An often-neglected autopsy component.  Curr Diag Path. 2007;13:357-365.
  21. Barry MJ.  Tattoos and identity (Letters to the journal).  Canad Med Ass J. 1963;89:1044.
  22. Grumet GW.  Psychodynamic implications of tattoos.  Amer. J. Orthopsychiat.  1983;53(3):482-492. 
  23. Burg BR.  Tattoo designs and locations in the Old U.S. Navy.  J Am Culture. 1995;18(1):69-75.
  24. Adatto M.  Living Skin, 1993: Basle: Editiones, Roche.
  25. Kandijeva J, Kamarashev J, Kadurina M, et al.  Unprofessional tattoos in Bulgaria – psychological aspects.  JEADV. 1995;4;254-259. 
  26. David Duke, running for governor proposes tattooing people with HIV.  AIDS policy & law. 1995(May 19); 53:7.  
  27. Baruchin AM, Schaf S.  Care of traumatic tattoos associated with gunpowder explosions and blast burns (Section V:  Chapter 54).  In M. Masellis & SWA Gunn.  The Management of Mass Burn Casualties and Fire Disasters. 1992  Netherlands:  Springer Kluwer Academic Publishers  292-295.  
  28. McCarron K.  Skin and self-indictment:  Prison tattoos, race, and heroin addiction.  ESC. 2008;34(1):85-102.
  29. Martinez RMA, Wetli CV.  Tattoos of the Marielitos.  Am J For Med & Path.  1989;10(4):315-325.
  30. Matsuzaka J, Aoki H, Banya Y, et al.  A foreign body of the corpus cavernosum in a patient with cleft glans penis: A case report.  Acta Urol. Jpn. 1994;545-547.
  31. Pehlivanov G, Kavaklieva S, Kazandjieve J, et al  Foreign-body granuloma of the penis in sexually active individuals (penile paraffinoma).  JEADV. 2008;22:845-851.
  32. Scholten E, Nanhekhan LV, Oudit DM, et al.  Scrotal and penile reconstruction after massive self-injection of liquid paraffin and petroleum jelly.  Plas & Recont. Surg.  2005;115(7):2168-2169. 
  33. Zargooshi J, Rahmanian E, Motaee H, et al.  Nonischemic priapism following penile tattooing.  J Sex Med. 2012; 9:844-848. 
  34. Lynn M, Synder CR. Uniqueness seeking.  In C.R. Snyder & S.J. Lopez (Eds).  Handbook of Positive Psychology.  New York:  Oxford University Press 2002;395-410.
  35. Taylor AJW.  Criminal tattoos.  Int.Rev. App Psychol.  1974; 29(2); 121-129.
  36. Wylie K.  The way forwards with the obession with the penis.  Sexologies.  2008; 17:S46.
  37. Tiggemann M, Golder F.  Tattooing: An appearance-based expression of uniqueness.  Body Image: Int J Research.  2006;3:309-315. 
  38. Guttmacher Institute.     Facts on American Teens‘ Sexual and Reproductive Health.  .  Retrieved 5/26/13 from www.guttmacher.org/pubs/FB-ATSRH.html
  39. Stirn A, Oddo S, Peregrinova L, et al.  Motivations for body piercings and tattooss—The role of sexual abuse and the frequency of body modifications.  Psych Res.  2011;190(2-3):359-363. 
  40. Susman J. Perspicacity, profiling, and prejudice. J Fam Prac. 2007;56(2):83.
  41. Titilayo CA, Balogun JA, Adefuye AS, et al.  Body art practices among inmates:  Implications for transmission of bloodborne infections.  Am J Inf Control. 2010;38(2):121-129. 

Biologics in Dermatology: A Brief Review

Authors
Iffat Hassan, Samia Aleem, Gousia Sheikh and Parvaiz Anwar
Article Citation and PDF Link
BJMP 2013;6(4):a629
Abstract / Summary
Abstract: 

With the advent of biologic therapy, the treatment of various systemic and cutaneous diseases, especially autoimmune diseases, has been revolutionized. Most of the treatment modalities available prior to biologics aimed at producing clinical improvement of the disease without targeting the actual causative factors. Biologics are protein molecules produced by recombinant DNA technology, which target the specific sites in the immune-pathogenesis pathway of the diseases. Because of the specific action on immune system, biologics are presumed to have lesser side effect profile compared to the traditional immune-suppressants. However, the use of biologics is still limited because of unknown long-term safety profile and various aspects of the biologics need to be thoroughly evaluated by conducting large scale studies worldwide. In this review we give a brief description of various biologic agents that are known till date.

Keywords: 
Biologics; proteins; autoimmune diseases.

INTRODUCTION

The US Food and Drug Administration (USFDA) considers the following as biologics: any therapeutic serum, toxin, antitoxin, vaccine, virus, blood, blood component or derivative, allergenic product, analogous product, or derivatives applicable to the prevention, treatment, or cure of injuries or disease of man1. However, generally, biologics refer to protein molecules therapeutically used in various diseases so as to target specific points in the inflammatory cascade of these disorders 2. Hope for an improved tolerability, convenience in usage and lasting remissions, combined with increased knowledge of immune-pathogenesis of various cutaneous diseases has lead to the introduction of biologics as alternative immune-modulating agents in the field of dermatology.

CLASSIFICATION

Biologics are generally divided into three major groups 3:

a) Monoclonal antibodies

b) Fusion antibody proteins

c) Recombinant human cytokines and growth factors

The main groups and the principal agents in each group are summarised in Box 1 and described below.

A) MONOCLONAL ANTIBODIES

Monoclonal antibodies target specific cell-surface receptors. In the early days of biologic therapy, purely murine monoclonal antibodies were used. However, due to the development of antimurine antibodies, which blocked their action, these could be given only for very short periods. The monoclonal antibodies used now have different amounts of murine sequences in the variable region. They may be categorised into three classes:

(a) chimeric antibodies comprising of 30% murine genes fused with human antibodies

(b) humanised antibodies, which have 10% murine sequences, and

(c) human antibodies, which are solely derived from human immunoglobulin genes 4.

Principal monoclonal antibodies with therapeutic relevance in Dermatology

The principal monoclonal antibodies known till date are enumerated in Box 1 and described briefly below.

Box 1: Classification of biologics
Monoclonal antibodies
    · Anti-TNFα: Infliximab, Adalimumab, Certolizumab, Golimumab
    · Anti-LFA1: Efalizumab
    · Anti-CD20: Rituximab
    · Anti-IL-12 and anti-IL-23 monoclonal antibody: Ustekinumab
    · Anti-CD2 antibody: Siplizumab
    · Anti-CD4 antibody: Orthoclone (OKTcdr4a)
    · Anti-CD25 antibodies: Basiliximab, Daclizumab
    · Anti-CD80r: Galiximab (IDEC 114)
    · Anti-IgE: Omalizumab
Fusion antibody proteins
    · Etanercept
    · Alefacept
    · Abatacept
    · Onercept
    · Denileukin Diftitox
Recombinant human cytokines and growth factors
  a) Interferons
    · Interferon α (IFNα)
    · Interferon γ (IFNγ)
    · Interleukin 1 Receptor antagonist (IL1Ra)
    · Interleukin 2 (IL-2)
    · Interleukin 4 (rhIL-4)
    · Interleukin 10 (rhIL-10)
    · Interleukin 11 (rhIL-11)
  b) Granulocyte macrophage colony stimulating factor (GM-CSF)
  c) Platelet derived growth factor (PDGF)

1. Infliximab

Infliximab (trade name Remicade) is a human-mouse monoclonal antibody that binds to and inhibits the activity of TNF-α, and also causes lysis of TNF-α producing cells5.

Important uses of Infliximab

Psoriasis: Infliximab is approved for the treatment of psoriatic arthritis and plaque psoriasis by FDA6, 7. Infliximab may also be of value in recalcitrant or unstable disease and in generalised pustular psoriasis. It is given as an IV (intravenous) infusion in doses of 5 or 10 mg/kg, over a period of 2 hours at weeks 0, 2, 6 and may be followed by repeat single infusions at 8-12 week intervals 8. In various controlled trials, improvement at 10 weeks has been noted in 87% of patients 7, 9.

Atopic dermatitis: Infliximab has also been evaluated in a study of atopic dermatitis 10. At 2 weeks, there was significant improvement in all patients. At 10, 14, and 30 weeks, variable response was seen.

Hidradenitis suppurativa: Long-term efficacy has also been evaluated in hidradenitis suppurativa. In one study, some patients had no evidence of recurrence after 2 years, while others relapsed within a mean of 8.5 months11.

2. Adalimumab

Adalimumab (Humira®) is a human IgG1 monoclonal antibody directed against TNF-α. Adalimumab is given in a dose of 40 mg subcutaneously (SC) every other week as self-injection 5, 12.

Important uses of Adalimumab

Psoriasis: Adalimumab rapidly reverses the decrease in epidermal Langerhans cell density in psoriatic plaques 13. In a trial, patients with psoriatic arthritis received adalimumab every other week for 24 week14. It was well-tolerated and helped improve joint and skin manifestations significantly.

Hidradenitis suppurativa: An increasing number of reports in refractory hidradenitis supprativa have shown successful control with adalimumab 15, 16, 17.

3. Basiliximab

Successful treatment for severe psoriasis and generalised pustular psoriasis has been reported with basiliximab, an interleukin-2 receptor (IL-2R; CD25) chimeric monoclonal antibody 18, 19.

4. Daclizumab

Daclizumab is a humanised monoclonal antibody thatbinds to the CD25 subunit of the IL-2 receptor onT-cells, thus blocking T-cell proliferation. It has been tried in recalcitrant psoriasis and HIV-associated psoriatic erythroderma with a mean reduction in PASI of 30% 20, 21 22.

5. Siplizumab

Siplizumab (Medi-507) is a humanisedmonoclonal antibody directed against CD2. It is designed to block stimulationby inhibiting the CD2–LFA-3 interaction. In earlyphase studies in psoriasis, significant response to therapy has been noted23.

6. Efalizumab

Efalizumab is a recombinant humanised monoclonal IgG1 antibody that binds to CD11a, a subunit of leukocyte function-associated antigen 1 (LFA-1) 24. It destabilises and decreases the trafficking of T-cells into dermal and epidermal tissues.

Important uses of Efalizumab

Psoriasis: Efalizumab was approved by the US FDA in October 2003 for the treatment of psoriasis5. It is currently the only biologic agent approved for continuous administration to adult patients24. The licensed dose of efalizumab is 1 mg/kg weekly as a subcutaneous self- administered injection for 12 weeks, following a first conditioning dose of 0.7 mg/kg 24.

Lichen planus: There is one case report of 3 months duration of treatment with efalizumab for lichen planus with resolution of skin lesions and pruritis 25.

7. Rituximab

Rituximab is a monoclonal humanised antibody directed again in the B cell-specific antigen CD20.

Important uses of Rituximab

Lymphoma: It has been used in patients with CD20-positive non-Hodgkin's lymphoma in a dosage of 375 mg/m2 for four infusions26.

SLE: In systemic lupus, dose escalation studies revealed no differences with respect to clinical outcome in patients who received either a single infusion of 100 mg/m2, a single infusion of 375 mg/m2, or four weekly infusions of 375 mg/m2 27.

Blistering diseases: For patients with blistering diseases, most patients receive the lymphoma dosage schedule. However, serious side effects were considerably higher.

There are reports of refractory pemphigus patients who received infusions of rituximab and had rapid resolution of lesions and a long lasting clinical remission 28, 29, 30.

8. Galiximab

Galiximab a humanised monoclonal antibody directedagainst CD80 and blocks its interaction with CD28 on the T cell, for T-cell stimulation 31. Clinicaldata for this drug are just beginning to emerge with 40% of patients achieving at least 50% reduction in PASI after receiving4 biweekly doses in a trial32, 33.

9. Ustekinumab

It is a fully human monoclonal antibody targeting IL-12 and IL-23, presently undergoing clinical trials for psoriasis and psoriatic arthropathy2, 34. In placebo-controlled studies, (PHOENIX 1) and (PHOENIX 2) have shown that ustekinumab could control plaque psoriasis with only four injections a year resulting in greater ease of use and more sustained relief 35, 36.

10. Certolizumab pegol

Certolizumab is the recombinant antibody Fab' fragment of a humanised TNF inhibitor monoclonal antibody. A study in chronic plaque psoriasis showed that certolizumab pegol, given subcutaneously every two weeks, over a period of 12 weeks shows significant improvement 37.

11. Golimumab

Golimumab, a fully human monoclonal antibody is at present undergoing Phase III clinical trials in psoriatic arthropathy 38.

12. Orthoclone or OkT4a

It is a humanised antihuman CD4 IgG4 monoclonal antibody preventing the recognition of the MHC-bound antigen by an appropriate T-cell receptor, hence T cells do not get activated 39. Several studies have found orthoclone to be effective in moderate to severe psoriasis 40, 41.

13. ABX-IL8

ABX-IL8 is a fully human monoclonalantibody designed to bind free IL-8, a key chemokine in psoriasis and deactivate it in theskin42, 43. In early trials, the drug has demonstrated good clinical responsein psoriasis 44, 45.

14. Omalizumab

Omalizumab is a recombinant, humanised, monoclonal antibody against immunoglobulin IgE. This agent acts as a neutralising antibody by binding IgE at the same site on IgE as its high-affinity receptor, FcεR I, thus inhibiting the biological effects before the generation of allergic symptoms 46. There are reports of the efficacy of omalizumab in chronic urticaria 47, cold urticaria 48 and atopic dermatitis 49.

15. Mepolizumab

Mepolizumab is a humanised monoclonal IgG antibody to the IL-5 molecule, which is essential for eosinophil growth and differentiation. Two weekly infusions showed significant clinical improvement in atopic dermatitisand clinical trials are underway for hyper-eosinophillic disorders 50, 51.

16. SMART Anti–IFN-γ

SMART anti–IFN- γ,a humanised monoclonal antibody, binds and inactivates IFN- γ, an important Th1 cytokinein psoriasis. Early phase studies are being performed at this time 52.

B) FUSION ANTIBODY PROTEINS

Fusion proteins, also known as chimeric proteins, are proteins which are created by the fusion of the receptor domain of a human protein with the constant region of human IgG. The resultant fusion protein binds specifically to a ligand or co-receptor 53. The most commonly used fusion proteins in dermatology are briefly described below and enumerated in Box 1.

1. Alefacept

Alefacept is a bivalent recombinant fusion protein composed of the first extracellular domain lymphocyte function antigen 3 (LFA-3), fused to the hinge domain of human IgG1. The LFA-3 portion of alefacept binds to CD2 receptors on T-cells, thereby blocking their natural interaction with LFA-3 on antigen presenting cells (APCs). The IgG1 portion of alefacept binds to FcγR receptor on natural killer cells to induce T-cell apoptosis 54.

Important uses of Alefacept

Psoriasis: The US FDA approved alefacept in January 2003 for treatment in adult patients with moderate to severe chronic plaque psoriasis. It is given by intramuscular or intravenous route with a dose of 10-15 mg IM weekly or 7.5 mg IV weekly and a 12 week course is recommended 5, 54. Two 12-week courses showed a75% or greater reduction in the PASI 55.

Alopecia areata: Case reports have shown that alefacept may be effective in the treatmentof AA56, 57.

Pyoderma gangrenosum: Alefacept has been used for pyoderma gangrenosum and improvement was shown in 25% of these patients 58.

Other Indications

Some of the off-label conditions where alefacept has been used with success are graft-versus-host disease (GVHD), lichen planus, alopecia areata, atopic dermatitis, mycosis fungoides,alopecia universalis, erosive lichen planus,Hailey-Hailey diseaseand hand dermatitis 58, 59, 60, 61, 62, 63.

2. Denileukin diftitox

Denileukin diftitox is a novel recombinant fusion protein consisting of fragments of diphtheria toxin linked to human interleukin-2 and works by targeting the high-affinity interleukin-2 receptors. It was tried in patients with recalcitrant psoriasis and the rate of improvement for treated patients was found to be significant 64.

3. Abatacept (Ctla4ig)

It is a fusion protein composed of the extracellular domain of CTLA4 and the Fc region of IgG4. It interferes with T-cell activation by competitively binding the B7.1 and B7.2 molecules on the surface of APC 65. In a study, patients with stable psoriasis vulgaris showed good improvement with IV infusion of abatacept 33. A second generation CTLA4Ig, Belatacept, is currently under Phase II clinical trial for allograft diseases 66.

4. Etanercept

Etanercept is a recombinant fully human dimeric fusion protein comprising of the human TNF-α p75 receptor and the Fc portion of human IgG1 molecule. It functions as a TNF inhibitor, thereby preventing interaction with its cell surface receptors on target cells and blocking its pro-inflammatory effects.

Important uses of Etanercept

Psoriasis: Etanercept (Enbrel®) is FDA approved for use as subcutaneous monotherapy in psoriasis. Several clinical trials have shown that it is effective67, 68, 69, 70. The adult dose is 50 mg/week, given subcutaneously for three months 5. The drug is also indicated for treatment of psoriatic arthritis 71, 72.

Hidradenitis supprivata:There is a study of etanercept in patients with severe hidradenitis with more than 50% score improvement12.

5. Onercept

Onercept is a recombinant human soluble p55 tumour necrosis factor binding protein under development for the potential treatment of psoriasis and psoriatic arthritis73.
C) RECOMBINANT HUMAN CYTOKINES AND GROWTH FACTORS

Cytokines are non-immunoglobulin proteins and glycoproteins produced by a wide variety of cells in the human body and released in response to any immune stimulus 74, 75. Recombinant cytokines or cytokine antagonists have been used as immunomodulators 76. The principal recombinant cytokines used in dermatology, enumerated in Box 1, are described below.

1. Interferons (IFNs)

IFNs, a family of related proteins, are produced by virus-infected leucocytes. They exhibit anti-proliferative, immune-modulatory and anti-neoplastic functions 77.

· Interferon α

Recombinant IFNα is given as a subcutaneous or intramuscular injection to treat verruca vulgaris 78 , condyloma acuminatum 79, cutaneous T cell lymphoma 80, Kaposi's sarcoma (AIDS related) 81, melanoma 82, basal cell carcinoma 83, squamous cell carcinoma 84, actinic keratosis 85, Behçet's disease 86, hemangiomas 87 and keloids 88.

The injections are usually given thrice weekly and the dose (depending on the condition being treated) varies from low-dose therapy for condyloma acuminatum to high-dose therapy for melanoma 89, 90. Of late, pegylated IFNα is being used for convenience, because it has a longer half-life and hence can be given once weekly 80.

· Interferon-γ

It is FDA approved for the treatment of chronic granulomatous disease 91 and has also been used in atopic dermatitis 92 and cutaneous T cell lymphoma 90.

2. Interleukin 1 receptor antagonist (IL1Ra, Anakinra)

Anakinra is the non-glycosylated form of human IL-1Ra and acts by blocking the functions of the naturally occurring IL-193. Good results have also been reported in Schnitzler's syndrome94, familial cold auto-inflammatory syndrome 95 and psoriatic arthropathy96. It is given by subcutaneous injection 100 mg once a day.

3. Interleukin 2

Recombinant IL-2 is an antitumour cytokine that has been used in cutaneous T cell lymphoma (CTCL) and metastatic melanoma 97. When given intravenously in high doses of 600,000-720,000 IU/kg in melanoma, IL-2 has produced a 15-20% overall response, with complete cure in 4-6% 98.

4. Interleukin 4 (rhIL-4)

In a dose-escalation study (0.5 to 5mg/kg given by subcutaneous injection thrice a week), IL-4 has been shown to cause improvement in psoriasis by inducing Th2 differentiation in human CD4 + T cells 99.

5. Interleukin 11 (rhIL-11, Oprelvekin)

It has also shown reasonably good results in the treatment of psoriasis at doses of 2.5 or 5mg/ kg, by subcutaneous injection 100.

6. Granulocyte macrophage colony stimulating factor (GM-CSF)

GM-CSF acts by stimulating stem cells to produce granulocytes, monocytes and macrophages 101. Recombinant human GM-CSF has been used to promote wound healing in ulcerated skin for example leg ulcers 102, 103, and for the treatment of melanoma 104 and Sezary syndrome 105.

7. Platelet derived growth factor (PDGF)

PDGF is a dimeric glycoprotein which regulates and promotes granulation tissue formation, re-epithelialisation and wound angiogenesis 106. Recombinant PDGF-BB topical gel (100ìg/g), applied once daily, has been approved by FDA for the treatment of diabetic foot ulcers 107, 108.

8. Recombinant Human IL-10

Recombinant Human IL-10 (Tenovil)can be given in subcutaneousinjections. Early phase clinical trials have shown thatrecombinant human IL-10 three times a week improved psoriasis 109, 110.

SIDE EFFECTS OF BIOLOGICS 5, 111, 112, 113

Some of the adverse effects of biologics are described below:

  • Allergic reaction and antibody formation: Mostly seen with TNF-α blockers.
  • Mild transient injection site reactions: Comprising of erythema, oedema and bruising, noted with etanercept in 10-20% of cases in the first month of therapy. Antibodies to etanercept may develop in 6% of patients.
  • Infusion reaction: Occurs during or within 1-2 hours of treatment and may affect up to 20% of all the patients treated with infliximab, rarely anaphylactic shock may occur.
  • Acute flu-like symptoms: Headache, chills, fever, nausea and myalgia may occur within 48 hours after administration of the first two doses of efalizumab and Interferon α.
  • Infections: Reactivation of tuberculosis may occur on treatment with anti-TNF-α agents and sepsis secondary to Listeria monocytogenes and Histoplasma capsulatum have been reported 113.
  • Malignancy: Patients previously treated with PUVA represent an at-risk group.
  • Demyelinating disease: Worsening of multiple sclerosis and demyelination reported with infliximab.
  • Thrombocytopenia: Occurs with efalizumab and warrants discontinuation of therapy.
  • Autoimmune haemolytic anemia: Occurs 4-6 months after the start of treatment with efalizumab.
  • Congestive cardiac failure: Worsening of congestive cardiac failure with TNF-α blockers is reported to occur.
  • Antinuclear antibodies and lupus-like syndrome: May develop during therapy with anti-TNF-α agents, but not associated with symptoms and signs of lupus in the majority.
  • Hepatitis: Reported following infliximab therapy, occurring from 2 weeks to more than a year after initiation of treatment. Treatment should be stopped in the event of jaundice and/ or marked elevations (>5 times the upper limit of normal) in liver enzymes.

PATIENT SCREENING FOR BIOLOGIC THERAPY 113, 114

All patients to be put on biologics should undergo a thorough evaluation including detailed clinical history, physical examination and relevant investigations with particular reference to known toxicity profile of the agent being considered. The investigations generally advised are113: full blood count, liver and renal function tests, screening for hepatitis and HIV infection, anti-nuclear antibodies, anti-ds DNA, urine analysis, chest X-ray and Tuberculin skin testing.

For efalizumab, haemogram (including platelet count) is recommended monthly for the first 3 months and then every 3 months. For TNF blockers, it is done at 3 months initially and repeated every 6 months.

Liver and renal function tests, serum electrolytes and urine analysis are done at 3 months initially and then every 6 months.

EXCLUSION CRITERIA/ CONTRAINDICATIONS

There are various contraindications for use of biologics, warranting their exclusion and precautions are to be exercised because of their immune-modulator properties. The main exclusion criteria are: active tuberculosis, severe congestive heart failure, patients having >200 treatments of PUVA (because of a risk of developing malignancies with anti- TNF agents), history of demyelinating disease or optic neuritis, hepatitis B and C positivity, HIV positivity, premalignant states, active infections and high risk states such as chronic leg ulcers, persistent or recurrent chest infections and indwelling urinary catheter infections, pregnancy and breast-feeding.

ASSESSMENT OF THE RESPONSE TO BIOLOGICS

Many scoring systems for assessing the severity of various dermatological diseases exist. These scoring systems and other indices can be used for assessment of response to the use of biologics. For example, for evaluation of improvement in psoriasis, PASI (psoriasis area and severity index) and DLQI (dermatology life quality index) are recommended at 3 months initially and then every 6 months 113. Reduction in baseline PASI score of >75% is the standard used by FDA to assess the efficacy of a new psoriasis agent 115. Similarly in atopic eczemas, improvement is monitored based on the Eczema Area and Severity Index, Pruritus Severity Assessment and DLQI. Reduction of the Eczema Area and Severity Index score by 50% is considered excellent, 30-49% moderate and <29% non-significant.

SUMMARY

To summarise, biologics represent the future of therapeutics, not only in dermatology but also in other fields of medicine. Among the various dermatological disorders where they are used, biologics have been most evaluated in psoriasis 116. However, the possibility of serious infections and the oncogenic potential combined with the high cost of the drugs limit their routine use at the present stage 117. Regular re-evaluation of efficacy and safety is essential if these agents are to be used to the maximum benefit of patients118.

Acknowledgements / Conflicts / Author Details
Acknowledgement: 
Peerzada Sajad and Konchok Dorjay
Competing Interests: 
None declared
Details of Authors: 
IFFAT HASSAN, MD, Professor and Head, Department of Dermatology, STD and Leprosy, Govt.Medical College Srinagar (University of Kashmir), J & K, India; SAMIA ALEEM, MBBS, Resident, Department of Dermatology, STD and Leprosy, Govt.Medical College Srinagar (University of Kashmir), J & K, India; GOUSIA SHEIKH, MBBS, Resident, Department of Dermatology, STD and Leprosy, Govt.Medical College Srinagar (University of Kashmir), J & K, India; PARVAIZ ANWAR, MD, Senior Resident, Department of Dermatology, STD and Leprosy, Govt.Medical College Srinagar (University of Kashmir), J & K, India.
Corresponding Author Details: 
Professor IFFAT HASSAN, Head of Department of Dermatology, STD and Leprosy, Govt.Medical College Srinagar (University of Kashmir), J & K, India.
Corresponding Author Email: 
hassaniffat@gmail.com
References
References: 
  1. Feldmann M, Steinman L. Design of effective immunotherapy for human autoimmunity. Nature 2005; 435: 612-9.
  2. Dogra A, Salonie S. Biologic therapy in psoriasis. Indian J Dermatol Venereol Leprol 2006: 72: 256-65.
  3. Stern DK, Tripp JM, Ho VC, Lebwohi M. The use of systemic immune moderators in dermatology. In: Maclean DI, Maddin WS, editors. Dermatologic clinics. Vol. 23. Elsevier; 2005. p. 275.
  4. Waldmann TA. Immunotherapy: Past, present and future. Nat Med 2003; 9: 269-77.
  5. Kormeili T, Lowe NJ, Yamuchi PS. Psoriasis: Immunopathogenesis and evolving immunomodulators and systemic therapies; U.S experiences. Br J Dermatol 2004; 151: 3-15.
  6. Antoni CE, Kruegere GG, de Vlam K, Birbara C, Beutler A, Guzzo C, et al. Infiliximab improves signs and symtoms of psoriatic arthritis the IMPACT 2 trial.Ann Rheum Dis 2005; 64: 1150-7.
  7. Gottlieb AB, Evans R, Li S, Dooley LT, Guzzo CA, Baker D, et al . Infliximab induction therapy for patients with severe plaque type psoriasis: A randomized, double-blind, placebo controlled trial. J Am Acad Dermatol 2004; 51: 534-42.
  8. Winterfield L, Menter A. Psoriasis and its treatment with infliximab-mediated tumour necrosis factor a blockade. Dermatol Clin 2004; 22: 437-44.
  9. Chaudhari U, Romano P, Mulcahy LD, Dooley LT, Baker DG, Gottlieb AB. Efficacy and safety of infliximab montherapy for plaque type psoriasis: A randomized trial. Lancet 2001; 35: 1842-7.
  10. Jacobi A, Antoni C, Manger B, Schuler G, Hertl M. Infliximab in the treatment of moderate to severe atopic dermatitis. J Am Acad Dermatol. 2005; 2: 522-6.
  11. Giamarellos-Bourboulis EJ, Pelekanou E, Antonopoulou A, Petropoulou H, Baziaka F, Karagianni V, et al. An open-label phase II study of the safety and efficacy of etanercept for the therapy of hidradenitis suppurativa. Br J Dermatol 2008; 158: 567-72.
  12. Scheinfeld N. Adalimumab (HUMIRA): A review. J Drugs Dermatol 2003; 2: 375-7.
  13. Gordon KB, Bonish BK, Patel T, Leonardi CL, Nickloff BJ. The tumour necrosis factor-alpha inhibitor adalimumab rapidly reverses the decrease in epidermal Langerhans cell density in psoriatic plaques. Br J Dermatol 2005; 153: 945-53.
  14. Mease PJ, Gladman DD, Ritchlin CT, Ruderman EM, Steinfeld SD, Choy EH, et al. Adalimumab for the treatment of patients with moderately to severely active psoriatic arthritis: Results of a double-blind, randomized, placebo-controlled trial. Arthritis Rheum 2005; 52: 3279-89.
  15. Blanco R, Martínez-Taboada VM, Villa I, González-Vela MC, Fernández-Llaca H, Agudo M, et al. Long-term Successful Adalimumab Therapy in Severe Hidradenitis Suppurativa. Arch Dermatol 2009; 145: 580-84.
  16. Moul DK, Korman NJ. Severe hidradenitis suppurativa treated with adalimumab. Arch Dermatol 2006; 142: 1110-2.
  17. Scheinfeld N. Treatment of coincident seronegative arthritis and hidradenitis suppurativa with adalimumab. J Am Acad Dermatol 2006; 55: 163-4.
  18. Owen CM, Harrison PV. Successful treatment of severe psoriasis with basilixima, an interleukin-2 receptor monoclonal antibody. Clin Exp Dermatol 2000; 25: 195-7.
  19. Salim A, Emerson RM, Dalziel KL. Successful treatment of severe generalized pustular psoriasis with basiliximab (interleukin-2 receptor blocker). Br J Dermatol 2000; 143: 1121-2.
  20. Wohlrab J, Fischer M, Taube KM, Marsch WC. Treatment of recalcitrant psoriasis with daclizumab. Br J Dermatol 2001; 144: 209-10.
  21. Dichmann S, Mrowietz U, Schopf E, Norgauer J. Humanized monoclonal anti-CD25 antibody as a novel therapeutic option in HIV-associated psoriatic erythroderma. J Am Acad Dermatol 2002; 47: 635-6.
  22. Krueger JG, Walters IB, Miyazawa M, Gilleaudeau P, Hakimi J, Light S, et al. Successful in vivo blockade of CD25 (high-affinity interleukin 2 receptor) on T cells by administration of humanized anti-Tac antibody to patients with psoriasis. J Am Acad Dermatol 2000; 43: 448-58.
  23. Langley R, Roenigk HH, McCall C, Stricklin G, Dingivan C. Phase I results of intravenous MEDI-507, an anti-T-cell monoclonal antibody, for the treatment of psoriasis. J Invest Dermatol 2001; 117: 817.
  24. Leonardi CL. Current concepts and review of efalizumab in the treatment of psoriasis. Dermatol Clin 2004; 22: 427-35.
  25. Bohm M, Luger T. Lichen planus responding to efalizumab. J Am Acad Dermatol 2007; 56: 92-3.
  26. Leandro M, Edwards J, Cambridge G. Clinical outcome in 22 patients with rheumatoid arthritis treated with B lymphocyte depletion. Ann Rheum Dis 2002; 61: 883-8.
  27. Anolik JH, Barnard J, Cappione A, Pugh-Bernard AE, Felgar RE, Looney RJ,  et al. Rituximab improves peripheral B cell abnormalities in human systemic lupus erythematous. Arthritis Rheum 2004; 50: 3580-90.
  28. Ahmed A, Spiegelman Z, Cavacini L, Posner M. Treatment of pemphigus vulgaris with rituximab and intravenous immune globulin. N Engl J Med 2006; 355: 1772-9.
  29. Faurschou A, Gniadecki R. Two courses of rituximab for recalcitrant pemphigus vulgaris. Int J Dermatol 2008; 47: 292-4.
  30. Schmidt E, Brockerm E, Goebeler M. Rituximab in treatment-resistant autoimmune blistering skin disorders. Clin Rev Allergy Immunol 2008; 34: 56-64.
  31. Mitra RS, Judge TA, Nestle FO, Turka LA, Nickoloff BJ. Psoriatic skin-derived dendritic cell function is inhibited by exogenous IL-10: differential modulation of B7-1 (CD80) and B7-2 (CD86) expression. J Immunol 1995; 154: 2668-77.
  32. Gottlieb A, Abdulghani A, Totoritis M, Lizambri R, Shuey S, Romano P, et al. Results of a single-dose, dose-escalating trial of an anti-B7 monoclonal antibody (IDEC-114) in patients with psoriasis. J Invest Dermatol 2000; 114: 840.
  33. Gottlieb AB, Kang S, Linden KG, Lebwohl M, Menter A, Abdulghani AA, et al. Results of a multiple-dose, multiple schedule trial of an anti-CD80 monoclonal antibody (IDEC-114) in patients with psoriasis. Poster presented at: annual meeting of the American Academy of Dermatology; March 2, 2001; Washington, DC.
  34. Krueger GG, Langley RG, Leonardi C.A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis. N Engl J Med 2007; 356: 580-92.
  35. Leonardi CL, Kimball AB, Papp KA, Yeilding N, Guzzo C, Wang Y, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX). Lancet 2008; 371: 1665-74.
  36. Papp KA, Langley RG, Lebwohl M, Krueger GG, Szapary P, Yeilding N, et al . Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo- controlled trial (PHOENIX). Lancet 2008; 371: 1675-84.
  37. Melmed GY, Targan SR, Yasothan U. Certolizumab pegol. Nat Rev Drug Discov 2008; 7: 641-2.
  38. Hutas G. Golimumab: A fully human monoclonal antibody against TNF alpha. Curr Opin Mol Ther 2008; 10: 393-406.
  39. Rao PE, Kroon DJ. Orthoclone OKT3: Chemical mechanisms and functional effects of degradation of a therapeutic monoclonal antibody. Pharm Biotechnol 1993; 5: 135-58.
  40. Bachelez H, Flageul B, Dubertret L, Fraitag S, Grossman R, Brousse N, et al. Treatment of recalcitrant plaque psoriasis with a humanized non-depleting antibody to CD4. J Autoimmun 1998; 11: 53-62.
  41. Gottlieb AB, Lebwohl M, Shirin S, Sherr A, Gilleaudeau P, Singer G, et al . Anti-CD4 monoclonal antibody treatment of moderate to severe psoriasis vulgaris: Results of a pilot, multicenter, multiple-dose, placebo-controlled study. J Am Acad Dermatol 2000; 43: 595-604.
  42. Barker JN, Jones ML, Mitra RS, et al. Modulation of keratinocyte-derived interleukin-8 which is chemotactic for neutrophils and T lymphocytes. Am J Pathol 1991; 139: 869-76.
  43. Biasi D, Carletto A, Caramaschi P, Bellavite P, Maleknia T, Scambi C, et al. Neutrophil functions and IL-8 in psoriaticarthritis and in cutaneous psoriasis. Inflammation 1998; 22: 533-43.
  44. Krueger GC, Lohner M, Roskos L, Hwang CC, Bell G, Schwab G. Clinical trials results: a fully human anti-IL-8 antibody in patients with moderate to severe psoriasis. Poster presented at: annual meeting of the American Academy of Dermatology; March 2, 2001; Washington, DC.
  45. Lohner M, Krueger J, Gottlieb A, Beutner K, Levy R, Wiesenhutter C, et al. Clinical trials of a fully human anti-IL-8 antibody for the treatment of psoriasis. Br J Dermatol 1999; 141: 989.
  46. Mankad VS. Omalizumab: Other indications and unanswered questions. Clin Rev Allergy Immunol 2005; 29: 17-30.
  47. Spector SL, Tan RA. Effect of omalizumab on patients with chronic urticaria. Ann Allergy Asthma Immunol 2007; 99: 190-3.
  48. Boyce JA. Successful treatment of cold-induced urticaria/anaphylaxis with anti-IgE J Allergy Clin Immunol 2006; 117: 1415-8.
  49. Forman SB, Garrett AB. Success of omalizumab as monotherapy in adult atopic dermatitis: Case report and discussion of the high-affinity immunoglobulin E receptor, Fcepsilon RI. Cutis 2007; 80: 38-40.
  50. Oldhoff JM, Darsow U, Werfel T, Katzer K, Wulf A, Laifaoui J, et al. Anti il-5 recombinant humanised monoclonal antibody for the treatment of atopic dermatitis. Allergy 2005; 60: 693-6.
  51. Simon D, Brathen LR, Simon HU. Anti-interleukin-5 antibody therapy in eosinophilic disorders. Pathabiol 2005; 72: 287-92.
  52. Prashant S, Dennis PW, Kenneth BG. Biologic Therapy for Psoriasis, The New Therapeutic Frontier. Arch Dermatol 2002; 138: 657-63.
  53. Krueger JG. The immunologic basis for the treatment of psoriasis with new biologic agents. J Am Acad Dermatol 2002; 46: 1-23.
  54. Krueger GG. Current concepts and review of alefacept in the treatment of psoriasis. Dermatol Clin 2004; 22: 407-26.
  55. Krueger GG, Papp KA, Stough DB, Loven KH, Gulliver WP, Ellis CN. Alefacept Clinical Study Group. A randomized, double-blind, placebo-controlled phase III study evaluating efficacy and tolerability of 2 courses of alefacept in patients with chronic plaque psoriasis. J Am Acad Dermatol 2002; 47: 821-33.
  56. Heffernan MP, Hurley MY, Martin KS, Smith DI, Anadkat MJ. Alefacept for alopecia areata. Arch Dermatol 2005; 141: 1513-6.
  57. Bui K, Polisetty S, Gilchrist H, Jackson SM, Frederic J. Successful treatment of alopecia universalis with alefacept: a case report and review of the literature. Cutis 2008; 81: 431-4.
  58. Foss C, Clark A, Inabinet R, Camacho F, Jorizzo J. An open label pilot study of alefacept for the treatment of pyoderma gangrenosum. J Eur Acad Derm Venerol 2008; 22: 943-9.
  59. Richardson S, Getfand J. Immunobiologicals, cytokines and growth factors in dermatology. In: Wolff K, Goldsmith LA, Katz SI, Gilchrest BA, Paller AS, Leffell DJ, editors. Fitzpatrick's dermatology in general medicine. 7 th ed. McGraw - Hill; 2008. p. 2223-31.
  60. Moul DK, Routhouska SB, Robinson MR, Korman NJ. Alefacept for moderate to severe atopic dermatitis: A pilot study in adults. J Am Acad Dermatol 2008; 58: 984-9.
  61. Green WH, Leicht SS, Youngberg GA. Patch-stage mycosis fungoides in remission after therapy with alefacept. J Am Acad Dermatol 2008; 58: 110-2.
  62. Chang AL, Badger J, Rehmus W. Alefacept for erosive lichen planus: A case series. J Drugs Dermatol 2008; 7: 379-83.
  63. Hurd DS, Johnston C, Bevins A. A case report of Hailey-Hailey disease treated with alefacept (Amevive). Br J Dermatol 2008; 158: 399-401.
  64. Bagel J, Garland WT, Breneman D, Holick M, Littlejohn TW, Crosby D, et al . Administration of DAB389IL-2 to patients with recalcitrant psoriasis: A double blind, phase II multicenter trial. J Am Acad Dermatol 1998; 3: 938-44.
  65. Moreland L, Bate G, Kirkpatrick P. Abatacept. Nat Rev Drug Discov 2006; 5: 185-6.
  66. Vincenti F, Kirk AD. What's next in the pipeline. Am J Transplant 2008; 8: 1972-81.
  67. Mease PJ, Goffe BS, Metz J, Vander SA, Finck B, Burge DJ. Etanercept in treatment of psoriatic arthritis and psoriasis: A randomized trial. Lancet 2000; 356: 385-90.
  68. Gottlieb AB, Matheson RT, Lowe N, Krueger GG, Kang S, Goffe BS, et al . A randomized trial of etanercept as monotherapy for psoriasis. Arch Dermatol 2003; 139: 1627-32.
  69. Leonardi CL, Powers JL, Matheson RT, Goffe BS, Zitnik R, Wang A, et al . Etanercept as monotherapy in patients with psoriasis. N Engl J Med 2003; 349: 2014-22.
  70. Papp KA, Tyring S, Lahfa M, Prinz J, Griffiths CE, Nakanishi AM, et al . A global Phase III randomized control trial of etanercept in psoriasis: Safety, efficacy and effect of dose reduction. Br J Dermatol 2005; 152: 1304-12.
  71. Goldsmith DR, Wagstaff AJ. Etanercept: A review of its use in the management of plaque psoriasis and psoriatic arthritis. Am J Clin Dermatol 2005; 6: 121-36.
  72. Yamuchi PS, Gindi V, Lowe NJ. The treatment of psoriasis and psoriatic arthritis with etanercept: Practical considerations on monotherapy, combination therapy and safety. Dermatol Clin 2004; 22: 449-59.
  73. Nikas SN, Drosos AA. Onercept. Serono. Curr Opin Investig Drugs 2003; 4: 1369-76.
  74. Holman DM, Kallaji AN. Cytokines in dermatology. J Drug Dermatol 2006; 5: 520-4.
  75. Oppenheim JJ. Cytokines: Past, present and future. Int J Haematol 2001; 74: 3.
  76. Trefzer U, Hofmann M, Sterry W, Asadullah K. Cytokine and anticytokine therapy in dermatology. Expert Opin Biol Ther 2003; 3: 733-43.
  77. Thiel DJ, le Du MH, Walter RL, D'Arcy A, Chène C, Fountoulakis M, et al . Observation of an unexpected third receptor molecule in the crystal structure of human interferon-receptor complex. Structure 2000; 8: 927-36.
  78. Cac NN, Ballas ZK. Recalcitrant warts, associated with natural killer cell dysfunction, treated with systemic IFN-alpha. J Allergy Clin Immunol 2006; 118: 526-8.
  79. Panja SK, Chowdhury A, Banerjee PK, Coondoo A, Nandi S, Saha NC, et al . Interferon Alpha-2b in genital warts. Indian J Dermatol 1991; 36: 5.
  80. Rook AH, Kuzel TM, Olsen EA. Cytokine therapy of cutaneous T-cell lymphoma:         Interferons, interleukin-12, and interleukin-2. Hematol Oncol Clin North Am 2003; 17: 1435-8.
  81. Potthoff A, Brockmeyer NH. HIV-associated Kaposi sarcoma: Pathogenesis and therapy. J Dtsch Dermatol Ges 2007; 5: 1091-4.
  82. Kalani AD, Jack A, Montenegro G, Degliuomini J, Wallack MK. Immunotherapy as an adjuvant therapy in the management of advanced, surgically resected melanoma. G Ital Dermatol Venereol 2008; 143: 59-70.
  83. Tucker SB, Polasek JW, Perri AJ, Goldsmith EA. Long-term follow-up of basal cell carcinomas treated with perilesional interferon alfa 2b as monotherapy. J Am Acad Dermatol 2006; 54: 1033-8.
  84. Edwards L, Berman B, Rapini RP, Whiting DA, Tyring S, Greenway HT Jr, et al . Treatment of cutaneous squamous cell carcinomas by intralesional interferon alpha 2b therapy. Arch Dermatol 1992; 128: 1486-9.
  85. Edwards L, Levine N, Widener M. Effect of intralesional alpha 2 interferon on Actinic keratosis. Arch Dermatol 1986; 122: 779.
  86. Tsambaos D, Eichelberg D, Goos M. Behçet's syndrome: Treatment with recombinant leucocyte alpha-interferon. Arch Dermatol Res 1986; 278: 335-6.
  87. Kim HJ, Colombo M, Frieden IJ. Ulcerated haemangiomas: Clinical characteristics and response to therapy. J Am Acad Dermatol 2001; 44: 962-72.
  88. Al-Attar A, Mess S, Thomassen JM., Kauffman CL, Davison SP. Keloid pathogenesis and treatment. Plast Reconstr Surg 2006; 117: 286-300.
  89. Gross G, Ikenberg H, Roussaki A, Drees N, Schöpf E. Systemic therapy of condylomata acuminata with recombinant alpha 2a: Low dose superior to the high dose regimen. Chemotherpay 1986; 32: 537-41.
  90. Mitchell MS, Abrams J, Thompson JA, Kashani-Sabet M, DeConti RC, Hwu WJ, et al . Randomized trial of an allogeneic melanoma lysate vaccine with low-dose interferon Alfa-2b compared with high-dose interferon Alfa-2b for Resected stage III cutaneous melanoma. J Clin Oncol 2007; 25: 2078-85.
  91. Bolinger A, Taeubel MA. Recombinant interferon gamma for treatment of chronic granulomatous disease and other disorders. Clin Pharm 1992; 11; 834-50.
  92. Hanifin JM, Schneider LC, Leung DY, Ellis CN, Jaffe HS, Izu AE, et al . Recombinant interferon gamma therapy for Atopic Dermatitis. J Am Acad Dermatol 1993; 28: 189-97.
  93. Pierer M, Baerwald C. Biological therapy for the treatment of rheumatic diseases. Internist (Berl) 2008; 49: 938-46.
  94. Frischmeyer-Guerrerio PA, Rachamalla R, Saini SS. Remission of Schnitzler syndrome after treatment with anakinra. Ann Allergy Asthma Immunol 2008; 100: 617-9.
  95. Ross JB, Finlayson LA, Klotz PJ, Langley RG, Gaudet R, Thompson K, et al . Use of anakinra (Kineret) in the treatment of familial cold autoinflammatory syndrome with a 16-month follow-up. J Cutan Med Surg 2008; 12: 8-16.
  96. Peddle L, Butt C, Snelgrove T. Interleukin (IL) 1alpha, IL1beta, IL receptor antagonist, and IL10 polymorphisms in psoriatic arthritis. Ann Rheum Dis 2005; 64: 1093-4.
  97. Marzec M, Halasa K, Kasprzycka M, Wysocka M, Liu X, Tobias JW, et al . Differential effects of interleukin-2 and interleukin-15 versus interleukin-21 on CD4 + cutaneous T-cell lymphoma cells. Cancer Res 2008; 68: 1083-91.
  98. Atkins MB, Lotze MT, Dutcher JP, Fisher RI, Weiss G, Margolin K, et al . High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: Analysis of 270 patients treated between 1985 and 1993. J Clin Oncol 1999; 17: 2105-16.
  99. Ghoreschi K, Thomas P, Breit S, Dugas M, Mailhammer R, van Eden W, et al . Interleukin-4 therapy of psoriasis induces Th2 responses and improves human autoimmune disease. Nat Med 2003; 9: 40-6.
  100. Trepicchio WL, Ozawa M, Walters IB, Kikuchi T, Gilleaudeau P, Bliss JL, et al . Interleukin-11 therapy selectively down regulates type I cytokine proinflammatory pathways in psoriasis lesions. J Clin Invest 1999; 104: 1527-37.
  101. Conti L, Gessani S. GM-CSF in the generation of dendritic cells from human blood monocyte precursors: Recent advances. Immunobiology 2008; 213: 859-70.
  102. Chaperot L, Chokri M, Jacob MC, Drillat P, Garban F, Egelhofer H, et al . Differentiation of antigen-presenting cells (dendritic cells and macrophages) for therapeutic application in patients with lymphoma. Leukemia 2000; 14: 1667-77.
  103. Meier K, Nanney LB. Emerging new drugs for wound repair. Expert Opin Emerg Drugs 2006; 11: 23-37.
  104. Alikhan MA, Carter G, Mehta P. Topical GM-CSF hastens healing of leg ulcers in sickle cell disease. Am J Hematol 2004; 76: 192.
  105. Elias EG, Zapas JL, McCarron EC, Beam SL, Hasskamp JH, Culpepper WJ. Sequential administration of GM-CSF (Sargramostim) and IL-2 ± autologous vaccine as adjuvant therapy in cutaneous melanoma: An interim report of a phase II clinical trial. Cancer Biother Radiopharm 2008; 23: 285-91.
  106. Bouwhuis SA, Markovic SN, McEvoy MT. Extracorporeal photopheresis and adjuvant aerosolized granulocyte-macrophage colony-stimulating factor for Sézary syndrome. Mayo Clin Proc 2002; 77: 197-200.
  107. Pinkas H, Fisch B, Rozansky G, Felz C, Kessler-Icekson G, Krissi H, et al . Platelet-derived growth factors (PDGF-A and -B) and their receptors in human fetal and adult ovaries. Mol Hum Reprod 2008; 14: 199-206.
  108. Embil JM, Papp K, Sibbald G. Recombinant human platelet-derived growth factor-BB (becaplermin) for healing chronic lower extremity diabetic ulcers: An open-label clinical evaluation of efficacy. Wound Repair Regen 2000; 8: 162-8.
  109. Asadullah K, Döcke WD, Ebeling M, Friedrich M, Belbe G, Audring H, et al. Interleukin 10 treatment of psoriasis: clinical results of a phase II trial. Arch Dermatol 1999; 135: 187-192.
  110. Smith CH, Anstey AV, Barker JN, Burden AD, Chalmers RJ, Chandler D, et al. British association of Dermatologists guidelines for use of biological interventions in psoriasis 2005. Br J Dermatol 2005; 153: 486-97.
  111. Sterry W, Barker J, Boehncke WH, Bos JD, Chimenti S, Christophers E, et al. Biological therapies in the systemic management of psoriasis: International Consensus Conference. Br J Dermatol 2004; 151: 3-17.
  112. Koo J, Khera P. Update on the mechanisms and efficacy of biological therapies for psoriasis. J Dermatol Sci 2005; 38: 75-87.
  113. Asadullah K, Sterry W, Ebeling M, Friedrich M, Leupold M, Jasulaitis D, et al. Clinical and immunological effects of IL-10 therapy in psoriasis. Br J Dermatol 1999; 141: 989.
  114. Sukal SA, Nadiminti L, Granstein RD. Etanercept and demyelinating disease in a patient with psoriasis. J Am Acad Dermatol 2006; 54: 160-4.
  115. Scheinfeld N. Adalimumab: A review of side effects. Exp Opin Drug Saf 2005; 4: 637-41.
  116. Thappa DM, Laxmisha C. Immunomodulators in the treatment of Psoriasis. Indian J Dermatol Venereol Leprol 2004; 70: 1-9.
  117. Lebowhl M, Clark LN. Recent advances in medical dermatology. Int J Dermatol 2007; 1: 197-202.
  118. Smith CH, Anstey AV, Barker JN, Burden AD, Chalmers RJ, Chandler DA, et al. British association of dermatologist guidelines for biological intervention for psoriasis 2009.Br J Dermatol 2009; 161: 987-1019.

Guttate psoriasis: A rare cause of diffuse rash.

Authors
Nauman Shahid, Muhammad Z Bawany, Ehsan Rafiq and Thomas Sodeman
Article Citation and PDF Link
BJMP 2013;6(4):a627
Abstract / Summary
Abstract: 

Guttate psoriasis is a variant of psoriasis presenting as small, erythematous papules and plaques on the skin. Streptococcal infection is a common inciting factor. We are reporting a case of a 53 years old male, who presented with a generalized rash without any history of a recent infection and was diagnosed with guttate psoriasis on skin biopsy.

Introduction

Psoriasis is a common skin disorder characterized by erythematous papules and plaque formation with silver scaling. Guttate psoriasis is much less common and many studies cite a prevalence of less than 30% among patients who have psoriasis. It refers to the acute appearance of multiple skin eruptions mostly in a patient with no preexisting psoriasis and less commonly in a patient with psoriasis. We report here a case of guttate psoriasis associated with a flare of psoriatic arthritis.

Case report

A 53 year old man presented with a generalized body rash and multiple joint pains. His symptoms started a week prior to presentation. The skin rash initially appeared on his back and flanks but gradually progressed to involve the thighs and arms. He had ‘sausage fingers’, bilateral knee and ankle swelling associated with pain and sporadic metatarsophalyngeal joint pain as manifestations of his arthritis. His past medical history included hypertension, diabetes mellitus, hyperlipidemia and psoriasis with psoriatic arthritis. He did not report any recent changes in his medication. The patient denied any history of fever, sore throat, weight loss, visual problems, dyspnea, cough, gastrointestinal complaints or recent travel. Physical examination revealed a diffuse, non-blanching, pruritic, maculopapular and maculopustular rash over the trunk. He also had a scaly and diffuse erythematic rash over the lower abdomen which was non-blanching and pruritic. Auspitz’s sign was positive. He had multiple painful joints including both knees, right wrist, left proximal interphalangeal joint, and both his ankles. Left knee arthrocentesis was done which revealed joint fluid consistent with inflammatory joint disease without any evidence of crystals. Laboratory tests, including red and white blood cell count, haemoglobin, cyclic citrullinated peptide antibody, rapid plasma regain, hepatitis panel, antinuclear antibody, rheumatoid factor, lyme markers and serum uric acid revealed no abnormalities. ASO titer level was positive at 196.  An x-ray of his left hand showed periarticular erosive changes along the distal aspect of the proximal phalanx. A skin biopsy was performed which revealed mild spongiosis and a perivascular lymphoplastic infiltrate. A diagnosis of guttate psoriasis was made. He was started on prednisone, methotrexate and folic acid and discharged from hospital. He was followed up in the rheumatology clinic 2 weeks after discharge and his rash had improved.

Discussion

Unlike psoriasis, guttate psoriasis is a much lesser known entity. It refers to the acute appearance of multiple skin eruptions, mostly in patients with no preexisting psoriasis and less commonly in chronic plaque psoriasis (guttate flare of chronic plaque psoriasis).

The characteristic skin lesions of guttate psoriasis are less than 1 cm in diameter, hence the name guttate (drop like). The lesions look like a shower of red, scaly tear drops that have fallen down on the body mainly involving the trunk, arms, thighs and face. Guttate psoriasis should be differentiated from diabetic dermopathy, also called shin spots, which typically begin as dull red, scaly papules or plaques and later develop into bilateral asymmetrical circumscribed shallow pigmented scars and/or brownish macular lesions with a fine scale. In diabetic dermopathy the lesions usually are greater than 4 cm in size.

Diagnosis is usually made on clinical examination; however skin biopsy is helpful in difficult cases. Histopathologic findings of guttate psoriasis vary with the age of the lesion. Findings in early lesions may be nonspecific and may include mild acanthosis, papillary dermal oedema and lymphocyte-predominant dermal infiltrate. Mature lesions exhibit parakeratosis alternating with hyperkeratosis, epidermal acanthosis, Munro microabscesses and dermal perivascular infiltrate containg neutrophils, lymphocytes and macrophages.

Streptococcal infections are well known to precipitate guttate psoriasis,1 however there have been no significant improvements in patients who were given penicillin or erythromycin when compared to those who were not treated.2 Other known precipitants are physical and psychological trauma.

The exact pathophysiologic mechanism is undetermined. The disease is believed to result from an immune reaction triggered by a previous streptococcal infection in a genetically susceptible host. Recent research points toward chromosome 6 as HLA-Cw*0602 allele positive patients are more prone to develop the guttate form.

Acknowledgements / Conflicts / Author Details
Competing Interests: 
None declared
Details of Authors: 
NAUMAN SHAHID, MD; MUHAMMAD Z BAWANY, MD; EHSAN RAFIQ, MD; THOMAS SODEMAN, MD, FACP; Internal Medicine department, University of Toledo Medical Center, Toledo, Ohio.
Corresponding Author Details: 
Nauman Shahid, 200 High Park Avenue, Goshen, IN 46526.
Corresponding Author Email: 
nshahid1980@gmail.com
References
References: 
  1. The role of streptococcal infection in initiation of Guttate Psoriasis ., Telfer ,Chalmer . Arch Dermatol 1992 Jan ;128 : 39-42   (Telfer NR, Chalmers RJG, Whale K, Colman G. The role of streptococcal infection in the initiation of guttate psoriasis. Arch Dermatol. 1992;128:39-42)
  2. Antistreptococcal treatment of guttate psoriasis: a controlled study.Dogan B, Karabudak O, Harmanyeri Y. Int J Dermatol. 2008 Sep;47(9):950-2.

Atopic Dermatitis for Family Physicians

Authors
Aly Khanbhai and Daljit Singh Sura
Article Citation and PDF Link
BJMP 2013;6(3):a626

Introduction

Atopic dermatitis (AD), also known as atopic eczema is a chronic, relapsing, inflammatory skin disease that can cause significant physical, psychological and social stress for patients and their families.1 This article focuses primarily on AD in adults. It is the most frequent inflammatory skin disease in the western world and is often characterized by chronic inflammation and pruritus interrupted by acute flares and bacterial infection.2,3 The majority of the care of AD is provided in primary care, with a minority of patients being referred to secondary care. There are currently extensive cost implications to the National Health Service (NHS) for both treating patients and for lost working days.4 AD can be a therapeutic challenge, especially in primary care, and there appears to be a great potential for improving the outcome and cost effectiveness of treatment in the community setting.4

Epidemiology and pathogenesis

AD typically begins in young infants or early childhood and subsides spontaneously by adolescence in approximately 90% of patients although it can persist into adulthood in about 10% of patients.5 The incidence of AD is generally considered to be increasing worldwide.6 AD affects both sexes equally, and in the United kingdom (UK) approximately 15-20% of school-aged children and 2-10% of adults will be affected by the condition at some stage.7

AD appears to result from a complex interplay between defects in skin barrier function, environmental agents, modified immune responses of the immune system to exogenous and endogenous factors, IgE-mediated mechanisms and other factors. However, the pathogenesis leading to the precise manifestation of AD is not completely understood.3,8

Diagnosis

There are no laboratory or diagnostic tests for AD. The diagnosis is based on visual assessment and clinical history. The UK diagnostic criteria (Table1) has been shown to be the most extensively validated for AD in comparison to the Hanifin and Rajka criteria, Schulz-Larsen criteria, Diepgen criteria, and Kang and Tian diagnostic criteria. Although several different diagnostic criteria have been developed they should mainly be used for research purposes as opposed to daily clinical management.4

Skin tests and laboratory investigations (specific IgE) may be helpful in the investigation of provocative factors such as food or environmental allergens. It is important to note that laboratory investigations should be interpreted in the context of the patient’s history.9

It is often difficult to differentiate AD from other skin conditions (e.g. seborrhoeic dermatitis, contact dermatitis, psoriasis, scabies). However, a family history of atopy and the clinical distribution of the lesions are helpful in making the diagnosis. Other conditions that need to be considered in the differential diagnosis of AD are metabolic and nutritional deficiencies, malignancies and immunodeficiency syndromes that present with skin manifestations.8

Table 1 – UK Working Party Diagnostic Criteria4

The patient must report an itchy skin condition (or parental report of scratching or rubbing in a child) in the last 12 months, plus three or more of the following:
  1. History of involvement of the skin creases (front of elbows, behind knees, fronts of ankles, around neck or around eyes)
  1. Personal history of asthma or hay fever (or history of atopic disease in first degree relative if child aged under four years)
  1. A history of generally dry skin in the last year
  1. Onset under the age of two years (not used if child aged under 4 years)
  1. Visible flexural dermatitis (including dermatitis affecting cheeks or forehead and outer aspects of limbs in children under four years)

Management

The management of AD should involve a combination that includes short-term treatment of flares and a long-term maintenance approach to prevent flares. For patients with mild to moderate AD, topical therapy may be sufficient to control the condition. Patients with more severe disease may require advanced therapy such phototherapy or systemic therapy.

Education

For optimal disease management, patients and/or their carers should be educated about the nature of AD, the need for continued adherence to prescribed treatment and about the appropriate use of topical therapies. Time spent educating patients and carers have been shown to have a positive effect on disease outcomes. Patients may also benefit from written information to reinforce learning.8

Emollients

Emollients soften the skin, aid in restoring the impaired barrier function, reduce itching, prevent moisture from leaving the skin and increase the efficacy of topical corticosteroids (TCS). They also replace the natural surface oils that are essential to preventing irritant materials, infection and allergy-inducing substances from entering the skin.4

Healthcare professionals should offer a range of emollients, and prescriptions should be reviewed frequently. To optimise adherence to emollient therapy, creams, lotions, and ointments, or a combination can be used depending on patient preference. Continued use of emollients during periods of disease quiescence can reduce the likelihood for exacerbations.10

When the treatment regimen involves both an emollient and TCS, there is no evidence on which to base the order of application. Patients should be advised to apply emollients liberally and frequently (at least 2-4 times a day). It is especially important to use emollients during or after bathing. The emollient should be applied in the general direction of growth of body hair in order to prevent accumulation at hair bases which might predispose to folliculitis. Emollients can become contaminated with bacteria and the use of pump dispensers minimises this risk. If the emollient is in a pot it should be removed with a clean spoon or spatula.4

Topical corticosteroid therapy (TCS)

TCS is considered first-line therapy for AD flares. Available preparations include ointments, creams, gels, lotions, liquids, and foams. Ointments and creams are generally the most effective in treating AD as they tend to be more moisturising.10 The least potent preparation required to control AD, particularly in sensitive areas, should be utilised. When possible the TCS should be stopped for short periods to reduce the risk of adverse events.8

TCS is categorised into four groups according to potency: mild, moderately potent, potent and very potent. The choice of TCS potency should be tailored to the age of the patient, the body region being treated, and the severity of inflammation. Patients should be advised to apply TCS once daily. If there is an inadequate response to once daily application, the frequency should be increased to twice daily.4 Once control has been achieved, twice weekly maintenance therapy with a TCS should be considered in patients with moderate to severe AD experiencing frequent relapses. The local adverse effects of TCS usage include skin thinning, bruising, perioral dermatitis, folliculitis, pruritus, allergic contact dermatitis and the spread of fungal infection. Patients being treated with intermittent courses of TCS should be reviewed regularly (depending on TCS potency and site of application) to determine response to therapy and assess skin for potentially reversible atrophic changes.4

Table 2 - The ‘fingertip unit’ (FTU) is a method of determining the amount of TCS to apply. It is described as “the amount of ointment expressed from a tube with a 5 mm diameter nozzle, applied from the distal skin crease to the tip of the palmar aspect of the index finger.” The following table is a guide to the use of FTU in adults.4

Skin area FTU per dose
Face and neck 2.5
Torso and abdomen 7
Back and buttocks 7
Entire arm and hand 4
A hand and fingers (front and back) 1
Entire leg and foot 8

Topical calcineurin inhibitors (TCIs)

TCIs are non-steroidal immunomodulating agents licensed for the treatment of AD.4 TCIs work by inhibiting the phosphatase activity of calcineurin to block expression of cytokines and are thought to represent a more targeted way to limit inflammation and avoid many of the adverse effects of TCS. TCIs may be used either as monotherapy, as a combination or as sequential therapy. TCS are generally less expensive and more effective than TCIs although individual clinical situations will arise in which TCIs are preferred.10

Two TCIs are available: tacrolimus (0.03% and 0.1% ointments) and 1% pimecrolimus cream. The tacrolimus 0.03% and 0.1% ointments are both licensed for moderate to severe eczema and the 0.03% ointment is licensed for use in children aged two years and over. The 1% pimecrolimus cream is licensed for mild to moderate eczema in patients aged two years and over. The use of tacrolimus should be limited to doctors with a specialist interest and experience in treating AD.4

TCIs should not be used as first line treatment unless there is a specific reason to avoid the use of TCS.4 Given the high cost of TCIs, and the fact that their long-term safety is not fully known, they are generally reserved for patients with persistent disease or frequent flares that would require continuous TCS treatment. They are also of use in patients with severe disease in sensitive skin areas (e.g. around the eyes, face, neck and genitals) where systemic absorption and the risk of skin atrophy with TCS are of concern. Considering the possibility that the normal immunological response to infection may be suppressed, TCIs should not be applied to skin which appears actively infected.4, 8

Dressings and wet wrap treatment

Patients with non-infected moderate to severe AD can be advised to cover affected areas with dry wrap dressings to provide a physical barrier to scratching and improve the retention of emollients. Wet wrapping generally consists of two layers of bandage applied over topical preparations. The bottom layer is soaked in warm water, squeezed out and then put onto the skin over the topical preparation. The top layer is dry. Wet wraps can be worn under nightwear or ordinary clothes and used during the day or night. They are available in bandage form or as garments.4 Disadvantages include a high cost, inconvenience, a need for specialised training, and an increased potential for adverse effects from occluded corticosteroids (such as systemic absorption, atrophy, striae), and increased incidence of skin infection.10 There is currently insufficient consistent evidence on which to base a recommendation for wet wrap use in the primary care setting.4

Antimicrobial measures

Skin lesions of around 90% of patients with AD are colonised compared to less than 5% of individuals with healthy skin. Staphylococci are the main organisms isolated although other organisms such as streptococci may also cause infection. The routine swabbing of skin is not indicated although swabs of potential Staphylococcus aureus carriage sites should be considered in patients with recurrent infection. Oral antibiotics are not recommended in the routine treatment of non-infected AD but patients with clinically infected AD can be prescribed short term oral antibiotic treatment based on local/regional antibiotic sensitivities. However, first- or second-generation cephalosporins or penicillins for 7 to 10 days are usually effective in managing bacterial infection. Macrolides are less useful alternatives due to resistant patterns in patients with AD. Patients with AD are also prone to recurrent viral infections. Eczema herpeticum is a severe disseminated herpes infection that is a serious risk in patients with widespread AD and may be misdiagnosed as a bacterial infection. Patients with eczema herpeticum normally require systemic antiviral treatment4, 10.

Antihistamines

Although first-generation antihistamines do not directly affect the itching associated with AD, the sedative effects have been found to help improve sleep. Therefore, short-term bedtime use of sedating antihistamines should be considered in patients with AD where there is debilitating sleep disturbance. Daytime use of first generation antihistamines should be avoided given their sedative effects. Non-sedating antihistamines appear to have limited value in patients with AD but they may provide some benefit in patients with allergic triggers.4, 8

Dietary interventions

Although there is an association between IgE mediated food allergy and AD severity in infants, it is unclear whether hypersensitivity to food is a major factor in causing and maintaining AD. Dietary exclusion is not recommended for managing AD in patients without confirmed food allergy. The exclusion of foods during pregnancy and breast feeding to prevent the development of AD in infants is not recommended. Breast feeding for three months or more may help prevent the development of infant eczema where there is a family history of atopy. However, current UK guidelines state that weaning should start at six months.4

Table 3 - Guidance on when to refer to secondary care4

Eczema herpeticum (widespread herpes simplex) – emergency referral
Uncertainty concerning the diagnosis
Poor control of the condition or failure to respond to appropriate topical treatments
Psychological upset or sleep problems
Recurrent secondary infection

Other therapies

Systemic corticosteroids are usually reserved for the acute treatment of severe AD exacerbations. Prolonged use of oral steroids is associated with potentially serious adverse effects and their long-term use should be avoided. Furthermore, relapses are common following discontinuation of oral corticosteroid therapy.8

Ultraviolet (UV) phototherapy may also be beneficial for the treatment of AD in adults. In addition, systemic therapies are available and may be broadly classified into traditional medications (e.g. cyclosporine, azathrioprine, methotrexate) and biologic agents (targeted monoclonal antibodies). These options are available for severe, refractory AD. These therapeutic options should generally be reserved for unique situations and require consultation with a dermatologist. These therapeutic options are beyond the scope of this article.8

Acknowledgements / Conflicts / Author Details
Competing Interests: 
None declared
Details of Authors: 
ALY KHANBHAI, MB ChB, DRCOG, GP VTS ST2 Trainee'Queen's Hospital, Rom Valley Way, Romford, Essex, RM7 0AG, UK. DALJIT SINGH SURA, MBBS BSc DRCOG DFSRH MRCGP, General Practitioner North Street Medical Care / Lawns Medical Care, Romford, Essex, RM1 4QJ, UK.
Corresponding Author Details: 
Dr Aly Khanbhai MB ChB DRCOG Queen's Hospital, Rom Valley Way, Romford, Essex, United Kingdom RM7 0AG.
Corresponding Author Email: 
ali.dh.kh@gmail.com
References
References: 
  1. Gelbard CM, Hebert AA. New and emerging trends in the treatment of atopic dermatitis. Patient Preference and Adherence 2008:2 387–392
  2. Ong PY, Leung DYM. The Infectious Aspects of Atopic Dermatitis. Immunology and Allergy Clinics of North America 2010 August; 30(3): 309–321
  3. Novaka N, Leung DYM. Advances in Atopic dermatitis. Current Opinion in Immunology 2011 December; 23(6): 778–783
  4. SIGN. Scottish Intercollegiate Guidelines Network. Management of atopic eczema in primary care. A national clinical guideline. March 2011Scottish Intercollegiate Guidelines Network
  5. Park MK, Park KY, Li K, et al. The Short Stature in Atopic Dermatitis Patients: Are Atopic Children Really Small for Their Age? Annals of Dermatology Vol. 25, No. 1, 2013
  6. Furue M, Chiba T, Takeuchi S. Current status of atopic dermatitis in Japan.Asian Pacific Journal of Allergy and Immunology 2011; 1: 64-72
  7. Green C, Colquitt JL, Kirby J, et al. Clinical and cost-effectiveness of once-daily versus more frequent use of same potency topical corticosteroids for atopic eczema: a systematic review and economic evaluation. Southampton, UK: Health Technology Assessment; 2004
  8. Watson W, Kapur S. Atopic dermatitis. Allergy, Asthma & Clinical Immunology 2011, 7(1):S4
  9. Bieber T. Atopic Dermatitis. Annals of dermatology 2010, Vol. 22, No. 2
  10. Walling HW, Swick BL. Update on the management of chronic eczema: new approaches and emerging treatment options. Clinical, Cosmetic and Investigational Dermatology. July 2010

Management of alopecia areata: an update

Authors
Imran Majid and Abid Keen
Article Citation and PDF Link
BJMP 2012;5(3):a530
Abstract / Summary
Abstract: 

Alopecia areata is a common, non-scarring, autoimmune disorder affecting any hair-bearing area. It is often psychologically devastating. This disorder occurs in both the sexes, in all age groups, and is characterized by the sudden appearance of circumscribed areas of hair loss on the scalp or other parts of the body. Various therapeutic approaches are presently available for managing alopecia areata including corticosteroids, contact sensitizers and immunosuppressants, but none have been shown to alter the course of the disease on a consistent basis.

Keywords: 
Alopecia areata, treatment, autoimmune, corticosteroids, recent advances, contact sensitizers

Introduction

Alopecia areata is a non-scarring autoimmune, inflammatory hair loss affecting the scalp and/or body. Although the etiopathogenesis of alopecia areata is still unknown, the most widely accepted hypothesis is that it is a T-cell mediated autoimmune condition that occurs in genetically predisposed individuals. The term ‘alopecia areata’ was first used for this disorder by Savages1.Alopecia areata has a reported incidence of 0.1-0.2%, with a life-time risk of 1.7%2-4. The disease can begin at any age, but the peak incidence is between 20 and 50 years of age5. Both the sexes are equally affected and there is no racial variation reportedClinically, alopecia areata may present as a single well demarcated patch of hair loss, multiple patches, or extensive hair loss in the form of total loss of scalp hair (alopecia totalis) or loss of entire scalp and body hair (alopecia universalis). Histopathologically, alopecia areata is characterized by an increase in the number of catagen and telogen follicles and the presence of perifollicular lymphocytic infiltrate around the anagen phase hair follicles. The condition is thought to be self-limited in majority of cases, but in some the disease has a progressive course and needs active treatment in the form of oral or topical therapeutic options. Progressive alopecia areata is associated with severe social and emotional impact.

Clinical features

Alopecia areata mostly presents as a sudden loss of hair in well demarcated localized areas. The lesion is usually a round or oval flat patch of alopecia with normal skin colour and texture involving the scalp or any other region of the body. The patch of alopecia may be isolated or there may be numerous patches. It usually has a distinctive border where normal hair demarcates the periphery of the lesion. In acute phases, the lesions can be slightly erythematous and oedematous.

The patches of alopecia areata are usually asymptomatic, although several patients may sometimes complain of local paraesthesia, pruritus or pain. The affected hairs undergo an abrupt conversion from anagen to telogen, clinically seen as localized shedding. Characteristic hairs, known as ‘exclamation point hairs’ may be seen within or around the areas of alopecia. The hairs are tapered towards the scalp end with thickening at the distal end. These hairs may also demonstrate deposition of melanin pigment in the distal extremity, also known as Wildy’s sign. Although not absolutely pathognomonic, it strongly suggests the diagnosis of alopecia areata. Hair pull test conducted at the periphery of the lesion may be positively correlated (six or more) with disease activity. In the chronic phases, the test is negative, since the hair is not plucked as easily as in the acute phases.

Another important clinical sign that can aid in the diagnosis is the presence of ‘cadaverous hair’. These are the hairs in which there occurs a fracture of the shaft inside the hair follicle, producing blackened points inside the follicular ostia resembling comedones. In alopecia areata, the hair loss progresses in a circumferential pattern. Often, distinct patches merge to form large patches. Upon regrowth, hairs will often initially lack pigment resulting in blonde or white hairs7.

Extrafollicular involvement in alopecia areata:

a) Nail changes: Nail changes are more frequent in children (12%) than in adults (3.3%)8.The prevalence of nail changes is greater in the more severe forms of alopecia areata such as alopecia universalis and alopecia totalisFinger nails are more commonly involved than the toe nails. Pitting is the most common finding. Other nail changes include koilonychias, onycholysis, onychomadesis, punctuate leukonychia, trachyonychia, Beau’s lines and red lunulae8-11.

b) Ocular changes: Various ocular changes have been reported to occur in alopecia areata. These include focal hypopigmentation of the retina12, lens opacities, posterior subcapsular cataracts13 decrease in visual acuity, Horner’s syndrome, heterochromia of the iris14, miosis and palpebral ptosis.

Treatment of alopecia areata

Treatment of alopecia areata is not mandatory in every affected patient because the condition is benign in majority and spontaneous remission is common. Treatment is mainly directed towards halting the disease activity as there is no evidence that the treatment modalities influence the ultimate natural course of the disease. Treatment modalities are usually tailored as per the extent of hair loss and the patient’s age. Addressing the impressive inflammatory process occurring in alopecia areata, corticosteroids have by far been the most commonly used treatment modality-16Few treatments have been subjected to randomized control trials and except for contact immunotherapy, there is a paucity of published data on their long term outcomes. Currently, new treatments targeting the immune system are being explored for the use in alopecia areata.

Topical treatments

Topical steroids

Intralesional steroid injections

Topical contact sensitizers

Anthralin

Minoxidil

Topical retinoids

Tacrolimus

Systemic treatments

Systemic corticosteroids

Sulfasalazine

Azathioprine

Methotrexate

Oral zinc sulphate

Photo-and photochemotherapy

PUVA

NBUVB

Excimer laser

Miscellaneous and Non-pharmacological treatment

Dermatography, wigs

Hypnotherapy etc

Topical treatment options

Topical corticosteroids:

Several topical corticosteroids with varying levels of efficacy have been used to treat alopecia areata. These include fluocinolone acetonide cream17, fluocinolone scalp gel, betamethasone valerate lotion18, clobetasol propionate ointment19, dexamethasone in a penetration-enhancing vehicle and halcinonide cream20. They are a good option in children because of their painless application and wide safety margin21. Topical corticosteroids are ineffective in alopecia totalis/universalisFolliculitis is a common side effect of corticosteroid treatment, appearing after a few weeks of treatment. Telangiectasia and local atrophy have also been reported. Treatment must be continued for a minimum of 3 months before regrowth can be expected and maintenance therapy often is sometimes necessary.

Intralesional corticosteroids:

Intralesional corticosteroids are widely used in the treatment of alopecia areata. In fact, they are the first-line treatment in localized conditions involving <50% of the scalp22. Hydrocortisone acetate (25mg/ml) and Triamcinolone acetonide (5-10mg/ml) are commonly used. Triamcinolone acetonide is administered usually in the concentration of 5mg/ml using a 0.5 inch long 30-gauge needle in multiple 0.1 ml injections approximately 1 cm apart22-23. The solution is injected in or just beneath the dermis and a maximum of 3 ml on the scalp in one visit is recommended23. Lower concentrations of 2.5mg/ml are used for eyebrows and face. Regrowth usually is seen within 4-6 weeks in responsive patients. Treatments are repeated every 3-6 weeks. Skin atrophy at the sites of injection is a common side effect, particularly if triamcinolone is used, but this usually resolves after a few months. Repeated injections at the same site or the use of higher concentrations of triamcinolone should be avoided as this may lead to prolonged skin atrophyPain limits the practicality of this treatment method in children who are less than 10 years of age. Severe cases of alopecia areata, alopecia totalis, alopecia universalis as well as rapidly progressive alopecia areata respond poorly to this form of treatment25.

Anthralin:

Dithranol (anthralin) or other irritants have been used in the treatment of alopecia areata. The exact mechanism of action is unknown, but is believed to be through immunosuppressant and anti-inflammatory properties with the generation of free radicals. It is used at concentrations ranging from 0.5 to 1 % for 20-30 minutes after which the scalp should be washed with shampoos in order to avoid excessive irritant effects. The applications are made initially every other day and later on daily. Adverse effects include pruritus, erythema, scaling, staining of treated skin and fabrics, folliculitis, and regional lymphadenopathy26-27. In an open study, 25% patients with severe alopecia areata were shown to respond positively to local applications of 0.5-1% anthralinMore placebo control studies are needed to justify the use of anthralin in alopecia areata.

Minoxidil:

Minoxidil appears to be effective in the treatment of alopecia areata. It’s mechanism of action has yet to be determined, but it is known to stimulate DNA synthesis in hair follicles and has a direct action on the proliferation and differentiation of the keratinocytes28. In one clinical study, hair growth was demonstrated in 38% and 81% of patients treated with 1% and 5% minoxidil respectively. Thus 5% minoxidil solution is usually recommended as a treatment option in alopecia areata. No more than 25 drops are applied twice per day regardless of the extent of the affected area. Initial regrowth can be seen within 3 months, but continued application is needed to achieve cosmetically acceptable regrowth. Minoxidil has also been studied in combination with anthralin29, topical betamethasone propionate30 and prednisolone31. Minoxidil is of little benefit to patients of severe alopecia areata, alopecia totalis or alopecia universalisThe possible side effects from minoxidil are allergic and irritant contact dermatitis and hypertrichosis which is usually reversible with the interruption of the treatment.

Topical immunotherapy:

Topical immunotherapy is the best documented treatment so far for severe and refractory cases of alopecia areata. Topical immunotherapy is defined as the induction and periodic elicitation of allergic contact dermatitis by applying a potent contact allergen33. In 1965, the alkylating agent triethyleneimino benzoquinone was the first topical sensitizer used to treat cutaneous disease, but it was abandoned on account of its mutagenic potential. Later nitrogen mustard, poison ivy, nickel, formalin, and primin were tried, mainly as topical immunotherapy, for alopecia areata and warts. Contact immunotherapy was introduced in 1976, by Rosenberge and Drake. Later, potent contact allergens namely dinitrochlorobenzene (DNCB) and diphenylcyclopropenone (DPCP) replaced the allergens that were used earlier33. DNCB is mutagenic against Salmonella tymphimurium in the Ames test and is no longer usedNeither SADBE, nor DPCP are mutagenic. DPCP is more stable in solution and is usually the agent of choice.

Mechanism of action: Topical immunotherapy acts by varied mechanisms of action. The most important mechanism is a decrease in CD4 to CD8 lymphocyte ratio which changes from 4:1 to 1:1 after contact immunotherapy. A decrease in the intra-bulbar CD6 lymphocytes and Langerhan cells is also noted. Happle et al, proposed the concept of ‘antigenic competition’, where an allergic reaction generates suppressor T cells that non-specifically inhibit the autoimmune reaction against a hair follicle constituent. Expression of class I and III MHC molecules, which are normally increased in areas affected by alopecia areata disappear after topical immunotherapy treatment34.A ‘cytokine inhibitor’ theory has also been postulated34.

Method of sensitization: The protocol for contact immunotherapy was first described by Happle et al in 1983 The scalp is the usual sensitization site. For the initial sensitization a cotton-tipped applicator saturated with 2% DPCP in acetone is applied to a small area. Patients are advised to avoid washing the area and protect it from sunlight for 48 hours. After 2 weeks 0.001% solution of DPCP is applied on the scalp and then the application of contact allergen is repeated weekly with increasing concentrations. The usual concentration of DPCP that ultimately causes mild contact eczema is 0.01-0.1% and this is repeated weekly till a response is seen. An eczematous response indicates that sensitization has taken place. Only 1-2% of the patients fail to sensitize. It is important to remember that DPCP is degraded by light and should thus be stored in the dark and the patient should also wear a wig or hat during the day after application of DPCP. DPCP immunotherapy has even been combined with oral fexofenadine treatment with good effect36.

Evaluation of efficacy: The clinical response after six months of treatment is rated as per the grading system proposed by Mcdonald Hull and Norris37:

Grade 1- Regrowth of vellus hair.

Grade 2- Regrowth of sparse pigmented terminal hair.

Grade 3- Regrowth of terminal hair with patches of alopecia.

Grade 4- Regrowth of terminal hair on scalp.

If no regrowth is observed within six months of treatment, the patient is considered to be a non-responder. Evaluation of plucked hair is done using light microscopy, for evaluation of anagen/telogen ratio.

A review of most of the published studies of contact immunotherapy concluded that 50-60% of patients achieve a worthwhile response but the range of response rates was very wide (9-87%)Patients with extensive hair loss are less likely to respond. Other reported poor prognostic factors include the presence of nail changes, early onset disease and a positive family history39.

Topical immunotherapy can lead to certain side effects such as persistent dermatitis, painfull cervical lymphadenopathy, generalized eczema, blistering, contact leukoderma, and urticarial reaction. Systemic manifestations such as fever, arthralgia and yellowish discoloration of hair are noted more often with DNCB.

In poor responders to DPCP, squaric acid dibutylester (SADBE) can be tried as a contact sensitizer. The method of application is the same as with DPCP but the applications are done once or twice weekly40.

Good care should be taken to avoid contact with the allergen by handlers, including pharmacy and nursing staff. Those applying the antigen should wear gloves and aprons. There is no available data on the safety of contact immunotherapy during pregnancy and it should not be used in pregnant women or in women intending to become pregnant.

Tacrolimus:

Tacrolimus is a topical calcineurin inhibitor that inhibits transcription following T-cell activation of several cytokines including IL-2, IFN-gamma and TNF-α. Yamamoto et al reported in their findings that tacrolimus stimulated hair growth in mice41, although subsequent studies have shown conflicting resultsRecently, Price et al reported an 11-patient study in which none of the patients had terminal hair growth in response to tacrolimus ointment 0.1 % applied twice daily for 24 weeks43.

Topical garlic

Garlic is a very commonly used home remedy in the treatment of alopecia areata in India and even in the rest of the world. One study analyzed the effect of a combination of topical garlic gel and betamethasone valerate ointment in alopecia areata in a double-blind study. The study found the combination useful in majority of the patients with a statistically significant difference between the treatment and control groups44.

Topical retinoids:

Among topical retinoids, tretinoin and bexarotene have been tried in alopecia areata with mixed results-46Irritation of the skin is a very common side effect and the efficacy is doubtful in the absence of double-blind randomized trials.

Prostaglandin analogs:

The propensity of certain prostaglandin analogues used as anti-glaucoma eye drops to cause hypertrichosis has been employed in the treatment of alopecia areata. These prostaglandin analogues include Latanoprost and Bimatoprost and they are used in the treatment of alopecia areata involving the eyelashes-48However, the results obtained with these drugs have not been really encouraging49.

Systemic treatments

Systemic treatments, as a rule, are used only in progressive forms of alopecia areata and going by the immune nature of the disease, majority of these treatment options are immunosuppressants or immunomodulators in nature.

Systemic corticosteroids:

The use of systemic corticosteroids for the treatment of alopecia areata is under much debate. Some authors support a beneficial role of systemic steroids on halting the progression of alopecia areata, but many others have had poor results with this form of therapy. The suggested dosages are 0.5-1mg/kg/day for adults and 0.1-1 mg/kg/day for children50. Treatment course ranges from 1-6 months, but prolonged courses should be avoided to prevent the side effects of corticosteroids. Side effects profile of corticosteroids in conjunction with the long-term treatment requirements and high relapse rates make systemic corticosteroids a more limited option. In addition to the daily oral administration of corticosteroids, there are several reports of high-dose pulsed corticosteroid treatments employing different oral and intravenous regimens51-53. Many of these regimens have been tried in alopecia areata with encouraging results but the majority of these studies have been non-blind open studies. One such pulsed administration employs a high dose oral corticosteroid on two consecutive days every week with a gap of 5 days between the two pulses. This modality of treatment is known as oral minipulse therapy (OMP) and it has been tried in many skin diseases in addition to alopecia areata like vitligo54-55 and lichen planusSome open label studies on corticosteroid OMP therapy have reported encouraging results in alopecia areata53.

Sulfasalazine:

Because of its immunomodulatory and immunosuppressive actions, sulfasalazine has shown good hair regrowth in the treatment of alopecia areata. The drug is administered orally usually as enteric coated tablets to minimize the gastrointestinal side effects. The treatment is started at a lower dose, usually in the range of 500 mg twice daily and then the dose is gradually increased to 1 g three times a dayAdverse effects include gastrointestinal distress, liver toxicity and haemotological side effects. Sulfasalazine helps in alopecia areata because it causes inhibition of T cell proliferation, and natural killer cell activity and also inhibits antibody production. It also inhibits the secretion of interleukin (IL)-2, IL-1, TNF- and IFN-gamma and even IL-667.

A number of clinical studies have documented a positive effect of sulfasalazine in alopecia areata. In one clinical study, 23% patients showed a really good response with satisfactory hair growth after sulfasalazine therapyOther studies have also shown a beneficial effect of this treatment option in resistant cases of alopecia areata66,69.

Azathioprine:

Azathioprine, being an immunosuppressive agent has also been tried in alopecia areata. The drug is used in many cutaneous disorders owing to its effect on circulating lymphocytes as well as Langerhan cells. In a limited study on 20 patients hair regrowth was demonstrated in about half of the patients with a dosage regimen of 2g/day70.

Cyclosporine:

This drug has proven effective in the treatment of alopecia areata because of its immunosuppressive and hypertrichotic properties. The side effect profile and high rate of recurrence render the drug a poor choice for the use in alopecia areata. So the drug is to be attempted only in severe forms of alopecia areata not responding to treatment71.

Methotrexate:

Methotrexate either alone or in combination with prednisolone has been used in the treatment of alopecia areata in various studies with variable success rates72.

Oral zinc sulphate

Serum zinc levels have been found to be lower in patients with alopecia areata than in control populationIn a study on 15 patients, hair regrowth was observed in 9 patients (67%) after oral zinc gluconate administration74.

Biological agents:

Tumour necrosis factor inhibitors such as Adalimumab, Infliximab and Etanercept have been tried in alopecia areata, but the results have not been encouraging-76Clinical trials conducted till now have failed to demonstrate the efficacy of any biological agent in alopecia areata.

Photo-and photochemotherapy

Photochemotherapy:

Several uncontrolled studies regarding PUVA therapy for the treatment of alopecia areata exist. All types of PUVA (oral PUVA, topical PUVA, local or whole body UVA irradiation) have been used with success rates of up to 60-65%57-59. The mechanism of action is considered to be the interference in the presentation of follicular antigens to T-lymphocytes by depletion of the Langerhan cells. The relapse rate following treatment is high, sometimes demanding repeated treatments for a prolonged period with implications for carcinogenic risks60. To mitigate the side effects of systemic psoralens, PUVA-turban therapy is used for alopecia areata involving the scalp. In this form of photochemotherapy, very dilute solutions of 8-methoxy psoralen are applied on the scalp by utilizing a cotton towel as a turban. The patient’s scalp is exposed to UVA after keeping the ‘turban’ in contact with the scalp for about 20 minutesThe efficacy of this form of PUVA therapy has been seen to be about 70%61.

Phototherapy

Although narrowband UVB is among the most effective treatment options in a number of immune mediated skin diseases, the same efficacy has not been found in alopecia areata. Properly designed randomized trials are needed to elucidate whether NBUVB has any role in the management of alopecia areata62-63.

Excimer laser and excimer light

Excimer laser and excimer light are two more recent additions to the phototherapeutic armamentarium for many skin and hair disorders. While the main use of these phototherapeutic modalities remains to be psoriasis and vitiligo, their immunomodulatory effect can be made use of in many other skin disorders. Some clinical studies have documented the efficacy of excimer laser and excimer light in alopecia areata64-65. In one such study, 41.5% patches were shown to respond to excimer laser therapy administered over 12 weeks64. Another study on childhood alopecia areata found regrowth in 60% lesions after a treatment period of 12 weeksThe treatment is well tolerated with erythema of the skin as the only adverse effect reported.

Miscellaneous therapies

Various non-conventional therapeutic agents have been used in alopecia areata with some degrees of success. These include fractional Er-Glass laser77, topical azelaic acid78, topical onion juice79, topical 5-fluorouracil ointment80 and photodynamic therapyThe efficacy and safety of these therapeutic agents need to be confirmed in large-scale, double-blind, placebo-controlled trials before they can be recommended for treatment of alopecia areata.

Non-pharmacological methods

Cosmetic treatments for patients with alopecia areata include the following:

a) Dermatography: It has been used to camouflage eyebrows of patients with alopecia areata. In this treatment tiny pigment dots of pigment are used on the skin on the region of the eyebrows to mask the underlying alopecia81.

b) Wigs or Hair pieces: These are useful for patients with extensive disease and allow them to carry on their usual social life.

Conclusion:

Alopecia areata is now regarded as an autoimmune disease involving the cellular immunity through the CD8 lymphocytes that act on follicular antigens. The pathogenesis of alopecia areata is being unravelled with various animal and human studies.

The localized forms often heal spontaneously or respond to simple treatments such as topical or intralesional corticosteroids. The severe forms have a reserved prognosis and are difficult to treat. In these cases the best results are achieved by topical immunotherapy technique.

Acknowledgements / Conflicts / Author Details
Competing Interests: 
None declared
Details of Authors: 
IMRAN MAJID, MD Dermatology and STD, Assistant Professor Dermatology, Govt Medical College, Srinagar, Kashmir, India. ABID KEEN, Postgraduate student Dermatology, Govt Medical College, Srinagar, Kashmir, India.
Corresponding Author Details: 
Dr IMRAN MAJID, CUTIS Skin and Laser Institute, Srinagar, Kashmir, India 190002.
Corresponding Author Email: 
imran54@yahoo.
References
References: 
  1. Dawber R. Alopecia areata. Monogr Dermatol 1989;2:89-102.
  2. Safavi K. Prevelance of alopecia areata in the First National Health and Nutrition Examination Survey. Arch Dermatol 1992;128:702.
  3. Safavi KH, Muller SA, Suman VJ, Moshell AN, Melton LJ 3rd. Incidence of alopecia areata in Olmsted Country,Minnesota, 1975 through 1989. Mayo Clin Proc 1995;70:628-33.
  4. Muller SA, Winkelmann RK, Alopecia areata. An evaluation of 736 patients. Arch Dermatol 1963;88:290-7.
  5. Sharma VK, Dawn G, Kumar B. Profile of alopecia areata inNorthern India. Int J Dermatol 1996;35:22-7.
  6. Dawber RPR, de Berker D, Wojnarowska F. Disorders of hair. In: Champion RH, Burton JL, Burns DA, Breathnach SM, editors. Textbook of Dermatology.Oxford: Blackwell Science; . P. 2919-27.
  7. Cline DJ. Changes in hair color. Dermatol Clin 1988;6:295-303.
  8. Tosti A, Morelli R, Bardazzi Peluso AM. Prevalence of nail abnormalities in children with alopecia areata. Padiatr Dermatol 1994;11:112-5.
  9. Tosti A, Fanti PA, Morelli R, Bardazzi F. Trachyonychia associated with alopecia areata: a clinical and pathological study. J Am Acad Dermatol 1991;25:266-70.
  10. Bergner T, Donhauser G, Ruzicka T. Red lunula in severe alopecia areata. Acta Dermato Venereol (Stockh) 1992;72:203-5.
  11. Sahn EE. Alopecia areata in childhood. Semin Dermatol 1995;14:9-14.
  12. Tosti A, Colombati S, Caponeri GM, Ciliberti C, Tosti G, Basi M, et al. Ocular abnormalities occurring with alopecia areata. Dermatologica 1985;170:69-73.
  13. Muller SA,Brunsting LA.Cataracts associated with dermatologic disorders. Arch Dermatol 1963;88:330-9.
  14. Hordinsky MA. Alopecia areata. In: Olsen EA, editor. Disorders of hair growth. Diagnosis and treatment.New York: MacGraw-Hill 1994. p.195-222.
  15. Alsantali A. Alopecia areata: a new treatment plan. Clin Cosmet Investig Dermatol 2011;4:107-15.
  16. Shimmer BP, Parker KL. Goodman’s and Gillman’s: The Pharmacological Basis of Therapeutics.New York: Mc Graw-Hill, 2001: 1661-1663.
  17. Pascher F, Kurtin S, Andrade R. Assay of 0.2 percent fluocinolone acetonide cream for alopecia areata and totalis: efficacy, side effects including histologic study ensuing localized acneform response. Dermatologica 1970; 141: 193-202.
  18. LeydenJL, Kligman AM. Treatment of alopecia areata with steroid solution. Arch Dermatol 1972;106:924.
  19. Tosti A, Piraccini BM, Pazzaglia M, et al. Clobetasol propionate 0.05% under occlusion in the treatment of alopecia totalis / universalis. J Am Acad Dermatol 2003;49:96-8.
  20. Montes LF. Topical halcinonide in alopecia areata and alopecia totalis. J Cutan Pathol 1997;4:47-50.
  21. Tan E,TayYK, Giam YCH. A clinical study of childhood alopecia areata inSingapore. Pediatr Dermatol 2002;19:298-301.
  22. Ross EK, Shapiro J. Management of hair loss. Dermatol Clin 2005;23:227-43.
  23. Madani S, Shapiro J. Alopecia areata update. J Am Acad Dermatol 2000;42:549-66.
  24. Whiting DA. The treatment of Alopecia Areata. Cutis 1987;40:247-50.
  25. Price VH. Treatment of hair loss. New Engl J Med 1999;341:964-73.
  26. Nelson DA, Spielvogel RL. Anthralin therapy for alopecia areata. Int J Dermatol 1985;24:606-7.
  27. Fiedler-Weiss VC, Buys CM. Evaluation of anthralin in the treatment of alopecia areata. Arch Dermatol 1987;123:1491-3.
  28. Buhl AE. Minoxidil's action in hair follicles. J Invest Dermatol 1991;96:73S-74S.
  29. Fiedler VC, Wendrow A, Szpunar GJ, et al. Treatment resistant alopecia areata. Response to combination therapy with minoxidil plus anthralin. Arch Dermatol 1990;126:756-9.
  30. Fiedler VC. Alopecia areata: Current therapy. J Invest Dernatol 1991;96:69S.
  31. Olsen EA,CarsonSC, Turney EA. Systemic steroids with or without 2% topical minoxidil in the treatment of alopecia areata. Arch Dermatol 1992;128:1467-73.
  32. Vanderveen EF, Ellis CN, Kang S, et al. Topical minoxidil for hair regrowth. J Am Acad Dermatol 1984;11:416-21.
  33.  Hoffmann R, Happle R. Topical immunotherapy in alopecia areata: What, how and why? Dermatol Clin 1996;14:739-44.
  34. Summer KH, Goggelman W. 1-chloro-2,4-dinitrobenzene depletes glutathione in rat skin and is mutagenic in Salmonella tymphimurium. Mutat Res 1980;77:91-3.
  35. Happle R, Hausen BM, Wiesner-Menzel L. Diphencyprone in the treatment of alopecia areata. Acta Derm Venereol 1983; 63:49–52.
  36. Inui S, Nakajima T, Toda N, Itami S. Fexofenadine hydrochloride enhances the efficacy of contact immunotherapy for extensive alopecia areata: Retrospective analysis of 121 cases.
  37. Contelessa C, Peris K, Caracciolo E, Mordenti C, Chimenti S. The use of topical diphenylcyclopropenone for the treatment of extensive alopecia areata. J Am Acad Dermatol 2001;44:73-6.
  38. Gordon PM,Aldrige RD, Mc Vittie E et al. Topical diphencyprone for alopecia areata: evaluation of 48 cases after 30 months follow-up. Br J Dermatol 1996;134:869-71.
  39. Van der Steen PH, Baar HM, Happle R et al. Prognostic factors in the treatment of alopecia areata with diphenylcyclopropenone. J Am Acad Dermatol 1991;24:227-30.
  40. Orecchia G, Malagoli P, Santagostino L. Treatment of severe alopecia areata with squaric acid dibutylester in pediatric patients. Pediatr Dermatol 1994;11:65-8.
  41. Yamamoto S, Jiang H, Kato R. Stimulation of hair growth by topical application of FK 506, a potent immunosuppressive agent. J Invest Dermatol 1994;102:160-4.
  42. Yamamoto S, Jiang H, Kato R. Induction of anagen in telogen mouse skin by topical application of FK 506, a potent immunosuppressant. J Invest Dermatol 1995;104:523-5.
  43. Price V, Willey A, Chen BK. Topical tacrolimus in alopecia areata. J Am Acad Dermatol 2005;52:138-9.
  44. Hajheydari Z, Jamshidi M, Akbari J, Mohammadpur R. Combination of topical garlic gel and betamethasone valerate cream in the treatment of localized alopecia areata: A double-blind randomized controlled study. Indian J Dermatol Venereol Leprol 2007;73:29-32.
  45. Das S, Ghorami RC, Chatterjee T, Banerjee G. Comparative assessment of topical steroids, topical tretinoin (0.05%) and dithranol paste in alopecia areata. Indian J Dermatol 2010;55:148-9.
  46. Talpur R, Vu J, Bassett R, Stevens V, Duvic M. Phase I/II randomized bilateral half-head comparison of topical bexarotene 1% gel for alopecia areata. J Am Acad Dermatol 2009;61:592-9e.
  47. Tosti A, Pazzaglia M, Voudouris S, Tosti G. Hypertrichosis of the eyelashes caused by bimatoprost. J Am Acad Dermatol 2004;51:149S-50S.
  48. Roseborough I, Lee H, Chwalek J, Stamper RL, Price VH. Lack of efficacy of topical latanoprost and bimatoprost ophthalmic solutions in promoting eyelash growth in patients with alopecia areata. J Am Acad Dermatol 2009;60:705-6.
  49. Coronel- Perez IM, Rodriguez-Rey EM, Camacho-Martinez FM. Latanoprost in the treatment of eyelash alopecia in alopecia areata universalis. J Eur Acad Dermatol Venereol 2010;24:481-5.
  50. Burton JL, Shuster S. Large doses of glucocorticoids in the treatment of alopecia areata. Acta Derm Venereol (Stockh) 1975;55:493-6.
  51. Sharma VK. Pulsed administration of corticosteroids in the treatment of alopecia areata. Int J Dermatol 1996;35:133-6.
  52. Ait Ourhroui M, Hassam B, Khoudri I. Treatment of alopecia areata with prednisolone in a once-monthly oral pulse. Ann Dermatol Venereol 2010;137:514-8.
  53. Sharma VK, Gupta S. Twice weekly 5-mg dexamethasone oral pulse in the treatment of extensive alopecia areata. J Dermatol 1999;26:562-5.
  54. Majid I, Masood Q, Hassan I, Khan D, Chisti M. Childhood vitiligo: Response to methylprednisolone oral minipulse therapy and topical fluticasone combination. Indian J Dermatol 2009;54:124-7.
  55. Pasricha JS, Khaitan BK. Oral minipulse therapy with betamethasone in vitiligo patients having extensive or fast spreading disease. Int J Dermatol 1993;31:753-7.
  56. Mittal R, Manchanda Y. Lichen planus treated with betamethasone oral minipulse therapy. Indian J Dermatol Venereol Leprol 2000;66:34-5.
  57. Claudy AL, Gagnaire D. PUVA treatment of alopecia areata. Arch Dermatol 1983;119:975-8.
  58. Mitchell AJ, Douglass MC. Topical photochemotherapy for alopecia areata. J Am Acad Dermatol 1985;12:644-9.
  59. Lassus A, Kianto U, Johansson E. PUVA treatment for alopecia areata. Dermatologica 1980;151:298-304.
  60. Vander Schaar WW, Silliis SJ. An evaluation of PUVA-therapy for alopecia areata. Dermatologica 1984;168:250-2.
  61. Behrens-Williams SC, Leiter U, Schiener R et al. The PUVA-turban therapy as a new option of applying a dilute psoralen solution selectively to the scalp of patients with alopecia areata. J Am Acad Dermatol 2001;44:248-52.
  62. Bayramqurler D, Demirsoy EO, Akturk AS, Kiran R. Narrowband ultraviolet B phototherapy for alopecia areata. Photodermatol Photoimmunol Photomed 2011;27:325-7.
  63. Veith W, Deleo V, Silverberg N. Medical phototherapy in childhood skin diseases. Minerva Pediatr 2011;63:327-33.
  64. Al-Mutairi N. 308-nm excimer laser for the treatment of alopecia areata. Dermatol Surg 2007;33:1483-7.
  65. Al-Mutairi N. 308-nm excimer laser for the treatment of alopecia areata in children. Pediatr Dermatol 2009;26:547-50.
  66. Ellis CN, Brown MF, Voorhess JJ. Sulfasalazine for alopecia areata. J Am Acad Dermatol 2012;46:541-4.
  67. Otberg N  Systemic treatment for alopecia areata. Dermatol Therap 2011;24:320-5.
  68. Rashidi T, Mahd AA. Treatment of persistent alopecia areata with sulfasalazine. Int J Dermatol 2008;47:850-2.
  69. Farshi S, Mansouri P, Safar F, Khiabanloo SR. could azathioprine be considered as a therapeutic alternative in alopecia areata? A pilot study. Int J Dermatol 2010;49:1188-93.
  70. Gupta A, Ellis C, Cooper K, et al. Oral cyclosporine for the treatment of alopecia areata: a clinical and immunohistochemical analysis. J Am Acad Dermatol 1990;22:242-50.
  71.  Chartaux E, Jolly P. Long-term follow-up of the efficacy of methotrexate alone or in combination with low dose of oral corticosteroids in the treatment of alopecia totalis or universalis. Ann Dermatol Venereol 2010;137:507-13.
  72. Bhat YJ, Manzoor S, Khan AR, Qayoom S. Trace element levels in alopecia areata. Indian J Dermatol Venereol Leprol 2009;75:29-31.
  73. Park H, Kim CW, Kim SS, Park CW. The therapeutic effect and the changed serum zinc level after zinc supplementation in patients with alopecia areata who had a low serum zinc level. Ann Dermatol 2009; 21(2): 142-146.
  74. Abramovits W, Losornio M. Failure of two TNF-alpha blockers to influence the course of alopecia areata. Skinmed 2006;5:177-81.
  75. Strober BE, Sin K, Alexis AF, et al. Etanercept does not effectively treat moderate to severe alopecia areata: an apen-label study. J Am Acad Dermatol 2005;52:1082-4.
  76. Yoo KH, Kim MN, Kim BJ, Kim CW. Treatment of alopecia areata with fractional photothermolysis laser. Int J Dermatol 2010;49:485-7.
  77. Sasmaz S, Arican O. Comparison of azelaic acid and anthralin for the therapy of patchy alopecia areata: a plot study. Am J Clin Dermatol 2005; 6(6): 403-406.
  78. Sharquie KE, Al –Obaidi HK. Onion juice (Allium cepa L.), a new topical treatment for alopecia areata. J Dermatol 2002; 29(6):343-346.
  79. Kaplan AL, Olsen EA. Topical 5-fluorouracil is ineffective in the treatment of extensive alopecia areata. J Am Acad Dermatol 2004;50:941-3.
  80. Fernandez-Guarino M, Harto A, Garcia-Morales I, Perez-Garcia B, Arrazola JM, Jean P. Failure to treat alopecia areata with photodynamic therapy. Clin Exp Dermatol 2008;33:585-7.
  81. van der Velden EM, Drost BH, Ijsselmuiden OE, Baruchin AM, Hulsebosch HJ. Dermatography as a new treatment for alopecia areata of the eyebrows. Int J Dermatol 1998;37:617-21.

Vitiligo Management: An Update

Authors
Imran Majid
Article Citation and PDF Link
BJMP 2010;3(3):a332
Abstract / Summary
Abstract: 

Vitiligo is one of the commonest skin disorders with a presumed autoimmune aetiology. The management options for this disease have undergone a sea of change over the last two or three decades and we are now in a much better position to treat this disease than in the past. Treatment options such as Narrowband Ultraviolet B (NB-UVB), Targeted Phototherapy, and Excimer laser on the medical front, in addition to epidermal cell transplantation and melanocyte culture transplants on the surgical front, have all revolutionized the management of this psychologically devastating disease.

Introduction

Vitiligo is one of the oldest and commonest skin disorders affecting approximately 1-2% of the human population.1 The disease shows no regard to the ethnic, racial or socioeconomic background of the affected sufferers. The cosmetic impact of this disease is tremendous and its psychological impact devastating particularly in coloured races.2,3,4 The aetiopathogenesis of this disease is now much better understood (table 1)5 compared with a decade earlier but much remains unknown. In parallel with these developments on the aetiological front, a lot of new advances have been made on the therapeutic front as well. With these new therapeutic options, we are currently in a much better position to treat this disease than we were a decade or two earlier. So, how far and how satisfactorily are we able to treat this disorder now? What are the new treatment options available for this disorder and how far have they helped a dermatologist to claim a cure for this disorder? These are some of the questions that will be addressed in this paper. 
 
New advances in management
 
Medical therapies
 
The most recent advances on the medical front have been Narrowband Ultraviolet B (NB-UVB) therapy, Targeted Ultraviolet B (UVB), Excimer laser therapies, topical immunomodulator treatment in the form of topical calcineurin inhibitors, topical pseudocatalase, and topical Vitamin D analogues in combination with Ultraviolet (UV) light.
 
NB-UVB
 
NB-UVB, using UV-lamps with a peak emission of around 311nm has now emerged as the treatment of first choice in generalized vitiligo as well as vitiligo vulgaris (patchy vitiligo).6,7,8 The efficacy of NB-UVB in vitiligo was first demonstrated by Westerhof and Nieuwboer-Krobotova in 1997.9 Since then there have been a large number of clinical studies that have demonstrated the therapeutic benefit of NB-UVB in vitiligo patients. The mechanism of action of NB-UVB in vitiligo is through induction of local immunosuppression and stimulation of the proliferation of melanocytes in the skin and the outer root sheath of hair follicles.6 There is a stimulatory effect on melanogenesis and on the production of Melanocyte Stimulating Hormone (MSH).Comparison studies have shown a significantly enhanced rate of repigmentation with NB-UVB compared with topical Psoralen and Ultraviolet A (PUVA) therapy.10 Furthermore, the incidence of adverse effects seen commonly with topical PUVA, such as phototoxicity, is significantly reduced with the use of NB-UVB.
 
NB-UVB has shown a number of advantages over PUVA in vitiligo patients in addition to its excellent efficacy. These advantages include its extremely low side-effect profile particularly on the systemic front, its established safety in children, and safety in pregnant females. NB-UVB also has considerably better patient compliance as there is no need to time the exposure with any drug intake or any need for eye protection beyond treatment exposure time. A recent double-blind randomized11 study comparing NB-UVB with PUVA demonstrated a much better efficacy with NB-UVB. The study found that repigmentation achieved with NB-UVB was much better with respect to colour matching with uninvolved skin, and this was also more persistent than that achieved with PUVA.11
 
In addition NB-UVB has been used in childhood vitiligo with excellent results.12 No additional adverse effects were seen in children with NB-UVB as compared with those in adults. Furthermore, given the long-term safety profile of NB-UVB in comparison with PUVA as far as skin malignancies are concerned,13 NB-UVB is now preferred over all other treatment options in the management of generalized vitiligo in both adults and children.
 
Table 1: Aetiological hypothesis of vitiligo5
Aetiological hypothesis
Brief explanation
Autoimmune hypothesis
Believes that vitiligo occurs because of destruction of melanocytes by an immune mechanism.
Most favoured theory at present, supported by many recent in-vitro studies.
Auto-cytotoxic hypothesis
Believes that vitiligo occurs because of accumulation of toxic metabolites in the melanocytes secondary to a defect in their metabolic clearance of the toxins.
Neurogenic hypothesis
Believes that vitiligo is because of an altered reaction to neuropeptides, catecholamines and their metabolites by epidermal melanocytes.
Biochemical hypothesis
Believes that over-secretion of hydrobiopterin, a cofactor of tyrosine hydroxylase results in accumulation of catecholamines that in turn results in formation of reactive oxygen species in the melanocytes. These reactive oxygen species are thought to cause destruction of affected melanocytes in vitiligo patients.
 
NB-UVB has been used in combination with different topical agents to increase its efficacy and thus shorten the total duration of treatment. Treatment options that have been used with NB-UVB in vitiligo till date include topical tacrolimus,14,15 pimecrolimus,16 Vitamin D analogues17,18 and even topical pseudocatalase.19 While some studies have shown a synergistic effect with these combinations, others have found the efficacy of the combinations to be similar to NB-UVB alone. In one half-body comparison study, topical placental extract was used in combination with NB-UVB but the combination was shown to offer no added benefit than NB-UVB alone.20 Therefore, the ideal topical agent to be combined with NB-UVB remains unknown.
 
Laser Therapy
 
Excimer laser, which uses Xenon-Chlorine (Xe-Cl) gas and produces a monochromatic laser light of 308nm wavelength, is another innovative treatment option for vitiligo. The laser system has been used with increasing frequency over the last few years for targeted treatment of individual vitiligo lesions.21 The laser is used either alone or in combination with topical immunomodulator or PUVA-sol therapy.22,23 Treatment with this laser is claimed to give extremely good and early results in both localized and segmental vitiligo. In a pilot study21 on 18 patients with 29 affected areas 57% of lesions showed varying degrees of repigmentation after just six exposures over two weeks. The figure was increased to 87% after 12 treatments over four weeks.21 Another recent study has reported a repigmentation of >75% in 61% of lesions after 30 treatments with Excimer laser. Repigmentation was found to be better on the face and trunk than on the extremities.24
 
Topical therapies, particularly topical tacrolimus, have been used in combination with Excimer laser. This combination has been claimed to be more effective than Excimer laser alone.22 In a randomized right-left comparison study22 with 14 patients, Excimer light monotherapy was compared with a combination of Excimer laser with topical tacrolimus. While 20% of lesions treated with Excimer laser alone achieved >75% repigmentation, the same degree of repigmentation was obtained in 70% lesions with the combination treatment.22 Topical methoxsalen has also been used in combination with Excimer laser phototherapy and this has been claimed to have worked better than laser therapy alone.23
 
The advantage of Excimer laser therapy over conventional UVB therapy is the targeted mode of treatment with no exposure of the uninvolved skin. Moreover, the onset of repigmentation is earlier with Excimer laser therapy than with UVB therapy.
 
Targeted UVB therapy
 
This is another recent innovation in vitiligo management that has arrived over the last few years. The beauty with this therapy is that it delivers high intensity UVB light only to the affected vitiliginous areas, avoiding any exposure to the uninvolved skin. This not only decreases the cumulative UVB dose received by an individual patient, but is also claimed to improve the efficacy of treatment quite significantly.
 
Targeted UVB therapy, as expected, finds its use more in the treatment of focal and segmental types of vitiligo. In fact, the first study25 with targeted UVB therapy was done on eight patients with segmental vitiligo. Five of these patients achieved >75% repigmentation of their lesions with this therapy.25
 
Targeted UVB therapy offers certain advantages over Excimer laser phototherapy. The treatment is safer and more efficacious compared with conventional UVB therapy, and almost as efficacious but much less costly than Excimer laser therapy.26
 
Systemic immunomodulator therapy
 
Vitiligo is thought to be an immune-mediated disease and thus immune-suppressive and immunomodulator agents have been used on a regular basis in this disease. Among the immunosuppressants, systemic steroids have been the most commonly used. However, systemic steroid therapy has always been associated with a high incidence of adverse effects especially in children which is the age-group most commonly affected. To overcome this limitation, steroids have been given in pulse or even in mini-pulse form. A prospective study involving 14 patients with progressive or static vitiligo showed cessation of disease activity and a repigmentation rate of 10-

60% after high-dose methylprednisolone pulse therapy administered on three consecutive days.27 Systemic steroids have also been administered in a mini-pulse form on two consecutive days every week, known as Oral Minipulse (OMP) therapy. The first study demonstrating the efficacy of OMP with oral betamethasone (0.1mg/kg with a maximum of 5mg) was described in 1991.28 In a later study29 on childhood vitiligo, betamethasone was replaced by oral methylprednisolone and combined with topical fluticasone ointment on the vitiligo lesions. The disease was arrested in >90% of patients, and >65% of children achieved good to excellent (>50%) repigmentation of their vitiligo lesions.29    

 
Topical Vitamin D analogues
 
Vitamin D analogues, particularly Calcipotriol, have been used topically either alone or in combination with topical steroids in the management of vitiligo. The basis for the use of these agents is that Vitamin D3 affects the growth and differentiation of both melanocytes and keratinocytes. This has been further proved by the demonstration of receptors for 1 alpha-dihydroxyvitamin D3 on the melanocytes. These receptors are believed to have a role in stimulating melanogenesis.29 Vitamin D analogues have given variable results in the treatment of vitiligo in different studies. These agents have also been used in combination with UV-light (including NB-UVB) and topical steroids with variable results.30,31,32
 
Topical immunomodulators
 
Topical immunomodulators, such as tacrolimus and pimecrolimus, have been the most promising recent additions to topical vitiligo therapy. In fact because of their efficacy and a remarkable safety profile the use of these agents in vitiligo has shown a consistently increasing trend over the last few years. These agents can be safely administered in young children, as they don’t cause any atrophy or telangiectasia of the skin even after prolonged use. There is also no risk of hypothalamic-pituitary-adrenal (HPA) axis suppression as seen with the widespread use of potent topical steroids.33 The first study that demonstrated the efficacy of tacrolimus in vitiligo was published in 2002.34 In this study tacrolimus was used in six patients with generalized vitiligo and five of them achieved >50% repigmentation of their lesions by the end of study period.34 Since then many additional studies have been published on this subject and have clearly demonstrated the role of topical tacrolimus in vitiligo. The best results with topical immunomodulator therapy have been seen on exposed parts of the body such as the face and neck and, as with any other therapy, the acral parts of the body respond the least.34,35 Similar results were obtained with the use of topical pimecrolimus in vitiligo patients.36

Pseudocatalase
 
Pseudocatalase has been used in combination with Dead Sea climatotherapy or UVB exposure for the treatment of vitiligo. The basis for the use of this agent in vitiligo is the evidence of oxidative stress and high H2O2 levels in the lesional skin.37 While some earlier studies37 demonstrated excellent results with this agent in inducing repigmentation in vitiligo, later studies have cast doubts on its efficacy.38 Pseudocatalase is used topically on the lesional skin, and this is followed by UVB exposure to the whole body or to the lesional skin. The combination is claimed to correct the oxidative stress on melanocytes in vitiligo patients and thus lead to correction of the depigmentation.    
 
Topical 5-Fluorouracil
 
Topical 5-fluorouracil is supposed to induce repigmentation of vitiligo lesions by overstimulation of follicular melanocytes which migrate to the epidermis during epithelialization.39 This form of topical therapy can be combined with spot dermabrasion of the vitiligo lesions to improve the repigmentation response. In a study by Sethi et al,40 a response rate of 73.3% was observed with a combination of spot dermabrasion and topical 5-fluorouracil after a treatment period of six months.40
 
Surgical therapies
 
Surgical therapies for vitiligo have further increased the percentage cure of the disease by an appreciable degree, with the consequent increase of their use in the management of unresponsive vitiligo both in India and abroad. These surgical therapies, as a rule, are indicated in those patients who have a stable (non-progressive) disease of at least one year and not responding to medical treatment. In general the most important advantage with these procedures is that the chances of repigmentation of lesions are in the range of 90-100%. Moreover, these interventions are becoming better and easier to perform with every passing day.
 
Different surgical therapies that have been attempted in the management of vitiligo include autologous suction blister grafting, split-thickness grafting, punch grafting, smash grafting, single follicular unit grafting, cultured epidermal suspensions and autologous melanocyte culture grafting. All these grafting procedures, except the melanocyte culture grafting, are easy to perform and do not require any sophisticated instruments. These grafting techniques have now been divided into two types, tissue grafts and cellular grafts, depending on whether whole epidermal/dermal tissue is transplanted or the individual cellular compartment.
 
Tissue grafting technique
 
Suction blister grafting
 
Here, thin epidermal grafts are taken from suction blisters on the donor site, usually on the buttocks or thighs. These suction blisters are produced by applying sufficient negative pressure on the skin at the donor site by using a suction apparatus or syringes with three-way cannulae. The epidermal grafts are then transplanted on to dermabraded vitiligo lesions. This leads to repigmentation of the recipient areas with an excellent cosmetic matching. The ease of the procedure, the high success rate and the excellent cosmetic results have all made suction blister grafting the procedure of choice in vitiligo grafting.41
 
Split thickness grafting
 
In this grafting technique a thin split thickness graft is taken from a donor site with the help of a dermatome, Humby’s knife, Silver’s knife or a simple shaving blade. This graft is then transplanted on to dermabraded recipient areas. This technique also gives excellent cosmetic matching after repigmentation and the incidence of repigmentation in this technique is also quite high. In fact, most comparison studies on grafting techniques in vitiligo have shown that maximum repigmentation is achieved with either suction blister grafting or split thickness grafting.41 The advantage of partial thickness grafting over the suction blister method is that a relatively larger area of vitiligo can be tackled in a single sitting. Both partial thickness skin grafting as well as suction blister grafting can be followed up by NB-UVB to achieve faster and better results.
 
Miniature punch grafting
 
Here full-thickness punch grafts of 1.0 to 2.0 mm diameter are taken from a suitable donor site and then transplanted on to similar punch shaped beds on the recipient vitiligo lesions. The recipient area is then treated with either PUVA/PUVA-sol or topical steroids leading to spread of pigment from the transplanted punches to the surrounding skin. With time the whole of the recipient area gets repigmented. The advantages of this procedure are that it is easy to perform and can take care of a relatively larger vitiligo area compared with the above two procedures. Also vitiligo lesions with irregular or geographical shapes can be treated with this procedure. However there are certain limitations. There is the risk of ‘cobblestone appearance’, ‘polka-dot appearance’, and hypertrophic changes at the recipient site.42 All these side effects can be minimized by proper patient selection and by use of smaller sized punches of 1.0 to 1.5 mm diameter. Miniature punch grafting is presently the commonest surgical procedure performed in India on vitiligo patients.
 
Follicular unit grafting
 
In this technique, single-hair follicular units are harvested/prepared from a suitable donor area as in the case of hair transplantation. These follicular units are then cut above the level of the follicular bulb and then transplanted into vitiligo lesions. The idea behind this technique is that the melanocytes in the follicular unit are ‘donated’ to the vitiliginous skin and serve as a source of pigment at the recipient site. The repigmentation process here simulates the normal process of repigmentation of vitiliginous skin quite closely and thus gives an excellent cosmetic result. This procedure combines the advantages of punch grafting with the excellent cosmetic results of split thickness or blister grafting techniques.43 The procedure is however tedious and needs good expertise on the part of the cosmetic surgeon.
 
Smash grafting
 
In this technique, a partial thickness graft is taken and is ‘smashed’, or cut into very small pieces, by means of a surgical blade on a suitable surface such as a glass slide. This ‘smashed’ tissue is then transplanted on to the dermabraded recipient skin and covered with a special powder or corrugated tube dressing so as to keep the smash-graft undisturbed on the recipient area. The advantage of this technique, over a simple partial thickness grafting, is that thicker grafts can be used with a good cosmetic result. The procedure has been indicated for those who are relatively inexperienced and cannot take an ideal, thin and transparent partial thickness graft from the donor area.44
 
Cellular grafting techniques
 
Non-cultured epidermal suspensions
 
Here a split-thickness graft is taken from a donor area and then incubated overnight. On the next day the cells are mechanically separated using trypsin-EDTA solution and then centrifuged to prepare a suspension. This cell suspension is then applied to the dermabraded vitiligo lesions, and a collagen dressing is applied to keep it in place. A relatively large area of vitiligo, about ten times the size of the donor graft can be taken care of with this procedure.45 The recipient area however has to be treated with either NB-UVB or PUVA for two to three months to achieve the desired pigmentation.
 
Melanocyte culture transplantation
 
This is a relatively more advanced grafting procedure where, once again, a split-thickness graft is taken from a donor area and incubated in an appropriate culture medium to grow the melanocytes or the keratinocytes-melanocyte combination in vitro. The cultured cells are then applied onto laser dermabraded, or even mechanically abraded, lesional skin.46,47 The procedure is obviously more difficult to perform, as it needs the advanced laboratory facilities for melanocyte culture. However the results with this procedure are excellent and a relatively large area of involved skin can be tackled by a single donor graft.
 
Summary
 
Table 2 summarises the above discussion of treatment options in vitiligo.
 
Table 2: New treatment options in vitiligo
Medical therapies and phototherapy
Surgical therapies
 
Narrowband UVB therapy either alone or in combination with immunomodulators, Vitamin D analogues etc.
Excimer laser therapy
Targeted UVB phototherapy
Topical immunomoulators
Topical Vitamin D analogues
Topical pseudocatalase with UVB
Oral minipulse steroid therapy
Suction blister skin grafting
Partial thickness skin grafting
Miniature punch grafting
Follicular skin grafting
Smash grafting
Non-cultured epidermal cell transplant
Melanocyte culture transplant

 

Acknowledgements / Conflicts / Author Details
Competing Interests: 
None Declared
Details of Authors: 
IMRAN MAJID, MD, MBBS. Assistantt Professor in Dermatology, Govt Medical College, Srinagar Kashmir, India
Corresponding Author Details: 
Dr Imran Majid, Department of Dermatology, Governmentt Medical College, Srinagar, India
Corresponding Author Email: 
imran54@yahoo.com
References
References: 

 1.      Lerner AB. Vitiligo. J Invest Dermatol 1959;32:285-310.

2.      Hautmann G, Panconesi E. Vitiligo: a psychologically influenced and influencing disease. Clin Dermatol 1997;15:875-78.
3.      Mattoo SK, Handa S, Kaur I, Gupta N, Malhotra R. Psychiatric morbidity in vitiligo: prevalence and correlates in India. J Eur Acad Dermatol Venereol 2002;16:573-8.
4.      Aghaei S, Sodaifi M, Jafari P, Mazharinia N, Finlay AY. DLQI scores in vitiligo: reliability and validity of the Persian version. BMC Dermatol 2004;4:8.
5.      Kostovic K, Pasic A. New treatment modalities for vitiligo: focus on topical immunomodulators. Drugs 2005;65:447-59.
6.      Njoo MD, Spuls PL Bos JD et al. Nonsurgical repigmentation therapies in vitiligo: meta-analysis of the literature. Arch Dermatol 1998;134:1532-40.
7.      Scherschun L, Kim JJ, Lim HW. Narrow-band ultraviolet B is a useful and well-tolerated treatment for vitiligo. J Am Acad Dermatol 2001;44:999-1003.   
8.      Njoo MD, Boss JD, Westerhof W. Treatment of generalised vitiligo in children with narrow-band (TL-01) UVB radiation therapy. J Am Acad Dermatol 2000;42:245-53.
9.      Westerhof W, Nieuweboer-Krobotova L. Treatment of vitiligo with UV-B radiation vs topical psoralen plus UV-A. Arch Dematol 1997;133:1525-28.
10.   Natta R, Somsak T, Wisuttida T, Laor L. Narrow-band ultraviolet B radiation therapy for recalcitrant vitiligo in Asians. J Am Acad Dermatol 2003;49:472-76.
11.   Yones SS, Palmer RA, Garibaldinos TM, et al. Randomized double-blind trial of treatment of vitiligo: Efficacy of psoralen-UVA therapy vs. narrowband-UVB therapy. Arch Dermatol 2007;143:578-84.
12.   Njoo Njoo MD, Boss JD, Westerhof W. Treatment of generalised vitiligo in children with narrow-band (TL-01) UVB radiation therapy. J Am Acad Dermatol 2000;42:245-53.
13.   Hearn RMR, Kerr AC, Rahim KF Ferguson J, Dawe RS. Incidence of skin cancers in 3867 patients treated with narrowband ultraviolet B phototherapy. Br J Dermatol 2008;159:931-5.
14.   Fai D, Cassano N, Vena GA. Narrowband UVB phototherapy combined with tacrolimus ointment in vitiligo: a review of 110 patients. J Eur Acad Dermatol Venereol 2007;21:916-20.
15.   Mehrabi D, Pandya AG. A randomized, placebo-controlled, double-blind trial comparing narrowband UV-B plus 0.1% tacrolimus ointment with narrowband UV-B plus placebo in the treatment of generalized vitiligo. Arch Dermatol 2006;142:927-9.
16.   Esfanduarpour I, Ekhlasi A, Farajedah S, Shamsadini S. The efficacy of 1% pimecrolimus cream plus ultraviolet B in the treatment of vitiligo: a double-blind, placebo-controlled clinical trial. J Dermatolog Treat 2009;20:14-18.
17.   Goktas EO, Aydin F, Senturk N, Canturk MT, Turanli AY. Combination of narrow-band UVB and topical calcipotriol for the treatment of vitiligo. J Eur Acad Dermatol Venereol 2006;20:553-7.
18.   Leone G, Pacifico P, Lacovelli P Vidolin A Paro, Picardo M. Tacalcitol and narrow-band phototherapy in patients with vitiligo. Clin Exp Dermatol 2006;31:200-05.
19.   Schallreuter KU, Wood JM, Lemke KR, Levenig C. Treatment of vitiligo with a topical application of pseudocatalase and calcium in combination with short-term UVB exposure: a case study on 33 patients. Dermatology 1995;190:181-82.
20.   Majid I. Topical placental extract: Does it increase the efficacy of Narrowband UVB therapy in vitiligo. Indian J Dermatol Venereol Leprol 2010;76:254-8.
21.   Baltas E, Csoma Z, Ignacz et al. Treatment of vitiligo with the 308nm xenon chloride excimer laser. Arch Dermatol 2002;138:1116-20.
22.   Kawalek AZ, Spencer JM, Phelps RG. Combined excimer laser and topical tacrolimus for the treatment of vitiligo: a pilot study. Dermatol Surg 2004; 30: 130-35.
23.   Grimes PE. Advances in the treatment of vitiligo: targeted phototherapy. Cosmet Dermatol 2003;140:1065-69.
24.   Zhang XY, He YL, Dong J, Xu JZ, Wang J. Clinical efficacy of a 308nm excimer laser in the treatment of vitiligo. Photodermatol Photoimmunol Photomed 2010;26:138-42. 
25.   Lotti TM, Menchini G, Andreasi L. UV-B radiation microphototherapy: an elective treatment for segmental vitiligo. J Eur Acad Dermatol Venereol 1999;113:102-08.
26.   Menchini G, Tsoureli-Nikita E, Hercogova J. Narrowband UV-B microphototherapy: a new treatment for vitiligo. J Eur Acad Dermatol Venereol 2003; 17: 171-77.
27.   Seiter S, Ugurel C, Pfohler W, Tilgen W, Reinhold U. Successful treatment of progressive vitiligo with high-dose intravenous methylprednisolone pulse therapy. Dermatology 1999;199:261-62
28.   Pasricha JS, Khaitan BK. Oral minipulse therapy with betamethasone in vitiligo patients having extensive or fast spreading disease. Int J Dermatol 1993;31:753-7.
29.   Majid I, Masood Q, Hassan I, Khan D, Chisti M. Childhood vitiligo: Response to methylprednisolone oral minipulse therapy and topical fluticasone combination. Indian J Dermatol 2009;54:124-7.
30.   Prasad D, Saini R, Nagpal R.Topical Calcipotriol in vitiligo: a preliminary study. Pediatr Dermatol 1999;16:317-20.
31.   Prasad D, Saini R, Verma N. Combination of PUVAsol and topical Calcipotriol in vitiligo. Dermatology 1998;197:167-70.
32.   Baysal V, Yildirim M, Erel A, Yilmaz E. Is the combination of Calcipotriol and PUVA effective in vitiligo? J Eur Acad Dermatol Venereol 2003;17:299-302.
33.   Plettenberg H, Assmann T, Ruzicka T. Childhood vitiligo and tacrolimus. Immunomodulatory treatment for an autoimmune disease. Arch Dermatol 2003;139:651-54.
34.   Grimes PE, Soriano T, Dytoc MT. Topical tacrolimus for the repigmentation of vitiligo. J Am Acad Dermatol 2002;47:789-91.
35.   Xu AE, Zhang DM, Wei XD, et al. Efficay and safety of tacrolimus cream 0.1% in the treatment of vitiligo. Int J Dermatol 2009;48:86-90.
36.   Mayoral FA, Gonzalez C, Shah NS, Arciniegas C. Repigmentation of vitiligo with pimecrolimus cream: a case report. Dermatology 2003;207:322-23.
37.   Schallreuter KU, Moore J, Behrens Williams S et al. Rapid initiation of repigmentation in vitiligo with Dead Sea climatotherapy in combination with pseudocatalase. Int J Dermatol 2002;41:482-87.
38.   Patel DC, Evans AV, Hawk JL. Topical pseudocatalase mousse and narrowband UVB phototherapy is not effective for vitiligo: an open, single-centre study. Clin Exp Dermatol 2003;28:562-63.
39.   Tsuji T, Hamada T. Topically administered fluorouracil in vitiligo.Arch Dermatol 1983;119:722-27.
40.   Sethi S; Mahajan BB; Gupta RR; Ohri A. Comparative evaluation of the therapeutic efficacy of dermabrasion, dermabrasion combined with topical 5% 5-fluorouracil cream, and dermabrasion combined with topial placentrex gel in localized stable vitiligo. Int J Dermatol 2007;46:875-9.
41.   Njoo MD, Westerhof W, Bos JD, Bossuyt PM. A systematic review of autologous transplantation methods in vitiligo. Arch Dermatol 1998;134:1543-49.
42.   Falabella R. Treatment of localized vitiligo by autologous minigrafting. Arch Dermatol 1988;124:169-55.
43.   Rusfianti M, Wirohadidjodjo YW. Dermatosurgical techniques for repigmentation of vitiligo. Int J Dermatol 2006;45:411-17.
44.   Patwardhan N. Conference report ACSICON 2008. J Cutan Aesthet Surg 2009;2:47-8.
45.   Mulekar SV. Long-term follow-up study of segmental and focal vitiligo treated by autologous, noncultured melanocyte-keratinocyte cell transplantation. Arch Dermatol 2004; 140: 1273-74.
46.   Chen YF, Yang PY, Hu DN et al. Treatment of vitiligo by transplantation of cultured pure melanocyte suspension: analysis of 120 cases. J Am Acad Dermatol 2004;51:68-74.
47.   Piangiani E, Risulo M, Andreassi A et al. Autologous epidermal cultures and narrow-band ultraviolet B in the surgical treatment of vitiligo. Dermatol Surg 2005;31:155-59.

share
Syndicate content