lyme disease update for the general dermatologist

Upload: elisa

Post on 28-Feb-2018

222 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    1/14

    T H E R A P Y I N P R A C T I C E

    Lyme Disease Update for the General Dermatologist

    Desiree A. Godar

    Valerie Laniosz

    David A. Wetter

    Springer International Publishing Switzerland 2015

    Abstract Lyme disease is an Ixodes tickborne illness

    that may arise from different species of the Borrelia spi-rochete and may be propagated in various hosts. Humans

    are considered dead-end hosts in this propagation cycle but

    may have a range of Lyme disease characteristics as a result

    of borrelial infection. Lyme disease has varied cutaneous

    manifestations, and the approach to diagnosis and treatment

    is based on the patient, the region, and suspected coinfec-

    tion with another tick-borne illness. An understanding of the

    distribution of the Ixodes tick, its vectors, and the most

    likely dermatologic presentation based on these factors

    allows the dermatologist to make appropriate testing and

    treatment recommendations. Our aim is to simplify this

    approach for the treating practitioner.

    Key Points

    The cutaneous manifestations of Lyme disease differ

    among the 3 stages of Lyme disease: solitary

    erythema migrans (stage 1), multiple erythema

    migrans (stage 2), borrelial lymphocytoma (stage 2),

    and acrodermatitis chronica atrophicans (stage 3).

    Erythema migrans is a clinical diagnosis and should

    be treated empirically with doxycycline 100 mg

    twice daily for 1421 days in adult, nonpregnantpatients and serologic testing is not indicated in

    patients with erythema migrans rash.

    Lyme prevention strategies focus around insect

    repellence and early tick detection and removal.

    1 Historical Perspective and Epidemiology

    The eruptions and arthritides associated with what is now

    known asLyme diseasehave been reported in the scientific

    literature for the past century. Specifically, the bulls eye

    rash known as erythema chronicum migrans was first

    described by Lipshutz in 1913 [1]. Lyme disease has been

    tracked since 1982 by a surveillance program of the Cen-

    ters for Disease Control and Prevention (CDC) after

    Burgdorfer et al. [2] isolated the spirochete Borrelia

    burgdorferi as a causative agent of Lyme disease.

    Lyme disease is the most prevalent tick-borne disease in

    the United States, and its incidence is increasing [3]. Chil-dren age 714 years and adults age 5570 years represent

    the majority of persons affected by Lyme disease [4]. Initial

    infection with B. burgdorferi occurs most often during

    summer, with the geographical distribution correlating with

    the distribution of its vector, the Ixodestick. In the wild, the

    white-footed mice and white-tailed deer serve as reservoirs

    for B. burgdorferi [1]. Global climate change has, and is

    projected to continue to, lead to a northward shift in the

    distribution of Lyme disease cases (Fig. 1) [5]. Currently,

    most US cases of Lyme disease occur in the northern

    Midwest and Northeastern regions. Worldwide, Lyme dis-

    ease also affects persons in central Europe and Asia.Table1 describes the worldwide distribution of Borrelia

    species, along with tick vectors and host reservoirs.

    The risk of a human becoming infected with Borrelia

    after a tick bite in an endemic area in Switzerland was

    estimated at 4.5 % and was similarly estimated at 3.2 % in

    Westchester County, New York [6, 7]. Overall, many

    studies cite a risk of acquiring Lyme disease from a tick

    bite to be 16 % [6]. This risk calculation is contingent, of

    course, on patients recognizing that they had a tick bite, as

    D. A. Godar V. Laniosz D. A. Wetter (&)

    Department of Dermatology, Mayo Clinic, 200 First St SW,

    Rochester, MN 55905, USA

    e-mail: [email protected]

    Am J Clin Dermatol

    DOI 10.1007/s40257-014-0108-2

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    2/14

    well as their geographic location. This detail is important

    because many tick bites go unrecognized. Statistically, the

    risk of infection can be estimated from knowing theprobability of being bitten by 1 tick, the number of tick

    bites per person, and the spirochete prevalence in the

    region [8]. Risk factors for infection include time spent

    outdoors, geographic location, duration of tick feeding, and

    time of year [1].

    Unfortunately, Ixodes ticks can be coinfected with other

    infectious agents, includingAnaplasma phagocytophilum and

    Babesia microti, the causative agents of human granulocytic

    ehrlichiosis and babesiosis, respectively [1, 9]. Detailed

    information regarding coinfection rates has been obtained for

    many states in North America, as reported by Swanson et al.

    [10]. As few as 1 % of ticks are coinfected with multiplepathogens in California, whereas as many as 28 % of ticks in

    New York may harbor more than 1 pathogen [10].

    2 Pathogenesis of Lyme Disease

    Ticks acquire Borrelia when they take a blood meal from

    an infected reservoir host. The spirochetes express the

    adhesion lipoproteins OspA and OspB that allow them to

    Fig. 1 Maps depicting current distribution of Lyme disease in the

    United States and the world. a Predominance of US cases in the

    Midwest and Northeast. b US distribution of babesiosis. c US

    distribution of anaplasmosis. d Worldwide distribution of the Ixodes

    tick vectors. e The projected shift by 2050 in Lyme disease

    distribution in the United States as a result of climate change.

    (Adapted from Centers for Disease Control and Prevention [25])

    D. A. Godar et al.

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    3/14

    adhere to the tick midgut epithelium [11]. The Borrelia

    organism then migrates to the salivary glands with a pro-

    tein called OspC[12], which is upregulated when the tick

    feeds and is essential for the infectivity of the organism

    [13]. B. burgdorferi is protected from antibody-mediated

    killing because of Salp15, a protein found in tick saliva that

    binds to the outer surface protein ofB. burgdorferi [14].

    Importantly, a study by Sood et al. [15] showed that the

    risk of Borrelia transmission increases with a duration of

    tick attachment greater than 72 h. Sood et al. showed poor

    correlation between patient-predicted duration of tick

    attachment and duration of attachment determined by

    scutula measurement, which was used for the study.

    After entering the human host, the Borrelia organism

    disseminates widely, activating toll-like receptor 2, mye-

    loid differentiation antigen 88, and CD14, and this acti-

    vation leads to subsequent cytokine production.

    Complement system activation also occurs; however, some

    Borrelia species have mechanisms to evade complement-

    mediated immunity [16]. OspC is downregulated after

    long-term infection has been established in the host, and

    the organism undergoes genetic recombination to evade

    host humoral responses [17]. Specifically, vlsE proteins

    have been implicated in this process that allows for per-

    sistent infection [17].

    3 Clinical Features

    Lyme disease has three stages: early localized, early dis-

    seminated, and chronic. Further details about the derma-

    tologic manifestations are included herein, and their

    associated Borrelia species are summarized in Table2.

    Early localized disease (stage 1) can present as erythema

    chronicum migrans, also known as erythema migrans(EM). Patients at this stage often have regional lymphad-

    enopathy, with up to two-thirds of patients having consti-

    tutional symptoms, such as fever, myalgias, arthralgias,

    malaise, headache, and fatigue [1]. EM is a common

    clinical presentation across Europe, Asia, and the United

    States.

    Early disseminated disease (stage 2) is characterized by

    involvement of internal organ systems, including the cen-

    tral nervous system (CNS), musculoskeletal system, and

    heart. Dissemination may occur within days to months after

    the initial infection [1]. Borrelial lymphocytoma is a

    cutaneous manifestation that may be seen at this stage, andpatients also may have multiple EM patches. Borrelial

    lymphocytoma is caused by Borrelia afzelii, which is

    endemic to Europe and Asia.

    Chronic Lyme disease (stage 3) is characterized by

    encephalopathy and chronic arthritis, with only rare cuta-

    neous manifestations. Acrodermatitis chronica atrophicans

    (ACA) is a cutaneous feature of stage 3 disease. This form

    of Lyme disease is not seen in the United States because it

    is caused by B. afzelii, endemic to Europe and Asia. Bor-

    relial infections also have been linked to morphea and

    lichen sclerosus, with rare and controversial associations

    with various granulomatous skin diseases such as sarcoid

    and granuloma annulare [1, 18].

    3.1 Erythema Chronicum Migrans

    Among persons with Lyme disease, 50 % of adults and

    90 % of children have EM [1]. EM typically appears

    714 days following infection but may occur as early as

    3 days or as late as 1 month after tick exposure [4]. EM is

    often associated with constitutional symptoms, including

    fever, malaise, fatigue, and headache [4]. It is characterized

    by an erythematous macule that begins at the site of a tick

    bite and spreads centrifugally to a diameter of at least 5 cm

    (Fig.2ad). A range of 550 cm (median 13 cm) has been

    reported in the literature [19]. The lesion can expand rap-

    idly, sometimes growing 3 cm in 1 day. As it expands, the

    center may clear, resulting in an annular or targetoid

    morphology.

    In the United States, less than 10 % of patients present

    with the classic targetoid morphology and instead have a

    homogeneous patch of erythema [4]. The annular mor-

    phologic pattern is more common in Europe [4]. Rare

    Table 1 Geographic Distribution of Borrelia, Tick Species, and

    Reservoirs

    Agent United States Europe Asia

    Borrelia

    species

    implicated

    in Lyme

    disease

    Borrelia

    burgdorferi

    Common

    Borrelia garinii

    Borrelia afzelii

    OccasionalB. burgdorferi

    Rare

    Borrelia

    lusitaniae

    Borrelia

    valaisiana

    Borrelia bissettii

    Borrelia

    spielmanii

    Common

    B. garninii

    B. afzelii

    RareB. valaisiana

    Tick vector Ixodes scapularis

    Ixodes pacificus

    Ixodes

    persulcatus

    Ixodes ricinus

    I. persulcatus

    Ixodes ovatus

    Host

    reservoir

    Northeast: white-

    footed mouse,

    white-tailed

    deer, and

    raccoons

    West: wood

    mouse

    Dusky-footed

    wood rat,

    cotton rat,

    cotton mouse,

    and many other

    rodents and

    birds

    Long-tailed

    shrew and

    wood

    mouse

    Data from Bhate and Schwartz [1]

    Lyme Disease Review

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    4/14

    vesicular, purpuric, and necrotic variants of EM have been

    reported [4, 20]. Lesions of the lower leg may have pete-

    chial qualities [21]. The most common EM locations cor-

    relate to the most frequent areas of tick bites: intertriginous

    areas in adults and head and neck in children [21]. EM

    lesions usually are asymptomatic, but up to one-half of

    patients report mild itching or burning [21]. EM lasts

    approximately 6 weeks if untreated, with treatment leading

    to EM resolution in a median of 18 days [ 22]. EM tends to

    be less inflammatory and more insidious in Europe [23].

    Although EM is traditionally thought to be an isolated

    lesion, up to 20 % of patients have multiple lesions as a

    result of spirochetemia, multiple tick bites, or lymphatic

    spread [24] (Fig. 2eg). The existence of multiple EM

    lesions is considered stage 2 disease, with lesions appear-

    ing either simultaneously or sequentially [1]. Conversely,

    disseminated disease may be present in the absence of

    multiple EM lesions [1]. Patients with multiple EM lesions

    typically have smaller lesions than patients with an isolated

    EM lesion, and the lesions are less likely to be annular [ 1].

    In addition, patients with multiple lesions are more likely

    to report extracutaneous symptoms than those with isolated

    lesions [21]. In patients who have EM resolution with

    persistent flulike symptoms, coinfection with another

    microorganism, such as B. microti or A. phagocytophilum,

    should be considered [25].

    3.2 Borrelial Lymphocytoma

    Lymphocytoma presents as a soft, nontender erythematous

    to violaceous nodule that may occur within an EM lesion

    [1] (Fig.2h). The earlobe frequently is an area of

    involvement in children; the nipple area is most frequently

    seen in adults [21,26]. Lymphocytoma is more common in

    Europe and in children and is essentially nonexistent in the

    United States [1,21]. Its lesions are not usually associated

    with systemic symptoms [21].

    3.3 Acrodermatitis Chronica Atrophicans

    ACA is a skin manifestation of chronic late-stage Borreliainfection, occurring months to even years after the initial

    infection and more commonly in elderly persons [1]. ACA

    is rare as sequelae of Lyme disease in the United States, but

    it has been reported in approximately 10 % of patients with

    Lyme borreliosis in Europe.

    ACA lesions favor the distal extensor surfaces of the

    extremities, presenting initially as edematous plaques and

    progressing to hyperpigmented atrophic plaques, resulting in

    a cigarette paper appearance of the skin if untreated

    (Fig. 2i, j). These plaques are progressive,coalescing to cover

    large areas over time. Some patients with ACA recall having

    EM in the same body region in the past [21]. ACA may beassociated with development of subcutaneous firm nodules

    that develop primarily on the elbows and knees [1,21].

    Dermatitis atrophicans maculosa is a variant of ACA

    that resembles anetoderma and frequently affects the but-

    tocks and thighs. Another variant of ACA is a pseudo-

    sclerodermatous presentation in which persons have

    indurated, white plaques within or near existing ACA

    lesions and favoring the distal extremities [21]. ACA is

    associated with peripheral neuropathy in up to 60 % of

    patients, often with allodynia [21].

    4 Diagnosis

    The diagnosis of Lyme disease requires presence of a

    lesion consistent with EM and history of tick exposure or

    laboratory evidence of Borrelia infection [27]. Thus, the

    diagnosis does not require that the patient recall a tick bite.

    The CDC and the Infectious Diseases Society of America

    state that any annular, erythematous lesion occurring

    within several hours of a tick bite likely represents a

    hypersensitivity reaction and not EM [3,27]. Few patients

    sustaining a tick bite have Lyme disease, although this

    occurrence depends on the local prevalence of infection in

    ticks; the tick must have been attached for at least 24 h for

    Borrelia transmission to occur [24]. If a patient does not

    have the characteristic EM lesions or cannot recall a tick

    bite, diagnosis of Lyme disease is more difficult.

    4.1 Pathologic Characteristics

    Although skin biopsy is not routinely obtained for the

    evaluation of Lyme disease, when it is performed, the

    Table 2 Borrelia species associated with major cutaneous manifes-

    tations of Lyme disease

    Cutaneous manifestation Borrelia

    species

    Geographic locationa

    Erythema migransb Borrelia

    afzelii

    Europe, Asia

    Borrelia

    garinii

    Europe, Asia

    Borrelia

    burgdorferi

    North America, Europe

    (uncommon)

    Borrelia

    spielmanii

    Europe

    Lymphocytoma B. afzelii Europe, Asia

    Acrodermatitis chronica

    atrophicans

    B. afzelii Europe, Asia

    a Geographic information from Bhate and Schwartz [1]b Homogeneous erythema is observed more commonly in North

    America; erythema migrans is observed more commonly with annular

    morphologic characteristics in Europe [4]

    D. A. Godar et al.

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    5/14

    Fig. 2 Cutaneous

    manifestations of Lyme disease.

    ac The classic single annular

    patch of erythema with central

    clearing. Annular erythema

    migrans surrounds tick in image

    (c). d Atypical presentation of

    erythema migrans with a

    homogeneous, expanding

    erythematous patch; in this case

    with central necrotic eschar,

    hemorrhage, and vesiculation.

    (Adapted from Wetter and Ruff

    [20]. Used with permission.) e

    g Multiple dusky patches of

    erythema migrans that may be

    associated with spirochetemia

    (eg) and which may become

    nearly confluent (g). h An

    infiltrated red plaque due to a

    lymphocytoma. i, j Cigarette-

    paper changes in acrodermatitis

    chronica atrophicans, rarely

    seen in the United States

    Lyme Disease Review

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    6/14

    histopathologic evaluation often shows nonspecific features

    (Table3; Fig. 3). If high clinical suspicion exists, Warthin-

    Starry and Steiner stains may be used to demonstrate

    Borreliaspirochetes in the skin or internal organs (Fig. 3d)

    and tissue culture may be performed on skin specimens

    obtained from EM lesions. Although borrelial organisms

    can be visualized on tissue samples with special stains, this

    is not a standard means for the diagnosis of Lyme disease.Because this is not a routinely used method of diagnosing

    EM, sensitivity and specificity information regarding

    visualization of Borrelia spirochetes in the skin is not

    readily available. Mitchell et al [28] described a case series

    in which spirochetes were able to be visualized in 24 of 34

    specimens of patients with confirmed Lyme disease.

    4.2 Tissue Testing

    4.2.1 Culture

    Culture to grow and detect the Borreliaspirochetes can beperformed on skin, blood, or cerebrospinal fluid (CSF).

    Tissue culture from skin requires an EM eruption and skin

    biopsy. The skin biopsy specimen should be obtained from

    the edge of the EM lesion [4].Borreliatypically is cultured

    on modified Kelly-Pettenkofer and Barbour-Stoenner-

    Kelly II media [29]. Culture is specific but the sensitivity is

    poorly defined [19,30]. Recovery of the organism is more

    likely when the EM lesion has been present for a short

    duration and generally cannot be recovered in patients

    taking oral antibiotics [30]. Yield rates on culture from skin

    samples reportedly range from 40 to 86 % [30]. Culture has

    limitations. It can only be performed on untreated patients

    [30]. Also, it is labor intensive and can take up to 3 months

    to yield negative results, which substantially limits its

    clinical utility [1, 30].

    4.2.2 Polymerase Chain Reaction

    Polymerase chain reaction (PCR) is an excellent test early

    in Lyme disease and can be performed using sera, CSF,

    synovial fluid, or even skin obtained through biopsy [31].

    Specificity approaches 100 % and the test may yield

    positive results in early Lyme disease, before the serologic

    testings are positive [3032]. Sensitivity ranges from 36 to88 % in samples taken from EM lesions [30]. PCR has

    reduced sensitivity due to destruction of the DNA during

    sample processing or transport [30]. To minimize this

    effect, DNA should be extracted shortly after collecting the

    sample [30]. PCR also has decreased sensitivity because of

    lack of conservation of target genes between different

    Borrelia species [3032]. PCR sensitivity is better when

    performed on skin and synovial fluid samples than blood or

    CSF samples [32]. Unfortunately, PCR may not be widely

    available and has a risk of unreliable results due to lack of

    standardization [32].

    4.3 Lyme Disease-Specific Serologic Testing

    Serologic testing is indicated when all of the following

    conditions are true [25]:

    1. Recent history of living in or traveling to an area

    endemic for Lyme disease

    2. Risk factor for tick exposure

    3. Symptoms concerning for disseminated Lyme disease

    (eg, neurologic symptoms, carditis, arthritis)

    Serologic testing is not indicated for patients with an

    EM rash or when the pretest probability exceeds 80 % [ 4].

    These patients should be treated empirically. Serologic

    testing should not be obtained for patients with nonspecific

    symptoms.

    Initial screening may be performed with an enzyme-

    linked immunosorbent assay (ELISA) or an indirect fluo-

    rescent antibody (IFA) test. If the results from initial

    screening tests are equivocal or positive, the diagnosis

    should be confirmed with a Western blot. Patients with a

    positive ELISA and negative Western blot should be con-

    sidered negative for Lyme disease [4]. ELISA and IFA

    tests are both limited by false-positive and false-negative

    results. Early in the disease course, these tests may be

    falsely negative. False-positive results occur in approxi-

    mately 5 % of cases and have been reported in patients

    with Epstein-Barr virus infection and syphilis infection, as

    well as autoimmune conditions [4,33]. Thus, testing is not

    recommended when the pretest probability of Lyme dis-

    ease is low [4]. The sensitivity of these tests is good; the

    specificity is excellent and is discussed in further detail

    herein. Serologic testing for Lyme disease has limitations.

    It often shows negative results early in the disease, cannot

    Table 3 Histopathologic features of Lyme disease

    Clinical finding Pathologic correlation

    Erythema migrans Thickened stratum corneum

    Epidermal atrophy

    Focal eosinophilic spongiosis

    Deep dermal perivascularlymphocytes

    Plasma cells

    Loss of pilosebaceous units

    Lymphocytoma Dense dermal lymphocytic

    infiltrate

    Infiltrate containing B and T cells

    Acrodermatitis chronica

    atrophicans

    Sclerosis

    Plasma cells

    Data from Bhate and Schwartz [1]

    D. A. Godar et al.

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    7/14

    reliably distinguish between active and past infection, and

    cannot be used to monitor treatment response [32]. Of note,

    more than 70 types of immunoassay have been approved

    by the US Food and Drug Administration for use in

    detecting B. burgdorferi [30]. The pros and cons of the

    different serologic tests are detailed in Table 4.

    4.3.1 Enzyme-Linked Immunosorbent Assay

    ELISA is the most commonly used initial test for Lymedisease. ELISA detects immunoglobulin M (IgM) and

    immunoglobulin G (IgG) antibodies toB. burgdorferi, with

    IgM antibodies usually detected 2 weeks after exposure

    and IgG positivity at 4 weeks after exposure [1, 34]. IgM

    testing should be used only during the first 4 weeks of

    infection [1, 35]. Prolonged IgM positivity may correlate

    with more severe disease [34]. IgG levels typically stay

    elevated for 216 weeks after remission of Lyme disease

    [34]. ELISA traditionally has targeted the entire organism;

    however, this targeting increases the rate of false positives

    because of antigens cross-reacting with other bacteria [30].

    Recently, more targeted assays with v1sE and C6 have

    been developed and have considerably enhanced the sen-

    sitivity and specificity of ELISA [36]. Several enzyme

    immunoassays are commercially available, and each differs

    slightly in its sensitivity and specificity. The specificity of

    the traditional 2-tier testing with sonicate ELISA and

    confirmatory Western blot has been estimated at 99 % [35].

    The sensitivity of the 2-tier testing with IgM or IgG is29 % during acute phase EM, 64 % during convalescent

    phase EM, and 100 % in patients with neurologic, cardiac,

    or joint manifestations of Lyme disease [35]. The sensi-

    tivity and specificity of the IgG v1sE C6 peptide was

    comparable in the discussion of Steere et al [35], but

    according to Liang et al [37], it was improved at 74, 8590,

    and 100 % for acute, convalescent, and late phase speci-

    mens, respectively. The advantage of ELISA testing is that

    it is automated and thereby less expensive [32]. ELISA

    Fig. 3 Histopathologic features of Lyme disease. a Superficial

    lymphoplasmacytic inflammation in erythema migrans (H&E 94).

    b Deep perivascular lymphoplasmacytic inflammation in erythema

    migrans (H&E 940). c Dense collections of lymphocytes in the

    dermis in a lymphocytoma (H&E 94).d Spirochetes (arrow) shown

    on WarthinStarry stain of cardiac tissue of a patient with Lyme

    carditis (WarthinStarry 9158). (Adapted from Centers for Disease

    Control and Prevention [CDC]. Three sudden cardiac deaths associ-

    ated with Lyme carditis: United States, November 2012July 2013.

    MMWR Morb Mortal Wkly Rep. 2013 Dec 13;62(49):9936. http://

    www.cdc.gov/mmwr/preview/mmwrhtml/mm6249a1.htm )

    Lyme Disease Review

    http://www.cdc.gov/mmwr/preview/mmwrhtml/mm6249a1.htmhttp://www.cdc.gov/mmwr/preview/mmwrhtml/mm6249a1.htmhttp://www.cdc.gov/mmwr/preview/mmwrhtml/mm6249a1.htmhttp://www.cdc.gov/mmwr/preview/mmwrhtml/mm6249a1.htm
  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    8/14

    results also are quantitative because they correlate with the

    amount of antibody response [30]. The interpretation of

    ELISA results is detailed in Table 5.

    4.3.2 Indirect Fluorescent Antibody Testing

    IFA testing uses immunofluorescence to test for the pre-

    sence of antibodies in the serum of affected patients. This

    test is not automated and thus its reliability is operator-

    dependent [32]. Therefore, ELISA is more sensitive than

    IFA and thus is generally the preferred initial screening test[34].

    4.3.3 Western Blot

    Western blot is more sensitive and more specific than both

    ELISA and IFA; however, given the greater costs and labor

    associated with performing this test, it currently is used as

    confirmatory and not as an initial test [32]. According to

    the CDC, at least 2 of 3 bands must be present for a

    positive IgM blot and at least 5 of 10 bands must be present

    for a positive IgG blot [32,38].

    4.4 Other Laboratory Findings

    According to the CDC, general laboratory findings may

    include elevated erythrocyte sedimentation rate, mildly

    elevated levels of hepatic transaminases, and microscopic

    hematuria or proteinuria. Complete blood cell count (CBC)

    and renal function tests usually show normal findings [4].

    In patients with neurologic symptoms (eg, headache, nervepalsies, neck pain or stiffness), CNS involvement should be

    suspected and a spinal tap performed. CSF typically

    demonstrates lymphocytic pleocytosis, an elevated protein

    level, and a low glucose level in patients with CNS

    involvement; however, glucose levels can also be normal

    [4].

    Approximately 15 % of patients with Lyme disease may

    be coinfected with a second tick-borne illness. Swanson

    et al. [10] showed that in patients with multiple tick-borne

    Table 4 Pros and cons of diagnostic tests for Lyme disease

    Diagnostic

    test

    Advantages Disadvantages Comments

    ELISA Strong sensitivity

    Cost-effective

    Fast

    Variation between strains of different

    geographic origin

    False positive with EBV, malaria, syphilis,

    other spirochetal illnesses, and

    autoimmune diseases

    False negative early in disease

    Does not reliably distinguish past and

    active infection

    False-positive IgM testing is more common

    than false-positive IgG testing

    5 % of normal US population test positive on

    ELISAa

    IFA Good sensitivity Variation between strains of different

    geographic origin

    False positive with EBV, syphilis, and

    autoimmune disease

    False negative early in disease

    Does not reliably distinguish past and

    active infection

    Operator-dependent

    Westernblot More specific than ELISA and IFAtest Expensive

    Tissue

    PCR

    Can be performed on various sample

    sources (eg, blood, CSF, synovial

    fluid, skin)

    Poor sensitivity

    Generally not recommended because of

    limited duration of circulating organisms

    in the blood

    Tissue

    culture

    High specificity Poor sensitivity

    Time- and labor-intensive; expensive

    May be helpful in cases of suspected

    reinfection because serologic testing may

    be difficult to interpret

    Histologic

    analysis

    Can rule out other dermatologic

    diseases on differential diagnosis

    Nonspecific findings WarthinStarry and Steiner stains can be used

    to confirm presence ofBorrelia

    CSFcerebrospinal fluid, EBVEpsteinBarr virus, ELISA enzyme-linked immunosorbent assay, IFA indirect fluorescent antibody, IgG immu-

    noglobulin G, IgMimmunoglobulin M, PCR polymerase chain reactiona

    Table4 describes standard ELISA testing. Comparison of v1sE testing and traditional 2-tier ELISA is discussed in the text

    D. A. Godar et al.

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    9/14

    illnesses, 81 % had both borreliosis and babesiosis, 9 %

    had borreliosis and anaplasmosis, and 5 % had 3 tick-borneillnesses. The decision regarding further laboratory testing

    for these other organisms should be based on which tick-

    transmitted organisms are endemic to the area. If a patient

    presents with EM and treatment is initiated empirically,

    then serologic testing to document coinfection with A.

    phagocytophilum is not necessary because the therapy is

    effective for both organisms. A CBC, peripheral blood

    smear, and liver function testing (LFT) help to determine

    whether a coinfection is highly likely [10]. Patients with

    anaplasmosis often have laboratory abnormalities, such as

    leukopenia, lymphopenia, granulocytopenia, and elevated

    LFTs [10].

    Patients with babesiosis often have evidence of anemia.

    BothB. microti and A. phagocytophilum can be visualized

    with Giemsa stain [10]. If coinfection is strongly suspected

    on the basis of CBC and LFT findings, then PCR for other

    tick-transmitted organisms can be obtained to confirm the

    diagnosis in patients whose specimen smears are negative

    [10].

    5 Differential Diagnosis

    Lyme disease must be distinguished from other tick-borne

    illnesses, such as ehrlichiosis, southern tick-associated rash

    illness, and babesiosis [1]. Fever, headache, and arthralgias

    are common symptoms seen in these conditions.

    The differential diagnosis of EM is broad (Table6). Enti-

    ties in the differential of EM include exaggerated arthropod

    reaction, cellulitis, granuloma annulare, subacute cutaneous

    lupus erythematosus, and many other considerations.

    The most important considerations are to distinguish

    EM from a tick-bite hypersensitivity reaction or cellulitis.

    Hypersensitivity reactions to tick bites generally occur

    within 48 h of tick detachment, are usually less than 5 cm

    in diameter, and begin to resolve within 2448 h of onset

    [3]. This response is in contrast to the EM lesion, which

    generally appears later, is larger than 5 cm by definition,

    and continues to expand rather than resolve [3]. Unlike

    cellulitis, EM lesions are not usually indurated or warm or

    painful to the touch. They also are not typically vesicular orpruritic, features that are more typical of acute allergic

    contact dermatitis. In addition, EM lesions are not usually

    scaly, a characteristic observed in cases of dermatitis or

    fungal infection.

    6 Management

    The first-line treatment of early localized disease is doxy-

    cycline at 100 mg orally every 12 h for 1421 days

    (Table7). Doxycycline should be avoided in children

    younger than 8 years and in pregnant women [3]. Of note,unfavorable fetal outcomes have been reported in pregnant

    women who acquire Lyme disease [32]. Children younger

    than 8 years should be treated with 50 mg/kg of amoxi-

    cillin per day and adults should be treated with 500 mg

    orally 3 times a day as an alternative [4]. In patients with

    allergies to penicillin or unable to take tetracyclines, ce-

    furoxime 500 mg orally twice a day for adults or erythro-

    mycin 250 mg orally 4 times a day may be used for the

    same duration [4]. Early disseminated disease should be

    treated for 21 days, and ACA is generally treated for

    1 month [4]. Approximately 15 % of patients may have a

    Jarisch-Herxheimer reaction within the first 24 h of treat-

    ment [4]. These patients had worsening malaise with

    sweating and rigors that resolved spontaneously [4].

    Patients who have persistent or recurrent EM, persistent

    borreliosis, or major sequelae after appropriate treatment

    should be re-treated [19]. Patients with a longer duration of

    EM lesions or lesions of larger size are more likely to have

    persistent positive antiborrelial IgG after antibiotic treat-

    ment [19]. Patients with previous Lyme infection have no

    protection against repeat infection. Conversely, patients

    who continue to experience subjective symptoms, such as

    pain, fatigue, or altered cognition, will not improve with

    recurrent antibiotic treatment because these symptoms are

    not caused by persistence of the spirochete [19, 39].

    Instead, they are considered to be part of postLyme dis-

    ease syndrome and may persist for a year or longer after

    treatment of the initial infection [19].

    For patients who have coinfection with A. phagocyto-

    philum, doxycycline 100 mg orally twice daily is the

    treatment of choice, even in young children [10]. Doxy-

    cycline should still be avoided in pregnancy. Rifampin and

    levofloxacin can be considered alternative treatments in

    Table 5 General interpretation of ELISA testing in Lyme disease

    ELISA finding Interpretationa

    IgM?, IgG- Early infection; isolated IgM? with no IgG

    seroconversion after repeat testing is a nonspecific

    finding and suggestive of a false-positive IgM

    IgM?, IgG? Suggestive of active infection; obtain confirmatory

    testing

    IgM-, IgG- Negative testing rules out Lyme disease; if testing

    obtained within 2 wk of tick bite, testing may be

    repeated 4 wk later, and if it is still negative,

    results are consistent with no disease

    IgM-, IgG? IgG levels may persist, indicating exposure, but do

    not necessarily indicate active disease

    ELISA enzyme-linked immunosorbent assay, IgG immunoglobulin G,

    IgMimmunoglobulin Ma The interpretation does not account for pretest probability and the

    sensitivity and specificity of the test performed for stage of disease

    Lyme Disease Review

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    10/14

    necessary situations; however, little to no data exist

    regarding their efficacy in a clinical setting [10]. Of note,

    A. phagocytophilum is resistant to b-lactams, macrolides,

    sulfa antibiotics, clindamycin, and aminoglycosides [10].

    Treatment for at least 14 days is recommended in cases of

    coinfection with B. burgdorferi and A. phagocytophilum

    [10].

    Patients with babesiosis often have a mild, self-limitedinfection without need for treatment [10]. If they are

    symptomatic, oral clindamycin 600 mg 3 times per day can

    be administered for adults and 2040 mg/kg per day

    divided in 3 daily doses given to children. In addition, oral

    quinine 650 mg 3 times per day for adults and 25 mg/kg

    per day divided in 3 daily doses for children can be con-

    sidered. For severely ill patients, intravenous clindamycin

    300600 mg every 6 h is recommended for adults; for

    severely ill children, 2040 mg/kg per day and oral quinine

    are recommended [10]. Nevertheless, a notable amount of

    morbidity is associated with this treatment, with approxi-mately 72 % of patients having adverse reactions, includ-

    ing tinnitus, diarrhea, and hearing loss [10]. An alternative

    regimen includes atovaquone 750 mg twice daily with

    Table 6 Differential diagnosis for the cutaneous manifestations of Lyme disease

    Differential diagnosis Characteristics that distinguish from Lyme disease

    Clinical Pathologic Laboratory

    Erythema migrans

    Tinea corporis Scaly annular plaque

    KOH test positive for fungal elements

    Fungal stains positive for fungal

    elements

    No significant laboratory

    findingsGranuloma annulare Smooth, indurated, nonscaly annular plaques

    Asymptomatic to mildly pruritic

    Palisading granulomas with altered

    collagen and mucin

    No abnormalities

    Cellulitis Tender, indurated plaques

    Associated fever or malaise

    Leukocyte infiltration, capillary

    dilatation, and bacterial invasion

    Leukocytosis

    Elevated levels of

    inflammatory markers

    Fixed drug eruption Dusky plaques

    Typically with medication history

    Vacuolar interface dermatitis with

    pigment incontinence

    None

    Hypersensitivity or

    exaggerated arthropod

    bite reaction

    Develops within 48 h of tick bitea

    Usually more pruritic or more painful than

    erythema migrans

    Resolves spontaneously in a few days

    Clears from periphery rather than centrally

    Increased risk in persons with hematologic

    cancers

    Eosinophilic spongiosis None

    Allergic contact dermatitis Lichenification or vesiculation, depending on

    acuity

    Geometric borders

    Pruritus

    Eosinophilic spongiosis None

    Erysipelas Usually associated with fever and chi lls

    Well-defined raised border

    Firm and painful to the touch

    Dermal edema, vascular dilatation,

    and neutrophilic inflammation

    Elevated ESR and CRP

    levels

    Leukocytosis

    Disseminated erythema migrans

    Tuberculoid and borderline

    forms of leprosy

    Well-circumscribed erythematous to

    violaceous or hypopigmented macules or

    plaques; lesions in borderline forms have a

    more punched-out shape

    History of exposure

    Lesions with associated anesthesia

    Lesions possibly associated with alopecia and

    scaling

    Affecting cooler body parts

    Perineural granulomas

    Foamy macrophages

    Positive results on Fite and AFBstains

    Serologic assays to detect

    phenolic glycolipid-1

    (specific for

    Mycobacterium leprae) andlipoarabinomannan

    (common in mycobacteria)

    SCLE Annular plaques with or without scale

    Photodistributed

    May be associated with arthralgias and malaise

    Interface dermatitis with

    perivascular and periadnexal

    lymphocytes

    Positive SSA antibodies

    D. A. Godar et al.

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    11/14

    Table 6 continued

    Differential diagnosis Characteristics that distinguish from Lyme disease

    Clinical Pathologic Laboratory

    Erythema infectiosum Slapped cheeks rash followed by

    development of a reticular maculopapular

    rash

    Rash often pruriticAssociated with joint pains, especially in

    adults

    Nonspecific Positive PCR or

    immunoassay testing for

    parvovirus

    Erythema multiforme Associated with herpes simplex virus,

    medications, and mycoplasma

    Targetoid lesions with mucosal involvement

    common

    Painful lesions

    Partial to full-thickness epidermal

    necrosis with vacuolar interface

    dermatitis and intraepidermal

    vesiculation

    None

    Erythema annulare

    centrifugum

    Annular lesions with trailing scale Tight perivascular lymphocytic

    inflammation

    None

    Urticarial vasculitis Urticarial plaques with burning sensations

    Purpura common

    Leukocytoclastic vasculitis May have

    hypocomplementemia

    Urticaria Transient and recurrent wheals

    Pruritus

    Superficial perivascular infiltrate

    with eosinophils and lymphocytes

    None

    Erythema marginatum Polycyclic, asymptomatic eruption

    Preceding streptococcal infection

    Additional manifestations of rheumatic fever

    (eg, carditis, arthritis, fever)

    Scant neutrophilic perivascular

    infiltrate without vasculitis

    Elevated ESR and CRP

    levels

    Prolonged PR interval on

    ECG

    Positive streptococcal

    culture or rising

    antistreptolysin O or anti-

    DNase B titer

    Lymphocytoma

    Lymphoma Firm plum-colored nodules Atypical lymphocytic infiltration Variable CBC and bone

    marrow biopsy findings

    Sarcoidosis Reddish-brown papules and plaques with

    minimal epidermal change

    Lesions in old scars

    Erythema nodosum

    Noncaseating granulomas Elevated ESR

    Hypercalciuria

    Leukopenia uncommon but

    possible

    Increased ACE level

    Acrodermatitis chronica atrophicans

    Morphea Insidious devel opment of indurat ed violaceous

    plaques

    Thickening and homogenization of

    collagen bundles, with loss of

    subcutaneous fat and trapping of

    eccrine glands

    Peripheral eosinophilia

    possible

    Antinuclear, anticentromere,

    anti-Scl70,

    antiphospholipid,

    antitopoisomerase,

    antihistone, antiMMP-1,

    anti-dsDNA and anti-

    ssDNA antibodies possible

    Lichen sclerosus et

    atrophicus

    Atrophic white plaques common in genital

    areas; possible in extragenital sites

    Often pruritic in genital locations

    Atrophic epidermis

    Papillary dermal pallor

    Lymphoid band beneath zone of

    pallor

    None

    Superficial

    thrombophlebitis

    Painful subcutaneous nodules Thrombi within veins, surrounded

    by inflammation

    Possible hypercoagulability

    ACEangiotensin-converting enzyme, AFB acid-fast bacteria,CBCcomplete blood cell count, CRP C-reactive protein, DEJdermal-epidermal junction,

    ECGelectrocardiography,ESR erythrocyte sedimentation rate, KOHpotassium hydroxide, MMP-1matrix metalloproteinase-1, PCR polymerase chain

    reaction,SCLEsubacute cutaneous lupus erythematosusa Information from Wormser et al. [3]

    Lyme Disease Review

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    12/14

    meals for adults and 20 mg/kg twice daily with meals for

    children [10]. Azithromycin 500 to 1,000 mg daily has also

    had success and is better tolerated [10]. Patients with

    severe hemolysis may require exchange transfusions [10].Of note, babesiosis treatment is not effective for Lyme

    disease or anaplasmosis.

    7 Prevention

    7.1 Tick Bite Avoidance

    Avoidance of tick bites with use of insect repellant and

    protective clothing is recommended for all patients who

    choose to engage in outdoor activities in endemic areas.

    Insect repellents most effective against ticks include trans-

    p-menthane-3,8-diol and N,N-diethyl-3-methylbenzamide

    (DEET) [40]. Synthetic repellants including picaridin and

    IR3535 are less effective [40]. Trans-p-menthane-3,8-diol

    is the most highly repellent substance for ticks, and

    repellency is long-lived, lasting for several days [40]. Its

    use is not associated with toxicity [40]. In contrast, labo-

    ratory testing of DEET has shown repellency of up to

    80100 % with concentrations of 30 %, but the effect is

    relatively short-lived, lasting 25 h [40]. DEET has been

    unpopular in the general public also because of reports of

    seizures in children; however, research suggests that this

    adverse effect is more of a concern with ingested DEET

    than topically applied DEET [40]. DEET has also been

    used safely in pregnancy [40].

    Protective clothing treated with permethrin has been

    effective in repelling ticks [40]. In a field study of outdoor

    workers, researchers found a 93 % reduction in the number

    of tick bites in persons wearing permethrin-treated clothing

    compared with controls [40]. It is best to buy clothing in

    which the permethrin is polymerized to the clothing fibers

    as this is more long lasting and resists washing compared

    with clothing dipped in permethrin, which must be

    retreated every 20 washes [40]. Clothing treated with

    DEET is generally less effective [40].

    7.2 Tick Removal

    Following exposure to a tick-infested area, complete

    inspection of skin and scalp should be performed and ticksremoved promptly. Patients should be advised to remove

    ticks immediately using a fine-tipped tweezers or forceps to

    grasp the tick as close to the skin as possible and with

    steady upward pulling, avoiding twisting movements [40].

    The forceps method is the recommended tick removal

    strategy from several organizations. Methods using petro-

    leum jelly, nail polish, or other substances to suffocate the

    tick are generally ineffective because the tick has a very

    low respiratory rate [40]. Of note, some laboratory studies

    have not found a significant difference in risk of trans-

    mission with methods that squeeze the tick vs pull the tick;

    however, it is still recommended to remove ticks with theforceps method [40,41].

    The guidelines of the Infectious Diseases Society of

    America cite that after a tick bite, a sole 200-mg dose of

    doxycycline should be administered to adults and to chil-

    dren age 8 years or older when all of the following situa-

    tions exist [3]:

    1. An attached tick is reliably identified as an adult or

    nymphal Ixodes scapularis and has been attached for

    an estimated 36 hours or more, based on the extent of

    its blood engorgement or certainty about the time of

    tick exposure2. Prophylaxis is started within 72 h of tick removal

    3. Ecologic information indicates a local infection rate of

    20 % or more for B. burgdorferi in ticks

    4. Doxycycline use is not contraindicated

    No recommended prophylaxis is available for children

    younger than 8 years or for pregnant women because no

    trials have been performed in these patient groups [4].

    Instead, they should be observed for EM development and

    should be treated if necessary [4].

    7.3 Vaccines

    Only 1 human-targeted Lyme disease vaccineLYMEr-

    ixhas ever been approved by the US Food and Drug

    Administration. LYMErix was a recombinant protein

    vaccine against OspA that became available commercially

    in 1998 [42]. The vaccine was efficacious and well toler-

    ated but required multiple boosters, and there were con-

    cerns regarding potential risk of autoimmunity; thus it was

    removed from the US market in 2002 because of poor sales

    [42].

    Table 7 Treatment regimens for Lyme disease

    Cutaneous

    manifestation

    First choice

    treatment

    Alternative

    treatment

    Early localized disease

    (solitary erythema

    migrans)

    Doxycycline

    100 mg PO Q12h

    for 1421 daysa

    Amoxicillin

    500 mg PO Q8h

    for 1421 days

    Early disseminated

    disease (multiple

    erythema migrans;

    lymphocytoma)

    Doxycycline

    100 mg PO Q12h

    for 1428 daysa

    Amoxicillin

    500 mg PO Q8h

    for 1428 days

    Chronic disease

    (acrodermatitis

    chronica atrophicans)

    Ceftriaxone 2 g IV

    once daily for

    1428 days

    Cefotaxime 2 g IV

    Q8h for

    1428 days

    IV intravenously, PO orally, Q8h every 8 h, Q12h every 12 ha Doxycycline should be avoided for children\8 years old and for

    pregnant women

    D. A. Godar et al.

  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    13/14

    Vaccines targeting the tick and its reservoir have been

    postulated and investigated; however, numerous obstacles

    have led to little progress on this front [42].

    8 Conclusions

    Lyme disease has varied cutaneous manifestations thatdiffer among the 3 stages of Lyme disease, and the

    approach to diagnosis and treatment is based on the patient,

    the region, and suspected coinfection with another tick-

    borne illness. Erythema migrans is the most common

    clinical presentation of Lyme disease and is a clinical

    diagnosis, although atypical cases require careful clinico-

    pathologic correlation to differentiate the cutaneous man-

    ifestations of Lyme disease from other dermatologic

    conditions. An understanding of the distribution of the

    Ixodes tick, its vectors, and the most likely dermatologic

    presentation based on these factors allows the dermatolo-

    gist to make appropriate testing and treatment recommen-dations for patients with suspected Lyme disease who are

    encountered in the general dermatologic setting.

    Acknowledgments No sources of funding were used to prepare this

    review. D. A. Godar, V. Laniosz, and D. A. Wetter have no conflicts

    of interest that are directly relevant to the content of this review.

    References

    1. Bhate C, Schwartz RA. Lyme disease: part I: advances and

    perspectives. J Am Acad Dermatol. 2011;64(4):61936.

    2. Burgdorfer W, Barbour AG, Hayes SF, Benach JL, Grunwaldt E,

    Davis JP. Lyme disease: a tick-borne spirochetosis? Science.

    1982;216(4552):13179.

    3. Wormser GP, Dattwyler RJ, Shapiro ED, Halperin JJ, Steere AC,

    Klempner MS, et al. The clinical assessment, treatment, and

    prevention of Lyme disease, human granulocytic anaplasmosis,

    and babesiosis: clinical practice guidelines by the Infectious

    Diseases Society of America. Clin Infect Dis. 2006;43(9):

    1089134 (Epub 2006 Oct 2. Erratum in: Clin Infect Dis. 2007

    Oct 1;45(7):941).

    4. Bratton RL, Whiteside JW, Hovan MJ, Engle RL, Edwards FD.

    Diagnosis and treatment of Lyme disease. Mayo Clin Proc.

    2008;83(5):56671.

    5. Brownstein JS, Holford TR, Fish D. Effect of climate change on

    Lyme disease risk in North America. Ecohealth. 2005;2(1):3846.

    6. Nahimana I, Gern L, Blanc DS, Praz G, Francioli P, Peter O. Risk

    of Borrelia burgdorferi infection in western Switzerland fol-

    lowing a tick bite. Eur J Clin Microbiol Infect Dis.

    2004;23(8):6038 (Epub 2004 Jul 24).

    7. Nadelman RB, Nowakowski J, Fish D, Falco RC, Freeman K,

    McKenna D, Tick Bite Study Group, et al. Prophylaxis with

    single-dose doxycycline for the prevention of Lyme disease

    after an Ixodes scapularis tick bite. N Engl J Med. 2001;345(2):

    7984.

    8. Ginsberg HS. Transmission risk of Lyme disease and implica-

    tions for tick management. Am J Epidemiol. 1993;138(1):6573.

    9. Krause PJ, McKay K, Thompson CA, Sikand VK, Lentz R, Le-

    pore T, Deer-Associated Infection Study Group, et al. Disease-

    specific diagnosis of coinfecting tickborne zoonoses: babesiosis,

    human granulocytic ehrlichiosis, and Lyme disease. Clin Infect

    Dis. 2002;34(9):118491 (Epub 2002 Apr 4).

    10. Swanson SJ, Neitzel D, Reed KD, Belongia EA. Coinfections

    acquired from Ixodes ticks. Clin Microbiol Rev. 2006;19(4):

    70827.

    11. Yang XF, Pal U, Alani SM, Fikrig E, Norgard MV. Essential role

    for OspA/B in the life cycle of the Lyme disease spirochete.

    J Exp Med. 2004;199(5):6418 (Epub 2004 Feb 23).

    12. Pal U, Yang X, Chen M, Bockenstedt LK, Anderson JF, Flavell

    RA, et al. OspC facilitates Borrelia burgdorferi invasion of

    Ixodes scapularis salivary glands. J Clin Invest. 2004;113(2):

    22030.

    13. Seemanapalli SV, Xu Q, McShan K, Liang FT. Outer surface

    protein C is a dissemination-facilitating factor ofBorrelia burg-

    dorferi during mammalian infection. PLoS One. 2010;5(12):

    e15830.

    14. Ramamoorthi N, Narasimhan S, Pal U, Bao F, Yang XF, Fish D,

    et al. The Lyme disease agent exploits a tick protein to infect the

    mammalian host. Nature. 2005;436(7050):5737.

    15. Sood SK, Salzman MB, Johnson BJ, Happ CM, Feig K, Carmody

    L, et al. Duration of tick attachment as a predictor of the risk of

    Lyme disease in an area in which Lyme disease is endemic.

    J Infect Dis. 1997;175(4):9969.

    16. van Dam AP, Oei A, Jaspars R, Fijen C, Wilske B, Spanjaard L,

    et al. Complement-mediated serum sensitivity among spirochetes

    that cause Lyme disease. Infect Immun. 1997;65(4):122836.

    17. Zhang JR, Norris SJ. Kinetics and in vivo induction of genetic

    variation of vlsE in Borrelia burgdorferi. Infect Immun. 1998;66

    (8):368997 (Erratum in: Infect Immun 1999 Jan;67(1):468).

    18. Eisendle K, Zelger B. The expanding spectrum of cutaneous

    borreliosis. G Ital Dermatol Venereol. 2009;144(2):15771.

    19. Glatz M, Golestani M, Kerl H, Mullegger RR. Clinical relevance

    of different IgG and IgM serum antibody responses to Borrelia

    burgdorferi after antibiotic therapy for erythema migrans: long-

    term follow-up study of 113 patients. Arch Dermatol. 2006;142

    (7):8628.

    20. Wetter DA, Ruff CA. Erythema migrans in Lyme disease. CMAJ.

    2011;183(11):1281 (Epub 2011 May 30).

    21. Mullegger RR, Glatz M. Skin manifestations of Lyme borreliosis:

    diagnosis and management. Am J Clin Dermatol. 2008;9(6):35568.

    22. Glatz M, Fingerle V, Wilske B, Ambros-Rudolph C, Kerl H,

    Mullegger RR. Immunoblot analysis of the seroreactivity to

    recombinant Borrelia burgdorferi sensu lato antigens, including

    VlsE, in the long-term course of treated patients with erythema

    migrans. Dermatology. 2008;216(2):93103 (Epub 2008 Jan 23).

    23. Jones KL, Muellegger RR, Means TK, Lee M, Glickstein LJ,

    Damle N, et al. Higher mRNA levels of chemokines and cyto-

    kines associated with macrophage activation in erythema migrans

    skin lesions in patients from the United States than in patients

    from Austria with Lyme borreliosis. Clin Infect Dis. 2008;46(1):

    8592.24. Bolognia JL, Jorizzo JL, Schaffer JV, editors. Dermatology. 3rd

    ed. Philadelphia: Elsevier Saunders; 2012.

    25. Centers for Disease Control and Prevention. Tickborne diseases

    of the United States: a reference manual for health care providers

    [Internet]. 2014 (cited 2014 Aug 22). http://www.cdc.gov/lyme/

    resources/tickbornediseases.pdf.

    26. Malane MS, Grant-Kels JM, Feder HM Jr, Luger SW. Diagnosis

    of Lyme disease based on dermatologic manifestations. Ann

    Intern Med. 1991;114(6):4908.

    27. Centers for Disease Control and Prevention. National notifiable

    diseases surveillance system (NNDSS): Lyme disease (Borrelia

    burgdorferi): 2008 Case Definition [Internet]. Atlanta (GA):

    Lyme Disease Review

    http://www.cdc.gov/lyme/resources/tickbornediseases.pdfhttp://www.cdc.gov/lyme/resources/tickbornediseases.pdfhttp://www.cdc.gov/lyme/resources/tickbornediseases.pdfhttp://www.cdc.gov/lyme/resources/tickbornediseases.pdf
  • 7/25/2019 Lyme Disease Update for the General Dermatologist

    14/14

    Centers for Disease Control and Prevention; 2008 (updated 2014

    May 8; cited 2014 Aug 22). http://wwwn.cdc.gov/NNDSS/script/

    casedef.aspx?CondYrID=751&DatePub=1/1/2008%2012:00:00%

    20AM.

    28. Mitchell PD, Reed KD, Vandermause MF, Melski JW. Isolation

    ofBorrelia burgdorferi from skin biopsy specimens of patients

    with erythema migrans. Am J Clin Pathol. 1993;99(1):1047.

    29. Coulter P, Lema C, Flayhart D, Linhardt AS, Aucott JN, Auw-

    aerter PG, et al. Two-year evaluation of Borrelia burgdorferi

    culture and supplemental tests for definitive diagnosis of Lyme

    disease. J Clin Microbiol. 2005;43(10):50804 (Erratum in: J

    Clin Microbiol. 2007 Jan;45(1):277).

    30. Aguero-Rosenfeld ME, Wang G, Schwartz I, Wormser GP.

    Diagnosis of Lyme borreliosis. Clin Microbiol Rev. 2005;18(3):

    484509.

    31. Dunaj J, Moniuszko A, Zajkowska J, Pancewicz S. The role of

    PCR in diagnostics of Lyme borreliosis. Przegl Epidemiol.

    2013;67(1):359, 11923.

    32. Bhate C, Schwartz RA. Lyme disease: part II: management and

    prevention. J Am Acad Dermatol. 2011;64(4):63953.

    33. Magnarelli LA, Miller JN, Anderson JF, Riviere GR. Cross-

    reactivity of nonspecific treponemal antibody in serologic tests

    for Lyme disease. J Clin Microbiol. 1990;28(6):12769.

    34. Craft JE, Grodzicki RL, Steere AC. Antibody response in Lyme

    disease: evaluation of diagnostic tests. J Infect Dis. 1984;149(5):

    78995.

    35. Steere AC, McHugh G, Damle N, Sikand VK. Prospective study

    of serologic tests for Lyme disease. Clin Infect Dis. 2008;47(2):

    18895.

    36. Bacon RM, Biggerstaff BJ, Schriefer ME, Gilmore RD Jr, Philipp

    MT, Steere AC, et al. Serodiagnosis of Lyme disease by kinetic

    enzyme-linked immunosorbent assay using recombinant VlsE1 or

    peptide antigens ofBorrelia burgdorferi compared with 2-tiered

    testing using whole-cell lysates. J Infect Dis. 2003;187(8):1187

    99 Epub 2003 Apr 2.

    37. Liang FT, Steere AC, Marques AR, Johnson BJ, Miller JN,

    Philipp MT. Sensitive and specific serodiagnosis of Lyme disease

    by enzyme-linked immunosorbent assay with a peptide based on

    an immunodominant conserved region of Borrelia burgdorferi

    vlsE. J Clin Microbiol. 1999;37(12):39906.

    38. Centers for Disease Control and Prevention. Recommendations

    for test performance and interpretation from the second national

    conference on serologic diagnosis of Lyme disease. MMWR

    1995;44:5901. http://www.cdc.gov/mmwr/preview/mmwrhtml/

    00038469.htm.

    39. Klempner MS, Hu LT, Evans J, Schmid CH, Johnson GM,

    Trevino RP, et al. Two controlled trials of antibiotic treatment in

    patients with persistent symptoms and a history of Lyme disease.

    N Engl J Med. 2001;345(2):8592.

    40. Due C, Fox W, Medlock JM, Pietzsch M, Logan JG. Tick bite

    prevention and tick removal. BMJ. 2013;9(347):f7123.

    41. Kahl O, Janetzki-Mittmann C, Gray JS, Jonas R, Stein J, de Boer

    R. Risk of infection with Borrelia burgdorferi sensu lato for a

    host in relation to the duration of nymphal Ixodesricinus feeding

    and the method of tick removal. Zentralbl Bakteriol. 1998;287

    (12):4152.

    42. Embers ME, Narasimhan S. Vaccination against Lyme disease:

    past,present,and future. Front Cell Infect Microbiol. 2013;12(3):6.

    D. A. Godar et al.

    http://wwwn.cdc.gov/NNDSS/script/casedef.aspx?CondYrID=751&DatePub=1/1/2008%2012:00:00%20AMhttp://wwwn.cdc.gov/NNDSS/script/casedef.aspx?CondYrID=751&DatePub=1/1/2008%2012:00:00%20AMhttp://wwwn.cdc.gov/NNDSS/script/casedef.aspx?CondYrID=751&DatePub=1/1/2008%2012:00:00%20AMhttp://www.cdc.gov/mmwr/preview/mmwrhtml/00038469.htmhttp://www.cdc.gov/mmwr/preview/mmwrhtml/00038469.htmhttp://www.cdc.gov/mmwr/preview/mmwrhtml/00038469.htmhttp://www.cdc.gov/mmwr/preview/mmwrhtml/00038469.htmhttp://wwwn.cdc.gov/NNDSS/script/casedef.aspx?CondYrID=751&DatePub=1/1/2008%2012:00:00%20AMhttp://wwwn.cdc.gov/NNDSS/script/casedef.aspx?CondYrID=751&DatePub=1/1/2008%2012:00:00%20AMhttp://wwwn.cdc.gov/NNDSS/script/casedef.aspx?CondYrID=751&DatePub=1/1/2008%2012:00:00%20AM