gms | German Medical Science

GMS German Medical Science — an Interdisciplinary Journal

Association of the Scientific Medical Societies in Germany (AWMF)

ISSN 1612-3174

Cutaneous Lyme borreliosis: Guideline of the German Dermatology Society

Kutane Lyme-Borreliose: Leitlinie der Deutschen Dermatologischen Gesellschaft

Guideline Dermatology

  • corresponding author Heidelore Hofmann - Klinik für Dermatologie und Allergologie der TU München, München, Germany
  • Volker Fingerle - Bayerisches Landesamt für Gesundheit und Lebensmittelsicherheit (LGL) Oberschleißheim, Germany
  • Klaus-Peter Hunfeld - Zentralinstitut für Labormedizin, Mikrobiologie & Krankenhaushygiene, Krankenhaus Nordwest, Frankfurt, Germany
  • Hans-Iko Huppertz - Professor-Hess-Kinderklinik Klinikum Bremen-Mitte, Bremen, Germany
  • Andreas Krause - Immanuel Krankenhaus Berlin, Berlin, Germany
  • Sebastian Rauer - Neurologische Universitätsklinik, Freiburg, Germany
  • Bernhard Ruf - Klinik für Infektiologie Klinik St Georg, Leipzig, Germany
  • Consensus group

GMS Ger Med Sci 2017;15:Doc14

doi: 10.3205/000255, urn:nbn:de:0183-0002553

Received: August 9, 2017
Published: September 5, 2017

© 2017 Hofmann et al.
This is an Open Access article distributed under the terms of the Creative Commons Attribution 4.0 License. See license information at http://creativecommons.org/licenses/by/4.0/.


Abstract

This guideline of the German Dermatology Society primarily focuses on the diagnosis and treatment of cutaneous manifestations of Lyme borreliosis. It has received consensus from 22 German medical societies and 2 German patient organisations. It is the first part of an AWMF (Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften e.V.) interdisciplinary guideline: “Lyme Borreliosis – Diagnosis and Treatment, development stage S3”.

The guideline is directed at physicians in private practices and clinics who treat Lyme borreliosis. Objectives of this guideline are recommendations for confirming a clinical diagnosis, recommendations for a stage-related laboratory diagnosis (serological detection of IgM and IgG Borrelia antibodies using the 2-tiered ELISA/immunoblot process, sensible use of molecular diagnostic and culture procedures) and recommendations for the treatment of the localised, early-stage infection (erythema migrans, erythema chronicum migrans, and borrelial lymphocytoma), the disseminated early-stage infection (multiple erythemata migrantia, flu-like symptoms) and treatment of the late-stage infection (acrodermatitis chronica atrophicans with and without neurological manifestations). In addition, an information sheet for patients containing recommendations for the prevention of Lyme borreliosis is attached to the guideline.

Zusammenfassung

Diese Leitlinie der Deutschen Dermatologischen Gesellschaft bezieht sich primär auf die Diagnostik und Therapie von kutanen Manifestationen der Lyme-Borreliose. Sie ist Teil 1 der geplanten interdisziplinären AWMF Gesamtleitlinie „Lyme-Borreliose – Diagnostik und Therapie Entwicklungsstufe S3“.

Sie wurde interdisziplinär von 22 deutschen Fachgesellschaften und 2 deutschen Patientenorganisationen konsentiert und richtet sich an Ärzte in Praxis und Klinik, die mit der Behandlung der Lyme-Borreliose befasst sind.

Ziel der Leitlinie ist es, Empfehlungen zur Absicherung der klinischen Diagnosen, Empfehlungen zur stadiengerechten Labordiagnostik (serologischer Nachweis von IgM- und IgG-Borrelienantikörpern mit dem 2-Stufenverfahren ELISA/Immunoblot sowie der sinnvolle Einsatz molekulardiagnostischer und kultureller Verfahren) und Empfehlungen zur Therapie der lokalisierten Frühmanifestationen (Erythema migrans, Erythema chronicum migrans und Borrelienlymphozytom), zur Therapie der disseminierten Frühmanifestationen (Multiple Erythemata migrantia und/oder grippeartige Symptomatik nach Zeckenstich) und zur Therapie der Spätmanifestationen (Acrodermatitis chronica atrophicans mit und ohne neurologische Manifestationen) zu geben. Außerdem werden Empfehlungen für Patienten zur Prävention der Lyme-Borreliose und zur Nachbeobachtung eines Zeckenstiches formuliert.


Preamble

This guideline primarily focuses on the diagnosis and treatment of cutaneous manifestations of Lyme borreliosis. It is the first part of the scheduled interdisciplinary guideline: “Lyme Borreliosis – Diagnosis and Treatment, No. 013-080, Development Stage S3”.

This part has already received consensus from 22 medical societies and 2 patient organisations. The German Cochrane Centre, Freiburg (Cochrane Germany) is currently conducting systematic review and assessment of the literature to develop this guideline to stage 3.

The interdisciplinary guideline group is currently preparing part 2 “Neuroborreliosis” which will be followed by part 3 “Lyme Arthritis, Lyme Carditis and Other Rare Manifestations”.

Synonyms

Cutaneous borreliosis, cutaneous manifestations of Lyme borreliosis, skin borreliosis, cutaneous Lyme borreliosis, cutaneous Lyme disease

Search terms

Borrelia burgdorferi infection, hard-bodied tick borreliosis, Lyme disease, cutaneous Lyme borreliosis, erythema migrans disease, erythema migrans, erythema chronicum migrans, lymphadenosis cutis benigna, borrelial lymphocytoma, multiple erythemata migrantia, multiple erythema migrans, acrodermatitis chronica atrophicans.

ICD-10-No: A69.2, L90.4

AWMF Register No 013-044


List of abbreviations

  • ACA – Acrodermatitis chronica atrophicans
  • BL – Borrelial lymphocytoma
  • EM – Erythema migrans
  • ECM – Erythema chronicum migrans
  • i.v. – Intravenous
  • BW – Body weight
  • LB – Lyme borreliosis
  • LTT – Lymphocyte transformation test
  • MEM – Multiple Erythemata migrantia
  • MiQ – Quality standards in microbiological-infectiological diagnostics
  • NAT – Nucleic acid amplification techniques
  • NSAID – Nonsteroidal anti-inflammatory drugs
  • PCR – Polymerase chain reaction
  • p.o. – Per os
  • PPI – Proton pump inhibitor
  • RCT – Randomised controlled trial
  • SNRI – Serotonin-norepinephrine reuptake inhibitor

1 Introduction

The infectious disease most frequently transmitted by ticks in Europe is Lyme borreliosis. The Borrelia are transferred to the skin during the blood sucking process of the hard-bodied tick Ixodes ricinus. There the Borrelia are either killed off by the (unspecific, innate) immune system, or a localised infection occurs which leads to illness in only a small percentage of those infected. Most often there is an inflammation of the skin, typically in the form of an erythema migrans or, seldom, as borrelial lymphocytoma. In the course of the infection the Borrelia can disseminate and attack various organs. They primarily affect the skin, joints and nervous system. Acrodermatitis chronica atrophicans can develop as a chronic or late-form of skin manifestation.

1.1 Target group

This guideline is directed at physicians in private practices and clinics who treat Lyme borreliosis.

1.2 Objectives of this guideline

  • Recommendations for confirming a clinical diagnosis
  • Recommendations for a stage-related laboratory diagnosis: serological detection of IgM and IgG Borrelia antibodies using the 2-tiered ELISA/immunoblot process; sensible use of molecular-diagnostic and culture procedures
  • Treatment of the localised, early-stage infection (erythema migrans, erythema chronicum migrans and borrelial lymphocytoma)
  • Treatment of the disseminated early-stage infection (multiple erythemata migrantia, flu-like symptoms)
  • Treatment of the late-stage infection (acrodermatitis chronica without neurological manifestations)
  • Treatment of the late-stage infection (acrodermatitis chronica with neurological manifestations)
  • Prevention of Lyme borreliosis
    Recommendations for observing the area around the tick bite
    Information sheet for patients (Annex 1 in Attachment 1 [Attach. 1])

1.3 Participating medical societies

Steering group

  • Responsible:
    Prof. Dr. med. Heidelore Hofmann – coordinator
    German Dermatology Society (DDG)
  • Prof. Dr. med. Sebastian Rauer – coordinator, deputy Dr. Stephan Kastenbauer
    German Society of Neurology (DGN)
  • Dr. med. Volker Fingerle
    German Society for Hygiene and Microbiology (DGHM)
  • Prof. Dr. med. Klaus-Peter Hunfeld
    The German United Society of Clinical Chemistry and Laboratory Medicine (DGKL) and INSTAND e.V.
  • Prof. Dr. med. Hans-Iko Huppertz
    German Society of Paediatrics and Adolescent Medicine (DGKJ) and German Society of Paediatric Infectology (DGPI)
  • Prof. Dr. med. Andreas Krause
    German Society of Rheumatology (DGRh)
  • Prof. Dr. med. Bernhard Ruf
    German Society of Infectious Diseases (DGI)

Consensus group

  • Prof. Dr. med. Elisabeth Aberer
    Austrian Society of Dermatology and Venerology (ÖGDV)
  • Prof. Dr. med. Karl Bechter
    The German Association of Psychiatry, Psychotherapy and Psychosomatics (DGPPN)
  • Prof. Dr. med. Michael H. Freitag
    German College of General Practitioners and Family Physicians (DEGAM)
  • PD Dr. med. Gudrun Goßrau
    German Pain Society (DGSS)
  • Prof. Dr. med. Gerd Gross
    Paul Ehrlich Society for Chemotherapy (PEG)
  • Prof. Dr. med. Rainer Müller
    German Society of Oto-Rhino-Laryngology, Head and Neck Surgery (DGHNOKHC)
  • Dr. med. Kurt Müller / PD Dr. med. Walter Berghoff
    German Borreliosis Society (DBG)
  • Prof. Dr. med. Mathias Pauschinger
    German Society of Cardiology and Cardiovascular Research (DGK)
  • Prof. Dr. med. Monika A. Rieger
    German Society for Occupational and Environmental Medicine (DGAUM)
  • PD Dr. med. Rainer Schäfert
    German Society of Psychosomatic Medicine and Medical Psychotherapy (DGPM) and the German College of Psychosomatic Medicine (DKPM)
  • Prof. Dr. med. Stephan Thurau
    German Ophthalmological Society (DOG)
  • Prof. Dr. rer. nat. Reinhard Wallich
    German Society for Immunology (DGI)
  • Dr. Hendrik Wilking
    Robert Koch Institute (RKI)

Patient supporting groups

  • Ursula Dahlem
    Action Alliance Against Tick-Borne Infections Germany (OnLyme-Aktion)
  • Ute Fischer / Karin Friz
    Borreliosis and FSME Association Germany (BFBD)

Moderation

  • Prof. Dr. med. Ina B. Kopp
    AWMF Institute for Medical Knowledge Management

1.4 Methods

This guideline is based on an update of AWMF Guideline No. 013-044 “Cutaneous Manifestations of Lyme Borreliosis”, development stage S1, which was created by a committee of experts in 2009.

The guideline was created in accordance with the methodological requirements of the Association of the Scientific Medical Societies in Germany (AWMF) for developing and further developing diagnosis and treatment guidelines. It is an S2k guideline in accordance with the AWMF’s three-stage concept. The composition of the guideline group was interdisciplinary (IDA) and the appointed mandate holders of the expert medical societies were informed of the scheduled update on 11/2/2014.

Uniform formulations are used in order to standardise the recommendations of the guideline.

The following gradations shall apply here:

  • Strong recommendation: “shall”
  • Recommendation: “should”
  • Open recommendation: “may be considered”
  • Recommendation against an intervention: “should not”
  • Strong recommendation against an intervention: “shall not”

2 Microbiology of the pathogen

In Europe, 5 human-pathogenic genospecies from the Borrelia burgdorferi sensu lato complex have so far been isolated: B. afzelii is the most frequent, followed by B. garinii, B. bavariensis, B. burgdorferi sensu stricto and B. spielmanii [1], [2], [3], [4].

The human pathogenicity is still unclear for B. valaisiana, B. lusitaniae and B. bissettii. All of the species that have been ascertained to be human-pathogenic are found in Europe. Only B. burgdorferi sensu stricto is present in the USA, and all of the species are present in Asia except for B. burgdorferi sensu stricto. The various genospecies of the Borrelia burgdorferi sensu lato complex are genetically very heterogenic [5] and exhibit an organotropism in human infections. Erythema migrans is triggered by all 5 genospecies. Almost only B. afzelii is detected with acrodermatitis chronica atrophicans, B. garinii and B. bavariensis are often present in neurological manifestations, and B. burgdorferi sensu stricto mainly affects the joints [6]. B. spielmanii has so far only been isolated from erythema migrans [2], [7].


3 Epidemiology

Lyme borreliosis mainly exists between the 40th and 60th parallels of the northern hemisphere in line with the presence of its vectors. Few relevant epidemiological investigations have been conducted in Europe. A population-based study in southern Sweden reveals an incidence of 69 per 100,000 inhabitants [8]. In a prospective, population-based study of the region around Würzburg over a 12 month period, 313 cases of Lyme borreliosis were reported, which corresponds to an incidence of 111 per 100,000 inhabitants [9]. In terms of early manifestations, a localised erythema migrans was diagnosed in 89% of the cases and a disseminated erythema migrans in a further 3% of cases. Borrelial lymphocytoma was established in 2% of cases, early-stage neuroborreliosis in 3%, and carditis in <1%. In terms of late-stage forms of the disease, Lyme arthritis appeared in 5% of patients and acrodermatitis chronica atrophicans in 1%. No chronic neuroborreliosis was detected.

Currently nine states in Germany have an obligation to report acute manifestations of Lyme borreliosis (see Annex 4 in Attachment 1 [Attach. 1]). Epidemiological data obtained through this partial obligation to report are only based on the clearly diagnosable manifestations, such as erythema migrans, acute neuroborreliosis and acute Lyme arthritis. Thus, it can be assumed that the rate of incidence is considerably underreported [10], [11]. Secondary data analyses of health insurance data based on the ICD 10 coding A 69.2 (G) result in much higher rates of incidence [12].

Therefore, it can be concluded that the epidemiological data currently available is not sufficient for a definitive clarification. Data published up until now in Germany indicates the incidence of Lyme borreliosis to be somewhere between 60,000 to >200,000 cases per year.

In a major nation-wide seroprevalence study of children (KIGGS) and adults (DEGGS) it was shown that the percentage of Borrelia-specific antibodies in serum increases with increasing age of the population and already has an incidence rate of 7% in the group of 14 to 17 year olds. In adults, this percentage of Borrelia antibodies is even higher. In the group of 70 to 79 year olds, 24.5% of men and 16.4% of women are seropositive (Figure 1 [Fig. 1]) [13].

A prospective investigation of the incidence of Lyme borreliosis in Finland and southern Sweden (2008–2009) revealed that 78 (5%) of the 1,546 people bitten by a tick had a Borrelia burgdorferi infection. In 45 of the cases (3%) only a seroconversion occurred; 33 (2%) resulted in illness. Erythema migrans was diagnosed in 28 people, one person had borrelial lymphocytoma, two people had an acute case of neuroborreliosis and 2 had unspecified symptoms which were diagnosed as Lyme borreliosis [14].


4 Transmission routes

B. burgdorferi is transmitted to birds, mammals and humans from hard-bodied ticks of the I. ricinus/I. persulcatus spp. complex during the blood meal. In Europe this transmission is primarily from I. ricinus, in Asia from I. persulcatus and in the USA predominantly from I. scapularis. Ticks suck blood in the course of their cycle of development from larva to nymph to adult tick, and before they lay eggs. It is at this time that they can acquire and/or transmit Borrelia. Small rodents – particularly mice – and birds are the main reservoirs. Birds contribute to the geographical propagation of the infected ticks. In Germany, ticks are ubiquitously infected with Borrelia, however percentages can vary heavily from region to region, even between areas very close in proximity (e.g. 4–21% [15]).

The successful transmission from tick to mammal is the result of a specific, highly complex vector-pathogen interaction. First the Borrelia are activated in the tick’s intestines. Then they travel to the salivary glands where they bind immunosuppressive salivary proteins to their surface [16]. Finally, they are secreted with the saliva in the bite wound where they are at least partially protected from the host’s immune system by immunomodulating substances from the tick’s saliva which probably allows them to reach a sufficiently high infection doses. A similar transmission through blood-sucking insects is therefore close to impossible due to the short blood sucking time (lack of vector competence in insects for B. burgdorferi). Xenobiotic tests reveal that it can take hours for the Borrelia to be transferred – depending on the species of Borrelia [17].

When there is an occupationally higher risk of tick bites, cases of Lyme borreliosis (occupational disease No. 3102, diseases transmitted from animals to humans) should be reported to the accident insurer by the attending physician or employer as a work-related illness as per Art. 202 of the Social Security Code VII (see Annex 4 in Attachment 1 [Attach. 1]).


5 Pathogenesis

The pathogenesis of the borrelial infection is primarily determined by two factors:

1.
The evasion strategies of the pathogen [18], [19], [20].
2.
The quality of the host’s immune response [21], [22], [23], [24], [25], [26], [27], [28].

Moreover, salivary proteins that are released in the course of the tick’s blood meal also show immunosuppressive effects [29], [30], [31], [32], [33], [34], [35], [36], [37].

Host-specific inflammatory reactions in the skin also influence the course of the infection [38], [39].

Some of the many strategies the Borrelia use to evade the host’s immune system include the ability to mask their cell surface with proteins/inhibitors from the tick or the host, and to modify their phenotype expression of cell surface proteins (outer surface protein: osp) according to their environment [40], [41], [42], [43].

Several Borrelia species form a resistance to complement-mediated lysis by binding the regulators of the complement cascade (factor H) to their surface [44], [45], [46], [47], [48]. By binding to plasminogens, Borrelia are capable of breaking down collagen, fibronectin and laminin [47], [49], [50], [51], [52] and disseminating in the skin.

The innate immune system recognises the Borrelia mainly by their surface proteins (osp lipoproteins) [53], [54], [55], [56], [57]. This interaction leads to the activation of soluble factors, such as the complement system, as well as to the activation of target cells, like macrophages and dendritic cells, and to the induction of inflammatory cytokines [58], [59], [60], [61]. As the infection progresses, specific immune responses are generated, particularly the activation of T helper cells and B lymphocytes, and the production of Borrelia-specific antibodies [20], [62], [63], [64]. In reservoir hosts, like wild mice, the antibodies that form during an infection are able to prevent disease, however they are not able to eliminate the pathogen. In contrast, the antibodies that form in patients are often unable to prevent the disease. However, antibodies against certain Borrelia antigens have also been shown to protect against subsequent infection in humans (see vaccines).

There is no permanent immunity in humans after wild-type infection. Thus reinfection can occur.


6 Clinical manifestations of Lyme borreliosis

Lyme borreliosis is an inflammatory multi-organ disease. It manifests itself initially as a localised infection of the skin called erythema migrans. Because of its light symptoms, this early-stage inflammation of the skin can be overlooked or not even be visible. The Borrelia can spread haematogenically which is recognised clinically by flu-like symptoms or disseminated erythemas of the skin. As the disease progresses, manifestations can appear in other organs, with the nervous system and the joints primarily affected. The disease progresses very differently depending on the individual. Therefore, it doesn’t make sense to classify the disease into stages. A distinction between early and late manifestations is preferable since the clinical picture determines both the diagnosis and the treatment (Table 1 [Tab. 1]). European studies show that Lyme borreliosis manifests itself as a skin disease in 80–90% of patients and in other organs in around 10–20% of patients [8], [9], [10], [14], [65], [66].

6.1 Localised cutaneous early-stage infection

6.1.1 Erythema migrans

The skin around the infectious tick bite can become infected anywhere from 3 to 30 days after the tick bite occurs [67]. The extent and duration of the rash varies considerably between individuals. If the diameter of the erythema is more than 5 cm, a diagnosis of erythema migrans can be made (Figure 2a and b [Fig. 2]) [68].

The clinical picture of a typical erythema migrans is a marginated erythema that centrifugally spreads out around the tick bite (Figure 2c and d [Fig. 2]).

Features of a typical solitary erythema migrans

  • Free time interval between the tick bite and start of the erythema that is typically 3 days to several weeks. (Consensus: 18/20)
  • Increasing centrifugal spreading of the erythema (crescendo reaction). (Consensus: 17/20)
  • Marginated, non-raised erythema that is at least 5 cm in diameter. (Strong consensus: 20/20)
  • A visible puncture site in the centre of the erythema. (Strong consensus: 20/20)
6.1.2 Variability of the erythema migrans (atypical erythema migrans)

Very often the initial skin infection cannot be definitively diagnosed clinically. Borrelia have been identified in homogenously red and non-migrating erythemas, spotty and infiltrated erythemas (Figure 3b [Fig. 3]), erysipelas-like flaming red erythemas (Figure 3a [Fig. 3]) and in centrally vesicular erythemas (Figure 3d [Fig. 3]) [69], [70]. The inflammation can completely disappear in the middle and fade to such as extent that the erythema is only visible around the edges – in the area of the migrating Borrelia – when heat is applied (Figure 3c [Fig. 3]). The erythema can also be haemorrhagic, particularly on the lower extremities (Figure 3 e and f [Fig. 3]). The centre can turn a dark purple colour (Figure 3f [Fig. 3]). The edge can be raised or urticarial. The former puncture site can be identified in the centre as a red papule (Figure 2a and b [Fig. 2]) [70], [71]. Without antibiotic treatment the Borrelia can persist for months or years in the skin and the erythema can slowly spread throughout the body. Often the red edge is the only evidence of the inflammatory reaction to the migrating Borrelia. If the erythema migrans persists for multiple weeks and months, it is referred to as erythema chronicum migrans ([66]: >4 weeks). In most cases (approx. 80%) serological detection of the IgG antibodies (sometimes even the IgM antibodies) is possible [72].

Erythema can disapear even without antibiotic treatment. Spontaneous healing is possible, however the Borrelia can persist even without a visible inflammatory reaction and, after a period of latency, this can lead to further organ manifestations.

Variability of the erythema migrans (atypical erythema migrans)

  • Non-migrating
  • Not marginated
  • Infiltrated instead of macular
  • Centrally vesicular
  • Haemorrhagic
  • Irregular blotches
  • Only visible when heat is applied to the skin
  • No visible tick puncture site
    (Strong consensus 20/20)

Concluding recommendation:

Due to the extraordinary variability of the clinical presentation, atypical erythema migrans is difficult for dermatologically inexperienced physicians to diagnose. Therefore, patients with atypical erythema should be referred to a dermatologist. (Strong consensus 19/20)

6.1.3 Borrelial lymphocytoma

Pseudolymphoma (cutaneous lymphoid hyperplasia) can occur in the early stages at the puncture site or in the migrating erythema migrans (Figure 4b [Fig. 4]). Mostly it is solitary, in rare cases it is also disseminated. Borrelial lymphocytoma occurs more frequently in children than in adults (7% in children and only 2% in adults with Lyme borreliosis, [8]). The favoured sites in children are the earlobes (Figure 4a and 4c [Fig. 4]), nipples and genital-anal area (Figure 4f [Fig. 4]) [73]. The disease was first described as lymphadenosis cutis benigna by Bäferstedt in 1944. B. burgdorferi s.l. can be detected in the pseudolymphomas [74]. Mostly it is a case of B. afzelii [75]. From a histological perspective, there are mixed B and T lymphocytic infiltrates. However purely B cell infiltrates can also occur which are difficult to differentiate from low-grade B cell lymphoma (Figure 4d and e [Fig. 4]) [70]. Borrelial lymphocytoma can also occur in the late stages as part of an acrodermatitis chronic atrophicans [73].

In the case of borrelial lymphocytoma, a substantial increase in the number of IgG antibodies can be detected in the serum regardless of the length of infection [65], [76]. In rare cases, multiple borrelial lymphocytomas can occur in the early disseminated stages or even in the late stages of the disease. In these cases, precise histological, immune-histochemical and molecular-genetic clarification is required in order to diagnostically differentiate them from malignant cutaneous lymphomas.

Significant features of borrelial lymphocytoma

  • Pseudolymphoma, mostly solitary, more frequent in children
  • Localised, above all on the earlobes, nipples or in the genital area
  • Purple subcutaneous nodules or plaque
  • Histologically mostly mixed B and T lymphocytic infiltrates
    (Strong consensus: 19/20)

6.2 Disseminated cutaneous early manifestation

Some of the patients experience haematogenous dissemination in the early stages of the disease which can be identified by flu-like symptoms such as a slight fever, arthralgia, myalgia, headaches, lymphadenopathy and multiple erythemata migrantia. This stage is very difficult to diagnose if no erythemas are visible, or cannot be identified due to an atypical morphology.

Multiple erythemata migrantia (MEM)

The haematogenous dissemination of the Borrelia in the skin is noticeable by the many sharply marginated, symptomless, oval erythemas of various sizes: multiple erythemata migrantia (Figure 5b and 5c [Fig. 5]) [69], [77], [78]. Children often experience symmetrical erythemas on their face, similar to fifth disease (parvovirus B 19 infection) (Figure 5a [Fig. 5]) [68], [70]. MEM can be associated with systemic symptoms and acute neurological symptoms [79]. The histological picture is initially atypical. The typical perivascular plasma-cellular infiltrates are not found until the advanced stage of the disease. There is usually a strong increase in IgM antibodies in the serum or the antibodies increase rapidly once treatment begins. There is usually an increase in IgG antibodies. Borrelia taken from skin lesions and, in rare cases, blood can be cultivated or their DNA can be detected using PCR [78], [80].

Significant features of multiple erythemata migrantia

  • Symptomless, disseminated, round or oval redness on the skin (Strong consensus: 19/20)
    • Without epidermal changes
    • Ring-shaped or homogenous
    • Often symmetrical erythemas on the face of children (similar to fifth disease)
    • Persisting over days or weeks
    • Recurring at the same places
    • Possible association with systemic or acute neurological symptoms
      (Strong consensus: 19/20)

6.3 Cutaneous late manifestations

Acrodermatitis chronica atrophicans (ACA)

The disease can manifest itself in various organs after varying periods of time, from months to years depending on the individual. A chronic skin infection mostly occurs in older people and more frequently in women [81]. Isolated cases have also been reported in children [82], [83], [84].

Oedematous infiltrative stage of ACA

Acrodermatitis initially manifests itself as pink reticular, then increasingly purple, oedematous infiltrated cushion-like erythemas, mostly on the extremities. The skin is inflamed, however there is initially no pain except for a feeling of heaviness of the extremity. This is the oedematous infiltrative stage of acrodermatitis chronica (Figure 6a and 6b [Fig. 6]). These purple infiltrates can also appear on the face and be confused with lupus erythematosus or a cutaneous malignant lymphoma [70].

Atrophic stage of ACA

In the course of the infection there is an increasing atrophy of all skin layers and skin appendages. Occasionally juxta-articular rough fibroid nodules and band-shape stripes appear (Figure 6f [Fig. 6]), e.g. rare but typical inflammatory ulnar stripes and swelling in the heel and Achilles tendon, or in other joints around the ACA (Figure 6b [Fig. 6]). This results in circumscribed fibrosis or pseudo-scleroderma in the area of the ACA which can be confused with circumscribed scleroderma. Arthritides, arthralgia and myalgia in the affected extremities are frequently associated with ACA [81].

A peripheral neuropathy occurs in 40–60% of patients in association with ACA. It is characterized by a feeling of numbness, a tingling sensation, burning and an increased sensitivity to pain (allodynia) [85], [86], [87].

Without antibiotic treatment living Borrelia can be detected for years in the skin and in the fibroid nodules [88]. In the course of the infection, all of the affected skin becomes atrophic and there is a loss of body hair, connective tissue and fatty tissue (Figure 6e and 6g [Fig. 6]). When the changes to the ACA-affected skin are symmetrical, they are difficult to differentiate clinically from age-related skin atrophy, acrocyanosis and chronic venous insufficiency. From a histological standpoint, acrodermatitis chronica atrophicans is characterised by a pronounced perivascular plasma-cell rich inflammatory infiltrate in all layers of the skin (Figure 6d [Fig. 6]) and, in the late stage, by an increasing atrophy of the epidermis, connective tissues and fatty tissues [89].

An ACA diagnosis is based on a typical clinical presentation, a typical histology and, as a rule, a high elevation of Borrelia IgG antibodies in the serum [81]. In unclear cases, particularly in the case of marginal elevation of antibody concentrations, the diagnosis has to be made by skin biopsy for histology and Borrelia DNA detection by NAT (PCR), or if possible through the cultivation of Borrelia from the skin.

Significant clinical features of acrodermatitis chronica atrophicans (ACA)

  • Initial oedematous infiltrative stage (plasma-cellular dermatitis) reddish colouring of the skin, mostly on one extremity
  • Transition to the atrophic stage in the course of the disease, purple to brown colouring of the skin, skin atrophy, loss of body hair, connective and fatty tissues, emergence of veins, juxta-articular fibrous nodules and joint involvement
  • Association with a peripheral neuropathy in around 50% of the cases
  • Older women more strongly affected (Strong consensus: 19/19)

6.4 Manifestations in the nervous system and joints associated with cutaneous borreliosis

An acute neuroborreliosis can simultaneously appear as part of early-stage borreliosis with erythema migrans. Arnez et al. found pleocytosis in the cerebrospinal fluid of 26% of the 214 children diagnosed with multilocular erythemata migrantia. Of these, 11% had clinically symptomatic lymphocytic meningitis [90]. Radiculoneuritis with characteristic nightly pain can also occur – in rare cases with paresis of the cranial nerves or peripheral nerves.

A peripheral neuropathy of the affected extremity occurs in 50% of patients with ACA [87].

Rheumatic symptoms, above all myalgia and arthralgia, can occur in relatively early stages of the disease alongside erythema migrans. Cardiac symptoms with dysrhythmia (AV block) should be watched for, which can occur during or after erythema migrans.

Lyme arthritis can either be the initial symptom or it can occur after a non-treated case of erythema migrans. Frequently the joint adjacent to the erythema migrans is affected. This manifests itself as acute intermittent arthritis with voluminous, at times, painful joint swelling, usually as mono or oligoarthritis. The knee joints are affected in 85% of the cases. The often massive swelling of the knee leads, unusually frequently and early on, to the development of popliteal cysts (Baker’s cysts). Ankle and elbow joints are less often affected, and almost never finger joints, especially in the form of a polyarthritis, have been observed. Lyme arthritis usually proceeds episodically, in other words, with repetitive inflammatory flare-ups that are interrupted by intervals of light to no symptoms.

6.5 Differential diagnoses for cutaneous Lyme borreliosis

The most frequent differential diagnoses for cutaneous Lyme borreliosis are listed in Table 2 [Tab. 2].

The variety of differential diagnoses shows that, except for typical erythema migrans, most of the cutaneous manifestations of Lyme borreliosis require careful dermatological diagnostic procedures. In particular, the lack of response to antibiotic treatment should not be uncritically interpreted as persistent borreliosis and treated for months with antibiotics.

It is, therefore, recommended to refer a patient with indistinct skin afflictions that persist after treatment to dermatologists or to dermatologically experienced paediatricians.

Recommendation:

  • Skin inflammations that were diagnosed as Lyme borreliosis and which have not healed after lege artis antibiotic treatment shall be referred to a dermatologist. (Strong consensus 11/12)

7 Diagnostics

7.1 Indirect pathogen detection (serodiagnostics, detection of antibodies)

Due to the complex characteristics of the pathogen, indirect pathogen detection using serological methods continues to play a pivotal role in the diagnosis of Lyme borreliosis in practical laboratory-based medical care. In accordance with the methods and standards required in Germany, the antibodies are detected in a serum using a two tiered diagnostic approach with a standardised screening test (immunoassay: ELISA, CLIA etc.) and a confirmation assay (immunoblot). This is to ensure that the diagnostic procedure has a uniformly high level of sensitivity and specificity (Table 3 [Tab. 3]).

In Europe, diagnostics tests for borrelial serology do not undergo any form of mandatory, extensive or independent clinical evaluation as part of the approval process. Thus a range of different test formats are on the market. In addition to various types of immunoassays, there are also a variety of test antigen preparations that use native and recombinant antigen combinations with, at times, different performance data. This partly explains the high degree of variability in the lab results which depend on the manufacturer and the test [12], [91]. Even though the principle testing procedures and the interpretation of serological test results are laid down as part of binding standards [92] in Germany, the interpretation of testing results, and in particular the evaluation criteria for immunoblot testing, are subject to manufacturer-dependent differences and have to be done in accordance with the respective manufacturer requirements as a result of the variability and insufficient standardisation of commercial test systems. This ongoing issue of insufficient testing standardisation is confirmed through meta-analytical investigations as part of external quality controls [12], [93]. In this respect, attending physicians should be aware of the qualifications of their diagnostic laboratory and the diagnostic assays and test specifications which it uses.

The course of the immune response and interpretation of the findings

In the course of a natural infection, specific IgM antibodies are usually detectable 3–6 weeks after the onset of the illness; IgG antibodies reach their peak more slowly (weeks to months).

It should be further noted that, after early, successful treatment of early manifestations, seroconversion can fail to appear under certain circumstances or, in the case of a positive detection of IgM antibodies, there doesn’t have to be a regular continuation of the immune response in the sense of a conversion from IgM to IgG. In contrast to textbook examples of the courses of immune response for many viral diseases, the antibody response to Lyme borreliosis often regresses very slowly both after an infection that is latent or cured, and after successful treatment. Thus, under certain circumstances, IgM reactivities or specific IgG values after such infections can remain detectable for months or even years. Often low positive borrelial-specific antibody values are a sign of a previous infection in the sense of persisting antibodies from a past infection (serological scar) [91]. However, a reinfection cannot be excluded in the case of such a lab result. Such findings have been detected in 20% of the people examined in serial investigations who belong to population groups that are frequently exposed, e.g. forestry workers, without there being or having been any symptoms of illness [94], [95]. Possible coincidences of these types of titres, with persisting antibodies from previous infections and unspecified findings, are also possible amongst the normal population [13], [96] and can be responsible for erroneous interpretations and diagnoses.

Detection of elevated IgM antibodies only (without IgG) effectively excludes a late manifestation of Lyme borreliosis in the case of immune-competent patients.

Diagnostic use of very sensitive early-phase antigens, such as VlsE, which enable the detection of a specific IgG response very early on in the course of the infection, means specific IgM antibody findings as part of Lyme borreliosis diagnostics are playing an increasingly limited role, especially since the IgM detection exhibits a poorer overall specificity than the IgG detection [97], [98]. However positive IgG findings can persist, in part, in high concentrations over longer periods of time so that no conclusions can be drawn regarding the activity of Lyme borreliosis or even the necessity for treatment in the absence of a classic activity marker, without additional clinical information and only on the basis of positive serological findings. At the same time, a statement can only be made about the significance of changes in findings if the comparison tests are carried out on serum samples that were taken at different times, ideally using a parallel approach as with the preserum, but, in any case, using the same test [91], [99].

An analysis using immunoblot within the framework of the stepwise diagnostic approach generally serves to not only specifically confirm the findings of the screening test, it also enables the immune response to be divided into an early and late stage so that a better correlation can be made between the lab findings and the clinical symptoms based on the characteristic band spectrum, particularly in the IgG immunoblot. Thus a narrow spectrum of bands with antibodies against early-phase antigens (e.g. VlsE, OspC, p41) is typically compatible with an early manifestation (e.g. erythema migrans, facial paresis) or a brief latent infection. However, it does not point to persistent clinical symptoms [91], [99], [100], [101]. In contrast, a wide band spectrum, including reactions to late-phase antigens (e.g. p100, p17/p18), fits in well with a late manifestation (e.g. arthritis, acrodermatitis) [100], [101], also with an asymptomatic persistence of antibodies (serological scar), however it primarily does not point to an early manifestation or a short course of infection. Reinfections are difficult to detect based only on serological test results without additional clinical information and can only be detected based on a clearly verifiable IgG increase in a parallel approach, or significant changes in the immunoblot band pattern in serum samples that are tested in parallel.

One major premise of serological testing for Lyme borreliosis is the fact that the referring physician needs to be aware that these types of tests should only be requested when there is reasonable clinical suspicion. Only when there is sufficiently high pre-test probability (prevalence of Lyme borreliosis in the patient cohort being investigated >20%) can a sufficiently utilisable positive predictive value of a positive test result even be assumed [102]. If the test is only ordered to exclude Lyme borreliosis in the case of unspecified or non-typical disease symptoms, the positive predictive value of the lab test drops to almost zero with respect to the possible confirmation of Lyme borreliosis. On the other hand, due to the relatively low overall prevalence and incidence of Lyme borreliosis in the general public, a negative test result, which excludes the disease in immune-competent patients with persisting symptoms, has an excellent negative predictive value.

Recommendation:

  • Serological diagnostics shall only be ordered when there is sufficient clinical suspicion. (Strong consensus: 19/19)
  • The diagnostics shall be conducted using a stepwise approach (screening test and confirmation test). (Consensus: 16/19)
  • Positive antibody detection is not proof of a clinically present Lyme borreliosis. (Strong consensus: 19/19)
  • Negative antibody detection almost entirely excludes Lyme borreliosis in healthy immune system patients with a protracted duration of illness. (Consensus: 16/19)
  • An isolated positive IgM detection argues against a late manifestation of Lyme borreliosis. (Consensus: 17/19)

Dissenting opinion (German Borreliosis Society)

  • There are no systematic studies on the frequency of Bb antibodies in the case of late-stage Lyme borreliosis. The view that an isolated IgM detection argues against a late manifestation of Lyme borreliosis has not been verified by the literature.

7.2 Direct pathogen detection

The respective microbiological diagnostic quality standards (MiQ Lyme borreliosis, MiQ PCR) apply in the direct pathogen detection of Lyme borreliosis using culture and PCR.

7.2.1 Culture

Direct detection by culture with the modified Barbour-Stoenner-Kelly medium is considered to be the gold standard and to be clear proof of an infection with B. burgdorferi [103], [104]. Direct detection of skin manifestations by culture are frequently successful. To a limited degree, detection by culture is also possible in liquor and, in very rare cases, in synovial fluid, synovial biopsies and blood. In individual cases, the detection of B. burgdorferi has also been achieved in other tissue samples, e.g. heart muscle and iris [105], [106]. Cultivating from patient samples using suitable media is time-consuming and materially intensive, and usually takes more than two weeks. The sensitivity of the methods in European studies is between 40% and 90% for erythema migrans and between 20% and 60% for ACA [107], [108], [109], [110]. Overview in: [111]. Because of the invasiveness of the sample taking, direct detection by culture should therefore be based on a clear indication and explicitly remain limited to specially identified reference laboratories, such as the National Reference Centre for Borrelia at the Bavarian State Office for Health and Food Safety in Oberschleissheim. In addition, further molecular-biological confirmation assays are required in positive cases.

Recommendations for direct detection by culture:

  • Direct detection by culture should only be used in differential-diagnostically ambiguous cases. (Strong consensus: 19/19)
  • The cultivation of Borrelia burgdorferi sensu lato should be limited to specialist laboratories. (Strong consensus: 19/19)
  • Positive culture results are to be confirmed using suitable molecular-biological methods. (Strong consensus: 18/19)
7.2.2 Direct detection using molecular-biological detection methods

The detection methods currently being used in Lyme borreliosis diagnosis should be regarded as having a low level of standardisation [112]. This applies to DNA isolation from suitable clinical materials, as well as to the reaction conditions and the selection of the reaction starter molecules (primers). In principle, the detection of Borrelia from a skin biopsy using nucleic acid amplification techniques (NAT, usually PCR) is very reliable and, in the case of early manifestations, is more sensitive than serological antibody detection. The diagnostic sensitivity of NAT is around 70% for detection from biopsies from erythema migrans and acrodermatitis chronica atrophicans [107], [113], [114], [115], [116]. However, positive results have to be confirmed through molecular-biological confirmation assays with regard to specificity (probe hybridisation, sequencing of the amplificate) and the results must be indicated in the findings.

After treatment, Borrelia DNA can still be detected for weeks – or even months – in the affected area of skin before conclusions can be drawn as to whether the therapy has failed [117], [118], [119]. Molecular-biological detection of pathogens without the simultaneous presence of typical disease manifestions is not clinically relevant. Direct molecular-biological detection from urine samples is not currently recommended due to ambiguous diagnostic sensitivity and specificity [113], [120], [121]. Because of the invasiveness of the sample taking, direct detection by culture should therefore be based on a clear indication (e.g. unexplained skin manifestation that has been differentially diagnosed) and explicitly remain limited to specially identified reference laboratories, such as the National Reference Centre for Borrelia at the Bavarian State Office for Health and Food Safety in Oberschleissheim. In addition, further molecular-biological confirmation assays are required in positive cases.

Recommendations for direct molecular-biological detection:

  • Direct molecular-biological detection (PCR) is not a screening test if there is suspicion of Lyme borreliosis. (Strong consensus: 19/19)
  • A negative PCR test result does not exclude Lyme borreliosis. (Strong consensus: 19/19)
  • A positive PCR test result shall be confirmed by further molecular-biological methods and the detected genospecies shall be indicated in the findings. (Strong consensus: 19/19)
  • A positive PCR test result after treatment with antibiotics in accordance with the guidelines or without typical clinical manifestation has no clinical relevance. (Consensus: 16/19)
  • Direct molecular-biological detection should be limited to ambiguous skin manifestations and reserved for specially identified microbiological laboratories. (Strong consensus: 20/20)

7.3 Diagnosis of clinical skin manifestations

7.3.1 Erythema migrans (typical)

If a clinically typical erythema migrans is present (see section on clinical manifestations) no further laboratory diagnostic confirmation needs to occur; antibiotic treatment should begin immediately (Figure 7 [Fig. 7]).

Recommendation:

  • If a typical erythema migrans is present (see section on clinical Manifestations) no further laboratory diagnostic confirmation (serological, cultural, molecular-biological) needs to occur. (Strong consensus: 20/20)
  • If a typical erythema migrans is present, antibiotic treatment shall begin immediately. (Strong consensus: 20/20)
7.3.2 Erythema migrans (atypical)

If an atypical erythema migrans is suspected, antibody and pathogen detection by PCR and culture is available. A serological test should be carried out in every case. If the findings remain ambiguous, the aim should be pathogen detection using PCR, if necessary also by culture (Figure 7 [Fig. 7]). A skin biopsy should be taken near the inflamed edge. After informing the patient and obtaining written consent, the selected area of the skin is numbed using local anaesthesia. After thorough disinfection of the skin, a 4 mm punch is used to remove the skin, which is put in a sterile vessel with 0.9% saline solution. Direct inoculation in the cultivation medium only makes sense when the sample can be processed in the lab within a few hours. Otherwise, fast growing skin bacteria can hamper the cultivation of the Borrelia.

A histological analysis rarely has a guiding nature in the case of erythema migrans. It can, however, make sense for differential-diagnostic clarification.

Recommendation:

  • In the case of an atypical clinical appearance of erythema migrans, suspicion shall be clarified through a serological test. (Consensus: 18/20)
  • If the serological test is negative and the clinical suspicion remains, direct cultural or molecular-biological detection from biopsy material shall be used for clarification. (Strong consensus: 20/20)
7.3.3 Multiple erythemata migrantia (MEM)

If multiple erythemata migrantia, also known as multilocular erythema migrans (MEM) is suspected, serological antibody detection and pathogen detection using PCR and culture from a skin biopsy are available. A serological test should be carried out in every case. If the findings remain ambiguous, the aim should be to detect the pathogen using PCR, if necessary also by culture (Figure 7 [Fig. 7]) (see 7.2.1). Clinical signs of extra-cutaneous symptoms should be watched for in the case of MEM (see Table 1 [Tab. 1] in the section on clinical manifestations).

Recommendations:

  • A serological test shall be carried out when MEM is suspected. (Strong consensus : 20/20)
  • If the serological test is negative and the clinical suspicion remains, direct cultural or molecular-biological detection in biopsy material shall be used for clarification. (Strong consensus: 20/20)
7.3.4 Borrelial lymphocytoma

Confirming the diagnosis through serological antibody detection is obligatory and, in most cases, antibodies against B. burgdorferi can be detected [38], [70], [122]. When the findings are still ambiguous, the patient shall be referred to a dermatologist in order to detect the pathogen by PCR or, if necessary, culture. Two skin biopsies should be taken from the abnormal skin (see 7.2.2): one for the histological test in a 4% formalin solution, and one for the culture and PCR test in sterile, physiological saline solution.

PCR and culture allow B. burgdorferi to be detected. Although there is limited data on the sensitivity of the methods used to identify borrelial lymphocytoma; a PCR detection success rate can be expected in around 70% of the cases [73], [122].

Recommendation:

  • If there is an unambiguous clinical presentation of borrelial lymphocytoma and a positive serology, further microbiological tests are not required. (Strong consensus: 20/20)
  • If there is an unambiguous clinical presentation of borrelial lymphocytoma, antibiotic treatment shall begin immediately. (Consensus 16/20)
  • If the clinical presentation is not unambiguous and the serology is negative, further tests (primarily histology, molecular-biology, possibly culture) shall be conducted for differential-diagnostic clarification. (Strong consensus 20/20)
7.3.5 Acrodermatitis chronica atrophicans

If ACA is clinically suspected, a Borrelia serology should be carried out first. High antibody values in the IgG screening test, combined with a broad spectrum of borrelial-specific bands in the IgG blot or similar tests (see Section 7.1), are indications of ACA. A negative IgG serology excludes, with high certainty, ACA in immune-competent patients.

In ambiguous cases the patient should be referred to a dermatologist for differential diagnosis. If uncertainty remains, two skin biopsies (see 7.2.2.) should be taken from the abnormal patch of skin: one for the histological test in a 4% formalin solution, and one for the culture and PCR test in physiological saline solution. In the case of ACA, B. burgdorferi DNA can be detected in around 70% of the cases.

Recommendation:

  • When ACA is clinically suspected, the diagnosis shall be confirmed through a serological test. (Strong consensus: 19/20)
  • High IgG antibody values in the screening test, combined with a broad band pattern in the IgG immunoblot test, indicate a suspected clinical diagnosis. (Strong consensus: 20/20)
  • A negative Borrelia serology excludes ACA with a high degree of certainty in immune-competent patients. (Strong consensus: 20/20)
  • The diagnosis shall be histologically confirmed in all cases. (Majority approval: 12/20)
  • When the clinical picture is ambiguous, further diagnostic clarification through biopsy and subsequent histological testing should be done. When the findings are unclear, direct detection by culture and molecular biology is recommended. (Strong consensus: 19/20)
7.3.6 Ambiguous dermatological pathologies with a suspicion of Lyme borreliosis

See Table 4 [Tab. 4].

Recommendation:

  • If a cutaneous manifestation of Lyme borreliosis is suspected and there is no unambiguous clinical presentation, a skin biopsy with a histological examination shall be conducted along with direct pathogen detection using culture and molecular-biological methods. (Strong consensus: 20/20)

7.4 Non-recommended diagnostic approaches

In addition to the traditional diagnostic methods listed above, which are used when Lyme borreliosis is suspected, the literature describes a whole series of diagnostic techniques that, in part, have been inconclusively evaluated. This includes the immuno-histochemical detection of B. burgdorferi in biopsies and of antigens from blood and urine, as well as functional tests that test for cellular immunity (lymphocyte transformation tests (LTT), cytokine detection). Currently there is a lack of scientific investigations that prove there is a diagnostic benefit. Because the available LTT methods lack specificity, they should not be used.

Methods that are not recommended for use in the diagnosis of cutaneous manifestations of Lyme borreliosis:

  • Immunohistochemical detection of Borrelia from tissue is currently not recommended. (Strong consensus: 19/19)
  • The lymphocyte transformation test (LTT) and the detection of specific cytokines is currently not recommended. (Strong consensus: 18/19)
  • Detection of Borrelia in engorged ticks is not recommended. (Strong consensus: 19/19)
  • Detection of Borrelia antigens from patient samples is currently not recommended. (Strong consensus: 19/19)
  • Direct detection of Borrelia in patient samples using light microscopy is currently not recommended. (Strong consensus: 18/19)
  • The detection of circulating immune complexes is currently not recommended. (Strong consensus: 18/19)

7.5 Quality control and quality assurance

According to the guidelines of the German Medical Association (Bundesärztekammer), diagnostic laboratories must currently participate in infection-related serological round robin tests twice a year. This applies to serological antibody detection and to direct molecular-biological detection of Borrelia when Lyme borreliosis is suspected. The results of the external quality assessment tests (EQA tests), which INSTAND e.V. has been carrying out for years, reveal extensive heterogeneity in the testing systems currently on the market. The pass rates for the conventional serological and molecular-biological test systems, which have been collected from meta-analytical data, show that, despite good analytical pass rates for immunoassays and molecular-biological tests, clinical diagnostic interpretation of the result constellations often proves difficult and can hamper medical treatment in daily clinical practice [12], [91]. Thus, when Lyme borreliosis is suspected, infection diagnostics are to be conducted in laboratories that meet the laboratory diagnostic standards in accordance with the diagnostic guidelines of the expert medical societies and the guidelines of the German Medical Association. These laboratories must regularly and successfully participate in external quality assurance tests (round robin tests). Physicians treating patients with Lyme borreliosis should query about and ensure that these prerequisites are met in the laboratories charged with carrying out their diagnostic testing. If questionable result constellations or implausible test results are produced, expert laboratories specialising in Lyme borreliosis diagnostics and the National Reference Centre for Borreliosis at the Bavarian State Office for Health and Food Safety in Oberschleissheim should be consulted.

Recommendation:

  • Attending physicians shall be aware of whether their diagnostic laboratory complies with the respective diagnostic standards and qualifications and the extent to which the diagnostic assays used there conform to guidelines. (Strong consensus: 18/19)

8 Treatment of cutaneous Lyme borreliosis

Recommendations for treating Lyme borreliosis have been published in numerous European and American guidelines since 2004 (see Annex 2 “Comparison of Guidelines and Therapies” in Attachment 1 [Attach. 1]).

Table 5 [Tab. 5] summarises the best-evaluated antibiotic therapies taken from American and European guidelines.

Doxycycline and amoxicillin are the antibiotics of choice in all guidelines.

Both antibiotics are very effective in the dosages listed in Table 5 [Tab. 5] and are usually tolerated well. Gastrointestinal complaints can occur during treatment with doxycycline. It is particularly important that they are not taken together with dairy products. Furthermore, patients should be informed of the risk of phototoxic skin reactions and use light stabilisers when taking the antibiotics.

During treatment with amoxicillin, non-allergenic skin exanthemas frequently appear on the 8th day on the torso. If they are light exanthemas, the treatment can continue. If itching occurs, symptoms can be treated with antihistamines and skin care products. Corticosteroids are not necessary.

Of the oral cephalosporins, only cefuroxime axetile has demonstrated an efficacy that is comparable to treatment with doxycycline and amoxicillin [123]. The absolute bioavailability of cefuroxime axetil is comparatively low (40–45%). The best resorption is achieved when it is taken directly after a meal.

Other 1st and 2nd generation cephalosporins are not effective enough [124].

In the case of disseminated early-stage infection, intravenous treatment with ceftriaxone does not achieve any better results than oral doxycycline treatment [125].

Of the macrolides, azithromycin has proven to be adequately effective [79], [126], [127], [128]. The long tissue half-life period is advantageous because of the long generation time of Borrelia. The efficacy of clarithromycin is regarded as controversial. Clarithromycin was compared with amoxicillin in one of the newer, open, randomised comparative studies of children with erythema migrans and was classified as equally effective [129]. Roxithromycin is not effective enough. Because of its uncertain resorption and indications of resistance, erythromycin is no longer a treatment of choice [28], [130].

Treatment with oral penicillin V is controversial. Austrian, Swedish and Slovenian studies show that it is sufficiently effective [90], [131], [132], [133].

It is particularly important that dosage and length of treatment are observed.

Cutaneous early manifestations should be treated for 10–21 days (Table 5 [Tab. 5]). The length of treatment depends on the duration and severity of the clinical symptoms; in the case of solitary erythema migrans without general symptoms, a 10 to 14 day treatment is sufficient. In a comparative study by Stupica et al. [134] the results of treating localised erythema migrans with doxycycline for 10 versus 14 days were evaluated. There were no differences in the way the erythema healed. In both treatment groups symptoms persisted no longer or more frequently than in healthy subjects. Treatment should last 21 days if there is evidence that the Borrelia has disseminated (indicated by a flu-like feeling), or in the case of multiple erythemata migrantia and borrelial lymphocytoma.

Taking doxycycline or amoxicillin orally for 30 days to treat cutaneous late manifestations (acrodermatitis chronica in the oedematous-infiltrative or atrophic stage) without neurological involvement is usually sufficient [81], [135]. However, if there are also neurological symptoms, intravenous treatment with penicillin G or 3rd generation cephalosporins ceftriaxone or cefotaxime may be necessary.

The cure rates – defined as the reinstatement of the body’s original condition with regression of the disease-specific symptoms after successful treatment – is between 95%–100% when the localised and disseminated early manifestations are treated in time [136], [137].

Treatment failure with evidence of the pathogens after therapy rarely occurs if the treatment is conducted lege artis [45], [138]; individual cases have been published [104], [105], [139], [140], [141], [142].

Two larger studies were able to show that new infections with other Borrelia strains were the reason why Lyme borreliosis returned in every case [143], [144].

Currently there are no indications of a development of secondary antibiotic resistance of B. burgdorferi to the antibiotics recommended in the guidelines [145], [146], [147], [148].

If the late manifestations remain untreated for a long period of time, there is a higher risk of the patient having persistent physical symptoms and of their skin, joints and nervous system not properly healing.

It is disputed whether repeated antibiotic treatment makes sense for these patients with persisting complaints. According to published randomised controlled trials (RCT), long-term antibiotic treatment is less than promising [149], [150], [151], [152], [153], [154].

A European RCT published in 2016 (PLEASE Study) looked at 280 patients whose complaints persisted for more than 2 years after their Lyme borreliosis had been treated with antibiotics (78 patients after erythema migrans, 15 patients after meningoradiculitis) and 153 seropositive patients with borreliosis-related complaints after a tick bite. The study compared the health-related effects of a 2-week compared to a 14-week round of antibiotics. First, all of the patients that had previously been treated with antibiotics were given 2 g of ceftriaxone i.v. for 2 weeks. Then the patients were randomly placed in 3 groups. Group 1 received doxycycline 200 mg/d p.o. for 12 weeks, Group 2 clarithromycin 2x 500 mg plus hydroxychloroquin 2x 200 mg/d for 12 weeks, and Group 3 a placebo for 12 weeks. Treatment success was assessed as health-based quality of life after 14 weeks and then up to 52 weeks using the RAND 36 Health Status Inventory. The aggregate score improved equally after treatment in all three groups without a significant difference. The assessment of the quality of life remained lower than in the general population in all three groups. No difference in treatment success between the short-term treatment and the two long-term treatments could be made. Patients receiving the long-term treatment had considerably more antibiotic-related side-effects (primarily photosensitivity (18.6%) and nausea (10.5%) in connection with doxycycline, and primarily nausea (10.4%), diarrhoea (9.4%) and allergic exanthemas (8.3%) in connection with clarithromycin/hydroxychloroquine.) Vision problems were the most frequent complaint of the placebo group (10% of the patients) [155], [156].

8.1 Treatment during pregnancy and nursing

Oral treatment with amoxicillin p.o. is recommended during pregnancy and nursing. Alternatively, penicillin G and ceftriaxone can be administered i.v. [157], [158]. If the patient has an identified allergy to penicillin, azithromycin or cefuroxime axetil can be prescribed after strong indication. Ceftriaxone can be taken intravenously under clinical surveillance since the risk of a cross allergy between penicillin and 3rd generation cephalosporins is around 1% [159].

8.2 Treatment of children

Children can be treated with 4 mg/kg KG/day (up to a maximum dosage of 200 mg/day) of doxycycline once their tooth enamel has completely formed at age 9 and over (>8 years). For children under 8, the treatment of choice is 50 mg/kg KG/day of amoxicillin (Table 5 [Tab. 5]). Taking it the required 3 times a day can be difficult for kindergarten- and school-aged children.

Alternatively, cefuroxime axetil 30 mg/kg KG/day, azithromycin 5–10 mg/kg KG/day or clarithromycin 15 mg/kg KG/day can be prescribed, which is taken twice daily [129].

8.3 Therapy adherence

In order to improve treatment adherence/therapy compliance, the patient should be informed before beginning the treatment of the aspects of taking prescribed antibiotics and the potential risks of undesired effects.

A frequent cause of treatment failure is the incorrect administration of doxycycline. It should be noted that resorption can be compromised when it is taken together with bivalent or trivalent cations, such as aluminium, calcium (milk, dairy products and fruit juice containing calcium), and magnesium, in antacids or through iron supplements, as well as through activated charcoal and colestyramine. Therefore, there should be a 2 to 3 hour time span between when the antibiotic is taken and the medicine or food is ingested.

Another reason for treatment failure is irregular administration e.g. forgetting to take the midday dose in the case of amoxicillin, or when the length of antibiotic treatment is insufficient e.g. due to a deterioration in symptoms as a result of a Herxheimer reaction, because of gastrointestinal complaints, or as a result of phototoxic skin reactions through increased sensitivity to light from doxycycline.

In the case of a disseminated infection, the patient should be informed about a possible Herxheimer reaction with a flare up of the erythemas, which occurs in approx. 10% of cases, a feeling of being very unwell, and a rise in temperature in approx. 2% of cases within 24 hours of taking the antibiotics [78], [129]. Occasionally this reaction is delayed. It is a temporary immunological reaction as a result of the upregulation of proinflammatory cytokines and can be treated, for example, with non-steroidal anti-inflammatory drugs (NSAID). Cortisone treatment is not necessary. The antibiotic should continue to be taken.

Recommendations for treating cutaneous Lyme borreliosis:

Antibiotics

  • Doxycycline or amoxicillin p.o. are the treatments of choice (Strong consensus: 17/18)
  • Treatment alternatives are cefuroxime, azithromycin, possibly also clarithromycin p.o. (Strong consensus: 17/18)
  • Ceftriaxone i.v. is the treatment of choice for cutaneous Lyme borreliosis with neurological manifestations (Majority approval: 10/19)
  • See the recommendations of other expert medical societies for antibiotic treatment of patients with cutaneous Lyme borreliosis with neurological or cardiological manifestations. Possibilities include ceftriaxone i.v., cefotaxime i.v., penicillin G i.v. or doxycycline p.o. (Strong consensus: 18/18)

Duration of treatment

  • The treatment of the early manifestations of cutaneous Lyme borreliosis shall last 14–21 days. (Exceptions are azithromycin 5–10 days; doxycycline 10–14 days in the case of solitary erythema migrans) (Consensus: 17/19)
  • The treatment of cutaneous late manifestations shall last 30 days. (Consensus: 17/19)
  • A general extension of treatment beyond the recommended amount of time is not recommended. (Consensus: 17/19)
  • Treatment can be extended in individual cases depending on the clinical progression and after a re-evaluation of the diagnosis. (Consensus: 17/19)
  • Treatment is renewed on a case-by-case basis when the pathogen has been confirmed. (Consensus: 16/19)
  • The diagnosis should be re-evaluated if the cutaneous symptoms persist or progress despite treatment with antibiotics in line with the guidelines. (Consensus: 17/19)

Recommendations for treating cutaneous Lyme borreliosis during pregnancy:

  • Amoxicillin p.o. shall be administered as the treatment of choice during pregnancy. (Strong consensus: 18/18)
  • Penicillin G i.v. and ceftriaxone i.v. represent alternative therapies during pregnancy. (Strong consensus: 18/18)
  • If the patient is allergic to penicillin, cefuroxime p.o., ceftriaxone i.v., cefotaxime i.v. or azithromycin p.o. should be used. (Consensus: 15/17)

Recommendations for treating cutaneous Lyme borreliosis in children:

  • Amoxicillin p.o. shall be administered as the treatment of choice in children under 8. (Strong consensus: 17/17)
  • Children aged 9 and over can take doxycycline p.o. (Strong consensus: 17/17)
  • Azithromycin, clarithromycin or cefuroxime p.o. represent alternative treatments for children (Strong consensus: 17/17)

Dissenting opinion (OnLyme Aktion)

  • When other causal factors can be excluded and cutaneous, illness-specific symptoms recur or do not regress, another suitable antibiotic can be considered, taking into account the patient’s individual situation.

Dissenting opinion (German Borreliosis Society)

  • There are no evidence-based studies on the efficacy of treating late-stage Lyme borreliosis, particularly ACA, with antibiotics. The paper by Aberer et al. (1996) [135], cited in the text, states that the efficacy of ceftriaxone needs to be reviewed in further studies. In terms of oral antibiotics, it has been established that the length of treatment is a more critical factor than the type of antibiotic (penicillin/doxycycline).

8.4 Persisting symptoms after treatment/post-treatment Lyme disease syndrome (PTLS)

After antibiotic treatment has been carried out in accordance with the guidelines, inflammatory reactions can persist and symptoms such as tiredness, joint and muscle pain, headaches, a general feeling of being unwell, irritability or paraesthesia can last for months. If the unspecific constitutional symptoms last for more than 6 months, it is considered by some authors to be post-Lyme syndrome (PLS) or post-treatment Lyme disease syndrome (PTLDS) [150], [160]. So-called PTLDS is a syndrome that has yet to be generally defined scientifically and therefore is not yet universally accepted. It can be diagnostically differentiated from diagnosed late manifestations of Lyme disease and symptoms resulting from persisting reproducible pathogens and from improper healing. The benefit of repeated and long-term treatment with antibiotics has not been verified.

In a controlled study of patients with erythema migrans, in which a control group containing individuals of similar age and gender was simultaneously studied, no increased incidence of post-therapeutic symptoms compared to the control group were identified [136].

Several studies indicate special immunological characteristics. Patients who have persisting symptoms for months to years after receiving antibiotic treatment were identified as frequently having anti-neural antibodies [161], as well as a weaker Th1-immune response with elevated interleukin 23 concentrations in serum [39].

Bockenstedt et al. were able to identify Borrelia DNA in mice when treatment was focussed near cartilage, however they did not find any living Borrelia [117]. Persisting DNA and RNA, as well as living Borrelia, were detected in rhesus monkeys through xenodiagnoses (transfer of tissue to laboratory animals) [162]. Since these were animal studies, no statement on what this means for human infections can currently be made.

8.5 Course of action for persisting skin changes and symptoms after antibiotic treatment

A primary incorrect diagnosis is a common reason for persisting skin changes and symptoms after treatment with antibiotics [163].

In the case of clinically diagnosed erythema migrans and multiple erythemata migrantia that do not heal within 6 weeks, a differential diagnosis of circumscribed scleroderma (morphea), granuloma annulare, sarcoidosis, erythema annulare et diutinum, tinea (with low epidermal involvement) or uticarial vasculitis should be considered.

Patients should be referred to a dermatologist for further diagnostics. Borrelial lymphocytoma often heals very slowly over many months. According to studies by Maraspin et al. on 85 patients, healing time was, on average, 28 days (7–270 days). The longer the borrelial lymphocytoma was present, the longer it took to heal [73]. If the knots persist for more than one year, or new knots appear, a skin biopsy should be carried out by a dermatologist for a histological diagnosis and Borrelia PCR. Cutaneous pseudolymphoma, Jessner’s lymphocytic infiltration or a malignant lymphoma should be considered in the differential diagnosis.

It takes years following antibiotic treatment for skin changes to slowly regress in the case of an acrodermatitis chronica atrophicans that has persisted for years. The atrophy of the skin, tissue and fat can be irreversible – especially in older people. This also applies to ACA-associated peripheral neuropathy. (See also the AWMF-S3 Guideline on Neuroborreliosis which is in progress.)

Age-related skin atrophy, chronic thermal damage to the skin, e.g. chilblains and heat melanosis, as well as chronic venous insufficiency with stasis dermatitis can be considered in the differential diagnosis.

Chronic neuropathic pain after adequate antibiotic treatment of acrodermatitis chronica atrophicans with peripheral neuropathy is treated in accordance with the DGN’s guideline “Neuropathic Pain” (AWMF – guidelines register no. 030/114).

All patients whose symptoms persist after antibiotic treatment of cutaneous Lyme borreliosis should undergo careful differential diagnostic clarification by respective specialists, above all, for internal medicine (infectiology, rheumatology, cardiology, endocrinology), psychosomatics, psychotherapy, psychiatry or palliative care, since chronic infections with another etiology, other internal medical disorders, autoimmune diseases, chronic pain syndrome, and depressive and somatoform disorders should also be considered in the differential diagnosis and need to be treated accordingly.

Recommendations for persisting symptoms after treatment in accordance with the guidelines:

  • If an erythema or multiple erythemas persist after treatment of erythema migrans (longer than 6 weeks), the patient shall be referred to a dermatologist for a differential diagnosis of circumscribed scleroderma (morphea), granuloma annulare, sarcoidosis, erythema annulare et diutinum, tinea or urticarial vasculitis. (Strong consensus: 17/17)
  • If a lymphocytoma persists or progresses after treatment the patient shall be referred to a dermatologist for a differential diagnosis (cutaneous pseudolymphoma, Jessner’s lymphocytic infiltration or malignant lymphoma). (Strong consensus: 17/17)
  • If the acrodermatitis chronica atrophicans persists after treatment the patient shall be referred to a dermatologist for further consultation and for a differential diagnosis (age-related skin atrophy, chronic thermal damage to the skin e.g. chilblains and heat melanosis, chronic venous insufficiency with stasis dermatitis). (Strong consensus: 16/16)

9 Prophylaxis

9.1 Preventing tick bites

The best prophylaxis is to prevent tick bites by wearing clothing that covers the body, and carefully checking the skin, including the scalp, after being outdoors. This is particularly important for children, who have an increased risk when playing outdoors between spring and autumn.

Insect repellents that are effective against ticks, e.g. diethyltoluamide (DEET), icaridin (1-(1-methylpropyl carbonyl)-2-(2-hydroxyethyl)piperidine), ethyl butylacetylaminopropionate (EBAAP, IR 3535) can also be used, however their effectiveness is limited to up to 4 hours [164], [165].

9.2 Preventing Lyme borreliosis

Removing the ticks before they become engorged with blood is very important. The risk of a Borrelia transfer increases with the length of time that the tick sucks [17]. Transmission within the first 12 hours has rarely been observed in laboratory animals. After being in a garden, park, field forest or meadow where there may have been contact with a tick, the body should be checked the same evening for ticks.

The ticks should be removed immediately with a tick tweezer or a tick card in order to prevent the transfer of the Borrelia. If parts of the suction organ remain in the skin, they can later be removed with a needle or a curettage [160]. If the head or the suction organ remains in the skin, this is not critical in terms of a Borrelia transfer. When nymphs and adult ticks are engorged with blood, their bodies should not be squeezed in order to prevent a possible transfer of the Borrelia.

Checking the tick that has been removed from the skin for borrelia does not make sense since the detection of the borrelia in the tick is not sufficiently predictive for whether the Borrelia has been transferred to the host and for the emergence of the disease.

After removing the tick, the patient should be informed of the necessity of observing the bite site over the subsequent 6 weeks (Annex 1: “Patient information after a tick bite” in Attachment 1 [Attach. 1]).

9.3 Prophylactic treatment after a tick bite

According to an American study, the risk of infection after a tick bite can be reduced through a one-time prophylactic administration of 200 mg of doxycycline (87% effectiveness) [166], [167]. The results, however, should be interpreted with caution since only one follow-up check took place after 6 weeks. Thus no statement can currently be made as to whether this is sufficiently effective with regard to a late infection.

In light of the low risk of infection, doxycycline would have to be administered unnecessarily many times in order to prevent a potential infection. According to projections of infection risk in endemic areas, 40–125 prophylaxes would have to be taken in order to prevent 1 infection [168]. Impact on the intestinal flora and a possible development of resistance through frequent prophylaxis is conceivable. Therefore, oral doxycycline prophylaxis in Europe is not recommended.

The prophylactic application of an antibiotic cream is also controversial. Animal studies with azithromycin cream reveal a good prophylactic efficacy [169], [170]. Placebo-controlled studies on the effectiveness in humans have yet to be published. This treatment is not currently recommended due to the lack of clinical data.

Recommendations on infection prophylaxis:

  • Clothing that covers the body should be worn to prevent tick bites.
  • Using tick repellents can be recommended with some reservations.
  • Skin should be inspected in the evening for ticks after being outside in an area where there is a possibility of the individual coming into contact with ticks.
  • Ticks should be removed early in order to prevent Lyme disease.
  • The site of the bite should be observed for up to six weeks.
    (Consensus: 15/16)

Not recommended:

  • Analysing the removed tick for Borrelia is not recommended. (Consensus: 15/16)
  • Local or systemic prophylactic antibiotic treatment after a tick bite is not recommended. (Consensus: 14/16)

9.4 Vaccines

No approved vaccine that can be used on humans is currently available.

A vaccination with recombinant lipidated Osp A has been evaluated in the USA as part of a major study and has shown to be effective [171], [172]. The vaccine has been approved in the USA since 1999; however, it was taken from the market by its manufacturer. The reason for this is not medical. Reports on undesired vaccine reactions in individuals who are genetically predispositioned were refuted by multiple qualified studies [173], [174], [175]. This monovalent vaccine is not suitable for Europe as it only protects against an infection with B. burgdorferi sensu stricto, and not against the genospecies B. afzelii and B. garinii that are frequently found in Europe.

A polyvalent Osp A vaccine is currently being developed for Europe [176], however approval is not expected in the foreseeable future.


Notes

Procedure for forming a consensus

The guideline was created using a modified Delphi process and was voted on in an extended consensus conference of the Interdisciplinary S3 Guideline Group, moderated by Prof. Ina Kopp, Head of the AWMF Institute for Medical Knowledge Management.

It was passed by the 22 expert medical societies and patient organisations involved.

The guideline is a part of the registered Interdisciplinary S3 Overall Guideline on the “Diagnosis and Treatment of Lyme Borreliosis”.

Support

This guideline was created without the influence or financial support of sponsors.

The funds required to create and to translate this guideline were provided by the German Society for Dermatology and the Society for Promotion of Quality Assurance in Medical Laboratories (INSTAND e. V.).

Travel expenses were provided by the respective expert medical societies.

Declaration of competing interests by the authors


References

1.
Baranton G, Postic D, Saint Girons I, Boerlin P, Piffaretti JC, Assous M, Grimont PA. Delineation of Borrelia burgdorferi sensu stricto, Borrelia garinii sp. nov., and group VS461 associated with Lyme borreliosis. Int J Syst Bacteriol. 1992 Jul;42(3):378-83. DOI: 10.1099/00207713-42-3-378 External link
2.
Fingerle V, Schulte-Spechtel UC, Ruzic-Sabljic E, Leonhard S, Hofmann H, Weber K, Pfister K, Strle F, Wilske B. Epidemiological aspects and molecular characterization of Borrelia burgdorferi s.l. from southern Germany with special respect to the new species Borrelia spielmanii sp. nov. Int J Med Microbiol. 2008 Apr;298(3-4):279-90. DOI: 10.1016/j.ijmm.2007.05.002 External link
3.
Richter D, Postic D, Sertour N, Livey I, Matuschka FR, Baranton G. Delineation of Borrelia burgdorferi sensu lato species by multilocus sequence analysis and confirmation of the delineation of Borrelia spielmanii sp. nov. Int J Syst Evol Microbiol. 2006 Apr;56(Pt 4):873-81. DOI: 10.1099/ijs.0.64050-0 External link
4.
Schaarschmidt D, Oehme R, Kimmig P, Hesch RD, Englisch S. Detection and molecular typing of Borrelia burgdorferi sensu lato in Ixodes ricinus ticks and in different patient samples from southwest Germany. Eur J Epidemiol. 2001;17(12):1067-74.
5.
Wallich R, Helmes C, Schaible UE, Lobet Y, Moter SE, Kramer MD, Simon MM. Evaluation of genetic divergence among Borrelia burgdorferi isolates by use of OspA, fla, HSP60, and HSP70 gene probes. Infect Immun. 1992 Nov;60(11):4856-66.
6.
Stanek G, Strle F. Lyme borreliosis. Lancet. 2003 Nov;362(9396):1639-47. DOI: 10.1016/S0140-6736(03)14798-8 External link
7.
Maraspin V, Ruzic-Sabljic E, Strle F. Lyme borreliosis and Borrelia spielmanii. Emerging Infect Dis. 2006 Jul;12(7):1177. DOI: 10.3201/eid1207.060077 External link
8.
Berglund J, Eitrem R, Ornstein K, Lindberg A, Ringér A, Elmrud H, Carlsson M, Runehagen A, Svanborg C, Norrby R. An epidemiologic study of Lyme disease in southern Sweden. N Engl J Med. 1995 Nov;333(20):1319-27. DOI: 10.1056/NEJM199511163332004 External link
9.
Huppertz HI, Böhme M, Standaert SM, Karch H, Plotkin SA. Incidence of Lyme borreliosis in the Würzburg region of Germany. Eur J Clin Microbiol Infect Dis. 1999 Oct;18(10):697-703. DOI: 10.1007/s100960050381 External link
10.
Fülöp B, Poggensee G. Epidemiological situation of Lyme borreliosis in germany: surveillance data from six Eastern German States, 2002 to 2006. Parasitol Res. 2008 Dec;103 Suppl 1:S117-20. DOI: 10.1007/s00436-008-1060-y External link
11.
Wilking H, Stark K. Trends in surveillance data of human Lyme borreliosis from six federal states in eastern Germany, 2009-2012. Ticks Tick Borne Dis. 2014 Apr;5(3):219-24. DOI: 10.1016/j.ttbdis.2013.10.010 External link
12.
Müller I, Freitag MH, Poggensee G, Scharnetzky E, Straube E, Schoerner C, Hlobil H, Hagedorn HJ, Stanek G, Schubert-Unkmeir A, Norris DE, Gensichen J, Hunfeld KP. Evaluating frequency, diagnostic quality, and cost of Lyme borreliosis testing in Germany: a retrospective model analysis. Clin Dev Immunol. 2012;2012:595427. DOI: 10.1155/2012/595427 External link
13.
Wilking H, Fingerle V, Klier C, Thamm M, Stark K. Antibodies against Borrelia burgdorferi sensu lato among Adults, Germany, 2008-2011. Emerging Infect Dis. 2015 Jan;21(1):107-10. DOI: 10.3201/eid2101.140009 External link
14.
Wilhelmsson P, Fryland L, Lindblom P, Sjöwall J, Ahlm C, Berglund J, Haglund M, Henningsson AJ, Nolskog P, Nordberg M, Nyberg C, Ornstein K, Nyman D, Ekerfelt C, Forsberg P, Lindgren PE. A prospective study on the incidence of Borrelia burgdorferi sensu lato infection after a tick bite in Sweden and on the Åland Islands, Finland (2008-2009). Ticks Tick Borne Dis. 2016 Feb;7(1):71-79. DOI: 10.1016/j.ttbdis.2015.08.009 External link
15.
Fingerle V, Bergmeister H, Liegl G, Vanek E, Wilske B. Prevalence of Borrelia burgdorferi sensu lato in Ixodes ricinus in Southern Germany. J Spiroch Tick Dis. 1994;1:41-5.
16.
Cotté V, Sabatier L, Schnell G, Carmi-Leroy A, Rousselle JC, Arsène-Ploetze F, Malandrin L, Sertour N, Namane A, Ferquel E, Choumet V. Differential expression of Ixodes ricinus salivary gland proteins in the presence of the Borrelia burgdorferi sensu lato complex. J Proteomics. 2014 Jan;96:29-43. DOI: 10.1016/j.jprot.2013.10.033 External link
17.
Gern L. Life cycle of Borrelia burgdorferi sensu lato and transmission to humans. Curr Probl Dermatol. 2009;37:18-30. DOI: 10.1159/000213068 External link
18.
de Taeye SW, Kreuk L, van Dam AP, Hovius JW, Schuijt TJ. Complement evasion by Borrelia burgdorferi: it takes three to tango. Trends Parasitol. 2013 Mar;29(3):119-28. DOI: 10.1016/j.pt.2012.12.001 External link
19.
Hartiala P, Hytönen J, Suhonen J, Leppäranta O, Tuominen-Gustafsson H, Viljanen MK. Borrelia burgdorferi inhibits human neutrophil functions. Microbes Infect. 2008 Jan;10(1):60-8. DOI: 10.1016/j.micinf.2007.10.004 External link
20.
Rogovskyy AS, Bankhead T. Variable VlsE is critical for host reinfection by the Lyme disease spirochete. PLoS ONE. 2013;8(4):e61226. DOI: 10.1371/journal.pone.0061226 External link
21.
Bachmann M, Horn K, Rudloff I, Goren I, Holdener M, Christen U, Darsow N, Hunfeld KP, Koehl U, Kind P, Pfeilschifter J, Kraiczy P, Mühl H. Early production of IL-22 but not IL-17 by peripheral blood mononuclear cells exposed to live Borrelia burgdorferi: the role of monocytes and interleukin-1. PLoS Pathog. 2010 Oct;6(10):e1001144. DOI: 10.1371/journal.ppat.1001144 External link
22.
Cruz AR, Moore MW, La Vake CJ, Eggers CH, Salazar JC, Radolf JD. Phagocytosis of Borrelia burgdorferi, the Lyme disease spirochete, potentiates innate immune activation and induces apoptosis in human monocytes. Infect Immun. 2008 Jan;76(1):56-70. DOI: 10.1128/IAI.01039-07 External link
23.
Hovius JW, Bijlsma MF, van der Windt GJ, Wiersinga WJ, Boukens BJ, Coumou J, Oei A, de Beer R, de Vos AF, van 't Veer C, van Dam AP, Wang P, Fikrig E, Levi MM, Roelofs JJ, van der Poll T. The urokinase receptor (uPAR) facilitates clearance of Borrelia burgdorferi. PLoS Pathog. 2009 May;5(5):e1000447. DOI: 10.1371/journal.ppat.1000447 External link
24.
Petzke MM, Brooks A, Krupna MA, Mordue D, Schwartz I. Recognition of Borrelia burgdorferi, the Lyme disease spirochete, by TLR7 and TLR9 induces a type I IFN response by human immune cells. J Immunol. 2009 Oct;183(8):5279-92. DOI: 10.4049/jimmunol.0901390 External link
25.
Salazar JC, Duhnam-Ems S, La Vake C, Cruz AR, Moore MW, Caimano MJ, Velez-Climent L, Shupe J, Krueger W, Radolf JD. Activation of human monocytes by live Borrelia burgdorferi generates TLR2-dependent and -independent responses which include induction of IFN-beta. PLoS Pathog. 2009 May;5(5):e1000444. DOI: 10.1371/journal.ppat.1000444 External link
26.
Skogman BH, Hellberg S, Ekerfelt C, Jenmalm MC, Forsberg P, Ludvigsson J, Bergström S, Ernerudh J. Adaptive and innate immune responsiveness to Borrelia burgdorferi sensu lato in exposed asymptomatic children and children with previous clinical Lyme borreliosis. Clin Dev Immunol. 2012;2012:294587. DOI: 10.1155/2012/294587 External link
27.
Stanek G, Wormser GP, Gray J, Strle F. Lyme borreliosis. Lancet. 2012 Feb;379(9814):461-73. DOI: 10.1016/S0140-6736(11)60103-7 External link
28.
Takeuchi O, Sato S, Horiuchi T, Hoshino K, Takeda K, Dong Z, Modlin RL, Akira S. Cutting edge: role of Toll-like receptor 1 in mediating immune response to microbial lipoproteins. J Immunol. 2002 Jul;169(1):10-4.
29.
Dai J, Narasimhan S, Zhang L, Liu L, Wang P, Fikrig E. Tick histamine release factor is critical for Ixodes scapularis engorgement and transmission of the lyme disease agent. PLoS Pathog. 2010 Nov;6(11):e1001205. DOI: 10.1371/journal.ppat.1001205 External link
30.
Dai J, Wang P, Adusumilli S, Booth CJ, Narasimhan S, Anguita J, Fikrig E. Antibodies against a tick protein, Salp15, protect mice from the Lyme disease agent. Cell Host Microbe. 2009 Nov;6(5):482-92. DOI: 10.1016/j.chom.2009.10.006 External link
31.
Horká H, Cerná-Kýcková K, Skallová A, Kopecký J. Tick saliva affects both proliferation and distribution of Borrelia burgdorferi spirochetes in mouse organs and increases transmission of spirochetes to ticks. Int J Med Microbiol. 2009 Jun;299(5):373-80. DOI: 10.1016/j.ijmm.2008.10.009 External link
32.
Hovius JW, Schuijt TJ, de Groot KA, Roelofs JJ, Oei GA, Marquart JA, de Beer R, van 't Veer C, van der Poll T, Ramamoorthi N, Fikrig E, van Dam AP. Preferential protection of Borrelia burgdorferi sensu stricto by a Salp15 homologue in Ixodes ricinus saliva. J Infect Dis. 2008 Oct;198(8):1189-97. DOI: 10.1086/591917 External link
33.
Kern A, Collin E, Barthel C, Michel C, Jaulhac B, Boulanger N. Tick saliva represses innate immunity and cutaneous inflammation in a murine model of Lyme disease. Vector Borne Zoonotic Dis. 2011 Oct;11(10):1343-50. DOI: 10.1089/vbz.2010.0197 External link
34.
Oliveira CJ, Sá-Nunes A, Francischetti IM, Carregaro V, Anatriello E, Silva JS, Santos IK, Ribeiro JM, Ferreira BR. Deconstructing tick saliva: non-protein molecules with potent immunomodulatory properties. J Biol Chem. 2011 Apr;286(13):10960-9. DOI: 10.1074/jbc.M110.205047 External link
35.
Radolf JD, Caimano MJ, Stevenson B, Hu LT. Of ticks, mice and men: understanding the dual-host lifestyle of Lyme disease spirochaetes. Nat Rev Microbiol. 2012 Jan;10(2):87-99. DOI: 10.1038/nrmicro2714 External link
36.
Ramamoorthi N, Narasimhan S, Pal U, Bao F, Yang XF, Fish D, Anguita J, Norgard MV, Kantor FS, Anderson JF, Koski RA, Fikrig E. The Lyme disease agent exploits a tick protein to infect the mammalian host. Nature. 2005 Jul;436(7050):573-7. DOI: 10.1038/nature03812 External link
37.
Ribeiro JM, Makoul GT, Levine J, Robinson DR, Spielman A. Antihemostatic, antiinflammatory, and immunosuppressive properties of the saliva of a tick, Ixodes dammini. J Exp Med. 1985 Feb;161(2):332-44.
38.
Mullegger RR. Dermatological manifestations of Lyme borreliosis. Eur J Dermatol. 2004 Sep-Oct;14(5):296-309.
39.
Strle K, Stupica D, Drouin EE, Steere AC, Strle F. Elevated levels of IL-23 in a subset of patients with post-lyme disease symptoms following erythema migrans. Clin Infect Dis. 2014 Feb;58(3):372-80. DOI: 10.1093/cid/cit735 External link
40.
Bankhead T, Chaconas G. The role of VlsE antigenic variation in the Lyme disease spirochete: persistence through a mechanism that differs from other pathogens. Mol Microbiol. 2007 Sep;65(6):1547-58. DOI: 10.1111/j.1365-2958.2007.05895.x External link
41.
Crowley JT, Toledo AM, LaRocca TJ, Coleman JL, London E, Benach JL. Lipid exchange between Borrelia burgdorferi and host cells. PLoS Pathog. 2013 Jan;9(1):e1003109. DOI: 10.1371/journal.ppat.1003109 External link
42.
Tilly K, Krum JG, Bestor A, Jewett MW, Grimm D, Bueschel D, Byram R, Dorward D, Vanraden MJ, Stewart P, Rosa P. Borrelia burgdorferi OspC protein required exclusively in a crucial early stage of mammalian infection. Infect Immun. 2006 Jun;74(6):3554-64. DOI: 10.1128/IAI.01950-05 External link
43.
Xu Q, McShan K, Liang FT. Identification of an ospC operator critical for immune evasion of Borrelia burgdorferi. Mol Microbiol. 2007 Apr;64(1):220-31. DOI: 10.1111/j.1365-2958.2007.05636.x External link
44.
Hartmann K, Corvey C, Skerka C, Kirschfink M, Karas M, Brade V, Miller JC, Stevenson B, Wallich R, Zipfel PF, Kraiczy P. Functional characterization of BbCRASP-2, a distinct outer membrane protein of Borrelia burgdorferi that binds host complement regulators factor H and FHL-1. Mol Microbiol. 2006 Sep;61(5):1220-36. DOI: 10.1111/j.1365-2958.2006.05318.x External link
45.
Hunfeld KP, Ruzic-Sabljic E, Norris DE, Kraiczy P, Strle F. Risk of culture-confirmed borrelial persistence in patients treated for erythema migrans and possible mechanisms of resistance. Int J Med Microbiol. 2006 May;296 Suppl 40:233-41. DOI: 10.1016/j.ijmm.2006.01.028 External link
46.
Kraiczy P, Rossmann E, Brade V, Simon MM, Skerka C, Zipfel PF, Wallich R. Binding of human complement regulators FHL-1 and factor H to CRASP-1 orthologs of Borrelia burgdorferi. Wien Klin Wochenschr. 2006 Nov;118(21-22):669-76. DOI: 10.1007/s00508-006-0691-1 External link
47.
Kraiczy P, Würzner R. Complement escape of human pathogenic bacteria by acquisition of complement regulators. Mol Immunol. 2006 Jan;43(1-2):31-44. DOI: 10.1016/j.molimm.2005.06.016 External link
48.
Rossmann E, Kitiratschky V, Hofmann H, Kraiczy P, Simon MM, Wallich R. Borrelia burgdorferi complement regulator-acquiring surface protein 1 of the Lyme disease spirochetes is expressed in humans and induces antibody responses restricted to nondenatured structural determinants. Infect Immun. 2006 Dec;74(12):7024-8. DOI: 10.1128/IAI.01028-06 External link
49.
Coleman JL, Roemer EJ, Benach JL. Plasmin-coated borrelia Burgdorferi degrades soluble and insoluble components of the mammalian extracellular matrix. Infect Immun. 1999 Aug;67(8):3929-36.
50.
Fuchs H, Simon MM, Wallich R, Bechtel M, Kramer MD. Borrelia burgdorferi induces secretion of pro-urokinase-type plasminogen activator by human monocytes. Infect Immun. 1996 Oct;64(10):4307-12.
51.
Grab DJ, Kennedy R, Philipp MT. Borrelia burgdorferi possesses a collagenolytic activity. FEMS Microbiol Lett. 1996 Oct;144(1):39-45. DOI: 10.1111/j.1574-6968.1996.tb08506.x External link
52.
Önder Ö, Humphrey PT, McOmber B, Korobova F, Francella N, Greenbaum DC, Brisson D. OspC is potent plasminogen receptor on surface of Borrelia burgdorferi. J Biol Chem. 2012 May;287(20):16860-8. DOI: 10.1074/jbc.M111.290775 External link
53.
Sarkar A, Tilly K, Stewart P, Bestor A, Battisti JM, Rosa PA. Borrelia burgdorferi resistance to a major skin antimicrobial peptide is independent of outer surface lipoprotein content. Antimicrob Agents Chemother. 2009 Oct;53(10):4490-4. DOI: 10.1128/AAC.00558-09 External link
54.
Shi Y, Xu Q, Seemanaplli SV, McShan K, Liang FT. Common and unique contributions of decorin-binding proteins A and B to the overall virulence of Borrelia burgdorferi. PLoS ONE. 2008 Oct;3(10):e3340. DOI: 10.1371/journal.pone.0003340 External link
55.
Srivastava SY, de Silva AM. Reciprocal expression of ospA and ospC in single cells of Borrelia burgdorferi. J Bacteriol. 2008 May;190(10):3429-33. DOI: 10.1128/JB.00085-08 External link
56.
Tilly K, Bestor A, Rosa PA. Lipoprotein succession in Borrelia burgdorferi: similar but distinct roles for OspC and VlsE at different stages of mammalian infection. Mol Microbiol. 2013 Jul;89(2):216-27. DOI: 10.1111/mmi.12271 External link
57.
Xu Q, McShan K, Liang FT. Essential protective role attributed to the surface lipoproteins of Borrelia burgdorferi against innate defences. Mol Microbiol. 2008 Jul;69(1):15-29. DOI: 10.1111/j.1365-2958.2008.06264.x External link
58.
Gautam A, Dixit S, Philipp MT, Singh SR, Morici LA, Kaushal D, Dennis VA. Interleukin-10 alters effector functions of multiple genes induced by Borrelia burgdorferi in macrophages to regulate Lyme disease inflammation. Infect Immun. 2011 Dec;79(12):4876-92. DOI: 10.1128/IAI.05451-11 External link
59.
Rupprecht TA, Kirschning CJ, Popp B, Kastenbauer S, Fingerle V, Pfister HW, Koedel U. Borrelia garinii induces CXCL13 production in human monocytes through Toll-like receptor 2. Infect Immun. 2007 Sep;75(9):4351-6. DOI: 10.1128/IAI.01642-06 External link
60.
Shin OS, Isberg RR, Akira S, Uematsu S, Behera AK, Hu LT. Distinct roles for MyD88 and Toll-like receptors 2, 5, and 9 in phagocytosis of Borrelia burgdorferi and cytokine induction. Infect Immun. 2008 Jun;76(6):2341-51. DOI: 10.1128/IAI.01600-07 External link
61.
Yang X, Izadi H, Coleman AS, Wang P, Ma Y, Fikrig E, Anguita J, Pal U. Borrelia burgdorferi lipoprotein BmpA activates pro-inflammatory responses in human synovial cells through a protein moiety. Microbes Infect. 2008 Oct;10(12-13):1300-8. DOI: 10.1016/j.micinf.2008.07.029 External link
62.
Codolo G, Bossi F, Durigutto P, Bella CD, Fischetti F, Amedei A, Tedesco F, D'Elios S, Cimmino M, Micheletti A, Cassatella MA, D'Elios MM, de Bernard M. Orchestration of inflammation and adaptive immunity in Borrelia burgdorferi-induced arthritis by neutrophil-activating protein A. Arthritis Rheum. 2013 May;65(5):1232-42. DOI: 10.1002/art.37875 External link
63.
Lee WY, Moriarty TJ, Wong CH, Zhou H, Strieter RM, van Rooijen N, Chaconas G, Kubes P. An intravascular immune response to Borrelia burgdorferi involves Kupffer cells and iNKT cells. Nat Immunol. 2010 Apr;11(4):295-302. DOI: 10.1038/ni.1855 External link
64.
Shen S, Shin JJ, Strle K, McHugh G, Li X, Glickstein LJ, Drouin EE, Steere AC. Treg cell numbers and function in patients with antibiotic-refractory or antibiotic-responsive Lyme arthritis. Arthritis Rheum. 2010 Jul;62(7):2127-37. DOI: 10.1002/art.27468 External link
65.
Glatz M, Resinger A, Semmelweis K, Ambros-Rudolph CM, Müllegger RR. Clinical spectrum of skin manifestations of Lyme borreliosis in 204 children in Austria. Acta Derm Venereol. 2015 May;95(5):565-71. DOI: 10.2340/00015555-2000 External link
66.
Oschmann P, Kraiczy P, Halperin JJ, Brade V. Lyme borreliosis and tick-borne encephalitis. Bremen: UNI-MED Verlag; 1999.
67.
Strle F, Lusa L, Ružic-Sabljic E, Maraspin V, Lotric Furlan S, Cimperman J, Ogrinc K, Rojko T, Videcnik Zorman J, Stupica D. Clinical characteristics associated with Borrelia burgdorferi sensu lato skin culture results in patients with erythema migrans. PLoS ONE. 2013;8(12):e82132. DOI: 10.1371/journal.pone.0082132 External link
68.
Stanek G, Fingerle V, Hunfeld KP, Jaulhac B, Kaiser R, Krause A, Kristoferitsch W, O'Connell S, Ornstein K, Strle F, Gray J. Lyme borreliosis: clinical case definitions for diagnosis and management in Europe. Clin Microbiol Infect. 2011 Jan;17(1):69-79. DOI: 10.1111/j.1469-0691.2010.03175.x External link
69.
Eriksson P, Schröder MT, Niiranen K, Nevanlinna A, Panelius J, Ranki A. The many faces of solitary and multiple erythema migrans. Acta Derm Venereol. 2013 Nov;93(6):693-700. DOI: 10.2340/00015555-1549 External link
70.
Hofmann H. Variabilität der kutanen Lyme-Borreliose. Diagnostik und Therapie [The variable spectrum of cutaneous Lyme borreliosis. Diagnosis and therapy]. Hautarzt. 2012 May;63(5):381-9. DOI: 10.1007/s00105-011-2256-0 External link
71.
Aberer E. Lyme borreliosis--an update. J Dtsch Dermatol Ges. 2007 May;5(5):406-14. DOI: 10.1111/j.1610-0387.2007.06285.x External link
72.
Ang CW, Brandenburg AH, van Burgel ND, Bijlmer HA, Herremans T, Stelma F, Lunel FV, van Dam AP; Dutch Working Group on Diagnosis of Lyme Borreliosis. A Dutch nationwide evaluation of serological assays for detection of Borrelia antibodies in clinically well-defined patients. Diagn Microbiol Infect Dis. 2015 Nov;83(3):222-8. DOI: 10.1016/j.diagmicrobio.2015.07.007 External link
73.
Maraspin V, Cimperman J, Lotric-Furlan S, Ruzic-Sabljic E, Jurca T, Picken RN, Strle F. Solitary borrelial lymphocytoma in adult patients. Wien Klin Wochenschr. 2002 Jul;114(13-14):515-23.
74.
Hovmark A, Asbrink E, Olsson I. The spirochetal etiology of lymphadenosis benigna cutis solitaria. Acta Derm Venereol. 1986;66(6):479-84.
75.
Lenormand C, Jaulhac B, De Martino S, Barthel C, Lipsker D. Species of Borrelia burgdorferi complex that cause borrelial lymphocytoma in France. Br J Dermatol. 2009 Jul;161(1):174-6. DOI: 10.1111/j.1365-2133.2009.09100.x External link
76.
Arnež M, Ružic-Sabljic E. Borrelial Lymphocytoma in Children. Pediatr Infect Dis J. 2015 Dec;34(12):1319-22. DOI: 10.1097/INF.0000000000000884 External link
77.
Arnez M, Pleterski-Rigler D, Luznik-Bufon T, Ruzic-Sabljic E, Strle F. Solitary and multiple erythema migrans in children: comparison of demographic, clinical and laboratory findings. Infection. 2003 Dec;31(6):404-9. DOI: 10.1007/s15010-003-4007-3 External link
78.
Arnež M, Ružic-Sabljic E. Borrelia burgdorferi sensu lato bacteremia in Slovenian children with solitary and multiple erythema migrans. Pediatr Infect Dis J. 2011 Nov;30(11):988-90. DOI: 10.1097/INF.0b013e318225b8c3 External link
79.
Arnez M, Pleterski-Rigler D, Luznik-Bufon T, Ruzic-Sabljic E, Strle F. Solitary erythema migrans in children: comparison of treatment with azithromycin and phenoxymethylpenicillin. Wien Klin Wochenschr. 2002 Jul;114(13-14):498-504.
80.
Liveris D, Schwartz I, McKenna D, Nowakowski J, Nadelman RB, DeMarco J, Iyer R, Cox ME, Holmgren D, Wormser GP. Quantitation of cell-associated borrelial DNA in the blood of Lyme disease patients with erythema migrans. Eur J Clin Microbiol Infect Dis. 2012 May;31(5):791-5. DOI: 10.1007/s10096-011-1376-x External link
81.
Asbrink E. Acrodermatitis chronica atrophicans. Clin Dermatol. 1993 Jul-Sep;11(3):369-75. DOI: 10.1016/0738-081X(93)90092-Q External link
82.
Andres C, Ziai M, Bruckbauer H, Ring J, Hofmann H. Acrodermatitis chronica atrophicans in two children. Int J Dermatol. 2010 Feb;49(2):180-3. DOI: 10.1111/j.1365-4632.2009.04194.x External link
83.
Brzonova I, Wollenberg A, Prinz JC. Acrodermatitis chronica atrophicans affecting all four limbs in an 11-year-old girl. Br J Dermatol. 2002 Aug;147(2):375-8. DOI: 10.1046/j.1365-2133.2002.04792.x External link
84.
Zalaudek I, Leinweber B, Kerl H, Müllegger RR. Acrodermatitis chronica atrophicans in a 15-year-old girl misdiagnosed as venous insufficiency for 6 years. J Am Acad Dermatol. 2005 Jun;52(6):1091-4. DOI: 10.1016/j.jaad.2005.01.125 External link
85.
Hopf HC. Peripheral neuropathy in acrodermatitis chronica atrophicans (Herxheimer). J Neurol Neurosurg Psychiatr. 1975 May;38(5):452-8. DOI: 10.1136/jnnp.38.5.452 External link
86.
Kindstrand E, Nilsson BY, Hovmark A, Pirskanen R, Asbrink E. Peripheral neuropathy in acrodermatitis chronica atrophicans - a late Borrelia manifestation. Acta Neurol Scand. 1997 Jun;95(6):338-45. DOI: 10.1111/j.1600-0404.1997.tb00222.x External link
87.
Kristoferitsch W, Sluga E, Graf M, Partsch H, Neumann R, Stanek G, Budka H. Neuropathy associated with acrodermatitis chronica atrophicans. Clinical and morphological features. Ann N Y Acad Sci. 1988;539:35-45. DOI: 10.1111/j.1749-6632.1988.tb31836.x External link
88.
Asbrink E, Hovmark A. Successful cultivation of spirochetes from skin lesions of patients with erythema chronicum migrans Afzelius and acrodermatitis chronica atrophicans. Acta Pathol Microbiol Immunol Scand B. 1985 Apr;93(2):161-3. DOI: 10.1111/j.1699-0463.1985.tb02870.x External link
89.
Brehmer-Andersson E, Hovmark A, Asbrink E. Acrodermatitis chronica atrophicans: histopathologic findings and clinical correlations in 111 cases. Acta Derm Venereol. 1998 May;78(3):207-13. DOI: 10.1080/000155598441558 External link
90.
Arnez M, Pleterski-Rigler D, Luznik-Bufon T, Ruzic-Sabljic E, Strle F. Children with multiple erythema migrans: are there any pre-treatment symptoms and/or signs suggestive for central nervous system involvement? Wien Klin Wochenschr. 2002 Jul;114(13-14):524-9.
91.
Hunfeld KP, Wichelhaus TA, Brade V. Borreliose. In: Thomas L, editor. Labor und Diagnose: Indikation und Bewertung von Laborbefunden für die medizinische Diagnostik. 8. Auflage. Frankfurt/Main: Th-Books-Verl.-Ges.; 2012. p. 1955-65.
92.
DIN 58969-44:2005-07. Medizinische Mikrobiologie - Serologische und molekularbiologische Diagnostik von Infektionskrankheiten - Teil 44: Immunoblot (IB); Spezielle Anforderungen für den Nachweis von Antikörpern gegen Borrelia burgdorferi. 2005.
93.
Hunfeld KP, Stanek G, Straube E, Hagedorn HJ, Schörner C, Mühlschlegel F, Brade V. Quality of Lyme disease serology. Lessons from the German Proficiency Testing Program 1999-2001. A preliminary report. Wien Klin Wochenschr. 2002 Jul;114(13-14):591-600.
94.
Cetin E, Sotoudeh M, Auer H, Stanek G. Paradigm Burgenland: risk of Borrelia burgdorferi sensu lato infection indicated by variable seroprevalence rates in hunters. Wien Klin Wochenschr. 2006 Nov;118(21-22):677-81. DOI: 10.1007/s00508-006-0694-y External link
95.
Münchhoff P, Wilske B, Preac-Mursic V, Schierz G. Antibodies against Borrelia burgdorferi in Bavarian forest workers. Zentralbl Bakteriol Mikrobiol Hyg A. 1987 Feb;263(3):412-9. DOI: 10.1016/S0176-6724(87)80101-3 External link
96.
Dehnert M, Fingerle V, Klier C, Talaska T, Schlaud M, Krause G, Wilking H, Poggensee G. Seropositivity of Lyme borreliosis and associated risk factors: a population-based study in Children and Adolescents in Germany (KiGGS). PLoS ONE. 2012;7(8):e41321. DOI: 10.1371/journal.pone.0041321 External link
97.
Branda JA, Strle F, Strle K, Sikand N, Ferraro MJ, Steere AC. Performance of United States serologic assays in the diagnosis of Lyme borreliosis acquired in Europe. Clin Infect Dis. 2013 Aug;57(3):333-40. DOI: 10.1093/cid/cit235 External link
98.
Nyman D, Willén L, Jansson C, Carlsson SA, Granlund H, Wahlberg P. VlsE C6 peptide and IgG ELISA antibody analysis for clinical diagnosis of Lyme borreliosis in an endemic area. Clin Microbiol Infect. 2006 May;12(5):496-7. DOI: 10.1111/j.1469-0691.2006.01374.x External link
99.
Hunfeld KP, Kraiczy P. When is the best time to order a Western blot and how should it be interpreted? Curr Probl Dermatol. 2009;37:167-77. DOI: 10.1159/000213074 External link
100.
Goettner G, Schulte-Spechtel U, Hillermann R, Liegl G, Wilske B, Fingerle V. Improvement of Lyme borreliosis serodiagnosis by a newly developed recombinant immunoglobulin G (IgG) and IgM line immunoblot assay and addition of VlsE and DbpA homologues. J Clin Microbiol. 2005 Aug;43(8):3602-9. DOI: 10.1128/JCM.43.8.3602-3609.2005 External link
101.
Hauser U, Lehnert G, Wilske B. Validity of interpretation criteria for standardized Western blots (immunoblots) for serodiagnosis of Lyme borreliosis based on sera collected throughout Europe. J Clin Microbiol. 1999 Jul;37(7):2241-7.
102.
Bunikis J, Barbour AG. Laboratory testing for suspected Lyme disease. Med Clin North Am. 2002 Mar;86(2):311-40. DOI: 10.1016/S0025-7125(03)00089-0 External link
103.
Barbour AG. Immunochemical analysis of Lyme disease spirochetes. Yale J Biol Med. 1984 Jul-Aug;57(4):581-6.
104.
Preac-Mursic V, Wilske B, Schierz G. European Borrelia burgdorferi isolated from humans and ticks culture conditions and antibiotic susceptibility. Zentralbl Bakteriol Mikrobiol Hyg A. 1986 Dec;263(1-2):112-8. DOI: 10.1016/S0176-6724(86)80110-9 External link
105.
Preac-Mursic V, Pfister HW, Spiegel H, Burk R, Wilske B, Reinhardt S, Böhmer R. First isolation of Borrelia burgdorferi from an iris biopsy. J Clin Neuroophthalmol. 1993 Sep;13(3):155-61; discussion 162.
106.
Stanek G, Klein J, Bittner R, Glogar D. Borrelia burgdorferi as an etiologic agent in chronic heart failure? Scand J Infect Dis Suppl. 1991;77:85-7.
107.
Cerar T, Ruzic-Sabljic E, Glinsek U, Zore A, Strle F. Comparison of PCR methods and culture for the detection of Borrelia spp. in patients with erythema migrans. Clin Microbiol Infect. 2008 Jul;14(7):653-8. DOI: 10.1111/j.1469-0691.2008.02013.x External link
108.
Kuiper H, Cairo I, Van Dam A, De Jongh B, Ramselaar T, Spanjaard L, Dankert J. Solitary erythema migrans: a clinical, laboratory and epidemiological study of 77 Dutch patients. Br J Dermatol. 1994 Apr;130(4):466-72. DOI: 10.1111/j.1365-2133.1994.tb03379.x External link
109.
Picken RN, Strle F, Picken MM, Ruzic-Sabljic E, Maraspin V, Lotric-Furlan S, Cimperman J. Identification of three species of Borrelia burgdorferi sensu lato (B. burgdorferi sensu stricto, B. garinii, and B. afzelii) among isolates from acrodermatitis chronica atrophicans lesions. J Invest Dermatol. 1998 Mar;110(3):211-4. DOI: 10.1046/j.1523-1747.1998.00130.x External link
110.
van Dam AP, Kuiper H, Vos K, Widjojokusumo A, de Jongh BM, Spanjaard L, Ramselaar AC, Kramer MD, Dankert J. Different genospecies of Borrelia burgdorferi are associated with distinct clinical manifestations of Lyme borreliosis. Clin Infect Dis. 1993 Oct;17(4):708-17. DOI: 10.1093/clinids/17.4.708 External link
111.
Aguero-Rosenfeld ME, Wang G, Schwartz I, Wormser GP. Diagnosis of lyme borreliosis. Clin Microbiol Rev. 2005 Jul;18(3):484-509. DOI: 10.1128/CMR.18.3.484-509.2005 External link
112.
van Dam AP. Molecular diagnosis of Borrelia bacteria for the diagnosis of Lyme disease. Expert Opin Med Diagn. 2011 Mar;5(2):135-49. DOI: 10.1517/17530059.2011.555396 External link
113.
Brettschneider S, Bruckbauer H, Klugbauer N, Hofmann H. Diagnostic value of PCR for detection of Borrelia burgdorferi in skin biopsy and urine samples from patients with skin borreliosis. J Clin Microbiol. 1998 Sep;36(9):2658-65.
114.
Moter SE, Hofmann H, Wallich R, Simon MM, Kramer MD. Detection of Borrelia burgdorferi sensu lato in lesional skin of patients with erythema migrans and acrodermatitis chronica atrophicans by ospA-specific PCR. J Clin Microbiol. 1994 Dec;32(12):2980-8.
115.
Muellegger R, Zoechling N, Schluepen EM, Soyer HP, Hoedl S, Kerl, Volkenandt M. Polymerase chain reaction control of antibiotic treatment in dermatoborreliosis. Infection. 1996 Jan-Feb;24(1):76-9. DOI: 10.1007/BF01780664 External link
116.
von Stedingk LV, Olsson I, Hanson HS, Asbrink E, Hovmark A. Polymerase chain reaction for detection of Borrelia burgdorferi DNA in skin lesions of early and late Lyme borreliosis. Eur J Clin Microbiol Infect Dis. 1995 Jan;14(1):1-5. DOI: 10.1007/BF02112610 External link
117.
Bockenstedt LK, Gonzalez DG, Haberman AM, Belperron AA. Spirochete antigens persist near cartilage after murine Lyme borreliosis therapy. J Clin Invest. 2012 Jul;122(7):2652-60. DOI: 10.1172/JCI58813 External link
118.
Iyer R, Mukherjee P, Wang K, Simons J, Wormser GP, Schwartz I. Detection of Borrelia burgdorferi nucleic acids after antibiotic treatment does not confirm viability. J Clin Microbiol. 2013 Mar;51(3):857-62. DOI: 10.1128/JCM.02785-12 External link
119.
Pícha D, Moravcová L, Vanousová D, Hercogová J, Blechová Z. DNA persistence after treatment of Lyme borreliosis. Folia Microbiol (Praha). 2014 Mar;59(2):115-25. DOI: 10.1007/s12223-013-0272-4 External link
120.
Aberer E, Bergmann AR, Derler AM, Schmidt B. Course of Borrelia burgdorferi DNA shedding in urine after treatment. Acta Derm Venereol. 2007;87(1):39-42. DOI: 10.2340/00015555-0172 External link
121.
Rauter C, Mueller M, Diterich I, Zeller S, Hassler D, Meergans T, Hartung T. Critical evaluation of urine-based PCR assay for diagnosis of Lyme borreliosis. Clin Diagn Lab Immunol. 2005 Aug;12(8):910-7. DOI: 10.1128/CDLI.12.8.910-917.2005 External link
122.
Colli C, Leinweber B, Müllegger R, Chott A, Kerl H, Cerroni L. Borrelia burgdorferi-associated lymphocytoma cutis: clinicopathologic, immunophenotypic, and molecular study of 106 cases. J Cutan Pathol. 2004 Mar;31(3):232-40. DOI: 10.1111/j.0303-6987.2003.00167.x External link
123.
Dattwyler RJ, Volkman DJ, Conaty SM, Platkin SP, Luft BJ. Amoxycillin plus probenecid versus doxycycline for treatment of erythema migrans borreliosis. Lancet. 1990 Dec;336(8728):1404-6. DOI: 10.1016/0140-6736(90)93103-V External link
124.
Hunfeld KP, Rödel R, Wichelhaus TA. In vitro activity of eight oral cephalosporins against Borrelia burgdorferi. Int J Antimicrob Agents. 2003 Apr;21(4):313-8. DOI: 10.1016/S0924-8579(03)00005-0 External link
125.
Dattwyler RJ, Luft BJ, Kunkel MJ, Finkel MF, Wormser GP, Rush TJ, Grunwaldt E, Agger WA, Franklin M, Oswald D, Cockey L, Maladorno D. Ceftriaxone compared with doxycycline for the treatment of acute disseminated Lyme disease. N Engl J Med. 1997 Jul 31;337(5):289-94. DOI: 10.1056/NEJM199707313370501 External link
126.
Barsic B, Maretic T, Majerus L, Strugar J. Comparison of azithromycin and doxycycline in the treatment of erythema migrans. Infection. 2000 May-Jun;28(3):153-6. DOI: 10.1007/s150100050069 External link
127.
Luft BJ, Dattwyler RJ, Johnson RC, Luger SW, Bosler EM, Rahn DW, Masters EJ, Grunwaldt E, Gadgil SD. Azithromycin compared with amoxicillin in the treatment of erythema migrans. A double-blind, randomized, controlled trial. Ann Intern Med. 1996 May;124(9):785-91. DOI: 10.7326/0003-4819-124-9-199605010-00002 External link
128.
Weber K, Wilske B, Preac-Mursic V, Thurmayr R. Azithromycin versus penicillin V for the treatment of early Lyme borreliosis. Infection. 1993 Nov-Dec;21(6):367-72. DOI: 10.1007/BF01728915 External link
129.
Nizic T, Velikanje E, Ružic-Sabljic E, Arnež M. Solitary erythema migrans in children: comparison of treatment with clarithromycin and amoxicillin. Wien Klin Wochenschr. 2012 Jul;124(13-14):427-33. DOI: 10.1007/s00508-012-0194-1 External link
130.
Hansen K, Hovmark A, Lebech AM, Lebech K, Olsson I, Halkier-Sørensen L, Olsson E, Asbrink E. Roxithromycin in Lyme borreliosis: discrepant results of an in vitro and in vivo animal susceptibility study and a clinical trial in patients with erythema migrans. Acta Derm Venereol. 1992 Aug;72(4):297-300.
131.
Aberer E, Kahofer P, Binder B, Kinaciyan T, Schauperl H, Berghold A. Comparison of a two- or three-week regimen and a review of treatment of erythema migrans with phenoxymethylpenicillin. Dermatology (Basel). 2006;212(2):160-7. DOI: 10.1159/000090657 External link
132.
Arnez M. Antibiotic treatment of children with erythema migrans. Clin Infect Dis. 2007 Apr;44(8):1133-4; author reply 1137-9. DOI: 10.1086/512978 External link
133.
Bennet L, Danell S, Berglund J. Clinical outcome of erythema migrans after treatment with phenoxymethyl penicillin. Scand J Infect Dis. 2003;35(2):129-31. infectious diseases. 2003;35(2):129-31. DOI: 10.1080/0036554021000027009 External link
134.
Stupica D, Lusa L, Ruzic-Sabljic E, Cerar T, Strle F. Treatment of erythema migrans with doxycycline for 10 days versus 15 days. Clin Infect Dis. 2012 Aug;55(3):343-50. DOI: 10.1093/cid/cis402 External link
135.
Aberer E, Breier F, Stanek G, Schmidt B. Success and failure in the treatment of acrodermatitis chronica atrophicans. Infection. 1996 Jan-Feb;24(1):85-7. DOI: 10.1007/BF01780666 External link
136.
Cerar D, Cerar T, Ruzic-Sabljic E, Wormser GP, Strle F. Subjective symptoms after treatment of early Lyme disease. Am J Med. 2010 Jan;123(1):79-86. DOI: 10.1016/j.amjmed.2009.05.011 External link
137.
Kowalski TJ, Tata S, Berth W, Mathiason MA, Agger WA. Antibiotic treatment duration and long-term outcomes of patients with early lyme disease from a lyme disease-hyperendemic area. Clin Infect Dis. 2010 Feb;50(4):512-20. DOI: 10.1086/649920 External link
138.
Weber K. Treatment failure in erythema migrans--a review. Infection. 1996 Jan-Feb;24(1):73-5. DOI: 10.1007/BF01780663 External link
139.
Breier F, Khanakah G, Stanek G, Kunz G, Aberer E, Schmidt B, Tappeiner G. Isolation and polymerase chain reaction typing of Borrelia afzelii from a skin lesion in a seronegative patient with generalized ulcerating bullous lichen sclerosus et atrophicus. Br J Dermatol. 2001 Feb;144(2):387-92. DOI: 10.1046/j.1365-2133.2001.04034.x External link
140.
Häupl T, Hahn G, Rittig M, Krause A, Schoerner C, Schönherr U, Kalden JR, Burmester GR. Persistence of Borrelia burgdorferi in ligamentous tissue from a patient with chronic Lyme borreliosis. Arthritis Rheum. 1993 Nov;36(11):1621-6. DOI: 10.1002/art.1780361118 External link
141.
Priem S, Burmester GR, Kamradt T, Wolbart K, Rittig MG, Krause A. Detection of Borrelia burgdorferi by polymerase chain reaction in synovial membrane, but not in synovial fluid from patients with persisting Lyme arthritis after antibiotic therapy. Ann Rheum Dis. 1998 Feb;57(2):118-21. DOI: 10.1136/ard.57.2.118 External link
142.
Schmidli J, Hunziker T, Moesli P, Schaad UB. Cultivation of Borrelia burgdorferi from joint fluid three months after treatment of facial palsy due to Lyme borreliosis. J Infect Dis. 1988 Oct;158(4):905-6. DOI: 10.1093/infdis/158.4.905 External link
143.
Jares TM, Mathiason MA, Kowalski TJ. Functional outcomes in patients with Borrelia burgdorferi reinfection. Ticks Tick Borne Dis. 2014 Feb;5(1):58-62. DOI: 10.1016/j.ttbdis.2013.09.002 External link
144.
Nadelman RB, Hanincová K, Mukherjee P, Liveris D, Nowakowski J, McKenna D, Brisson D, Cooper D, Bittker S, Madison G, Holmgren D, Schwartz I, Wormser GP. Differentiation of reinfection from relapse in recurrent Lyme disease. N Engl J Med. 2012 Nov;367(20):1883-90. DOI: 10.1056/NEJMoa1114362 External link
145.
Hunfeld KP, Brade V. Antimicrobial susceptibility of Borrelia burgdorferi sensu lato: what we know, what we don't know, and what we need to know. Wien Klin Wochenschr. 2006 Nov;118(21-22):659-68. DOI: 10.1007/s00508-006-0693-z External link
146.
Hunfeld KP, Ruzic-Sabljic E, Norris DE, Kraiczy P, Strle F. In vitro susceptibility testing of Borrelia burgdorferi sensu lato isolates cultured from patients with erythema migrans before and after antimicrobial chemotherapy. Antimicrob Agents Chemother. 2005 Apr;49(4):1294-301. DOI: 10.1128/AAC.49.4.1294-1301.2005 External link
147.
Morgenstern K, Baljer G, Norris DE, Kraiczy P, Hanssen-Hübner C, Hunfeld KP. In vitro susceptibility of Borrelia spielmanii to antimicrobial agents commonly used for treatment of Lyme disease. Antimicrob Agents Chemother. 2009 Mar;53(3):1281-4. DOI: 10.1128/AAC.01247-08 External link
148.
Ruzic-Sabljic E, Podreka T, Maraspin V, Strle F. Susceptibility of Borrelia afzelii strains to antimicrobial agents. Int J Antimicrob Agents. 2005 Jun;25(6):474-8. DOI: 10.1016/j.ijantimicag.2005.02.007 External link
149.
Fallon BA, Keilp JG, Corbera KM, Petkova E, Britton CB, Dwyer E, Slavov I, Cheng J, Dobkin J, Nelson DR, Sackeim HA. A randomized, placebo-controlled trial of repeated IV antibiotic therapy for Lyme encephalopathy. Neurology. 2008 Mar;70(13):992-1003. DOI: 10.1212/01.WNL.0000284604.61160.2d External link
150.
Feder HM Jr, Johnson BJ, O'Connell S, Shapiro ED, Steere AC, Wormser GP; Ad Hoc International Lyme Disease GroupAgger WA, Artsob H, Auwaerter P, Dumler JS, Bakken JS, Bockenstedt LK, Green J, Dattwyler RJ, Munoz J, Nadelman RB, Schwartz I, Draper T, McSweegan E, Halperin JJ, Klempner MS, Krause PJ, Mead P, Morshed M, Porwancher R, Radolf JD, Smith RP Jr, Sood S, Weinstein A, Wong SJ, Zemel L. A critical appraisal of "chronic Lyme disease". N Engl J Med. 2007 Oct;357(14):1422-30. DOI: 10.1056/NEJMra072023 External link
151.
Kaplan RF, Trevino RP, Johnson GM, Levy L, Dornbush R, Hu LT, Evans J, Weinstein A, Schmid CH, Klempner MS. Cognitive function in post-treatment Lyme disease: do additional antibiotics help? Neurology. 2003 Jun;60(12):1916-22. DOI: 10.1212/01.WNL.0000068030.26992.25 External link
152.
Klempner MS, Baker PJ, Shapiro ED, Marques A, Dattwyler RJ, Halperin JJ, Wormser GP. Treatment trials for post-Lyme disease symptoms revisited. Am J Med. 2013 Aug;126(8):665-9. DOI: 10.1016/j.amjmed.2013.02.014 External link
153.
Klempner MS, Hu LT, Evans J, Schmid CH, Johnson GM, Trevino RP, Norton D, Levy L, Wall D, McCall J, Kosinski M, Weinstein A. Two controlled trials of antibiotic treatment in patients with persistent symptoms and a history of Lyme disease. N Engl J Med. 2001 Jul;345(2):85-92. DOI: 10.1056/NEJM200107123450202 External link
154.
Krupp LB, Hyman LG, Grimson R, Coyle PK, Melville P, Ahnn S, Dattwyler R, Chandler B. Study and treatment of post Lyme disease (STOP-LD): a randomized double masked clinical trial. Neurology. 2003 Jun;60(12):1923-30. DOI: 10.1212/01.WNL.0000071227.23769.9E External link
155.
Berende A, ter Hofstede HJ, Donders AR, van Middendorp H, Kessels RP, Adang EM, Vos FJ, Evers AW, Kullberg BJ. Persistent Lyme Empiric Antibiotic Study Europe (PLEASE)--design of a randomized controlled trial of prolonged antibiotic treatment in patients with persistent symptoms attributed to Lyme borreliosis. BMC Infect Dis. 2014 Oct;14:543. DOI: 10.1186/s12879-014-0543-y External link
156.
Berende A, ter Hofstede HJ, Vos FJ, van Middendorp H, Vogelaar ML, Tromp M, van den Hoogen FH, Donders AR, Evers AW, Kullberg BJ. Randomized Trial of Longer-Term Therapy for Symptoms Attributed to Lyme Disease. N Engl J Med. 2016 Mar;374(13):1209-20. DOI: 10.1056/NEJMoa1505425 External link
157.
Lakos A, Solymosi N. Maternal Lyme borreliosis and pregnancy outcome. Int J Infect Dis. 2010 Jun;14(6):e494-8. DOI: 10.1016/j.ijid.2009.07.019 External link
158.
Maraspin V, Ružic-Sabljic E, Pleterski-Rigler D, Strle F. Pregnant women with erythema migrans and isolation of borreliae from blood: course and outcome after treatment with ceftriaxone. Diagn Microbiol Infect Dis. 2011 Dec;71(4):446-8. DOI: 10.1016/j.diagmicrobio.2011.07.017 External link
159.
Buonomo A, Nucera E, Pecora V, Rizzi A, Aruanno A, Pascolini L, Ricci AG, Colagiovanni A, Schiavino D. Cross-reactivity and tolerability of cephalosporins in patients with cell-mediated allergy to penicillins. J Investig Allergol Clin Immunol. 2014;24(5):331-7.
160.
Wormser GP, Dattwyler RJ, Shapiro ED, Halperin JJ, Steere AC, Klempner MS, Krause PJ, Bakken JS, Strle F, Stanek G, Bockenstedt L, Fish D, Dumler JS, Nadelman RB. 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 Nov;43(9):1089-134. DOI: 10.1086/508667 External link
161.
Chandra A, Wormser GP, Klempner MS, Trevino RP, Crow MK, Latov N, Alaedini A. Anti-neural antibody reactivity in patients with a history of Lyme borreliosis and persistent symptoms. Brain Behav Immun. 2010 Aug;24(6):1018-24. DOI: 10.1016/j.bbi.2010.03.002 External link
162.
Embers ME, Barthold SW, Borda JT, Bowers L, Doyle L, Hodzic E, Jacobs MB, Hasenkampf NR, Martin DS, Narasimhan S, Phillippi-Falkenstein KM, Purcell JE, Ratterree MS, Philipp MT. Persistence of Borrelia burgdorferi in rhesus macaques following antibiotic treatment of disseminated infection. PLoS ONE. 2012;7(1):e29914. DOI: 10.1371/journal.pone.0029914 External link
163.
Puéchal X, Sibilia J. What should be done in case of persistent symptoms after adequate antibiotic treatment for Lyme disease? Curr Probl Dermatol. 2009;37:191-9. DOI: 10.1159/000213077 External link
164.
Faulde MK, Rutenfranz M, Keth A, Hepke J, Rogge M, Görner A. Pilot study assessing the effectiveness of factory-treated, long-lasting permethrin-impregnated clothing for the prevention of tick bites during occupational tick exposure in highly infested military training areas, Germany. Parasitol Res. 2015 Feb;114(2):671-8. DOI: 10.1007/s00436-014-4232-y External link
165.
Lupi E, Hatz C, Schlagenhauf P. The efficacy of repellents against Aedes, Anopheles, Culex and Ixodes spp. - a literature review. Travel Med Infect Dis. 2013 Nov-Dec;11(6):374-411. DOI: 10.1016/j.tmaid.2013.10.005 External link
166.
Leenders AC. Single-dose doxycycline for the prevention of Lyme disease. N Engl J Med. 2001 Nov;345(18):1349; author reply 1349-50.
167.
Nadelman RB, Nowakowski J, Fish D, Falco RC, Freeman K, McKenna D, Welch P, Marcus R, Agüero-Rosenfeld ME, Dennis DT, Wormser GP; Tick Bite Study Group. Prophylaxis with single-dose doxycycline for the prevention of Lyme disease after an Ixodes scapularis tick bite. N Engl J Med. 2001 Jul;345(2):79-84. DOI: 10.1056/NEJM200107123450201 External link
168.
Wilske B. Epidemiology and diagnosis of Lyme borreliosis. Ann Med. 2005;37(8):568-79. DOI: 10.1080/07853890500431934 External link
169.
Knauer J, Krupka I, Fueldner C, Lehmann J, Straubinger RK. Evaluation of the preventive capacities of a topically applied azithromycin formulation against Lyme borreliosis in a murine model. J Antimicrob Chemother. 2011 Dec;66(12):2814-22. DOI: 10.1093/jac/dkr371 External link
170.
Piesman J, Hojgaard A, Ullmann AJ, Dolan MC. Efficacy of an experimental azithromycin cream for prophylaxis of tick-transmitted lyme disease spirochete infection in a murine model. Antimicrob Agents Chemother. 2014;58(1):348-51. DOI: 10.1128/AAC.01932-13 External link
171.
Steere AC, Sikand VK, Meurice F, Parenti DL, Fikrig E, Schoen RT, Nowakowski J, Schmid CH, Laukamp S, Buscarino C, Krause DS. Vaccination against Lyme disease with recombinant Borrelia burgdorferi outer-surface lipoprotein A with adjuvant. Lyme Disease Vaccine Study Group. N Engl J Med. 1998 Jul;339(4):209-15. DOI: 10.1056/NEJM199807233390401 External link
172.
Wallich R, Kramer MD, Simon MM. The recombinant outer surface protein A (lipOspA) of Borrelia burgdorferi: a Lyme disease vaccine. Infection. 1996 Sep-Oct;24(5):396-7. DOI: 10.1007/BF01716093 External link
173.
Abbott A. Lyme disease: uphill struggle. Nature. 2006 Feb;439(7076):524-5. DOI: 10.1038/439524a External link
174.
Kalish RS, Wood JA, Golde W, Bernard R, Davis LE, Grimson RC, Coyle PK, Luft BJ. Human T lymphocyte response to Borrelia burgdorferi infection: no correlation between human leukocyte function antigen type 1 peptide response and clinical status. J Infect Dis. 2003 Jan;187(1):102-8. DOI: 10.1086/346059 External link
175.
Nigrovic LE, Thompson KM. The Lyme vaccine: a cautionary tale. Epidemiol Infect. 2007 Jan;135(1):1-8. DOI: 10.1017/S0950268806007096 External link
176.
Barrett PN, Portsmouth D. A novel multivalent OspA vaccine against Lyme borreliosis shows promise in Phase I/II studies. Expert Rev Vaccines. 2013 Sep;12(9):973-5. DOI: 10.1586/14760584.2013.824704 External link
177.
Brouqui P, Bacellar F, Baranton G, Birtles RJ, Bjoërsdorff A, Blanco JR, Caruso G, Cinco M, Fournier PE, Francavilla E, Jensenius M, Kazar J, Laferl H, Lakos A, Lotric Furlan S, Maurin M, Oteo JA, Parola P, Perez-Eid C, Peter O, Postic D, Raoult D, Tellez A, Tselentis Y, Wilske B; ESCMID Study Group on Coxiella, Anaplasma, Rickettsia and Bartonella; European Network for Surveillance of Tick-Borne Diseases. Guidelines for the diagnosis of tick-borne bacterial diseases in Europe. Clin Microbiol Infect. 2004 Dec;10(12):1108-32. DOI: 10.1111/j.1469-0691.2004.01019.x External link
178.
Canadian Public Health Laboratory Network. The laboratory diagnosis of Lyme borreliosis: Guidelines from the Canadian Public Health Laboratory Network. Can J Infect Dis Med Microbiol. 2007 Mar;18(2):145-8. DOI: 10.1155/2007/495108 External link
179.
Evison J, Aebi C, Francioli P, Péter O, Bassetti S, Gervaix A, Zimmerli S, Weber R. Borréliose de Lyme 2e partie: clinique et traitement [Lyme disease Part 2: clinic and treatment]. Rev Med Suisse. 2006 Apr;2(60):925-8, 930-4.
180.
Evison J, Aebi C, Francioli P, Péter O, Bassetti S, Gervaix A, Zimmerli S, Weber R. Borréliose de Lyme 3e partie: prévention, grossesse, états d'immunodéficience, syndrome post-borréliose de Lyme [Lyme disease Part 3: prevention, pregnancy, immunodeficient state, post-Lyme disease syndrome]. Rev Med Suisse. 2006 Apr;2(60):935-6, 938-40.
181.
Flisiak R, Pancewicz S; Polish Society of Epidemiology and Infectious Diseases. Diagnostyka i leczenie boreliozy z Lyme. Rekomendacje Polskiego Towarzystwa Epidemiologów i Lekarzy Chorób Zakaznych [Diagnostics and treatment of Lyme borreliosis. Recommendations of Polish Society of Epidemiology and Infectious Diseases]. Przegl Epidemiol. 2008;62(1):193-9.
182.
Ljøstad U, Mygland A. Lyme-borreliose hos voksne [Lyme borreliosis in adults]. Tidsskr Nor Laegeforen. 2008 May;128(10):1175-8.
183.
Mygland A, Ljøstad U, Fingerle V, Rupprecht T, Schmutzhard E, Steiner I; European Federation of Neurological Societies. EFNS guidelines on the diagnosis and management of European Lyme neuroborreliosis. Eur J Neurol. 2010 Jan;17(1):8-16, e1-4. DOI: 10.1111/j.1468-1331.2009.02862.x External link
184.
Société de pathologie infectieuse de langue française. 16e Conférence de consensus en thérapeutique anti-infectiese de la Spilf. Borréliose de Lyme: démarches diagnostiques, thérapeutiques et préventives. Texte long [Lyme borreliose: diagnostic, therapeutic and preventive approaches--long text]. Med Mal Infect. 2007 Dec;37 Suppl 3:S153-74.
185.
Cameron DJ, Johnson LB, Maloney EL. Evidence assessments and guideline recommendations in Lyme disease: the clinical management of known tick bites, erythema migrans rashes and persistent disease. Expert Rev Anti Infect Ther. 2014 Sep;12(9):1103-35. DOI: 10.1586/14787210.2014.940900 External link