Journal of Clinical & Experimental Dermatology Research

Journal of Clinical & Experimental Dermatology Research
Open Access

ISSN: 2155-9554

+44 1478 350008

Commentary - (2016) Volume 7, Issue 2

Lyme Disease: Beyond Erythema Migrans

Herbert B. Allen*, Harina Vin, Catherine Warner and Suresh Joshi
Department of Dermatology, Drexel University College of Medicine, Philadelphia, PA, USA
*Corresponding Author: Herbert B. Allen, Department of Dermatology, Drexel University College of Medicine, 219 North Broad Street, 4th Floor, Philadelphia, PA 19107, USA, Tel: 215-762-5550 Email:

Abstract

Background: With the recent discoveries of Borrelia burgdorferi and other spirochetes in the brains of Alzheimer’s patients and with a recent analysis showing the very same pathology in both syphilitic and Alzheimer’s dementia it seems both rational and urgent to consider all aspects of Lyme disease in a new light, especially the concept of “overdiagnosis”. The very presence of the organisms in the brains following supposedly effective treatment for Lyme disease is contradictory and should be the starting point for diagnosis and treatment. Also for consideration is the reliance on erythema migrans and serologies in the diagnosis of Lyme disease inasmuch as they occur in less than half the patients.

<

Keywords: Lyme disease; Borrelia burgdorferi ; Erythema migrans; Spirochetes; Arthritis; Neuroborreliosis

Introduction

Lyme disease is a tick-born infection causing major public health problems worldwide. Ixodes ticks, especially in their nymphal stage, are the transmitting agent of Lyme disease. A feeding tick, whose host is the white-tailed deer, must be attached for 24-48 hours before transmitting disease. Risk of infection after bite is 1%-3% [1-6]. In addition to their disease transmission, these ticks are thought to inject immunosuppressive, anticoagulant, and anti-inflammatory saliva into their hosts while feeding, allowing injected pathogens an opportunity to evade the immune system [7].

There are about 30,000 confirmed cases per year in the United States, but the Centers for Disease Control and Prevention recently estimates that the true number of cases may be 300,000 given underreporting of the disease. This number may also be incorrect; a recent analysis reveals the “true” number may be 1 million.8 nationwide surveys. In Maryland, an endemic region, the annual economic impact of chronic illness associated with Lyme disease if $16,199 per patient [9]. It causes multi-organ infection with dermatological, rheumatological, neurological, and cardiac manifestation.

Clinical manifestation

Borrelia burgdorferi is the primary etiologic agent of Lyme disease, and is a spirochete like T. pallidum . It is 10-30 microns in length (twice the size of the syphilis spirochete) and irregularly coiled [10] B. afzelii , and B. garinii are other less common causative species [11] that occur in Europe and are thought to be associated with Borrelia lymphocytoma and acrodermatitis chronica atrophicans, which are late cutaneous manifestations of Lyme disease [12]. Infection occurs in three stages, although infected patients may be asymptomatic. The gold standard of clinical diagnosis in early infection is the erythema (chronicum) migrans (EM) rash, which occurs in 80% of patients [13]. However, the true incidence of EM is unknown: it ranges from 27% to 40% to the 80% which is the CDC number [14,15]. One of the reasons for this discrepancy is the multiple clinical presentations of EM: these include the classic “bull’s eye”, multiple lesions with dusky centers, red oval shaped plaque (cellulitis-like), an expanding rash with central clearing, a dusky blue-purple rash with no central clearing, and a redblue lesion with central clearing. The “bull’s eye”, the one signature lesion of Lyme disease, is only one of many of the dermatologic findings [16]. The less familiar lesions may lead to marked underreporting and under-recognition of this disease.

The EM “bulls-eye” lesion typically begins as a pink papulethat expands centrifugally over days to become a plaque, classically with central clearing. B. burgdorferi organisms are found both in the center and periphery of erythema migrans rash. The largest diameter must be at least 5 cm. They are usually solitary but multiple EM-like lesions occur in up to 17% of patients. The rash may be accompanied by localized pain, localized pruritus, or lymphadenopathy but may also be asymptomatic. Stage 2 is early disseminated infection involving the nervous system, joints, and/or heart [17], and occurs within weeks of the rash. Weeks to months later, if left untreated, late disseminated infection occurs including persistent arthritis and acrodermatitis [13]. Involvement of the nervous system is termed neuroborreliosis [18] and is seen in up to 40% of infected individuals [18,19].

Diagnosis

Per the Centers for Disease Control (CDC), confirmed cases may be [1] EM with a known tick exposure, [2] EM with laboratory evidence of infection and without a known tick exposure, or [3] at least one late manifestation of disease with laboratory evidence of infection. The CDC recommends serologic testing for Lyme disease follow a 2-test approach. The first step is an enzyme-linked immunosorbent assay (ELISA). If the initial ELISA is positive or equivocal, antibodies are tested, with positive result showing at least 2 of 5 and 5 of 10 bands for IgM and IgG, respectively. If symptoms have persisted <28 days, positive IgM or IgG are sufficient; if symptoms have been present >28 days, positive IgG is required [20]. In late or persistent infection, detection of antibodies to B. burgdorferi is highly specific and sensitive. Unfortunately, sensitivity of this testing in patients with early LD is only 29%-40% [21].

There are a growing number of cases of Lyme disease that do not meet the criteria set by the CDC which include erythema migrans, lymphocytic meningitis, Bell’s palsy, arthritis, and heart block. This definition does not cover encephalopathy, polyneuropathy [22], neuropsychiatric disease, and chronic manifestations. Additionally, neither the ELISA testing nor the antibody testing is particularly sensitive. In a study of 46 patients with culture-proven erythema migrans, only 33% had a positive ELISA result and 43% had a positive IgM immunoblot. During follow-up, around 90% of the patients had IgG antibodies but using the IgG immunoblot criteria of the CDC showed a positive IgG in only 22% of patients [23].

The diagnostic gold standard is the isolation of Borrelia by culture with subsequent quantitative PCR (qPCR)-based confirmation. Borrelia can be cultured in 49% of skin biopsies of erythema migrans. Moreover, this method is not routinely used particularly since results are not available for 2-6 weeks, which is problematic because delay of treatment increases likelihood of long-term sequela of Lyme disease. Therefore, even when positive, this test is not useful for clinical decision-making.

Borrelia burgdorferi organisms can be detected in 80% of skin biopsy specimens by Qpcr [24]. Anther study also confirmed that skin biopsy qPCR is positive (78%) in more samples than are culture positive (55%) [25]. In CSF testing for Lyme neuroborreliosis, qPCR for Borrelial DNA was detected in 15% of CSF samples [26]. In synovial fluid testing for Lyme arthritis, Borrelial DNA was detected in 85% of synovial fluid samples [27].

The C6 peptide reproduces the sequence of invariable region 6 in the variable surface antigen VlsE (Vmp-like sequence, expressed) of B. burgdorferi . Detection of IgM and IgG antibodies of C6 peptides using ELISA has a sensitivity of >70% for detecting Lyme disease. Pomelova et al showed that of their 146 Lyme disease patients, 51% had seroconversion of IgM antibodies, and 88% had seroconversion of IgG antibodies [28]. Liange et al showed that in acute infected patients, including early localized and early disemminated infection) sensitivity of the C6 ELISA was 74%, In late phase disease, sensitivity was 100%. In both cases, specificity was 99% [29]. Importantly, C6 peptide ELISA has no cross-reactivity with anti- recombinant outer surface protein A (OspA) antibodies, which can be seen in vaccinated patients [30].

Evasion of immune system

Mechanisms of B. burgdorferi immune evasion, dormancy, and activation in hosts are best studied in the neurologic manifestations of B. burgdorferi . There are three recognized forms of neuropsychiatric disease in Lyme disease. The meningovascular form is associated with cerebrovascular infarcts [31,32]. The atrophic form is associated with cortical atrophy and gliosis, [31] and the central nervous form with neuropsychiatric symptoms [33]. It has been postulated that the pathophysiology of chronic Lyme disease involves a failure of the adaptive immune system leading to persistent infection by B. burgdorferi . Suggested mechanism include deception of alternative complement pathway by surface antigens, dampening the plasminogen activation system, or continuously varying surface antigens [34]. In patient with neurologic manifestations of Lyme disease proinflammatory cytokines IL6 and TNF alpha are increased in the cerebrospinal fluid [35] and IL6 levels have been shown to correlate to disease activity in neuroborreliosis [36]. Additionally, the outer membrane of the Borrelial cell well expresses pro-inflammatory lipoproteins from the spirochetes [37]. These lipoproteins are up to 500-fold more active inducers of cytokines than lipoproteins of other organisms including Escherichia coli [38] and can disseminate from the periphery to the brain [39].

Treatment

When a patient presents with ring-shaped erythema that is consistent with erythema migrans and exposure in an endemic region, treatment is initiated. In cases with the rash is not classically consistent with EM, a non-endemic region warrants observation whereas an endemic region merits two-step ELISA [40].

Antibiotic efficacy against B. burdorferi has been studied in vitro and in animal models. The generally accepted treatment for erythema migrans Lyme disease is a 14 day course of doxycycline. Prophylaxis with a single dose of 200 mg doxycycline after a tick bite was 87% effective in reducing erythema migrans in a randomized control trial [41]. However, caution must be used when providing prophylaxis as it could lead to a seronegative state that can delay diagnosis [42]. Amoxicillin is an alternative often used in children and pregnant women.

In beagle dogs infected by Ixodes ticks with confirmed positive skin cultures, spirochete levels were undetectable after treatment with either doxycycline or amoxicillin. However, skin biopsies taken from all antibiotic-treated dogs were PCR-positive for 23srRNA and ospA at one or more timepoints, indicating persistence of spirochetal DNA following antibiotic treatment [43]. In mice, low levels of spirochete DNA persisted in ceftriaxone-treated mice, although these mine did not have positive skin cultures or other evidence of disease [44]. In rhesus monkies treated for chronic Lyme disease with 28 days of oral doxycycline, Borrelia antigen, DNA, and RNA were persist after 90 days. Xenodiagnosis also recovered small numbers of intact spirochetes in treated monkeys [45].

This persistence of spirochetal DNA may be clinically significant. 34% of patients treated with a course of antibiotics had long-term sequelae of Lyme disease 6 years after treatment [46]. Asch et al conducted a retrospective cohort study of 215 patients diagnosed with Lyme disease based on the CDC criteria and followed the patients for 3.2 years. At follow-up, relapse with major organ involvement was seen in 28% of patient. 53% of the patient had “persistent symptoms of arthralgia, arthritis, cardiac, or neurologic involvement.” Notably, however, antibiotic treatment initiated within 4 weeks of disease onset was more likely to result in complete recovery [47].

Persistent infection despite treatment is recognized as posttreatment Lyme disease syndrome (PTLDS). It is characterized by “fatigue, musculoskeletal pain, and cognitive complaints that persist for 6 months or longer after completion of antibiotic therapy” [48]. The primary identified risk factor is delayed diagnosis. These patients tend to show a low level of persistent anti-C6 titers compared to untreated patients who showed higher titers. The C6 antibodies occur on a region of the B. burgdorferi VlsE lipoprotein and are common to all infectious variants. Decline in the C6 antibody titer as measured by ELISA may serve as a predictor of treatment outcome [49]. There is evidence that Borrelia burgdorferi is capable of forming biofilm in vitro as it secretes extracellular polymeric substance which likely also contributes to its persistence despite treatment [50]. Further, there is evidence of spirochetal biofilm formation in the brains of Alzheimer’s disease [51].

Impact

Per the CDC, Lyme disease is likely much more common than reported. They estimate over 300,000 new cases each year in the United States alone. Insurance coverage of Lyme disease requires strict criteria. Both clinical diagnosis and fulfillment of CDC two-step diagnostic criteria is required. This is problematic because of the 40% sensitivity of the test. Many insurance companies specifically deny coverage of the more sensitive test - C6 peptide ELISA - due to “inadequate scientific evidence” [52]. Even when CDC criteria are met, antibiotics are covered for only 28 days, which has been shown to be too short a course to prevent long term sequela in about 28-34% of patients [46,47]. Furthermore because no randomized control trials have been performed for antibiotic use in post Lyme disease syndrome, additional antibiotic therapy in PTLDS patients is considered “experimental and investigational” [52]. Where the pathology of syphilis and Alzheimer’s disease has been shown to be similar and where Borrelia spirochetes have been shown to be present in the brains of Alzheimer’s disease, it seems no coincidence that patients with untreated syphilis have a 35% incidence of tertiary findings (including dementia) [53] and patients treated with doxycycline for Lyme disease have a 35% chance of tertiary disease. It seems as if treatment with doxycycline is like no treatment at all. Stricker and Johnson called for a “Manhattan Project” to combat the Lyme disease epidemic which causes untold suffering and which is many times more prevalent than HIV/AIDS [54]. Nothing less should be advocated in this scourge which represents failure on every level.

References

  1. Miklossy J (2011) Alzheimer’s disease - a neurospirochetosis. Analysis of the evidence following Koch's and Hill's criteria. J Neuroinflammation 8: 90.
  2. Riviere GR, Riviere KH, Smith KS (2002) Molecular and immunological evidence of oral Treponema in the human brain and their association with Alzheimer’s disease. Oral MicrobiolImmunol 17: 113-118.
  3. Macdonald AB (2006) Plaques of Alzheimer’s disease originate from cysts of Borreliaburgdorferi, the Lyme disease spirochete. Medical Hypotheses 67: 592-600.
  4. Miklossy J (2015) Historic evidence to support a causal relationship between spirochetal infections and Alzheimer’s disease. Front Aging Neurosci 7: 1-12.
  5. Steere AC, Taylor E, McHugh GL, Logigian EL (1993) The overdiagnosis of Lyme disease. JAMA 269: 1812-1816.
  6. Sood SK, Salzman MB, Johnson BJ, Happ CM, Feig K, et al. (1997) Duration of Tick Attachment as a Predictor of the Risk of Lyme Disease in an Area in which Lyme Disease Is Endemic. J Infect Dis 175: 996-999.
  7. Francischetti IM, Sa-Nunes A, Mans BJ, Santos IM, Ribeiro JM (2009) The role of saliva in tick feeding. Front Biosci 14: 2051–2088.
  8. Hook S, Nelson C, Mead P (2013) Self-reported Lyme disease diagnosis, treatment, and recovery: Results from 2009, 2011, & 2012 HealthStyles nationwide surveys. Presented at the 13th International Conference on Lyme Borreliosis and other Tick-Borne Diseases, Boston, MA, August .
  9. Zhang X, Meltzer MI, Pena CA, et al. Economic impact of Lyme disease. Emer Infect Dis 2006; 12: 653-660.
  10. Dombrowski C, Kan W, Motaleb A, et al.The elastic basis for the shape of Borreliaburgdorferi. Biophys J 2009; 96 (11): 4409-4417.
  11. Dombrowski C, Kan W, Motaleb MA, Charon NW, Goldstein RE, et al. Delineation of BorreliaburgdorferiSensuStricto, Borreliagarinii sp. nov., and Group VS461 Associated with Lyme Borreliosis. IntJ SystBacteriol 1992; 42: 378-383.
  12. Bingham PM, Galetta SL, Athreya B, Sladky J (1995) Neurologic manifestations in children with Lyme disease. Pediatrics 96: 1053-1056
  13. Johnson L, Wilcox S, Mankoff J, Stricker RB (2014) Severity of chronic Lyme disease compared to other chronic conditions: a quality of life survey. PeerJ 2: e322
  14. Allen HB (2010) Lyme disease. In Dermatology Terminology, London: Springer-Verlag : 55.
  15. Meier C, Grehl H (1988) Vasculitic neuropathy in the Garin-Bujadoux-Bannwarth syndrome. A contribution to the understanding of the pathology and pathogenesis of the neurological complications in Lyme borreliosis. Dtsch Med Wochenschr 113: 135–138.
  16. Veenendaal-Hilbers JA1, Perquin WV, Hoogland PH, Doornbos L (1988) Basal meningovasculitis and occlusion of the basilar artery in two cases of Borreliaburgdorferi infection. Neurology 38: 1317–1319.
  17. Fallon BA, Nields JA (1994) Lyme disease: a neuropsychiatric illness. Am. J. Psychiatry 151: 1571–1583.
  18. Centers for Disease Control and Prevention (CDC) (1995) Recommendations for test performance and interpretation from the Second National Conference on Serologic Diagnosis of Lyme Disease. MMWR Morb. Mortal Wkly Rep 44: 590–591.
  19. Aguero-Rosenfeld ME, Wang G, Schwartz I, Wormser GP (2005) Diagnosis of Lyme borreliosis. ClinMicrobiol Rev 18: 484-509.
  20. Logigian EL, Kaplan RF,Steere AC (1990) Chronic Neurologic Manifestations of Lyme Disease.N Engl J Med 323: 1438-1444.
  21. Aguero-Rosenfeld ME, Nowakowski J, Bittker S, Cooper D, Nadelman RB, et al. (1996) Evolution of the serologic response to Borreliaburgdorferi in treated patients with culture-confirmed erythema migrans. J ClinMicrobiol34: 1-9.
  22. Liveris D, Wang G, Giroa G, et al. (2002) Quantitative detection of Borreliaburgdorferi in 2-millimeter skin samples of erythema migrans lesions: correlation of results with clinical and laboratory findings. J ClinMicrobiol40: 1249-53
  23. Liveris D, Wang G, Girao G, Byrne DW, Nowakowski J et al. (2013) Quantitative Detection of Borreliaburgdorferisensulato in Erythema Migrans Skin Lesions Using Internally Controlled Duplex Real Time PCR. PLoS ONE8: e63968.
  24. Cerar T, Ogrinc K, Cimperman J (2008) Validation of Cultivation and PCR Methods for Diagnosis of Lyme Neuroborreliosis. J ClinMicrobiol 46: 3375–3379.
  25. James J Nocton, Frank Dressler, Barbara J Rutledge, Paul N Rys, et al. (1994) Detection of Borreliaburgdorferi DNA by polymerase chain reaction in synovial fluid from patients with Lyme arthritis. N Engl JMed 1994 330:229–234.
  26. Pomelova VG, Korenberg EI, Kuznetsova TI, Bychenkova TA, Bekman NI, et al. (2015) C6 Peptide-Based Multiplex Phosphorescence Analysis (PHOSPHAN) for Serologic Confirmation of Lyme Borreliosis. PLOS ONE 10:e0130048.
  27. Liang FT, Steere AC, Marques AR, Johnson BJ, Miller JN, et al. (1999) Sensitive and specific serodiagnosis of Lyme disease by enzyme-linked immunosorbent assay with a peptide based on an immunodominant conserved region of BorreliaburgdorferivlsE. J ClinMicrobio 37: 3990-3996.
  28. Marques AR, Martin DS, Philipp MT (2002) Evaluation of the C6 Peptide Enzyme-Linked Immunosorbent Assay for Individuals Vaccinated with the Recombinant OspA Vaccine. J. Clin. Microbiol 40: 2591-2593.
  29. Miklossy J (2012) Chronic or Late Lyme Neuroborreliosis: Analysis of Evidence Compared to Chronic or Late Neurosyphilis. Open Neurol J6: 146–157.
  30. Back T, Grünig S, Winter Y, Bodechtel U, Guthke K, et al. (2013) Neuroborreliosis-associated cerebral vasculitis: long-term outcome and health-related quality of life. J Neurol 260: 1569–1575.
  31. BransfieldRC (2012) The psychoimmunology of lyme/tick-borne diseases and its association with neuropsychiatric symptoms. Open Neurol J 6: 88-93
  32. Berndtson K (2013)Review of evidence for immune evasion and persistent infection in Lyme disease. Int J Gen Med 6: 291–306.
  33. Widhe M, Grusell M, Ekerfelt C (2002) Cytokines in Lyme borreliosis: lack of early tumour necrosis factor-alpha and transforming growth factor-beta1 responses are associated with chronic neuroborreliosis. Immunology107: 46-55.
  34. Weller M, Stevens A, Sommer N, Wiethölter H, Dichgans J (1991) Cerebrospinal fluid interleukins, immunoglobulins, and fibronectin in neuroborreliosis. Arch Neurol 48: 837-841.
  35. Fraser CM, Casjens S, Huang WM, Sutton GG, Clayton R, et al. (1997) Genomic sequence of a Lyme disease spirochaete, Borreliaburgdorferi. Nature 390: 580-586.
  36. Weis JJ, Ma Y, Erdile LF (1994)Biological activities of native and recombinant Borreliaburgdorferi outer surface protein A: dependence on lipid modification. Infect Immun 62: 4632-4636.
  37. Londoño D,Cadavid D (2010) Bacterial lipoproteins can disseminate from the periphery to inflame the brain. Am J Pathol 176: 2848-2857.
  38. Nadelman RB, Horowitz HW, Hsieh TC, Wu JM, Aguero-Rosenfeld ME, et al. (1997) Simultaneous human granulocytic ehrlichiosis and Lyme borreliosis. N Engl J Med 337: 27–30.
  39. Cameron DJ, Johnson, LB, Maloney EL (2014) 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 Ther12: 1103-1135.
  40. R K Straubinger, B A Summers, Y F Chang, M J Appel J (1997) Persistence of Borreliaburgdorferi in experimentally infected dogs after antibiotic treatment. J ClinMicrobiol 1997 35: 111-116.
  41. Bockenstedt LK, Mao J, Hodzic E, Barthold SW, Fish D (2002) Detection of attenuated, noninfectious spirochetes in Borreliaburgdorferi-infected mice after antibiotic treatment. J Infect Dis 186: 1430-1437.
  42. Monica E Embers , Stephen W Barthold, Juan T Borda, Lisa Bowers, Lara Doyle, et al (2012) Persistence of Borreliaburgdorferi in rhesus macaques following antibiotic treatment of disseminated infection. PloS One2012; 7: e29914.
  43. Shadick NA, Phillips CB, Logigian EL, Steere AC, Kaplan RF et al. (1994)The long-term clinical outcomes of Lyme disease. A population-based retrospective cohort study. Ann Intern Med. 1994; 121: 560-567.
  44. Asch ES, Bujak D, Weiss M, Peterson MG, Weinstein A et al. (1994) Lyme disease: an infectious and postinfectious syndrome.J Rheumatol 21: 454-461.
  45. Aucott JN (2015) Posttreatment Lyme disease syndrome. Infect Dis Clin NorthAm 29: 309-323.
  46. Fleming RV, Marques AR,Klempner MS (2004)Pre-treatment and post-treatment assessment of the C(6) test in patients with persistent symptoms and a history of Lyme borreliosis. Eur J ClinMicrobiol Infect Dis 23: 615-618.
  47. Eva Sapi , Scott L Bastian, Cedric M Mpoy, Shernea Scott , et al. (2012) Characterization of Biofilm Formation by Borreliaburgdorferi In Vitro. PLoS ONE7: e48277.
  48. Hanaway M, Katsetos C, Cusack CA, Allen HB (2015) Tertiary treponematosis. J Am AcadDermatol 72: AB 130 suppl1.
  49. Gjestland T (1955)The Oslo study of untreated syphilis. ActaDermVenereol (Stockh)1955: 35.
  50. Stricker RB, Johnson L (2014) Lyme Disease: Call for a ‘‘Manhattan Project’’ to Combat the Epidemic. PLoSPathog 10(1): e1003796.
Citation: Allen HB, Vin H, Warner C, Joshi S (2016) Lyme Disease: Beyond Erythema Migrans. J Clin Exp Dermatol Res 7:330.

Copyright: © 2016 Allen HB, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top
https://www.olimpbase.org/1937/