Tick-Borne Encephalitis Vaccine: Recommendations of the Advisory Committee on Immunization Practices, United States, 2023

Susan L. Hills, MBBS1; Katherine A. Poehling, MD2; Wilbur H. Chen, MD3; J. Erin Staples, MD, PhD1 (View author affiliations)

View suggested citation
Article Metrics
Altmetric:
Citations:
Views:

Views equals page views plus PDF downloads

Related Materials

 

Summary

Tick-borne encephalitis (TBE) virus is focally endemic in parts of Europe and Asia. The virus is primarily transmitted to humans by the bites of infected: Ixodes species ticks but can also be acquired less frequently by alimentary transmission. Other rare modes of transmission include through breastfeeding, blood transfusion, solid organ transplantation, and slaughtering of viremic animals. TBE virus can cause acute neurologic disease, which usually results in hospitalization, often permanent neurologic or cognitive sequelae, and sometimes death. TBE virus infection is a risk for certain travelers and for laboratory workers who work with the virus. In August 2021, the Food and Drug Administration approved Ticovac TBE vaccine for use among persons aged ≥1 year. This report summarizes the epidemiology of and risks for infection with TBE virus, provides information on the immunogenicity and safety of TBE vaccine, and summarizes the recommendations of the Advisory Committee on Immunization Practices (ACIP) for use of TBE vaccine among U.S. travelers and laboratory workers.

The risk for TBE for most U.S. travelers to areas where the disease is endemic is very low. The risk for exposure to infected ticks is highest for persons who are in areas where TBE is endemic during the main TBE virus transmission season of April–November and who are planning to engage in recreational activities in woodland habitats or who might be occupationally exposed. All persons who travel to areas where TBE is endemic should be advised to take precautions to avoid tick bites and to avoid the consumption of unpasteurized dairy products because alimentary transmission of TBE virus can occur. TBE vaccine can further reduce infection risk and might be indicated for certain persons who are at higher risk for TBE. The key factors in the risk-benefit assessment for vaccination are likelihood of exposure to ticks based on activities and itinerary (e.g., location, rurality, season, and duration of travel or residence). Other risk-benefit considerations should include 1) the rare occurrence of TBE but its potentially high morbidity and mortality, 2) the higher risk for severe disease among certain persons (e.g., older persons aged ≥60 years), 3) the availability of an effective vaccine, 4) the possibility but low probability of serious adverse events after vaccination, 5) the likelihood of future travel to areas where TBE is endemic, and 6) personal perception and tolerance of risk.

ACIP recommends TBE vaccine for U.S. persons who are moving or traveling to an area where the disease is endemic and will have extensive exposure to ticks based on their planned outdoor activities and itinerary. Extensive exposure can be considered based on the duration of travel and frequency of exposure and might include shorter-term (e.g., <1 month) travelers with daily or frequent exposure or longer-term travelers with regular (e.g., a few times a month) exposure to environments that might harbor infected ticks. In addition, TBE vaccine may be considered for persons who might engage in outdoor activities in areas where ticks are likely to be found, with a decision to vaccinate made on the basis of an assessment of their planned activities and itinerary, risk factors for a poor medical outcome, and personal perception and tolerance of risk. In the laboratory setting, ACIP recommends TBE vaccine for laboratory workers with a potential for exposure to TBE virus.

Introduction

Tick-borne encephalitis (TBE) virus is a tickborne flavivirus that is focally endemic in a geographic region extending from western and northern Europe through to northern and eastern Asia (1). The most recognized clinical manifestation of TBE virus infection is acute neurologic disease, which usually results in hospitalization, often permanent neurologic or cognitive sequelae, and sometimes death. TBE is rare among travelers and laboratory workers.

In August 2021, the Food and Drug Administration (FDA) approved an inactivated TBE vaccine (Ticovac) for use in persons aged ≥1 year. The current adult formulation of TBE vaccine has been used since 2001 in Europe; however, before 2021, no TBE vaccine was licensed in the United States, and the Advisory Committee on Immunization Practices (ACIP) had no recommendations for use of TBE vaccine (2,3). This report provides information on TBE and TBE vaccine and describes the new ACIP recommendations for use of TBE vaccine among U.S. travelers and laboratory workers. Health care providers can use these guidelines to discuss the risks and benefits of TBE vaccination during pretravel consultations with persons traveling abroad to areas with TBE virus transmission risk and in occupational health consultations with laboratory workers with potential for exposure to TBE virus. The TBE vaccine recommendations will be reviewed and updated as needed if new data become available or if additional TBE vaccines are licensed in the United States.

Background

TBE Virus and Its Subtypes

TBE virus is a single-stranded RNA virus in the genus Flavivirus, family Flaviviridae (4). TBE virus is closely related to Powassan virus, a tickborne flavivirus transmitted in parts of the United States (5). The three main antigenic subtypes of TBE virus (i.e., European, Siberian, and Far Eastern) differ in the severity of disease they cause and geographic distribution (6). The principal geographic distribution of the European subtype virus is in parts of western and northern Europe through to the eastern European countries; the Siberian subtype virus is in Siberia and the Ural and European parts of Russia; and the Far Eastern subtype virus is in Japan, China, Mongolia, and the eastern parts of Russia; however, subtype virus distributions overlap substantially (69). Genomic studies have indicated two additional minor subtype viruses (i.e., Baikalian and Himalayan) (10,11).

Modes of TBE Virus Transmission

TBE virus is primarily transmitted to humans by the bites of infected Ixodes sp. ticks but can also be acquired less frequently by alimentary transmission. Other rare modes of transmission include through breastfeeding, blood transfusion, solid organ transplantation, and slaughtering of viremic animals. Nymphs and adult ticks are believed to be responsible for causing most human infections. Approximately 60%–70% of persons with TBE recall a bite (1219). Because TBE virus is present in the saliva of an infected tick, transmission likely occurs early during feeding (1,20).

Transmission by Ticks

Ixodes ricinus is the main vector for the European subtype TBE virus and Ixodes persulcatus for the Siberian and Far Eastern subtype viruses (21). I. ricinus is found in most of continental Europe and the United Kingdom and I. persulcatus in an area extending east from northeastern Europe through to China and Japan (22,23). The distributions of the two species overlap in certain countries, including Estonia, Finland, Latvia, and the European part of Russia (7,21,2327).

The preferred habitats for the vector ticks are woodland environments. The main habitats are deciduous forests for I. ricinus and coniferous forests for I. persulcatus (23,28). Ticks can be found either within the forest or on forest edges, where the forest transitions to grasslands, meadows, or marshlands, and they favor areas with low-growing dense brush and plant litter (23,2932). Recreational activities with increased risk for exposure to ticks include hiking, camping, cycling in woodland areas, hunting, fishing, birdwatching, and collecting mushrooms or berries (3336). Persons in certain occupations (e.g., farmers, forestry workers, military personnel, and researchers undertaking field work in rural areas) also might be at higher risk for exposure to infected ticks (17,37,38). Humans must enter a tick habitat to be at risk for infection because ticks do not, unaided, disperse widely (39,40). TBE virus infections acquired in urban areas (e.g., city parks) are occasionally reported; however, risk in urban areas is considered to be low (38,41,42).

The enzootic transmission cycle of TBE virus involves ticks and vertebrate hosts. Ticks are both virus vectors and reservoirs. A tick can become infected when feeding on a viremic host or through nonviremic transmission when co-feeding in close proximity to an infected tick (4345). After becoming infected, ticks remain infected through their various life stages and can transmit the virus sexually to other ticks and transovarially to their offspring (28,46,47). The main amplifying reservoir hosts are small mammals, particularly rodents (e.g., mice and voles). Larger forest animals (e.g., boar and deer) and domestic animals (e.g., cattle, dogs, goats, and sheep) do not have an important role in the maintenance of the virus in nature. However, deer and cattle have an important role in maintaining tick populations (22,23,40,48). Humans are incidental, dead-end hosts in the transmission cycle because they do not develop a level or duration of viremia sufficient to infect ticks or have sufficient numbers of attached ticks at one time to allow co-feeding (22,4951).

Other Modes of Transmission

Alimentary transmission is a less frequent means of acquisition of TBE virus and occurs after ingestion of unpasteurized dairy products (e.g., milk and cheese) from infected cattle, goats, or sheep; transmission from goats is most commonly reported (5260). Large outbreaks linked to infected dairy products have been reported from areas where TBE is endemic, including one with approximately 600 cases (58,61,62). Approximately 47 laboratory-acquired TBE virus infections have occurred globally (6365). TBE virus transmission from infected breastfeeding women to their infants has been described in at least two published reports; one infant remained healthy and the other had severe sequelae (30,66). Other rare modes of transmission include blood transfusion, solid organ transplantation, and slaughtering of viremic animals (6769).

Epidemiology of TBE

Geographic Distribution and Spread

TBE virus is focally endemic in a geographic region extending from western and northern Europe through to northern and eastern Asia (https://www.cdc.gov/tick-borne-encephalitis/geographic-distribution/index.html). Although the geographic range of TBE virus is restricted by the presence of the tick vectors, areas of TBE virus transmission are more limited and focal than the tick distribution. TBE virus-infected ticks typically are found in discrete areas (i.e., foci) confined by the presence of environmental conditions that allow maintenance of the natural transmission cycle rather than being distributed evenly across a region (13,70,71). Natural foci can be small, with locations <1 square mile (72). Multiple factors are required for maintenance of virus circulation (e.g., favorable microclimatic conditions, interactions of ticks and vertebrate hosts, and local vegetation), which likely contribute to the focal occurrence (73). Within affected areas, tick population density and TBE virus infection rates can be highly variable. Infection rates in ticks typically range from 0.1% to 5%, although rates of approximately 40% in I. persulcatus ticks have been reported (21,7477).

During recent decades, TBE virus has emerged in new geographical foci in countries where the disease is endemic, and the overall area of recognized transmission has expanded westward and northward (1,26,7889). Since 2016, three countries have reported their first autochthonous cases (Belgium, England, and the Netherlands) (9092). New TBE foci also have been detected at higher altitudes, reaching elevations up to 2,100 meters (6,890 feet) above sea level (52,81,9395). Concurrently, in certain countries, a reduction in virus transmission and possible loss of recognized geographical foci have been documented (81,93). Various factors might be contributing to the changing distribution, including changes in climatic and ecologic conditions altering tick habitats and transmission cycles and dispersal of ticks into new areas by birds, deer, or other animals (22,40,79,96103).

Incidence

In areas where TBE is endemic, approximately 5,00010,000 new cases are reported annually (9,104). However, this figure likely represents an underestimate of the actual number of cases because of underdiagnosis, underreporting, or both in certain countries (13,105108).

Incidence rates differ from country to country and depend on the local ecology and geographic distribution of the virus within the country. However, national incidence rates are not directly comparable because of variable approaches to surveillance, the extent of human and laboratory resources applied to surveillance, and the population vaccination coverage (109). Higher incidence rates are most commonly reported from the Baltic states (Estonia, Latvia, and Lithuania), Slovenia, and the Czech Republic (110).

Annual variability in countries’ incidence rates is typical (86,109,110). The reasons for this variability are not completely understood but reflect the complex interactions among factors that affect risk for infection, including tick density, presence of animal hosts, ecologic conditions, weather, and human behavior (35,76,109112). Longer-term fluctuations in TBE incidence also occur (81). Incidence of reported cases has increased in multiple countries in recent decades while remaining stable or decreasing in others (27,111118). In addition to the factors affecting annual variability in the short term, socioeconomic factors (e.g., political instability and poverty) can affect disease incidence over the longer term (79,93,119123). Other factors that can lead to observed increases include improved awareness of TBE, increased access to laboratory diagnostics, and better surveillance (1,13,89,109,110,124). Of note, TBE became a reportable disease in the European Union in 2012 (125). Reduced incidence in certain countries is related to increased vaccine uptake over time; for example, in Austria, a vaccination program resulted in TBE incidence decreasing approximately sixfold from 5.7 cases per 100,000 population during 1972–1981 to 0.9 during 2002–2011 (126).

Demographic and Seasonal Patterns

TBE can occur in persons of all ages, with encephalitis reported in one infant as young as 17 days (127). Incidence is typically low in children and increases with age, generally peaking in the 60–69 years age group and then decreasing in the ≥70 years age group (17,86,110,111,115,128130). TBE is more common in males, with reported incidence rates often 1.5–2 times higher than in females, likely reflecting a greater risk for tick exposure (14,35,37,67,110112,115,131,132).

The main TBE virus transmission season is April–November when ticks are most active because of warmer weather in the Northern Hemisphere (12,110,112,133,134). Peak transmission generally occurs for multiple weeks during the warm, humid summer months, typically during JuneAugust in European countries. However, in central and northern Europe, two peaks might occur in summer and early fall (12,15,16,23,37,111,112,115,135137). Unlike mosquitoborne diseases, large outbreaks of tickborne diseases do not occur. Occasional cases are reported during winter because tick activity is still possible at temperatures close to freezing (23,28,109,110,138).

Clinical Manifestations and Diagnosis

Symptoms and Signs

Approximately three fourths of TBE virus infections are asymptomatic (67,139,140). Among patients who develop clinical symptoms after a bite from an infected tick, the incubation period is typically 7–14 days (range = 2–28 days) (17,90,141). For TBE acquired through the alimentary route, the typical incubation period is shorter, usually <2 weeks and often 2–4 days (53,57,60,142).

The most recognized clinical presentation of TBE is central nervous system infection (i.e., aseptic meningitis, meningoencephalitis, or meningoencephalomyelitis) (Box 1). Overall, meningitis is reported in approximately 35%–45% of patients, meningoencephalitis in 45%–55%, and meningoencephalomyelitis in 10% (14,17,18,37,137). However, younger patients (i.e., aged approximately ≤15 years) more frequently have meningitis, and the percentage of patients with more severe clinical presentations usually increases with age (12,14,17,18,131,133,143148). Relatively mild forms of disease (e.g., undifferentiated febrile illness) can occur (36,149). A chronic form of disease has been reported from Russia linked to infection with the Siberian subtype virus and, rarely, the Far Eastern subtype virus and is possibly associated with long-term viral persistence (18,36,150153). Progressive, slow development of neurologic symptoms often occurs, with or without an initial acute illness. In certain patients, the incubation period can be prolonged and symptoms can first manifest many years after a tick bite. A chronic relapsing form of disease also has been reported in Russia (152,153). Immunity after TBE virus infection is considered to be lifelong (49).

TBE can have a monophasic or biphasic illness course (i.e., an isolated neurologic illness alone or neurologic disease after an initial nonspecific illness). The monophasic disease course is the most common in infections caused by the Far Eastern and Siberian subtype viruses. The biphasic course is the most frequent in patients infected with the European subtype virus; approximately 65%–75% of patients infected with the European subtype virus have a biphasic illness course (14,17,18,36,133,148,151,154). When biphasic illness occurs, the initial phase includes nonspecific symptoms (e.g., fever, headache, malaise, myalgia, nausea, and vomiting). These symptoms usually last for a median of approximately 4 days (range = 1–10 days), followed by a period of remission of approximately 7 days (range = 1–33 days), followed by the second (neurologic) phase (12,17,18,133,155).

Neurologic signs and symptoms of TBE vary but can include meningeal signs, altered mental status, cognitive dysfunction (e.g., decreased concentration and memory impairment), ataxia, rigidity, tremors, and cranial nerve and limb paresis or palsies. Limb involvement is more typically unilateral than bilateral, and the upper extremities are more often affected than the lower extremities (14). Seizures are not common with TBE caused by the European subtype virus (17,153,156,157).

Increasing age is a key risk factor for more severe disease. Other risk factors include infection with the Far Eastern subtype virus and being immunocompromised, and certain studies have found a correlation with the monophasic illness course (16,18,151,158163).

Among the limited number of published case reports of women infected during pregnancy, the clinical spectrum of illness appears similar to that of the nonpregnant population (164168). Apart from two reports from the 1960s in which the diagnostic methods used to confirm maternal infection were unclear, all infants born to infected mothers were reported to be healthy at birth and transplacental transmission of TBE virus had not been confirmed.

Clinical Laboratory Findings

Clinical laboratory findings with TBE are nonspecific. In the initial phase of a biphasic illness, findings can include leukopenia, thrombocytopenia, or elevated hepatic enzymes (145,149,154). In the neurologic phase of disease, findings can include a peripheral leukocytosis, an elevated erythrocyte sedimentation rate, and increased C-reactive protein levels (12,17,37,133,146). Cerebrospinal fluid (CSF) testing usually indicates a pleocytosis, typically lymphocytic, with moderately elevated protein levels (12,17,37,131,133). However, early in disease, neutrophils can predominate in CSF.

Neuroimaging and Electroencephalogram Investigations

Magnetic resonance imaging (MRI) occasionally detects abnormalities in the brain or spinal cord; however, sensitivity is low for diagnosis of TBE (169). In one prospective study in Germany, 18% (18 of 102) of patients with MRI results had abnormal findings, and in a retrospective study from Austria, 9% (four of 45) of patients with MRI results had abnormalities considered TBE related (17,170). Changes, when present, are most commonly observed in the thalamus, often bilaterally, and less often in the cerebellum, basal ganglia, brainstem, or other locations (17,147,171174). In patients with myelitis, radiculitis, or both, either alone or in association with encephalitis, spinal MRI can indicate changes such as T2-hyperintensities in the anterior horns of the cervical cord (171,175179). Computerized tomography scans do not usually identify any abnormalities (169). Abnormal electroencephalogram findings are common and can include diffuse slowing and focal abnormalities (17,132,133,147,173).

Laboratory Diagnosis

The laboratory diagnosis of TBE usually is based on detection of virus-specific immunoglobulin M (IgM) antibody in CSF or serum (180). An IgM enzyme-linked immunosorbent assay is routinely used for testing samples and usually is positive when neurologic symptoms are present. However, cross-reactivity with other flavivirus antibodies can occur because TBE virus shares common antigenic sites within its E protein with multiple other flaviviruses (49). Plaque reduction neutralization tests can be performed to discriminate between cross-reacting antibodies attributable to another primary flavivirus infection or to confirm recent TBE virus infection on the basis of a fourfold or higher increase in virus-specific neutralizing antibodies between acute- and convalescent-phase serum specimens. However, in patients who have been infected previously by another flavivirus or vaccinated with a different flavivirus vaccine (e.g., Japanese encephalitis or yellow fever vaccine), cross-reactive antibodies can make identifying a specific etiologic agent difficult (180). Vaccination history, date of symptom onset, and information about other flaviviruses known to circulate in the geographic area that might cross-react in serologic assays should be considered when interpreting results. In addition, possible antibody persistence from a previous TBE virus infection should be considered; serum IgM antibodies typically are detectable for approximately 3–4 months after infection but can persist for ≥3 years (12,181,182).

TBE virus occasionally has been isolated, or TBE viral RNA has been detected by nucleic acid amplification tests (NAATs), in serum, whole blood, urine, or CSF samples when a patient has neurologic illness (50,51,67,92,127,168,183187). Although these methods are insufficiently sensitive for routine diagnostic purposes, NAATs can be of value in patients who are immunocompromised (158,188,189). In addition, if testing is done during the initial febrile (viremic) phase of illness before neurologic symptoms develop and antibodies are measurable, RNA often can be detected; however, patients usually only are tested after neurologic disease manifests (49,169,184). In fatal encephalitis cases, TBE virus RNA has been detected in brain tissue (68,184).

No commercially available tests for TBE virus infection are available in the United States. Diagnostic testing can be performed at CDC. Clinicians should contact their state or local health department or the Arboviral Diseases Branch, Division of Vector-Borne Diseases (970-221-6400) for assistance with diagnostic testing.

Treatment and Management

No specific antiviral treatment for TBE is available. Patient management consists of supportive care, treatment of symptoms, and interventions to prevent secondary complications (e.g., aspiration pneumonia or urinary tract infection) (153,169). Patients with meningoencephalitis should be closely observed because coma or neuromuscular paralysis leading to respiratory failure can develop rapidly (36).

An anti-TBE virus intravenous immunoglobulin (IVIG) preparation was previously used in Europe for postexposure prophylaxis or treatment. However, no effectiveness data from controlled clinical trials are available, and IVIG use was discontinued after reports of suspected antibody-dependent enhancement of infection resulting in a more severe course of disease (190193). In Russia and Kazakhstan, specific anti-TBE virus IVIG preparations continue to be used; information on their effectiveness is published primarily in the non-English literature (153).

Outcome and Sequelae

The outcome of TBE largely depends on the patient’s age, clinical form of the disease, and virus subtype (194). Among patients with neurologic disease and infected with the European subtype virus, the case fatality rate is usually <2% (14,17,18,37,86,110,111,114,146,195,196). Fatality rates from infection with the Siberian subtype virus are higher but rarely exceed 6%–8% (151). Rates of 20%–40% were historically described with the Far Eastern subtype virus, although the extent of study methodology and patient inclusion criteria as contributing factors is unclear (62,151,197). In China, where the Far Eastern subtype virus is found, case-fatality rates were >25% in the 1950s; however, rates of <10% have been reported since the 1980s, purportedly related to improved disease awareness and quality of medical care (151,198). In Russia, where the Far Eastern and Siberian subtype viruses predominate, TBE mortality rates of approximately 2% have recently been reported (153).

Studies to assess frequency of sequelae have used variable symptom definitions, types of cohorts, investigation approaches, and durations of follow-up after illness to measure outcomes, making interpretation and comparison of studies difficult. A limitation of multiple studies is incomplete follow-up among the persons in the cohort, potentially biasing results. Among patients infected with the European subtype virus, sequelae have been reported in 20%–40% overall, including neurologic sequelae (e.g., limb paresis or paralysis) in up to 10% (17,18,155,194,196). Sequelae have been reported with higher frequency after infection with the Far Eastern and Siberian subtype viruses, but the reported differences might be a result of methodologic differences in published reports.

The severity of reported sequelae ranges from mild symptoms with limited to no effect on quality of life to severe sequelae that interfere with activities of daily living. Reported serious outcomes of infection include permanent limb or cranial nerve palsies or paralysis, ataxia, and dysphasia (155,194). Milder symptoms include cognitive impairment (e.g., difficulties with memory or concentration), headaches, fatigue, tremors, hearing loss, emotional lability, or minor problems with balance or coordination (17,18,37,196). In a case-control study in Sweden with 92 patients and 58 controls with follow-up conducted from 2 to 15 years (median = 5.5 years) after TBE virus infection, patients scored significantly lower than controls in the domains of memory and learning, executive function (i.e., initiative and motivation), vigilance (i.e., concentration, attention, and fatigue), and physical impairment (i.e., fine motor skills, coordination, and balance) (199).

Certain symptoms can improve or resolve during the weeks to months after hospitalization (200). In one study, the median time to recovery was 13 weeks (range = 2–156 weeks) among the patients who recovered completely or had only unrelated ongoing health issues (63%; 72 of 114) (196). However, patients occasionally have worsening of sequelae over time (18,173).

Severe outcomes are more frequent with increasing age and might be of particular concern for persons aged approximately ≥60 years (201). Older age has been correlated with a longer duration of hospitalization, lengthier time to recovery, higher case-fatality rate, and increased risk for sequelae (17,86,111,131,194,196). The association between older age and poor outcomes is likely related to immunosenescence with increasing age (202). Among children, deaths are rare and neurologic sequelae occur at low rates (<3%) (12,14,129,133,148,203). However, permanent, severe neurologic deficits can occur and subtle deficits (e.g., cognitive problems, headache, fatigue, or irritability) might be common (19,127,173,204).

TBE Among Travelers

TBE is rare among U.S. travelers to areas where the disease is endemic. Twelve TBE cases have been reported among U.S. adult and pediatric civilian (i.e., nonmilitary) travelers, including one case in 1979 and 11 cases during 2001–2021 (180,205,206) (Table 1). During 2001–2021, the mean was <1 reported case (range = 0–2 cases) annually. However, TBE cases might not have been identified if the illness was diagnosed overseas or if a clinician did not consider TBE in the differential diagnosis for a returning traveler with a compatible illness. On the basis of approximately 20–25 million U.S. citizen trips to countries with TBE risk each year, and a mean of <1 diagnosed TBE case each year, the overall incidence of TBE among U.S. civilian travelers is low (207). However, certain persons who travel abroad will be at increased risk for infection because of location and season of travel, their activities, and other factors (Box 2).

Among the 12 TBE cases diagnosed in U.S. civilian travelers during 1979–2021, a total of 10 (83%) occurred in males, the median age was 38 years (range = 4–79 years), and infections were acquired in Europe, Russia, or China. Travel, and thus exposure to TBE virus, for all patients occurred during May–August. Among 11 travelers for whom information was available on duration of travel in areas where TBE is endemic, the median travel duration was 18 days (range = 7–69 days). All eight travelers with available data reported activities with risk for tick exposure, including hiking, camping, fishing, and trail running. Clinical illness occurred in a biphasic manner in eight (67%) patients. Eight (67%) patients had meningoencephalitis and four (33%) had meningitis, and no deaths occurred. Seven (58%) patients recovered completely, two (17%) had mild cognitive sequelae, one (8%) recovered but information on possible sequelae was unavailable, one (8%) had severe neurologic sequelae including dysarthria and mild limb bradykinesia, and one (8%) was discharged from acute care to a rehabilitation facility but their clinical outcome was unknown.

In addition to the 12 cases among civilian travelers, 12 TBE cases were diagnosed among U.S. military personnel (n = 8) or their dependent children (n = 4) during 2012–2021 (208210) (Table 2). One case occurred in 2012, and the remaining 11 occurred during 2017–2021. At the time of infection, all persons were living in Germany; nine persons had specific information available and all were living in Baden-Württemberg or Bavaria, Germany’s two states with the highest number of reported annual TBE cases (86). On the basis of a mean of 1.2 cases per year among approximately 50,000 U.S. military personnel and dependents living in Germany, the TBE risk was similar to that of the local population of Baden-Württemberg and Bavaria where annual TBE incidence during 2012–2018 ranged from 0.7 to 2.0 cases per 100,000 population (86). Among the 12 TBE cases, 10 (83%) were in males, the median age was 33 years (range = 2–47 years), illness onsets occurred during April–November, and nine (75%) had neurologic illness. Five (42%) patients recovered, including two who had short-term sequelae before complete recovery; four (33%) had no outcome information reported; and three (25%) experienced moderate sequelae.

In Europe, a median of 36 traveler cases (range = 25–65 cases) wase reported to the European Centre for Disease Prevention and Control each year during 2014–2020 among the approximately 2,000–3,800 TBE cases reported annually (211). However, because TBE is endemic in multiple areas of Europe and TBE vaccines are available, the number of traveler cases prevented by vaccination is unknown (212). Most cases occurred among persons who reported undertaking activities with risk for tick exposure, with only rare case reports of travelers with TBE acquired through ingestion of unpasteurized dairy products (142,213216). Local population TBE incidence data for multiple countries where the disease is endemic in Europe are published annually by the European Centre for Disease Prevention and Control; however, infection risk for a traveler cannot be inferred from these data because the data might be influenced by surveillance methods, reporting practices, and vaccination coverage (104).

TBE Among Laboratory Workers

At least 47 laboratory-acquired TBE virus infections have been reported globally, and approximately all occurred before 1980 (6365,67). Among these 47 infections, 37 (79%) resulted in disease, and the remainder were asymptomatic infections. At least four of the infections occurred among U.S. laboratory workers; three cases were overt disease with two deaths reported, one was an asymptomatic infection, and all occurred before 1980. None of the infected laboratory workers was known to have received TBE vaccine. Limited information was available on transmission routes; however, all 10 cases with information reported were attributed to aerosolization during laboratory procedures or handling of infected animal waste. Transmission through accidental percutaneous or mucosal exposures is possible. Work with TBE virus typically is restricted to biosafety level (BSL)-4 facilities and practices (217).

Methods

The ACIP TBE Vaccine Work Group was formed in September 2020 to 1) review information on the epidemiology, clinical presentation, diagnosis, treatment, and outcome of TBE; 2) review data on disease risk and burden for travelers and laboratory workers; 3) review data on TBE vaccine safety, immunogenicity, and effectiveness; and 4) draft evidence-based vaccination recommendations for ACIP’s review. The work group included two ACIP members, one ex officio member from each of the National Institutes of Health and the FDA, liaison representatives from the American Academy of Pediatrics and the International Society of Travel Medicine, and technical advisors. The expertise represented by these members included infectious diseases, pediatrics, travel medicine, public health, arbovirology, entomology, vaccinology, and vaccine policy. The work group met via conference call 27 times during September 2020–March 2022 with participation from CDC consultants and staff members from various CDC divisions.

The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach was used to review and evaluate available data (218). Details on the methods used, including the policy question developed, ranking of outcomes, systematic review protocol, inclusion criteria, evidence summary, and certainty assessment are provided in GRADE for TBE vaccine (219). In developing the TBE vaccine recommendations, the work group also assessed additional factors as outlined in the ACIP Evidence to Recommendations (EtR) framework, including the public health importance of TBE, population values, stakeholder acceptability, resource use considerations, health equity issues, and feasibility of implementation (220). Detailed information is available in the ACIP EtR framework for TBE vaccination for persons who travel abroad and ACIP EtR framework for TBE vaccination for laboratory workers (221,222).

Work group members or the manufacturer gave presentations on vaccine immunogenicity, safety, and other topics related to the development of the TBE vaccine recommendations at ACIP meetings during October 2020–January 2022. The work group presented the TBE vaccine EtR for persons who travel abroad and for laboratory workers and preliminary vaccination recommendations to ACIP during its January 12, 2022, meeting. ACIP voting members unanimously approved the TBE vaccine recommendations for persons who travel abroad and for laboratory workers at the February 23–24, 2022, ACIP meeting (223).

Summary of Findings on TBE Vaccine

Manufacture and Licensure

On August 13, 2021, FDA approved a TBE vaccine manufactured by Pfizer, Inc. for use in persons aged ≥1 year (224). The vaccine is licensed in the United States under the trade name Ticovac; in Europe, it is also marketed as FSME-Immun (3). The vaccine is an inactivated, whole virus vaccine, prepared using a European subtype TBE virus. The vaccine has an adult (0.5 mL) formulation for use in persons aged ≥16 years and a pediatric (0.25 mL) formulation for use in children aged 1–15 years. In both age groups, the schedule includes a 3-dose primary series; a booster can be administered ≥3 years after completion of the primary series if ongoing exposure or re-exposure to TBE virus is expected. Pfizer’s TBE vaccine is the only U.S.-licensed TBE vaccine; other TBE vaccines are manufactured in China, Europe, and Russia but are not available in the United States (153).

An earlier formulation of Pfizer’s TBE vaccine was first licensed in Austria in 1976 (2). Changes to the vaccine formulation over time have included removal of thimerosal and a transition of the production virus seed from mouse brain suspension to chick embryo fibroblast cells. The current adult and pediatric formulations became available in Europe in 2001 and 2003, respectively (3).

Vaccine Protection for TBE

Measurement of Protection

No randomized controlled trials (RCTs) to demonstrate the efficacy of TBE vaccine for prevention of clinical disease have been conducted; the low incidence of TBE in areas where the disease is endemic would make such trials infeasible. Vaccine effectiveness (VE) studies for TBE vaccine using a different schedule, with a previous vaccine formulation or with VE assessed in combination with another TBE vaccine or both, have been published (126,130,225230); however, no studies assessing VE of the TBE vaccine alone in its current formulation and according to the U.S. licensed schedule exist. Clinical trials have been conducted using immunogenicity endpoints. TBE virus neutralizing antibodies are believed to confer protection against disease on the basis of animal and human studies (231,232). A neutralizing antibody titer of ≥10 is considered a seropositive result and is generally used to indicate protection. However, a correlate of protection has not been formally established, and no standardized reference reagents are available (232).

Level of Cross-Protection for TBE Virus Subtypes and Related Viruses from the TBE Vaccine

In addition to the genetic and antigenic similarity of the three TBE subtype viruses, data from human and animal studies suggest that cross-protection for heterologous subtype viruses from the European subtype-based TBE vaccine is likely. However, data are limited, and VE has not been demonstrated.

In one comparative sequence analysis, phylogenetic relations were determined for 25 TBE virus strains from the three genetic lineages (6). Variability among strains in the envelope protein, the main target for neutralizing antibodies at the amino acid level, was ≤2% within and 4%–6% among subtype viruses. In older human studies using the previous vaccine formulation, neutralizing antibody titers were similar against various European, Far Eastern, and Siberian TBE virus strains; in animal studies, generally good protection was demonstrated for vaccinated mice challenged with strains from the three TBE virus subtypes (233235). In a more recent study, 41 adults were vaccinated with the current formulation of the TBE vaccine, and the ability of postvaccination sera to neutralize representative European (n = 2), Far Eastern (n = 2), and Siberian (n = 1) subtype TBE pseudoviruses (created using a West Nile virus backbone and encoding the membrane and envelope proteins of the various TBE virus strains) was investigated. The sera successfully neutralized all the pseudoviruses, with no statistically significant differences between mean neutralizing antibody titers (236). In a mouse study, no statistically significant differences were found in mean neutralizing antibody titers against European and Far Eastern TBE pseudoviruses (237). In addition, the mice were protected against challenge with both strains in a dose-dependent manner, and the 50% protective dose values were not significantly different. In another mouse study, postvaccination neutralizing antibody titers measured using Far Eastern (n = 1), European (n = 1), and Siberian (n = 1) TBE virus strains were similar (238). Ten mice vaccinated with various dilutions of the TBE vaccine also underwent viral challenge with a recent Siberian subtype virus isolated in 2010. Although the percentage of lethal outcomes varied, the minimum vaccine dose protecting 50% of the mice was considered to be within acceptable limits.

Data are limited on potential cross-protection of TBE vaccine for Powassan virus infection. One study demonstrated limited neutralization of Powassan virus by sera from persons vaccinated with a European subtype virus TBE vaccine (239). A study of mice vaccinated with a Far Eastern subtype virus TBE vaccine demonstrated minimal to no neutralization of Powassan virus by the mice sera, and the mice were not protected against a lethal Powassan virus challenge (240). These data suggest minimal to no cross-protection against Powassan virus afforded by TBE vaccination.

Vaccine Effectiveness

VE studies have been conducted in four countries in Europe (Austria, Germany, Latvia, and Switzerland) where both the U.S.-licensed vaccine and another non–U.S.-licensed European subtype vaccine are in use (126,130,225230). The European subtype is the only TBE virus circulating in three of these countries; in Latvia, all three virus subtypes circulate with seeming predominance of the European subtype virus (241,242). No studies have investigated VE for the Far Eastern or Siberian subtype viruses. The studies all have one or more limitations, particularly when considering VE for the U.S.-licensed vaccine (e.g., inclusion of persons who received an older formulation of the vaccine, use of different dosing schedules to the U.S. schedule [e.g., >1 booster dose], unknown or potentially inaccurate estimates of market share for each vaccine, limited data by age group including from persons aged >50 years, or weaknesses in methods used to estimate vaccination coverage) (243).

Overall, VE estimates for the European subtype virus TBE vaccines against disease caused by the European subtype virus after ≥3 doses of vaccine are 91%–99% for prevention of various outcomes (e.g., all TBE disease and hospitalization) (225229). One study in Germany covering the period 2018–2020 used a case-control study design to estimate VE specifically for the U.S.-licensed TBE vaccine; VE was 93% (95% CI = 87%–97%) for prevention of neurologic or nonneurologic disease after ≥3 doses in adults and children; however, the percentage of persons who received only 3 primary doses and 1 booster dose (i.e., in accordance with the U.S.-licensed schedule) was unavailable (228). In a study that included data from southern Germany and Latvia, VE estimates for prevention of neurologic or nonneurologic TBE after 3 doses when the interval after the third dose was <3 years were 95% (95% CI = 93%–97%) and 99% (95% CI = 98%–100%), respectively (226). Estimates also were available by age group; in Germany and Latvia, VE estimates were 94% (95% CI = 83%–98%) and 98% (95% CI = 87%–100%) for persons aged ≤17 years, 97% (95% CI = 95%–98%) and 99% (95% CI = 97%–100%) for persons aged 18–59 years, and 88% (95% CI = 80%–93%) and 100% for persons aged ≥60 years, respectively. A study conducted in Austria demonstrated that VE to prevent hospitalization among patients aged ≥1 year after ≥1 dose was 95% (95% CI = 95%–96%) regardless of whether vaccinations were administered according to schedule (130). VE was slightly lower to prevent severe disease compared with mild disease, with estimates of 95% (95% CI = 94%–96%) and 97% (95% CI = 96%–98%), respectively. The lower VE was most evident for children aged 1–16 years, with estimates of 83% (95% CI = 70%–90%) for severe disease compared with 95% (95% CI = 91%–97%) for mild disease.

Immunologic Response After a 3-Dose Primary Series in Adults and Children

The pivotal adult immunogenicity study with the current formulation of the vaccine was conducted in Poland among adults aged 16–64 years (224,244). Among 416 adults seronegative at baseline, 411 (99%) were seropositive at 21–28 days after dose 3 of the primary series with a geometric mean titer (GMT) of 259 (95% CI = 235–285). In a follow-up study, the seropositivity rate at 2 years after dose 3 was 96% (242 of 252) and at 3 years after dose 3, immediately before the booster dose, was 94% (229 of 243) (245,246). Although the seropositivity rates at 2 and 3 years were high, GMTs had declined to 62 (95% CI = 53–71) at 2 years and 49 (95% CI = 42–56) at 3 years after completion of the primary series.

Key pediatric data came from a study conducted in Austria, Germany, and Poland among children aged 1–15 years (245,247). At 35–42 days after dose 3 of the primary series, 358 (99%) of 360 children were seropositive and GMT was 382 (95% CI = 351–415). Seropositivity rates were ≥99% in all age groups, but GMT was significantly lower among children aged 7–15 years (307 [95% CI = 272–347]) than among children aged 3–6 years (469 [95% CI = 399–552]) and 1–2 years (568 [95% CI = 527–612). In a follow-up study, seropositivity rates at 2 and 3 years after completion of the 3-dose primary series for all age groups combined were 98% at both time points (i.e., 352 of 358 children seropositive at year 2 and 346 of 353 at year 3; intent-to-treat data set) (245,248). When comparing GMTs by age group at these later time points, they were again lower in the group aged 7–15 years compared with younger age groups. GMTs for the children aged 7–15 years, 3–6 years, and 1–2 years at 2 years were 111 (95% CI = 95–129), 204 (95% CI = 165–252), and 154 (95% CI = 125–189), respectively, and at 3 years were 97 (95% CI = 83–113), 188 (95% CI = 150–236), and 166 (95% CI = 135–205), respectively. GMTs among children demonstrated a moderate decline between 1 month and 2 years after the primary series, then remained relatively stable between 2 and 3 years.

Immunologic Response After a 3-Dose Primary Series in Older Adults

Data are limited on the immunologic response measured by neutralizing antibody titer to the primary series of TBE vaccine among older adults, particularly those aged ≥65 years. A study conducted in Poland compared seropositivity rates among adults aged 16–49 years (n = 170) and 50–79 years (n = 170, including 31 adults aged ≥65 years) (3,245). The seropositivity rates at 21 days after dose 3 among younger and older age groups were 100% (144 of 144 [95% CI = 98%–100%]) and 99% (151 of 153 [95% CI = 95%–100%]), respectively. However, GMT among persons aged 16–49 years (208 [95% CI = 174–247]) was significantly higher than GMT among adults aged 50–79 years (104 [95% CI = 86–127]). A second study investigated the immune response in healthy, initially seronegative, older adults in Switzerland; all were aged ≥70 years but an age range was not provided (249). At 4 weeks after dose 3, the seropositivity rate was 99% (136 of 137) and GMT was 71 (249). In the pivotal adult immunogenicity study conducted in Poland, a post hoc analysis compared seropositivity rates and GMTs among younger persons (aged 18–50 years [n = 211]) and older persons (aged 51–67 years [n = 41, including 15 aged >60 years]) (245,246). At 21–28 days after dose 3 of the primary series, seropositivity rates were 100% in both groups but GMT was significantly higher for the younger age group (303 [95% CI = 269–341] versus 122 [95% CI = 87–173). At 3 years after dose 3, the seropositivity rate in the younger age group was significantly higher (97% [95% CI = 93%–99%]) than in the older age group (83% [95% CI = 68%–93%]), and GMT also was significantly higher (55 [95% CI = 47–64] versus 26 [95% CI = 18–36]).

Overall, seropositivity rates immediately after completion of the 3-dose primary series are high among older persons but levels of neutralizing antibodies are lower than among younger adults. Over time postvaccination, lower neutralizing antibody levels continue to be observed among older adults and they are more likely to become seronegative.

Immunologic Response After a Booster Dose in Adults and Children

The immunologic response to a booster dose administered 3 years after the 3-dose primary series was investigated in adults aged 18–67 years at the time of the booster dose (245,246). At 21–35 days after a booster dose, all 240 persons were seropositive. GMT increased ninefold from the prebooster titer of 49 (95% CI = 42–56) to the postbooster titer of 428 (95% CI = 394–464). In a follow-up study, regular blood draws were conducted to investigate the neutralizing antibody response for 10 years after the booster dose. The seropositivity rate at 5 years after a booster dose was 94% (209 of 222) and at 10 years was 85% (189 of 222). GMT decreased to 99 (95% CI = 87–112) at 2 years postbooster, then slowly declined to 32 (95% CI = 24–43) at the 10-year time point (250,251).

A pediatric booster dose study was conducted among children who had been enrolled in a vaccine immunogenicity study at ages 1–15 years; at the time of the booster study 3 years later most received the pediatric booster dose (0.25 mL) but approximately 21% were aged 16–18 years and received the adult dose (0.5 mL) for the booster (248). At 21–35 days after a booster, the seropositivity rate was 100% (172 of 172), and GMT increased sixfold from 59 (95% CI = 51–68) prebooster to 359 (95% CI = 316–409) postbooster. In a 10-year follow-up study, neutralizing antibody levels were measured among children who had received a booster dose 3, 4, or 5 years after the 3-dose primary series; approximately 85%, 14%, and <1% of the full cohort received the booster dose at these respective intervals (248). Seropositivity rates at 5 years and 10 years after a booster dose administered 3–5 years after the primary series were 99% (155 of 156) and 90% (140 of 155), respectively. GMTs demonstrated an initial moderate decrease from 381 approximately 1 month postbooster to 162 at 3 years postbooster, with a subsequent gradual decline to 54 at 10 years postbooster. Data on seropositivity rates and GMTs were not provided separately for the groups with boosters administered at 3, 4, or 5 years. At 10 years after the booster, the proportions seropositive by age group were 86% (25 of 29), 92% (23 of 25), 93% (57 of 61), and 88% (35 of 40) for children who had begun the primary series at age 1–2 years, 3–6 years, 7–11 years, and 12–15 years, respectively; none of these differences were statistically significant. GMTs at 10 years postbooster for these age group were 34, 80, 66, and 43, respectively; for each age group, the lower bound of the 95% CI for GMT was >10, and differences between GMTs were not statistically significant (Pfizer, Philadelphia, PA, unpublished data, 2021).

Immunologic Response After a Booster Dose in Older Adults

Data are limited on the immunologic response measured by neutralizing antibody titer to a booster dose among older adults. At 21–35 days after a booster dose administered 3 years after the 3-dose primary series to 41 adults aged 51–67 years, 100% were seropositive (245,246). GMT increased twelvefold from the prebooster titer of 26 (95% CI = 18–36) to the postbooster titer of 303 (95% CI = 228–403). GMTs in 199 adults aged 18–50 years were significantly higher prebooster (55 [95% CI = 47–64]) and postbooster (459 [95% CI = 424–497]).

No data are available for older adults on the longer-term immunologic response measured by neutralizing antibody titer after a booster dose of the current formulation of the TBE vaccine. Limited data are available from a booster dose study in which the primary series for approximately 75% of adults was a 3-dose series of the U.S.-licensed TBE vaccine and for 25% was 2 doses of another European TBE vaccine and 1 dose of the U.S.-licensed TBE vaccine; all persons received a booster dose with the U.S.-licensed TBE vaccine (250). Overall, 54 adults were aged 50–60 years and 10 were aged 61–70 years at the time of the booster dose. Seropositivity rates 5 years after a booster dose were 92% (47 of 51) for adults aged 50–60 years and 75% (six of eight) for adults aged 61–70 years; GMTs were 76 (95% CI = 53–110) and 24 (95% CI = 5–121), respectively. At 10 years, seropositivity rates were 75% (38 of 51) and 38% (three of eight), and GMTs were 26 (95% CI = 13–50) and 1 (95% CI = 0.1–20).

The limited sample sizes make interpretation of these data difficult. Results from additional studies on immunogenicity among older adults, although not directly relevant to the U.S-licensed TBE vaccine, have suggested a reduced immunologic response or steeper decline in neutralizing antibodies among older adults after a booster dose, likely related to immunosenescence (202,252254). However, in the absence of measurable neutralizing antibodies, vaccinated persons can usually still mount a rapid anamnestic response to infection, and T-cells also likely play a key part in the response. As a result, low neutralizing antibody levels might not necessarily indicate lack of protection (254,255).

Immunologic Response After an Incomplete Primary Series (1 or 2 Doses) in Adults and Children

Data on the short-term immunologic response after 1 dose of TBE vaccine were available from a study investigating kinetics of the immune response among adults when the first 2 vaccine doses were administered 10–14 days apart (3,245). In this study conducted in Poland, the seropositivity rate at approximately 12 days after dose 1 among persons aged 16–49 years was 52% (79 of 153) and among adults aged 50–79 years was 27% (43 of 159). GMTs for the younger and older groups were 11 (95% CI = 10–13) and eight (95% CI = 7–10), respectively.

Multiple studies among adults investigated the immunologic response after 2 doses of TBE vaccine. The seropositivity rates among healthy adults at approximately 3–4 weeks after dose 2 ranged from 83% to 100% in six studies (3,245,249,256259). Seropositivity rates were 72% and 87% at 5 months and 27% at 11 months on the basis of three studies that provided results at one of these time points (3,244,245,259). One of these studies reported a lower seropositivity rate among adults aged 50–79 years (65%) than adults aged 16–49 years (79%) (3,245). In studies among adults that measured GMTs immediately before and 3–4 weeks after dose 3 of the primary series, GMTs were close to 10 before dose 3 but administration of the final dose of the primary series resulted in a greater than tenfold increase in GMT (3,244,245,256).

Studies among pediatric populations also measured the immunologic response after 2 doses of TBE vaccine, and results were similar to those of studies among adults. Among healthy children and adolescents, seropositivity rates at 3–5 weeks after dose 2 ranged from 84% to 100% in three studies (245,247,260). Rates were 95% at 5 months and 38% at 9 months on the basis of two studies that reported results at one of these time points (245,261,262).

Estimates of VE after 2 doses of vaccine have been published, with similar limitations to those that assessed VE after ≥3 doses (i.e., no studies of VE for the Far Eastern or Siberian subtype viruses or for the U.S.-licensed vaccine alone, inclusion of persons vaccinated with an older vaccine formulation, and uncertainties with vaccine coverage estimates). VE estimates against TBE caused by the European subtype virus in studies with the European vaccines were 83%–99% overall after 2 doses of vaccine when the interval after dose 2 was ≤12 months (225228). In recent studies in southern Germany and Latvia, estimates by age group in each location were 94% (95% CI = 84%–98%) and 92% (95% CI = 74%–97%), respectively, for persons aged ≤17 years, 97% (95% CI = 94%–99%) and 98% (95% CI = 97%–99%) for those aged 18–59 years, and 99% (95% CI = 92%–100%) and 100% for those aged ≥60 years (226). If the interval after dose 2 was >12 months, VE was lower; the lowest VE estimate was 81% (95% CI = 39%–94%) among children aged ≤17 years in Latvia (226).

VE for Alimentary TBE

VE against TBE acquired through the alimentary route has been estimated on the basis of a 2017 TBE outbreak investigation in Germany that was identified after a patient with a history of consumption of raw goat milk was hospitalized with meningoencephalitis (263). Among 20 persons who had consumed the raw milk and were included in the investigation, 65% (n = 13) had laboratory-confirmed TBE presenting as nonspecific febrile illness. Among vaccinated persons, the attack rate was 17% (one of six); the infected person’s last TBE vaccine dose had been >15 years previously. Among unvaccinated persons, the attack rate was 86% (12 of 14). Although the number of included persons was limited, the estimated VE was 81% on the basis of these attack rates.

Concomitant Administration of TBE Vaccine with Other Vaccines

Data are limited on co-administration of TBE vaccine with other vaccines. One open label, nonrandomized clinical trial investigated concomitant administration of TBE vaccine and yellow fever vaccine (Stamaril) among adults aged 18–55 years (264). Among persons who received TBE and yellow fever vaccines concomitantly (n = 38), TBE vaccine alone (n = 19) or yellow fever vaccine alone (n = 20), no significant differences were identified between groups in TBE neutralizing antibody titers at 30 days after dose 3 of TBE vaccine, or in final yellow fever neutralizing antibody titers (taken at day 210 in the concomitant group and day 60 in the yellow fever vaccine alone group).

TBE Vaccine Safety

Although the TBE vaccine was only licensed in the United States in 2021, it is available in approximately 30 other countries, primarily in Europe, and during 2001–2021 at least 75 million doses of the current formulation of the vaccine were administered (251). On the basis of this experience, the TBE vaccine is considered to have a good safety profile, with adverse events reported more commonly after the primary series doses than after a booster dose (265).

Common Local and Systemic Adverse Events with TBE Vaccine in Adults

In clinical studies submitted for the vaccine’s licensure in the United States, 4,427 persons aged ≥16 years received at least one 0.5-mL dose of the TBE vaccine. The pivotal safety study was a single blind RCT conducted among adults aged 16–64 years in which persons were randomized to receive 2 doses of the TBE vaccine administered 21–35 days apart; in a follow-up study, persons received the third dose 6 months after dose 1 (224,244,245). Persons in the control group received another non–U.S.-licensed TBE vaccine. Study participants filled in diaries to document solicited local and systemic reactions for 4 days after each vaccine dose. A total of 2,977 persons received ≥1 dose of the U.S-licensed TBE vaccine. The most common reported solicited local reactions within 4 days of vaccination with the U.S.-licensed vaccine were injection site tenderness (30%) and pain (13%). The most common reported solicited systemic reactions were fatigue (6%), headache (6%), and myalgia (5%). In a later study after a booster dose administered 3 years after the primary series, the most common reported local reactions within 4 days of the booster dose among 240 adults were injection site tenderness (5%) and pain (4%) (224,245,246). The most common reported systemic reactions after the booster dose were headache, muscle pain, and malaise (all <1%).

Common Local and Systemic Adverse Events with TBE Vaccine in Children

In clinical studies submitted for vaccine licensure in the United States, 3,240 children aged 1–15 years received at least one 0.25-mL dose of the TBE vaccine. The pivotal pediatric safety study was an observational study that measured rates of adverse events after each of the 3 doses in the primary series (224,247). Caregivers of study participants filled in study diaries to document solicited local and systemic reactions for 4 days after each vaccine dose. Among 2,417 children or adolescents aged 1–15 years who received ≥1 vaccine dose, the most common local reactions reported within 4 days of a dose were injection site tenderness (18%) and pain (11%). The most common systemic reactions were headache (11%) and fever (10%). In addition, restlessness was reported among 9% (53 of 584) children aged 1–5 years after dose 1. Rates of fever were analyzed by age group. After the first dose of TBE vaccine, a fever occurred in 36% (66 of 186) of children aged 1–2 years, 13% (71 of 563) of those aged 3–6 years, and 6% (95 of 1,668) of those aged 7–15 years. Approximately half of all fevers were 100.4°F–101.1°F (38°C–38.4°C), and none were >104°F (40°C). Fever rates were three- to fivefold lower in each age group after doses 2 and 3. In a later booster dose study among 156 children, the most common local reactions were injection site pain (15%) and tenderness (10%) (224,248). The most common systemic reactions were headache (3%) and myalgia (3%). Fever was not reported among children or adolescents after the booster dose.

Serious Adverse Events After TBE vaccine

Serious adverse event (SAE) data were reported in 16 studies including four RCTs, nine observational studies, and three postmarketing surveillance studies (219). Among 3,562 adult and 3,350 pediatric participants who received ≥1 dose of vaccine in the primary series, no SAEs were considered vaccine related. Among 240 adult and 202 pediatric participants who received a booster dose of vaccine, no SAEs were considered vaccine related. Among 687 adult and 1,992 pediatric participants who received a primary series dose or booster dose in three active postmarketing surveillance studies, one SAE was considered possibly vaccine related by study investigators. A child aged 12 months with concomitant rhinopharyngitis, gastroenteritis, and otitis media diagnosed postvaccination was hospitalized for fever at 1 day after vaccination and had a febrile convulsion the following day (266). Although published case reports have occasionally suggested potential vaccine-related SAEs, the absence of reported concerns overall from Europe where millions of doses have been administered indicates SAEs after the TBE vaccine are likely rare (267,268).

Vaccination of Pregnant or Breastfeeding Persons

No adequate, controlled human studies have investigated the safety or immunogenicity of TBE vaccine in pregnant persons, and no animal developmental and reproductive toxicity studies have been conducted. Multiple European countries have recommendations that indicate vaccine can be administered during pregnancy if clearly indicated based on risk (269). Transplacental transfer of TBE virus antibody from vaccinated mothers has been reported and is likely protective for an infant (270). ACIP best practice guidelines for vaccination indicate that no evidence exists for risk to the fetus from vaccinating pregnant women with inactivated viral vaccines (271).

No studies have assessed the safety or immunogenicity of TBE vaccine in breastfeeding persons, and no data are available on excretion in human milk. No inactivated vaccine has been indicated to cause harm in a breastfeeding infant (272). ACIP best practice guidelines for vaccination indicate inactivated vaccines administered to breastfeeding women do not affect the safety of breastfeeding for these women or their infants (271).

Cost-Effectiveness of TBE Vaccine

Among populations in areas where TBE is endemic, results of cost-effectiveness analyses have been variable. TBE vaccination often has been considered not to be cost-effective, with incidence and vaccine cost being important variables in predicting cost-effectiveness (196,273275). Cost-effectiveness analyses for TBE vaccination of travelers have not been conducted. On the basis of the low risk for disease and the vaccine’s cost, vaccination would not be expected to be cost-effective for U.S travelers. However, cost-effectiveness considerations are less relevant when vaccination is for an individual traveler rather than the population. Travel vaccines are not covered by the Vaccines for Children program and usually are not covered by insurance. Travelers’ decisions on purchasing vaccine will likely be based on their willingness and ability to pay and personal perceptions and tolerance of risk.

TBE Prevention and Vaccine Recommendations

Rationale for TBE Vaccine Recommendations for Persons Who Travel Abroad

For most U.S. travelers to areas where TBE is endemic, the risk for TBE is low. However, certain persons who travel abroad are at increased risk for infection because of the season and location of travel and their activities. The risk for exposure to infected ticks is highest for persons who will be in areas where TBE is endemic during the main transmission season (April–November) and who are planning to undertake recreational activities in woodland habitats (e.g., hiking, camping, cycling, hunting, fishing, birdwatching, or collecting mushrooms or berries) or who might be occupationally exposed (e.g., farmers, forestry workers, field researchers, and military personnel) (Box 2).

Taking measures to reduce the risk for tick bites decreases the risk for infection. TBE vaccine can further reduce infection risk and might be indicated for certain persons who are at higher risk for TBE. The risk-benefit assessment for vaccination should consider multiple factors, including the likelihood of exposure to TBE virus-infected ticks on the basis of activities and itinerary (e.g., location, rurality, season, and duration of travel or residence). Persons with extensive exposure to ticks are likely to be at highest risk. Extensive exposure can be considered based on the duration of travel and frequency of exposure and might include shorter-term (e.g., <1 month) travelers with daily or frequent exposure or longer-term travelers with regular (e.g., a few times a month) exposure to environments that might harbor infected ticks. Other risk-benefit considerations should include the 1) rare occurrence but potentially high morbidity and mortality of TBE, 2) higher risk for severe disease among certain persons (e.g., persons aged ≥60 years), 3) availability of an effective vaccine, 4) possibility but low probability of SAEs after vaccination, 5) likelihood of future travel to areas where TBE is endemic, and 6) personal perception and tolerance of risk.

Recommendations for Prevention of TBE Among Persons Who Travel Abroad

Personal Protective Measures

All persons who travel to areas where TBE is endemic should be advised to take precautions to avoid tick bites. Preventive measures include using insect repellent registered by the Environmental Protection Agency, wearing protective clothing, and inspecting the body and clothing for ticks during and after outdoor activities. Travelers to areas where TBE is endemic also should be advised to avoid the consumption of unpasteurized dairy products.

Recommendations for the Use of TBE Vaccine Among Persons Who Travel Abroad

  • TBE vaccine is recommended for persons who are moving or traveling to an area where TBE is endemic and will have extensive exposure to ticks because of their planned outdoor activities and itinerary.
  • TBE vaccine may be considered for persons traveling or moving to an area where TBE is endemic who might engage in outdoor activities in areas where ticks are likely to be found. The decision to vaccinate should be based on an assessment of their planned activities and itinerary, risk factors for a poor medical outcome, and personal perception and tolerance of risk.

An algorithm to assist health care providers with decision-making for TBE vaccination for U.S. travelers is provided (Figure). The evidence for these recommendations can be found in the GRADE and EtR documents available at https://www.cdc.gov/vaccines/acip/recs/grade/table-refs.html and https://www.cdc.gov/vaccines/acip/recs/grade/etr.html.

Recommendations for Prevention of TBE Among Laboratory Workers

TBE virus transmission through aerosolization has been documented in the laboratory setting, and transmission through accidental percutaneous or mucosal exposures is possible. Although no published reports of laboratory-associated TBE virus infections in vaccinated laboratory workers are available, the level of protection that TBE vaccination provides against infection via these transmission routes is unknown.

Best Practices for Working with TBE Virus in the Laboratory

Laboratory work with TBE virus is generally restricted to BSL-4 facilities and practices. Recommendations for best practices for the safe conduct of work in biomedical and clinical laboratories are available (217).

Recommendations for the Use of TBE Vaccine Among Laboratory Workers

  • TBE vaccine is recommended for laboratory workers with a potential for exposure to TBE virus.

A local institutional biosafety committee should undertake a risk assessment of the potential for exposure to TBE virus for each laboratory worker working with TBE virus. The committee should consider the type of work to be performed and the biosafety level at which work will be conducted. Vaccination is not required for workers handling routine clinical samples.

Laboratory workers should receive a standard 3-dose primary series and 1 booster dose. If the potential for ongoing laboratory exposure after receipt of these doses exists, a local biosafety committee should provide guidance on the need for periodic checks of TBE virus-specific neutralizing antibody titers.

Administration of TBE Vaccine

Vaccine Composition, Presentation, Storage, and Handling

The TBE vaccine is an inactivated, whole virus vaccine prepared using a European subtype of TBE virus called the Neudoerfl strain, which was originally isolated from ticks in Austria (230). The vaccine is available as a 0.5-mL presentation for persons aged ≥16 years and a 0.25-mL presentation for children and adolescents aged 1–15 years (224). Each 0.5-mL dose contains 2.4 μg, and each 0.25-mL dose contains 1.2 μg of inactivated TBE virus. The vaccine is produced on chick embryo fibroblast cells with aluminum hydroxide as an adjuvant and human serum albumin as a stabilizer. No thimerosal or other preservatives are included. A list of additional components and substances used in manufacturing that might be found in the final product is available (Table 3).

TBE vaccine is supplied in a prefilled syringe. The tip cap and plunger are not made with natural rubber latex. The vaccine should be stored at 36°F–46°F (2°C–8°C) and should not be frozen. It should be protected from light. Before administration, the vaccine should be brought to room temperature and shaken well. After shaking, it should be an off-white, homogenous, opalescent suspension. Vaccine should not be administered if particulate matter or discoloration remains after shaking.

Dosage, Schedule, and Administration

Primary Vaccination Series

The vaccination dose and timing for the 3-dose primary TBE vaccine series vary by age (Table 3).

  • 1–15 years: 3 doses (0.25 mL each) administered intramuscularly (IM) with the first 2 doses administered 1–3 months apart and the third dose administered 5–12 months after dose 2.
  • ≥16 years: 3 doses (0.5 mL each) administered IM with the first 2 doses administered 14 days to 3 months apart and the third dose administered 5–12 months after dose 2.

For all age groups, the 3-dose primary vaccination series should be completed at least 1 week before potential exposure to TBE virus. For persons (e.g., travelers) who cannot complete the 3-dose primary series, see Immunologic Response After an Incomplete Primary Series (1 or 2 Doses) in Adults and Children.

Booster Dose

For all age groups, a booster dose (i.e., fourth dose) can be administered at least 3 years after completion of the primary 3-dose TBE vaccine series if ongoing exposure or re-exposure to TBE virus is expected. No ACIP recommendations are made on the need for subsequent booster doses.

  • 1–15 years: 0.25-mL dose administered IM ≥3 years after completion of primary series.
  • ≥16 years: 0.5-mL dose administered IM ≥3 years after completion of primary series.

Concomitant Administration of Other Vaccines or Drugs

Data are limited on co-administration of TBE vaccine with other vaccines or drugs. One clinical trial in which the first dose of TBE vaccine was administered concomitantly with yellow fever vaccine indicated no interference with the immune response to TBE vaccine or yellow fever vaccine (264). If TBE vaccine and other vaccines are administered concomitantly, they should be administered with separate syringes and at different anatomic sites (271).

Contraindications for the Use of TBE Vaccine

Allergy to Vaccine Components

A severe allergic reaction (e.g., anaphylaxis) to any component of the TBE vaccine, including substances remaining from the manufacturing process (e.g., protamine sulfate, neomycin, gentamicin, and chick protein), is a contraindication to vaccination (224) (Table 3). Although TBE vaccine is prepared from TBE virus propagated in chick embryo fibroblast cells, the ultracentrifugation process removes all but trace amounts of chick protein. No reports have been published related to TBE vaccine and egg allergy. However, a known severe hypersensitivity to egg or chicken protein (e.g., anaphylaxis after oral ingestion) is a contraindication to TBE vaccination. As with any vaccine, all persons should be vaccinated in settings with the capacity to manage allergic reactions (271).

Precautions for the Use of TBE Vaccine

Altered Immune States

Having an immunocompromising condition or being immunosuppressed are precautions for vaccination. The immunogenicity and safety of TBE vaccine in persons with altered immune status has not been well characterized. Available information suggests the vaccine can be safely administered, but the immune response might be diminished (224,276278). Additional information on vaccination in persons with altered immunocompetence can be found in the ACIP general guidelines for best vaccination practices (271).

Special Populations

Pregnant persons: Pregnancy is not a contraindication or precaution to vaccination. TBE virus infection can pose a risk for severe illness in pregnant persons; thus, the benefits of vaccinating pregnant persons when the likelihood of infection is high likely outweigh the potential risks.

Breastfeeding persons: Breastfeeding is not a contraindication or precaution to vaccination with TBE vaccine.

Infants aged <1 year: Safety and effectiveness of TBE vaccine have not been established for infants aged <1 year.

Persons in older age groups: Data on safety and immunogenicity of TBE vaccine among persons aged >50 years are limited but suggest there are no safety concerns. Immunologic data suggest the response to TBE vaccine is reduced among adults aged >50 years, particularly among those aged ≥65 years. Levels of neutralizing antibodies are lower postvaccination in older adults, and older adults are more likely to become seronegative than younger adults in the years after their last dose of vaccine.

Reporting of Vaccine Adverse Events

Surveillance for adverse events associated with administration of TBE vaccine is important. An adverse event that occurs after vaccine administration, even if a causal relation to vaccination is not certain, should be reported to the Vaccine Adverse Event Reporting System (VAERS) at https://vaers.hhs.gov or by calling 1-800-822-7967.

Future Research

The ACIP Tick-Borne Encephalitis Vaccine Work Group identified multiple key areas for further research. One important topic is the level of protection afforded by the U.S.-licensed TBE vaccine, based on a European subtype TBE virus, for non-European TBE virus subtypes.

Evaluation of potential immunological interactions between TBE vaccine and other flaviviruses or flavivirus vaccines (e.g., yellow fever or Japanese encephalitis vaccines) is needed. A small study investigating the effect of previous yellow fever vaccination found that TBE virus GMTs were significantly lower in persons who previously received yellow fever vaccine compared with flavivirus-naïve persons (279). In vaccine trials, persons with evidence of previous flavivirus exposure were usually excluded; therefore, determining whether previous exposure to other flaviviruses endemic in the United States (e.g., the tickborne Powassan virus or the mosquitoborne West Nile virus) or to flaviviruses travelers might have been exposed to (e.g., dengue and Zika viruses) might affect the immune response to TBE vaccine would be useful. Likelihood of interference is low when TBE vaccine is administered simultaneously with other vaccines, medications, or substances because it is an inactivated vaccine; however, potential immunological interactions could be addressed by appropriate studies.

Data are limited on TBE vaccine use among certain populations. Data on immunogenicity of TBE vaccine among older adults are limited and suggest the immunologic response to TBE vaccine is reduced. Additional data on vaccine protection among older adults would be valuable, particularly the need for and timing of any additional booster doses. Further research is warranted to assess the clinical spectrum of TBE virus infection during pregnancy to better understand the extent of risk for this population. Although no data are available to suggest concerns with use of this inactivated vaccine among pregnant or breastfeeding persons, systematic collection of additional data in these populations would be beneficial.

Additional Information

Additional information about TBE is available from CDC at https://www.cdc.gov/tick-borne-encephalitis. Additional licensure information for TBE vaccine is available from FDA (https://www.fda.gov/vaccines-blood-biologics/ticovac).

Acknowledgment

Ann Powers provided advice in development of the tick-borne encephalitis vaccine recommendations for laboratory workers.

Members of the Advisory Committee on Immunization Practices

ACIP member rosters for October 2020–February 2022 are available at https://www.cdc.gov/vaccines/acip/members/members-archive.html.

ACIP Tick-Borne Encephalitis Vaccine Work Group

Membership as of March 2, 2022

Chair: Katherine A. Poehling, MD, Wake Forest School of Medicine, Winston-Salem, North Carolina.

Members: Rodolfo M. Alarcon, PhD, National Institutes of Health, Bethesda, Maryland; Alan Barrett, PhD, University of Texas Medical Branch, Galveston, Texas; Lin Chen, MD, Mount Auburn Hospital, Cambridge, Massachusetts; Wilbur H. Chen, MD, University of Maryland School of Medicine, Baltimore, Maryland; Ihid Carneiro Leão, MD, PhD, Food and Drug Administration, Silver Spring, Maryland; Anthony A. Marfin, MD, PATH, Seattle, Washington; Bruce McClenathan, MD, U.S. Department of Defense, Fort Bragg, North Carolina; Kayvon Modjarrad, MD, PhD, Walter Reed Army Institute of Research, Silver Spring, Maryland; Mark Sawyer, MD, American Academy of Pediatrics and University of California San Diego School of Medicine, La Jolla, California; Steven Schofield, PhD, Canadian Forces Health Services, National Defence, Ottawa, Canada; Bryan Schumacher, MD, U.S. Department of Defense, Atlanta, Georgia; David R. Shlim, MD, International Society of Travel Medicine and Jackson Hole Travel and Tropical Medicine, Jackson, Wyoming; Mary Wilson, MD, University of California San Francisco, San Francisco, California.

CDC Contributors: Grace D. Appiah, MD, Atlanta, Georgia; Susan Y. Chu, PhD, Atlanta, Georgia; Caitlin M. Cossaboom, DVM, PhD, Atlanta, Georgia; Stacey Martin, MSc, Fort Collins, Colorado; Michael M. McNeil, MD, Atlanta, Georgia; J. Erin Staples, MD, PhD, Fort Collins, Colorado; Stephen Waterman, MD, San Juan, Puerto Rico.

Methodology Contributors: Doug Campos-Outcalt, MD; Rebecca Morgan, PhD.

Work Group Secretariat: Susan L. Hills, MBBS, CDC, Fort Collins, Colorado.

Corresponding author: Susan L. Hills, National Center for Emerging and Zoonotic Infectious Diseases, CDC. Telephone: 970-221-6400; Email: shills@cdc.gov.


1Division of Vector-Borne Diseases, National Center for Emerging and Zoonotic Infectious Diseases, CDC, Fort Collins, Colorado; 2Wake Forest School of Medicine, Winston-Salem, North Carolina; 3University of Maryland School of Medicine, Baltimore, Maryland

Conflicts of Interest

All authors have completed and submitted the International Committee of Medical Journal Editors form for disclosure of potential conflicts of interest. No conflicts of interest were reported.

References

  1. Dobler G, Gniel D, Petermann R, Pfeffer M. Epidemiology and distribution of tick-borne encephalitis. Wien Med Wochenschr 2012;162:230–8. https://doi.org/10.1007/s10354-012-0100-5 PMID:22699708
  2. Barrett PN, Schober-Bendixen S, Ehrlich HJ. History of TBE vaccines. Vaccine 2003;21(Suppl 1):S41–9. https://doi.org/10.1016/S0264-410X(02)00814-9 PMID:12628813
  3. Loew-Baselli A, Poellabauer EM, Pavlova BG, et al. Prevention of tick-borne encephalitis by FSME-IMMUN vaccines: review of a clinical development programme. Vaccine 2011;29:7307–19. https://doi.org/10.1016/j.vaccine.2011.07.089 PMID:21843576
  4. Heinz FX. Molecular aspects of TBE virus research. Vaccine 2003;21(Suppl 1):S3–10. https://doi.org/10.1016/S0264-410X(02)00820-4 PMID:12628809
  5. Calisher CH, Karabatsos N, Dalrymple JM, et al. Antigenic relationships between flaviviruses as determined by cross-neutralization tests with polyclonal antisera. J Gen Virol 1989;70:37–43. https://doi.org/10.1099/0022-1317-70-1-37 PMID:2543738
  6. Ecker M, Allison SL, Meixner T, Heinz FX. Sequence analysis and genetic classification of tick-borne encephalitis viruses from Europe and Asia. J Gen Virol 1999;80:179–85. https://doi.org/10.1099/0022-1317-80-1-179 PMID:9934700
  7. Demina TV, Dzhioev YP, Verkhozina MM, et al. Genotyping and characterization of the geographical distribution of tick-borne encephalitis virus variants with a set of molecular probes. J Med Virol 2010;82:965–76. https://doi.org/10.1002/jmv.21765 PMID:20419810
  8. Tkachev SE, Babkin IV, Chicherina GS, et al. Genetic diversity and geographical distribution of the Siberian subtype of the tick-borne encephalitis virus. Ticks Tick Borne Dis 2020;11:101327. https://doi.org/10.1016/j.ttbdis.2019.101327 PMID:31767494
  9. Erber W, Schmitt H, Janković T. TBE—epidemiology by country—an overview [Chapter 12a]. In: Dobler G, Erber W, Bröker M, Schmitt H, eds. The TBE book. 4th ed. Singapore: Global Health Press; 2021:154–170.
  10. Dai X, Shang G, Lu S, Yang J, Xu J. A new subtype of eastern tick-borne encephalitis virus discovered in Qinghai-Tibet Plateau, China. Emerg Microbes Infect 2018;7:74. https://doi.org/10.1038/s41426-018-0081-6 PMID:29691370
  11. Kovalev SY, Mukhacheva TA. Reconsidering the classification of tick-borne encephalitis virus within the Siberian subtype gives new insights into its evolutionary history. Infect Genet Evol 2017;55:159–65. https://doi.org/10.1016/j.meegid.2017.09.014 PMID:28919548
  12. Krbková L, Štroblová H, Bednářová J. Clinical course and sequelae for tick-borne encephalitis among children in South Moravia (Czech Republic). Eur J Pediatr 2015;174:449–58. https://doi.org/10.1007/s00431-014-2401-8 PMID:25199479
  13. Tonteri E, Kurkela S, Timonen S, et al. Surveillance of endemic foci of tick-borne encephalitis in Finland 1995–2013: evidence of emergence of new foci. Euro Surveill 2015;20. https://doi.org/10.2807/1560-7917.ES.2015.20.37.30020 PMID:26535471
  14. Kohlmaier B, Schweintzger NA, Sagmeister MG, et al.; The Eu-Tick-Bo Study Group. Clinical characteristics of patients with tick-borne encephalitis (TBE): a European multicentre study from 2010 to 2017. Microorganisms 2021;9:1420. https://doi.org/10.3390/microorganisms9071420 PMID:34209373
  15. Zavadska D, Odzelevica Z, Karelis G, et al. Tick-borne encephalitis: a 43-year summary of epidemiological and clinical data from Latvia (1973 to 2016). PLoS One 2018;13:e0204844. https://doi.org/10.1371/journal.pone.0204844 PMID:30422984
  16. Radzišauskienė D, Urbonienė J, Kaubrys G, et al. The epidemiology, clinical presentation, and predictors of severe tick-borne encephalitis in Lithuania, a highly endemic country: a retrospective study of 1040 patients. PLoS One 2020;15:e0241587. https://doi.org/10.1371/journal.pone.0241587 PMID:33211708
  17. Kaiser R. The clinical and epidemiological profile of tick-borne encephalitis in southern Germany 1994–98: a prospective study of 656 patients. Brain 1999;122:2067–78. https://doi.org/10.1093/brain/122.11.2067 PMID:10545392
  18. Mickiene A, Laiskonis A, Günther G, Vene S, Lundkvist A, Lindquist L. Tickborne encephalitis in an area of high endemicity in Lithuania: disease severity and long-term prognosis. Clin Infect Dis 2002;35:650–8. https://doi.org/10.1086/342059 PMID:12203160
  19. Fowler Å, Forsman L, Eriksson M, Wickström R. Tick-borne encephalitis carries a high risk of incomplete recovery in children. J Pediatr 2013;163:555–60. https://doi.org/10.1016/j.jpeds.2013.01.037 PMID:23452585
  20. Alekseev AN, Chunikhin SP. [The experimental transmission of the tick-borne encephalitis virus by ixodid ticks (the mechanisms, time periods, species and sex differences)]. Parazitologiia 1990;24:177–85. PMID:2216530
  21. Katargina O, Russakova S, Geller J, et al. Detection and characterization of tick-borne encephalitis virus in Baltic countries and eastern Poland. PLoS One 2013;8:e61374. https://doi.org/10.1371/journal.pone.0061374 PMID:23650497
  22. Michelitsch A, Wernike K, Klaus C, Dobler G, Beer M. Exploring the reservoir hosts of tick-borne encephalitis virus. Viruses 2019;11:669. https://doi.org/10.3390/v11070669 PMID:31336624
  23. Kahl O, Gray JS. The biology of Ixodes ricinus with emphasis on its ecology. Ticks Tick Borne Dis 2023;14:102114. https://doi.org/10.1016/j.ttbdis.2022.102114 PMID:36603231
  24. Lundkvist K, Vene S, Golovljova I, et al. Characterization of tick-borne encephalitis virus from Latvia: evidence for co-circulation of three distinct subtypes. J Med Virol 2001;65:730–5. https://doi.org/10.1002/jmv.2097 PMID:11745938
  25. Golovljova I, Vene S, Sjölander KB, Vasilenko V, Plyusnin A, Lundkvist A. Characterization of tick-borne encephalitis virus from Estonia. J Med Virol 2004;74:580–8. https://doi.org/10.1002/jmv.20224 PMID:15484275
  26. Jääskeläinen AE, Tonteri E, Sironen T, Pakarinen L, Vaheri A, Vapalahti O. European subtype tick-borne encephalitis virus in Ixodes persulcatus ticks. Emerg Infect Dis 2011;17:323–5. https://doi.org/10.3201/eid1702.101487 PMID:21291624
  27. Zlobin V, Esyunina M, Syrochkina M. TBE in Russia [Chapter 12b]. In: Dobler G, Erber W, Bröker M, Schmitt H, eds. The TBE book. 4th ed. Singapore: Global Health Press; 2021:298–309.
  28. Chitimia-Dobler L, Mackenstedt U, Kahl O. Transmission/natural cycle [Chapter 3]. In: Dobler G, Erber W, Bröker M, Schmitt H, eds. The TBE book. 4th ed. Singapore: Global Health Press; 2021:47–63.
  29. Wongnak P, Bord S, Jacquot M, et al. Meteorological and climatic variables predict the phenology of Ixodes ricinus nymph activity in France, accounting for habitat heterogeneity. Sci Rep 2022;12:7833. https://doi.org/10.1038/s41598-022-11479-z PMID:35552424
  30. Süss J. Epidemiology and ecology of TBE relevant to the production of effective vaccines. Vaccine 2003;21(Suppl 1):S19–35. https://doi.org/10.1016/S0264-410X(02)00812-5 PMID:12628811
  31. Zając Z, Kulisz J, Bartosik K, Woźniak A, Dzierżak M, Khan A. Environmental determinants of the occurrence and activity of Ixodes ricinus ticks and the prevalence of tick-borne diseases in eastern Poland. Sci Rep 2021;11:15472. https://doi.org/10.1038/s41598-021-95079-3 PMID:34326447
  32. Schulz M, Mahling M, Pfister K. Abundance and seasonal activity of questing Ixodes ricinus ticks in their natural habitats in southern Germany in 2011. J Vector Ecol 2014;39:56–65. https://doi.org/10.1111/j.1948-7134.2014.12070.x PMID:24820556
  33. Blasko-Markic M, Socan M. Tick-borne encephalitis in Slovenia: data from a questionnaire survey. Vector Borne Zoonotic Dis 2012;12:496–502. https://doi.org/10.1089/vbz.2011.0871 PMID:22448719
  34. Bušová A, Dorko E, Feketeová E, et al. Serostatus and risk factors of tick-borne encephalitis. Cent Eur J Public Health 2018;26(Suppl):S56–60. https://doi.org/10.21101/cejph.a5289 PMID:30817875
  35. Rezza G, Farchi F, Pezzotti P, et al.; TBE Virology Group. Tick-borne encephalitis in north-east Italy: a 14-year retrospective study, January 2000 to December 2013. Euro Surveill 2015;20. https://doi.org/10.2807/1560-7917.ES.2015.20.40.30034 PMID:26537646
  36. Dumpis U, Crook D, Oksi J. Tick-borne encephalitis. Clin Infect Dis 1999;28:882–90. https://doi.org/10.1086/515195 PMID:10825054
  37. Czupryna P, Moniuszko A, Pancewicz SA, Grygorczuk S, Kondrusik M, Zajkowska J. Tick-borne encephalitis in Poland in years 1993–2008—epidemiology and clinical presentation. A retrospective study of 687 patients. Eur J Neurol 2011;18:673–9. https://doi.org/10.1111/j.1468-1331.2010.03278.x PMID:21143706
  38. Schielein L, Tizek L, Biedermann T, Zink A. Tick bites in different professions and regions: pooled cross-sectional study in the focus area Bavaria, Germany. BMC Public Health 2022;22:234. https://doi.org/10.1186/s12889-021-12456-3 PMID:35120477
  39. Lane RS, Mun J, Stubbs HA. Horizontal and vertical movements of host-seeking Ixodes pacificus (Acari: Ixodidae) nymphs in a hardwood forest. J Vector Ecol 2009;34:252–66. https://doi.org/10.1111/j.1948-7134.2009.00034.x PMID:20352083
  40. Medlock JM, Hansford KM, Bormane A, et al. Driving forces for changes in geographical distribution of Ixodes ricinus ticks in Europe. Parasit Vectors 2013;6:1. https://doi.org/10.1186/1756-3305-6-1 PMID:23281838
  41. Makenov M, Karan L, Shashina N, et al. First detection of tick-borne encephalitis virus in Ixodes ricinus ticks and their rodent hosts in Moscow, Russia. Ticks Tick Borne Dis 2019;10:101265. https://doi.org/10.1016/j.ttbdis.2019.101265 PMID:31447316
  42. Oechslin CP, Heutschi D, Lenz N, et al. Prevalence of tick-borne pathogens in questing Ixodes ricinus ticks in urban and suburban areas of Switzerland. Parasit Vectors 2017;10:558. https://doi.org/10.1186/s13071-017-2500-2 PMID:29121976
  43. Labuda M, Jones LD, Williams T, Danielova V, Nuttall PA. Efficient transmission of tick-borne encephalitis virus between cofeeding ticks. J Med Entomol 1993;30:295–9. https://doi.org/10.1093/jmedent/30.1.295 PMID:8433342
  44. Randolph SE. Transmission of tick-borne pathogens between co-feeding ticks: Milan Labuda’s enduring paradigm. Ticks Tick Borne Dis 2011;2:179–82. https://doi.org/10.1016/j.ttbdis.2011.07.004 PMID:22108009
  45. Bakhvalova VN, Chicherina GS, Potapova OF, et al. Tick-borne encephalitis virus diversity in ixodid ticks and small mammals in south-western Siberia, Russia. Vector Borne Zoonotic Dis 2016;16:541–9. https://doi.org/10.1089/vbz.2015.1834 PMID:27303915
  46. Slovák M, Kazimírová M, Siebenstichová M, et al. Survival dynamics of tick-borne encephalitis virus in Ixodes ricinus ticks. Ticks Tick Borne Dis 2014;5:962–9. https://doi.org/10.1016/j.ttbdis.2014.07.019 PMID:25131151
  47. Kondrashova ZN, Filippovets RV. [Infectability of Ixodes persulcatus ticks and questions concerning transovarial transmission during dose regulated infection with tick-borne encephalitis virus]. Vopr Virusol 1970;15:703–8. PMID:5518645
  48. Stanko M, Derdáková M, Špitalská E, Kazimírová M. Ticks and their epidemiological role in Slovakia: from the past till present. Biologia (Bratisl) 2022;77:1575–610. Published online September 17, 2021. https://doi.org/10.1007/s11756-021-00845-3 PMID:34548672
  49. Holzmann H. Diagnosis of tick-borne encephalitis. Vaccine 2003;21(Suppl 1):S36–40. https://doi.org/10.1016/S0264-410X(02)00819-8 PMID:12628812
  50. Puchhammer-Stöckl E, Kunz C, Mandl CW, Heinz FX. Identification of tick-borne encephalitis virus ribonucleic acid in tick suspensions and in clinical specimens by a reverse transcription-nested polymerase chain reaction assay. Clin Diagn Virol 1995;4:321–6. https://doi.org/10.1016/0928-0197(95)00022-4 PMID:15566853
  51. Saksida A, Jakopin N, Jelovšek M, et al. Virus RNA load in patients with tick-borne encephalitis, Slovenia. Emerg Infect Dis 2018;24:1315–23. https://doi.org/10.3201/eid2407.180059 PMID:29912706
  52. Holzmann H, Aberle SW, Stiasny K, et al. Tick-borne encephalitis from eating goat cheese in a mountain region of Austria. Emerg Infect Dis 2009;15:1671–3. https://doi.org/10.3201/eid1510.090743 PMID:19861072
  53. Hudopisk N, Korva M, Janet E, et al. Tick-borne encephalitis associated with consumption of raw goat milk, Slovenia, 2012. Emerg Infect Dis 2013;19:806–8. https://doi.org/10.3201/eid1905.121442 PMID:23697658
  54. Balogh Z, Ferenczi E, Szeles K, et al. Tick-borne encephalitis outbreak in Hungary due to consumption of raw goat milk. J Virol Methods 2010;163:481–5. https://doi.org/10.1016/j.jviromet.2009.10.003 PMID:19836419
  55. Caini S, Szomor K, Ferenczi E, et al. Tick-borne encephalitis transmitted by unpasteurised cow milk in western Hungary, September to October 2011. Euro Surveill 2012;17:20128. https://doi.org/10.2807/ese.17.12.20128-en PMID:22490310
  56. Kerlik J, Avdičová M, Štefkovičová M, et al. Slovakia reports highest occurrence of alimentary tick-borne encephalitis in Europe: analysis of tick-borne encephalitis outbreaks in Slovakia during 2007–2016. Travel Med Infect Dis 2018;26:37–42. https://doi.org/10.1016/j.tmaid.2018.07.001 PMID:30012472
  57. Ličková M, Fumačová Havlíková S, Sláviková M, Klempa B. Alimentary infections by tick-borne encephalitis virus. Viruses 2021;14:56. https://doi.org/10.3390/v14010056 PMID:35062261
  58. Buczek AM, Buczek W, Buczek A, Wysokińska-Miszczuk J. Food-borne transmission of tick-borne encephalitis virus—spread, consequences, and prophylaxis. Int J Environ Res Public Health 2022;19:1812. https://doi.org/10.3390/ijerph19031812 PMID:35162837
  59. Martello E, Gillingham EL, Phalkey R, et al. Systematic review on the non-vectorial transmission of tick-borne encephalitis virus (TBEv). Ticks Tick Borne Dis 2022;13:102028. https://doi.org/10.1016/j.ttbdis.2022.102028 PMID:36030646
  60. Elbaz M, Gadoth A, Shepshelovich D, Shasha D, Rudoler N, Paran Y. Systematic review and meta-analysis of foodborne tick-borne encephalitis, Europe, 1980–2021. Emerg Infect Dis 2022;28:1945–54. https://doi.org/10.3201/eid2810.220498 PMID:36149234
  61. Hennechart-Collette C, Gonzalez G, Fourniol L, et al. Method for tick-borne encephalitis virus detection in raw milk products. Food Microbiol 2022;104:104003. https://doi.org/10.1016/j.fm.2022.104003 PMID:35287822
  62. Smorodintsev AA. Tick-borne spring-summer encephalitis. Prog Med Virol 1958;1:210–47. PMID:13579009
  63. Avsic-Zupanc T, Poljak M, Maticic M, et al. Laboratory acquired tick-borne meningoencephalitis: characterisation of virus strains. Clin Diagn Virol 1995;4:51–9. https://doi.org/10.1016/0928-0197(94)00062-Y PMID:15566827
  64. Laboratory safety for arboviruses and certain other viruses of vertebrates. The Subcommittee on Arbovirus Laboratory Safety of the American Committee on Arthropod-Borne Viruses. Am J Trop Med Hyg 1980;29:1359–81. https://doi.org/10.4269/ajtmh.1980.29.1359 PMID:6778230
  65. Nebogatkin I, Onishchuk O, Oleksandr H, Erber W, Vuković-Janković T. TBE in Ukraine [Chapter 12b]. In: Dobler G, Erber W, Bröker M, Schmitt H, eds. The TBE book. 4th ed. Singapore: Global Health Press; 2021:348–52.
  66. Kerlik J, Avdičová M, Musilová M, Bérešová J, Mezencev R. Breast milk as route of tick-borne encephalitis virus transmission from mother to infant. Emerg Infect Dis 2022;28:1060–1. https://doi.org/10.3201/eid2805.212457 PMID:35447060
  67. Wahlberg P, Saikku P, Brummer-Korvenkontio M. Tick-borne viral encephalitis in Finland. The clinical features of Kumlinge disease during 1959–1987. J Intern Med 1989;225:173–7. https://doi.org/10.1111/j.1365-2796.1989.tb00059.x PMID:2703799
  68. Lipowski D, Popiel M, Perlejewski K, et al. A cluster of fatal tick-borne encephalitis virus infection in organ transplant setting. J Infect Dis 2017;215:896–901. https://doi.org/10.1093/infdis/jix040 PMID:28453842
  69. Süss J. Tick-borne encephalitis 2010: epidemiology, risk areas, and virus strains in Europe and Asia—an overview. Ticks Tick Borne Dis 2011;2:2–15. https://doi.org/10.1016/j.ttbdis.2010.10.007 PMID:21771531
  70. Zubriková D, Wittmann M, Hönig V, et al. Prevalence of tick-borne encephalitis virus and Borrelia burgdorferi sensu lato in Ixodes ricinus ticks in Lower Bavaria and Upper Palatinate, Germany. Ticks Tick Borne Dis 2020;11:101375. https://doi.org/10.1016/j.ttbdis.2020.101375 PMID:31983627
  71. Zeman P. Objective assessment of risk maps of tick-borne encephalitis and Lyme borreliosis based on spatial patterns of located cases. Int J Epidemiol 1997;26:1121–9. https://doi.org/10.1093/ije/26.5.1121 PMID:9363536
  72. Agergaard CN, Rosenstierne MW, Bødker R, Rasmussen M, Andersen PHS, Fomsgaard A. New tick-borne encephalitis virus hot spot in Northern Zealand, Denmark, October 2019. Euro Surveill 2019;24:1900639. https://doi.org/10.2807/1560-7917.ES.2019.24.43.1900639 PMID:31662158
  73. Dobler G, Tkachev S. General epidemiology of TBE [Chapter 11]. In: Dobler G, Erber W, Bröker M, Schmitt H, eds. The TBE book. 4th ed. Singapore: Global Health Press; 2021:141–153.
  74. Laaksonen M, Sajanti E, Sormunen JJ, et al. Crowdsourcing-based nationwide tick collection reveals the distribution of Ixodes ricinus and I. persulcatus and associated pathogens in Finland. Emerg Microbes Infect 2017;6:e31. https://doi.org/10.1038/emi.2017.17 PMID:28487561
  75. Jääskeläinen AE, Sironen T, Murueva GB, et al. Tick-borne encephalitis virus in ticks in Finland, Russian Karelia and Buryatia. J Gen Virol 2010;91:2706–12. https://doi.org/10.1099/vir.0.023663-0 PMID:20660147
  76. Kupča AM, Essbauer S, Zoeller G, et al. Isolation and molecular characterization of a tick-borne encephalitis virus strain from a new tick-borne encephalitis focus with severe cases in Bavaria, Germany. Ticks Tick Borne Dis 2010;1:44–51. https://doi.org/10.1016/j.ttbdis.2009.11.002 PMID:21771510
  77. Korenberg EI, Kovalevskii YV. Main features of tick-borne encephalitis eco-epidemiology in Russia. Zentralbl Bakteriol 1999;289:525–39. https://doi.org/10.1016/S0934-8840(99)80006-1 PMID:10652719
  78. Skarpaas T, Ljøstad U, Sundøy A. First human cases of tickborne encephalitis, Norway. Emerg Infect Dis 2004;10:2241–3. https://doi.org/10.3201/eid1012.040598 PMID:15663873
  79. Jaenson TG, Hjertqvist M, Bergström T, Lundkvist A. Why is tick-borne encephalitis increasing? A review of the key factors causing the increasing incidence of human TBE in Sweden. Parasit Vectors 2012;5:184. https://doi.org/10.1186/1756-3305-5-184 PMID:22937961
  80. Fomsgaard A, Christiansen C, Bodker R. First identification of tick-borne encephalitis in Denmark outside of Bornholm, August 2009. Euro Surveill 2009;14:19325. https://doi.org/10.2807/ese.14.36.19325-en PMID:19758543
  81. Heinz FX, Stiasny K, Holzmann H, et al. Emergence of tick-borne encephalitis in new endemic areas in Austria: 42 years of surveillance. Euro Surveill 2015;20:9–16. https://doi.org/10.2807/1560-7917.ES2015.20.13.21077 PMID:25860391
  82. Rieille N, Bally F, Péter O. Tick-borne encephalitis: first autochtonous case and epidemiological surveillance in canton Valais, Switzerland [French]. Rev Med Suisse 2012;8:1916–8, 1920. PMID:23130421
  83. Herpe B, Schuffenecker I, Pillot J, et al. Tick-borne encephalitis, southwestern France. Emerg Infect Dis 2007;13:1114–6. PMID:18214196
  84. Botelho-Nevers E, Gagneux-Brunon A, Velay A, et al. Tick-borne encephalitis in Auvergne-Rhône-Alpes region, France, 2017–2018. Emerg Infect Dis 2019;25:1944–8. https://doi.org/10.3201/eid2510.181923 PMID:31538929
  85. Perfilyeva YV, Shapiyeva ZZ, Ostapchuk YO, et al. Tick-borne pathogens and their vectors in Kazakhstan—a review. Ticks Tick Borne Dis 2020;11:101498. https://doi.org/10.1016/j.ttbdis.2020.101498 PMID:32723625
  86. Hellenbrand W, Kreusch T, Böhmer MM, et al. Epidemiology of tick-borne encephalitis (TBE) in Germany, 2001–2018. Pathogens 2019;8:29. https://doi.org/10.3390/pathogens8020042 PMID:30934855
  87. Soleng A, Edgar KS, Paulsen KM, et al. Distribution of Ixodes ricinus ticks and prevalence of tick-borne encephalitis virus among questing ticks in the Arctic Circle region of northern Norway. Ticks Tick Borne Dis 2018;9:97–103. https://doi.org/10.1016/j.ttbdis.2017.10.002 PMID:29030314
  88. Tokarevich NK, Tronin AA, Blinova OV, et al. The impact of climate change on the expansion of Ixodes persulcatus habitat and the incidence of tick-borne encephalitis in the north of European Russia. Glob Health Action 2011;4:8448. https://doi.org/10.3402/gha.v4i0.8448 PMID:22028678
  89. Kunze U; ISW TBE. Tick-borne encephalitis: the impact of epidemiology, changing lifestyle, and environmental factors. Conference report of the 12th Annual Meeting of the International Scientific Working Group on Tick-Borne Encephalitis (ISW-TBE). Vaccine 2011;29:1355–6. https://doi.org/10.1016/j.vaccine.2010.12.048 PMID:21199705
  90. de Graaf JA, Reimerink JH, Voorn GP, et al. First human case of tick-borne encephalitis virus infection acquired in the Netherlands, July 2016. Euro Surveill 2016;21:30318. https://doi.org/10.2807/1560-7917.ES.2016.21.33.30318 PMID:27562931
  91. Kreusch TM, Holding M, Hewson R, et al. A probable case of tick-borne encephalitis (TBE) acquired in England, July 2019. Euro Surveill 2019;24:1900679. https://doi.org/10.2807/1560-7917.ES.2019.24.47.1900679 PMID:31771699
  92. Stoefs A, Heyndrickx L, De Winter J, et al. Autochthonous cases of tick-borne encephalitis, Belgium, 2020. Emerg Infect Dis 2021;27:2179–82. https://doi.org/10.3201/eid2708.211175 PMID:34111382
  93. Lukan M, Bullova E, Petko B. Climate warming and tick-borne encephalitis, Slovakia. Emerg Infect Dis 2010;16:524–6. https://doi.org/10.3201/eid1603.081364 PMID:20202437
  94. Zeman P, Bene C. A tick-borne encephalitis ceiling in Central Europe has moved upwards during the last 30 years: possible impact of global warming? Int J Med Microbiol 2004;293(Suppl 37):48–54. PMID:15146984
  95. Briggs BJ, Atkinson B, Czechowski DM, et al. Tick-borne encephalitis virus, Kyrgyzstan. Emerg Infect Dis 2011;17:876–9. https://doi.org/10.3201/eid1705.101183 PMID:21529400
  96. Ostfeld RS, Brunner JL. Climate change and Ixodes tick-borne diseases of humans. Philos Trans R Soc Lond B Biol Sci 2015;370:1665. https://doi.org/10.1098/rstb.2014.0051 PMID:25688022
  97. Hasle G. Transport of ixodid ticks and tick-borne pathogens by migratory birds. Front Cell Infect Microbiol 2013;3:48. https://doi.org/10.3389/fcimb.2013.00048 PMID:24058903
  98. Waldenström J, Lundkvist A, Falk KI, et al. Migrating birds and tick-borne encephalitis virus. Emerg Infect Dis 2007;13:1215–8. https://doi.org/10.3201/eid1308.061416 PMID:17953095
  99. Pfeffer M, Dobler G. Emergence of zoonotic arboviruses by animal trade and migration. Parasit Vectors 2010;3:35. https://doi.org/10.1186/1756-3305-3-35 PMID:20377873
  100. Weidmann M, Frey S, Freire CCM, et al. Molecular phylogeography of tick-borne encephalitis virus in central Europe. J Gen Virol 2013;94:2129–39. https://doi.org/10.1099/vir.0.054478-0 PMID:23784447
  101. Knap N, Avšič-Županc T. Factors affecting the ecology of tick-borne encephalitis in Slovenia. Epidemiol Infect 2015;143:2059–67. https://doi.org/10.1017/S0950268815000485 PMID:25918865
  102. Andreassen A, Jore S, Cuber P, et al. Prevalence of tick borne encephalitis virus in tick nymphs in relation to climatic factors on the southern coast of Norway. Parasit Vectors 2012;5:177. https://doi.org/10.1186/1756-3305-5-177 PMID:22913287
  103. Klaus C, Gethmann J, Hoffmann B, Ziegler U, Heller M, Beer M. Tick infestation in birds and prevalence of pathogens in ticks collected from different places in Germany. Parasitol Res 2016;115:2729–40. https://doi.org/10.1007/s00436-016-5022-5 PMID:27048511
  104. European Centre for Disease Prevention and Control. Tick-borne encephalitis. Annual Epidemiologic Report for 2020. Stockholm, Sweden; 2022. https://www.ecdc.europa.eu/en/publications-data/tick-borne-encephalitis-annual-epidemiological-report-2020
  105. Velay A, Solis M, Kack-Kack W, et al. A new hot spot for tick-borne encephalitis (TBE): a marked increase of TBE cases in France in 2016. Ticks Tick Borne Dis 2018;9:120–5. https://doi.org/10.1016/j.ttbdis.2017.09.015 PMID:28988602
  106. Yoshii K, Takahashi-Iwata I, Shirai S, Kobayashi S, Yabe I, Sasaki H. A retrospective epidemiological study of tick-borne encephalitis virus in patients with neurological disorders in Hokkaido, Japan. Microorganisms 2020;8:1–8. https://doi.org/10.3390/microorganisms8111672 PMID:33126600
  107. Haglund M, Settergren B, Heinz FX, Günther G; ISW-TBE Study Group. Report of the Meningitis Program of the International Scientific Working Group on TBE. Serological screening of patients with viral CNS-infection of unknown etiology in search of undiagnosed TBE cases. Vaccine 2003;21(Suppl 1):S66–72. https://doi.org/10.1016/S0264-410X(02)00816-2 PMID:12628817
  108. Stefanoff P, Zielicka-Hardy A, Hlebowicz M, et al.; TBE Enhanced Surveillance Working Group. New endemic foci of tick-borne encephalitis (TBE) identified in districts where testing for TBE was not available before 2009 in Poland. Parasit Vectors 2013;6:180. https://doi.org/10.1186/1756-3305-6-180 PMID:23777675
  109. Donoso Mantke O, Escadafal C, Niedrig M, Pfeffer M; Working Group For Tick-Borne Encephalitis Virus. Tick-borne encephalitis in Europe, 2007 to 2009. Euro Surveill 2011;16:29. https://doi.org/10.2807/ese.16.39.19976-en PMID:21968423
  110. Beauté J, Spiteri G, Warns-Petit E, Zeller H. Tick-borne encephalitis in Europe, 2012 to 2016. Euro Surveill 2018;23:11. https://doi.org/10.2807/1560-7917.ES.2018.23.45.1800201 PMID:30424829
  111. Schuler M, Zimmermann H, Altpeter E, Heininger U. Epidemiology of tick-borne encephalitis in Switzerland, 2005 to 2011. Euro Surveill 2014;19:20756. https://doi.org/10.2807/1560-7917.ES2014.19.13.20756 PMID:24721541
  112. Zöldi V, Juhász A, Nagy C, Papp Z, Egyed L. Tick-borne encephalitis and Lyme disease in Hungary: the epidemiological situation between 1998 and 2008. Vector Borne Zoonotic Dis 2013;13:256–65. https://doi.org/10.1089/vbz.2011.0905 PMID:23428090
  113. Kuitunen I, Renko M. Changes in the epidemiology of zoonotic infections in children: a nationwide register study in Finland. Pediatr Infect Dis J 2022;41:e113–9. https://doi.org/10.1097/INF.0000000000003440 PMID:34966137
  114. Slunge D, Boman A, Studahl M. Burden of tick-borne encephalitis, Sweden. Emerg Infect Dis 2022;28:314–22. https://doi.org/10.3201/eid2802.204324 PMID:35075997
  115. Grgič-Vitek M, Klavs I. High burden of tick-borne encephalitis in Slovenia—challenge for vaccination policy. Vaccine 2011;29:5178–83. https://doi.org/10.1016/j.vaccine.2011.05.033 PMID:21620916
  116. Nah K, Magpantay FMG, Bede-Fazekas Á, et al. Assessing systemic and non-systemic transmission risk of tick-borne encephalitis virus in Hungary. PLoS One 2019;14:e0217206. https://doi.org/10.1371/journal.pone.0217206 PMID:31163042
  117. Suss J. Tick-borne encephalitis in Europe and beyond—the epidemiological situation as of 2007. Euro Surveill 2008;13:18916. https://doi.org/10.2807/ese.13.26.18916-en PMID:18761916
  118. Stiasny K, Santonja I, Holzmann H, et al. The regional decline and rise of tick-borne encephalitis incidence do not correlate with Lyme borreliosis, Austria, 2005 to 2018. Euro Surveill 2021;26:2002108. https://doi.org/10.2807/1560-7917.ES.2021.26.35.2002108 PMID:34477056
  119. Randolph SE. Tick-borne encephalitis incidence in Central and Eastern Europe: consequences of political transition. Microbes Infect 2008;10:209–16. https://doi.org/10.1016/j.micinf.2007.12.005 PMID:18316221
  120. Godfrey ER, Randolph SE. Economic downturn results in tick-borne disease upsurge. Parasit Vectors 2011;4:35. https://doi.org/10.1186/1756-3305-4-35 PMID:21406086
  121. Randolph SE; EDEN-TBD Sub-Project Team. Human activities predominate in determining changing incidence of tick-borne encephalitis in Europe. Euro Surveill 2010;15:24–31. https://doi.org/10.2807/ese.15.27.19606-en PMID:20630144
  122. Sumilo D, Asokliene L, Bormane A, Vasilenko V, Golovljova I, Randolph SE. Climate change cannot explain the upsurge of tick-borne encephalitis in the Baltics. PLoS One 2007;2:e500. https://doi.org/10.1371/journal.pone.0000500 PMID:17551580
  123. Zeman P, Pazdiora P, Benes C. Spatio-temporal variation of tick-borne encephalitis (TBE) incidence in the Czech Republic: is the current explanation of the disease’s rise satisfactory? Ticks Tick Borne Dis 2010;1:129–40. https://doi.org/10.1016/j.ttbdis.2010.05.003 PMID:21771520
  124. Kunze U; ISW-TBE. Tick-borne encephalitis-still on the map: report of the 18th annual meeting of the International Scientific Working Group on Tick-Borne Encephalitis (ISW-TBE). Ticks Tick Borne Dis 2016;7:911–4. https://doi.org/10.1016/j.ttbdis.2016.04.009 PMID:27189584
  125. Amato-Gauci A, Zeller H. Tick-borne encephalitis joins the diseases under surveillance in the European Union. Euro Surveill 2012;17:20299. https://doi.org/10.2807/ese.17.42.20299-en PMID:23098821
  126. Heinz FX, Stiasny K, Holzmann H, et al. Vaccination and tick-borne encephalitis, central Europe. Emerg Infect Dis 2013;19:69–76. https://doi.org/10.3201/eid1901.120458 PMID:23259984
  127. Jones N, Sperl W, Koch J, Holzmann H, Radauer W. Tick-borne encephalitis in a 17-day-old newborn resulting in severe neurologic impairment. Pediatr Infect Dis J 2007;26:185–6. https://doi.org/10.1097/01.inf.0000253056.34894.5f PMID:17259888
  128. Radzišauskienė D, Žagminas K, Ašoklienė L, et al. Epidemiological patterns of tick-borne encephalitis in Lithuania and clinical features in adults in the light of the high incidence in recent years: a retrospective study. Eur J Neurol 2018;25:268–74. https://doi.org/10.1111/ene.13486 PMID:29053888
  129. Stähelin-Massik J, Zimmermann H, Gnehm HE. Tick-borne encephalitis in Swiss children 2000–2004: five-year nationwide surveillance of epidemiologic characteristics and clinical course. Pediatr Infect Dis J 2008;27:555–7. https://doi.org/10.1097/INF.0b013e318165c195 PMID:18434938
  130. Santonja I, Stiasny K, Essl A, Heinz FX, Kundi M, Holzmann H. Tick-borne encephalitis in vaccinated patients: a retrospective case-control study and analysis of vaccination field effectiveness in Austria from 2000 to 2018. J Infect Dis 2023;227:512–21. https://doi.org/10.1093/infdis/jiac075 PMID:35235953
  131. Logar M, Arnez M, Kolbl J, Avsic-Zupanc T, Strle F. Comparison of the epidemiological and clinical features of tick-borne encephalitis in children and adults. Infection 2000;28:74–7. https://doi.org/10.1007/s150100050050 PMID:10782391
  132. Barp N, Trentini A, Di Nuzzo M, Mondardini V, Francavilla E, Contini C. Clinical and laboratory findings in tick-borne encephalitis virus infection. Parasite Epidemiol Control 2020;10:e00160. https://doi.org/10.1016/j.parepi.2020.e00160 PMID:32637663
  133. Lesnicar G, Poljak M, Seme K, Lesnicar J. Pediatric tick-borne encephalitis in 371 cases from an endemic region in Slovenia, 1959 to 2000. Pediatr Infect Dis J 2003;22:612–7. https://doi.org/10.1097/00006454-200307000-00009 PMID:12867836
  134. Beran J. Tick-borne encephalitis in the Czech Republic. Euro Surveill 2004;8:2493. https://doi.org/10.2807/esw.08.26.20493-3en
  135. Kaiser R. Epidemiology and progress of early summer meningoencephalitis in Baden-Württemberg between 1994 and 1999. A prospective study of 731 patients [German]. Dtsch Med Wochenschr 2000;125:1147–53. https://doi.org/10.1055/s-2000-7668 PMID:11075241
  136. Borde JP, Kaier K, Hehn P, Böhmer MM, Kreusch TM, Dobler G. Tick-borne encephalitis virus infections in Germany. Seasonality and in-year patterns. A retrospective analysis from 2001–2018. PLoS One 2019;14:e0224044. https://doi.org/10.1371/journal.pone.0224044 PMID:31671121
  137. Bogovič P, Lotrič-Furlan S, Avšič-Županc T, Lusa L, Strle F. Factors associated with severity of tick-borne encephalitis: a prospective observational study. Travel Med Infect Dis 2018;26:25–31. https://doi.org/10.1016/j.tmaid.2018.10.003 PMID:30296483
  138. Furness RW, Furness EN. Ixodes ricinus parasitism of birds increases at higher winter temperatures. J Vector Ecol 2018;43:59–62. https://doi.org/10.1111/jvec.12283 PMID:29757511
  139. Gustafson R, Svenungsson B, Forsgren M, Gardulf A, Granström M. Two-year survey of the incidence of Lyme borreliosis and tick-borne encephalitis in a high-risk population in Sweden. Eur J Clin Microbiol Infect Dis 1992;11:894–900. https://doi.org/10.1007/BF01962369 PMID:1486884
  140. Leonova GN, Belikov SI, Kondratov IG, Takashima I. Comprehensive assessment of the genetics and virulence of tick-borne encephalitis virus strains isolated from patients with inapparent and clinical forms of the infection in the Russian Far East. Virology 2013;443:89–98. https://doi.org/10.1016/j.virol.2013.04.029 PMID:23735441
  141. Henningsson AJ, Lindqvist R, Norberg P, et al. Human tick-borne encephalitis and characterization of virus from biting tick. Emerg Infect Dis 2016;22:1485–7. https://doi.org/10.3201/eid2208.151962 PMID:27434395
  142. Brockmann SO, Oehme R, Buckenmaier T, et al. A cluster of two human cases of tick-borne encephalitis (TBE) transmitted by unpasteurised goat milk and cheese in Germany, May 2016. Euro Surveill 2018;23:17–00336. https://doi.org/10.2807/1560-7917.ES.2018.23.15.17-00336 PMID:29667575
  143. Kunze U. The International Scientific Working Group on Tick-Borne Encephalitis (ISW TBE): review of 17 years of activity and commitment. Ticks Tick Borne Dis 2016;7:399–404. https://doi.org/10.1016/j.ttbdis.2015.12.018 PMID:26795231
  144. Bogovič P, Lotrič-Furlan S, Avšič-Županc T, Lusa L, Strle F. Factors associated with severity of tick-borne encephalitis: a prospective observational study. Travel Med Infect Dis 2018;26:25–31. https://doi.org/10.1016/j.tmaid.2018.10.003 PMID:30296483
  145. Logar M, Bogovic P, Cerar D, Avsic-Zupanc T, Strle F. Tick-borne encephalitis in Slovenia from 2000 to 2004: comparison of the course in adult and elderly patients. Wien Klin Wochenschr 2006;118:702–7. https://doi.org/10.1007/s00508-006-0699-6 PMID:17160611
  146. Krawczuk K, Czupryna P, Pancewicz S, Ołdak E, Moniuszko-Malinowska A. Comparison of tick-borne encephalitis between children and adults—analysis of 669 patients. J Neurovirol 2020;26:565–71. https://doi.org/10.1007/s13365-020-00856-x PMID:32524423
  147. Lenhard T, Ott D, Jakob NJ, et al. Predictors, neuroimaging characteristics and long-term outcome of severe European tick-borne encephalitis: a prospective cohort study. PLoS One 2016;11:e0154143. https://doi.org/10.1371/journal.pone.0154143 PMID:27111657
  148. Fritsch P, Gruber-Sedlmayr U, Pansi H, et al. Tick-borne encephalitis in Styrian children from 1981 to 2005: a retrospective study and a review of the literature. Acta Paediatr 2008;97:535–8. https://doi.org/10.1111/j.1651-2227.2008.00763.x PMID:18394095
  149. Bogovič P, Kastrin A, Lotrič-Furlan S, et al. Clinical and laboratory characteristics and outcome of illness caused by tick-borne encephalitis virus without central nervous system involvement. Emerg Infect Dis 2022;28:291–301. https://doi.org/10.3201/eid2802.211661 PMID:35075993
  150. Gritsun TS, Frolova TV, Zhankov AI, et al. Characterization of a Siberian virus isolated from a patient with progressive chronic tick-borne encephalitis. J Virol 2003;77:25–36. https://doi.org/10.1128/JVI.77.1.25-36.2003 PMID:12477807
  151. Gritsun TS, Nuttall PA, Gould EA. Tick-borne flaviviruses. Adv Virus Res 2003;61:317–71. https://doi.org/10.1016/S0065-3527(03)61008-0 PMID:14714436
  152. Volok VP, Gmyl LV, Belyaletdinova IK, Karganova GG, Dekonenko EP. Progressive course of chronic tick-borne encephalitis manifesting as amyotrophic lateral sclerosis-like syndrome 35 years after the acute disease. Brain Sci 2022;12:1020. https://doi.org/10.3390/brainsci12081020 PMID:36009082
  153. Ruzek D, Avšič Županc T, Borde J, et al. Tick-borne encephalitis in Europe and Russia: review of pathogenesis, clinical features, therapy, and vaccines. Antiviral Res 2019;164:23–51. https://doi.org/10.1016/j.antiviral.2019.01.014 PMID:30710567
  154. Lotric-Furlan S, Strle F. Thrombocytopenia—a common finding in the initial phase of tick-borne encephalitis. Infection 1995;23:203–6. https://doi.org/10.1007/BF01781197 PMID:8522376
  155. Günther G, Haglund M, Lindquist L, Forsgren M, Sköldenberg B. Tick-bone encephalitis in Sweden in relation to aseptic meningo-encephalitis of other etiology: a prospective study of clinical course and outcome. J Neurol 1997;244:230–8. https://doi.org/10.1007/s004150050077 PMID:9112591
  156. Haglund M, Günther G. Tick-borne encephalitis—pathogenesis, clinical course and long-term follow-up. Vaccine 2003;21(Suppl 1):S11–8. https://doi.org/10.1016/S0264-410X(02)00811-3 PMID:12628810
  157. Hansson ME, Orvell C, Engman ML, et al. Tick-borne encephalitis in childhood: rare or missed? Pediatr Infect Dis J 2011;30:355–7. https://doi.org/10.1097/INF.0b013e3181fe3b5a PMID:21412206
  158. Steininger PA, Bobinger T, Dietrich W, et al. Two cases of severe tick-borne encephalitis in rituximab-treated patients in Germany: implications for diagnosis and prevention. Open Forum Infect Dis 2017;4:ofx204. https://doi.org/10.1093/ofid/ofx204 PMID:29670929
  159. Chmelík V, Chrdle A, Růžek D. Fatal tick-borne encephalitis in an immunosuppressed 12-year-old patient. J Clin Virol 2016;74:73–4. https://doi.org/10.1016/j.jcv.2015.11.029 PMID:26679831
  160. Knight A, Pauksens K, Nordmark G, Kumlien E. Fatal outcome of tick-borne encephalitis in two patients with rheumatic disease treated with rituximab. Rheumatology (Oxford) 2017;56:855–6. https://doi.org/10.1093/rheumatology/kew495 PMID:28130421
  161. Bogovič P, Lotrič-Furlan S, Avšič-Županc T, et al. Comparison of clinical, laboratory and immune characteristics of the monophasic and biphasic course of tick-borne encephalitis. Microorganisms 2021;9:796. https://doi.org/10.3390/microorganisms9040796 PMID:33920166
  162. Belikov SI, Kondratov IG, Potapova UV, Leonova GN. The relationship between the structure of the tick-borne encephalitis virus strains and their pathogenic properties. PLoS One 2014;9:e94946. https://doi.org/10.1371/journal.pone.0094946 PMID:24740396
  163. Grascenkov NI. Tick-borne encephalitis in the USSR. Bull World Health Organ 1964;30:187–96. PMID:14153408
  164. Divé I, Veje M, Dobler G, et al. Tick-borne encephalitis virus (TBEV) infection in pregnancy: absence of virus transmission to the fetuses despite severe maternal disease—a case study. Ticks Tick Borne Dis 2020;11:101491. https://doi.org/10.1016/j.ttbdis.2020.101491 PMID:32723645
  165. Weinmayr LM, Kanz D, Eckenweiler M, et al. Acute tick-borne encephalitis during pregnancy—an alarming combination. Ticks Tick Borne Dis 2020;11:101512. https://doi.org/10.1016/j.ttbdis.2020.101512 PMID:32993932
  166. Bjonholm E, Soderholm S, Stephansson O, Askling HH. Tick-borne encephalitis in pregnant women: a mini narrative review. New Microbes New Infect 2022;48:101017. https://doi.org/10.1016/j.nmni.2022.101017 PMID:36176540
  167. Dobler G, Stoma I. TBE in special clinical situations [Chapter 7]. In: Dobler G, Erber W, Bröker M, Schmitt H, eds. The TBE book. 4th ed. Singapore: Global Health Press; 2021:98–104.
  168. Velay A, Janssen-Langenstein R, Kremer S, et al. Tick-borne encephalitis in pregnant woman and long-term sequelae. Emerg Infect Dis 2023;29:669–71. https://doi.org/10.3201/eid2903.221328 PMID:36823716
  169. Taba P, Schmutzhard E, Forsberg P, et al. EAN consensus review on prevention, diagnosis and management of tick-borne encephalitis. Eur J Neurol 2017;24:1214-e61. https://doi.org/10.1111/ene.13356 PMID:28762591
  170. Pichler A, Sellner J, Harutyunyan G, et al. Magnetic resonance imaging and clinical findings in adults with tick-borne encephalitis. J Neurol Sci 2017;375:266–9. https://doi.org/10.1016/j.jns.2017.02.003 PMID:28320144
  171. Horger M, Beck R, Fenchel M, et al. Imaging findings in tick-borne encephalitis with differential diagnostic considerations. AJR Am J Roentgenol 2012;199:420–7. https://doi.org/10.2214/AJR.11.7911 PMID:22826407
  172. von Stülpnagel C, Winkler P, Koch J, et al. MRI-imaging and clinical findings of eleven children with tick-borne encephalitis and review of the literature. Eur J Paediatr Neurol 2016;20:45–52. https://doi.org/10.1016/j.ejpn.2015.10.008 PMID:26586340
  173. Schmolck H, Maritz E, Kletzin I, Korinthenberg R. Neurologic, neuropsychologic, and electroencephalographic findings after European tick-borne encephalitis in children. J Child Neurol 2005;20:500–8. https://doi.org/10.1177/088307380502000606 PMID:15996399
  174. Marjelund S, Tikkakoski T, Tuisku S, Räisänen S. Magnetic resonance imaging findings and outcome in severe tick-borne encephalitis. Report of four cases and review of the literature. Acta Radiol 2004;45:88–94. https://doi.org/10.1080/02841850410003356 PMID:15164786
  175. Marjelund S, Jaaskelainen A, Tikkakoski T, Tuisku S, Vapalahti O. Gadolinium enhancement of cauda equina: a new MR imaging finding in the radiculitic form of tick-borne encephalitis. AJNR Am J Neuroradiol 2006;27:995–7. PMID:16687530
  176. Bender A, Schulte-Altedorneburg G, Walther EU, Pfister HW. Severe tick borne encephalitis with simultaneous brain stem, bithalamic, and spinal cord involvement documented by MRI. J Neurol Neurosurg Psychiatry 2005;76:135–7. https://doi.org/10.1136/jnnp.2004.040469 PMID:15608016
  177. Beer S, Brune N, Kesselring J. Detection of anterior horn lesions by MRI in central European tick-borne encephalomyelitis. J Neurol 1999;246:1169–71. https://doi.org/10.1007/s004150050537 PMID:10653310
  178. Stich O, Reinhard M, Rauer S. MRI scans of cervical cord provide evidence of anterior horn lesion in a patient with tick-borne encephalomyeloradiculitis. Eur J Neurol 2007;14:e5–6. https://doi.org/10.1111/j.1468-1331.2007.01774.x PMID:17539929
  179. Cruciatti B, Beltrame A, Ruscio M, Viale P, Gigli GL. Neurological manifestation of tick-borne encephalitis in North-Eastern Italy. Neurol Sci 2006;27:122–4. https://doi.org/10.1007/s10072-006-0612-0 PMID:16816910
  180. Hills SL, Broussard KR, Broyhill JC, et al. Tick-borne encephalitis among US travellers, 2010–20. J Travel Med 2022;29:taab167. https://doi.org/10.1093/jtm/taab167 PMID:34741518
  181. Günther G, Haglund M, Lindquist L, Sköldenberg B, Forsgren M. Intrathecal IgM, IgA and IgG antibody response in tick-borne encephalitis. Long-term follow-up related to clinical course and outcome. Clin Diagn Virol 1997;8:17–29. https://doi.org/10.1016/S0928-0197(97)00273-0 PMID:9248655
  182. Hofmann H, Kunz C, Heinz FX, Dippe H. Detectability of IgM antibodies against TBE virus after natural infection and after vaccination. Infection 1983;11:164–6. https://doi.org/10.1007/BF01641297 PMID:6309676
  183. Mohareb E, Christova I, Soliman A, Younan R, Kantardjiev T. Tick-borne encephalitis in Bulgaria, 2009 to 2012. Euro Surveill 2013;18:20635. https://doi.org/10.2807/1560-7917.ES2013.18.46.20635 PMID:24256889
  184. Saksida A, Duh D, Lotric-Furlan S, Strle F, Petrovec M, Avsic-Zupanc T. The importance of tick-borne encephalitis virus RNA detection for early differential diagnosis of tick-borne encephalitis. J Clin Virol 2005;33:331–5. https://doi.org/10.1016/j.jcv.2004.07.014 PMID:15919235
  185. Veje M, Studahl M, Norberg P, et al. Detection of tick-borne encephalitis virus RNA in urine. J Clin Microbiol 2014;52:4111–2. https://doi.org/10.1128/JCM.02428-14 PMID:25165082
  186. Nagy A, Nagy O, Tarcsai K, Farkas Á, Takács M. First detection of tick-borne encephalitis virus RNA in clinical specimens of acutely ill patients in Hungary. Ticks Tick Borne Dis 2018;9:485–9. https://doi.org/10.1016/j.ttbdis.2017.12.017 PMID:29373305
  187. Haglund M, Vene S, Forsgren M, et al. Characterisation of human tick-borne encephalitis virus from Sweden. J Med Virol 2003;71:610–21. https://doi.org/10.1002/jmv.10497 PMID:14556277
  188. Kapadia RK, Staples JE, Gill CM, et al. Severe arboviral neuroinvasive disease in patients on rituximab therapy: a review. Clin Infect Dis 2023;76:1142–8. https://doi.org/10.1093/cid/ciac766 PMID:36103602
  189. Caracciolo I, Bassetti M, Paladini G, et al. Persistent viremia and urine shedding of tick-borne encephalitis virus in an infected immunosuppressed patient from a new epidemic cluster in North-Eastern Italy. J Clin Virol 2015;69:48–51. https://doi.org/10.1016/j.jcv.2015.05.019 PMID:26209378
  190. Waldvogel K, Bossart W, Huisman T, Boltshauser E, Nadal D. Severe tick-borne encephalitis following passive immunization. Eur J Pediatr 1996;155:775–9. https://doi.org/10.1007/BF02002905 PMID:8874110
  191. Valdueza JM, Weber JR, Harms L, Bock A. Severe tick borne encephalomyelitis after tick bite and passive immunisation. J Neurol Neurosurg Psychiatry 1996;60:593–4. https://doi.org/10.1136/jnnp.60.5.593 PMID:8778279
  192. Kluger G, Schöttler A, Waldvogel K, et al. Tick-borne encephalitis despite specific immunoglobulin prophylaxis. Lancet 1995;346:1502. https://doi.org/10.1016/S0140-6736(95)92527-9 PMID:7491037
  193. Bröker M, Kollaritsch H. After a tick bite in a tick-borne encephalitis virus endemic area: current positions about post-exposure treatment. Vaccine 2008;26:863–8. https://doi.org/10.1016/j.vaccine.2007.11.046 PMID:18206274
  194. Czupryna P, Grygorczuk S, Krawczuk K, et al. Sequelae of tick-borne encephalitis in retrospective analysis of 1072 patients. Epidemiol Infect 2018;146:1663–70. https://doi.org/10.1017/S0950268818002005 PMID:30047354
  195. Wahlberg P, Carlsson SA, Granlund H, et al. TBE in Aland Islands 1959–2005: Kumlinge disease. Scand J Infect Dis 2006;38:1057–62. https://doi.org/10.1080/00365540600868297 PMID:17148077
  196. Haglund M, Forsgren M, Lindh G, Lindquist L. A 10-year follow-up study of tick-borne encephalitis in the Stockholm area and a review of the literature: need for a vaccination strategy. Scand J Infect Dis 1996;28:217–24. https://doi.org/10.3109/00365549609027160 PMID:8863349
  197. Silber LA, Soloviev VD. Far Eastern tick-borne spring-summer (spring) encephalitis. Am Rev Sov Med 1946;(Suppl):1–80. PMID:21002377
  198. Xing Y, Schmitt HJ, Arguedas A, Yang J. Tick-borne encephalitis in China: a review of epidemiology and vaccines. Vaccine 2017;35:1227–37. https://doi.org/10.1016/j.vaccine.2017.01.015 PMID:28153343
  199. Veje M, Nolskog P, Petzold M, et al. Tick-borne encephalitis sequelae at long-term follow-up: a self-reported case-control study. Acta Neurol Scand 2016;134:434–41. https://doi.org/10.1111/ane.12561 PMID:26810689
  200. Bogovič P, Stupica D, Rojko T, et al. The long-term outcome of tick-borne encephalitis in Central Europe. Ticks Tick Borne Dis 2018;9:369–78. https://doi.org/10.1016/j.ttbdis.2017.12.001 PMID:29275872
  201. Varnaitė R, Gredmark-Russ S, Klingström J. Deaths from tick-borne encephalitis, Sweden. Emerg Infect Dis 2022;28:1471–4. https://doi.org/10.3201/eid2807.220010 PMID:35731187
  202. Jelenik Z, Keller M, Briggs B, et al. Tick-borne encephalitis and golden agers: position paper of the International Scientific Working Group on Tick-Borne Encephalitis (ISW-TBE). Wien Med Wochenschr 2010;160:247–51. https://doi.org/10.1007/s10354-010-0758-5 PMID:20632153
  203. Steffen R. Tick-borne encephalitis (TBE) in children in Europe: epidemiology, clinical outcome and comparison of vaccination recommendations. Ticks Tick Borne Dis 2019;10:100–10. https://doi.org/10.1016/j.ttbdis.2018.08.003 PMID:30241699
  204. Engman ML, Lindström K, Sallamba M, et al. One-year follow-up of tick-borne central nervous system infections in childhood. Pediatr Infect Dis J 2012;31:570–4. https://doi.org/10.1097/INF.0b013e31824f23c0 PMID:22333696
  205. Cruse RP, Rothner AD, Erenberg G, Calisher CH. Central European tick-borne encephalitis: an Ohio case with a history of foreign travel. Am J Dis Child 1979;133:1070–1. PMID:495602
  206. CDC. Tick-borne encephalitis among U.S. travelers to Europe and Asia—2000–2009. MMWR Morb Mortal Wkly Rep 2010;59:335–8. PMID:20339345
  207. International Trade Administration. U.S. outbound travel. Washington, DC: US Department of Commerce, International Trade Administration, National Travel and Tourism Office. https://www.trade.gov/travel-and-tourism-research
  208. Mancuso JD, Bazaco S, Stahlman S, Clausen SS, Cost AA. Tick-borne encephalitis surveillance in U.S. military service members and beneficiaries, 2006–2018. MSMR 2019;26:4–10. PMID:31804845
  209. Mease LE, Maddox SA, Noss MR, Whitman S. Case report: tick-borne encephalitis virus infection in beneficiaries of the U.S. military healthcare system in southern Germany. MSMR 2019;26:12–5. PMID:31804846
  210. Stahlman S. Surveillance snapshot: tick-borne encephalitis in Military Health System beneficiaries, 2012–2021. MSMR 2022;29:23. PMID:36250851
  211. European Centre for Disease Prevention and Control. Annual epidemiological reports (AERs). Stockholm, Sweden: European Centre for Disease Prevention and Control. https://www.ecdc.europa.eu/en/publications-data/monitoring/all-annual-epidemiological-reports
  212. Steffen R. Epidemiology of tick-borne encephalitis (TBE) in international travellers to Western/Central Europe and conclusions on vaccination recommendations. J Travel Med 2016;23:taw018. PMID:27087558
  213. Aendekerk RP, Schrivers AN, Koehler PJ. Tick-borne encephalitis complicated by a polio-like syndrome following a holiday in central Europe. Clin Neurol Neurosurg 1996;98:262–4. https://doi.org/10.1016/0303-8467(96)00030-3 PMID:8884102
  214. Reusken C, Reimerink J, Verduin C, Sabbe L, Cleton N, Koopmans M. Case report: tick-borne encephalitis in two Dutch travellers returning from Austria, Netherlands, July and August 2011. Euro Surveill 2011;16:20003. https://doi.org/10.2807/ese.16.44.20003-en PMID:22085619
  215. Pontali E, Bobbio N, Cassola G. Tick-borne encephalitis (TBE) during a “grand-tour” of Europe clinically manifesting in a TBE-free region. J Travel Med 2010;17:143–5. https://doi.org/10.1111/j.1708-8305.2009.00383.x PMID:20412185
  216. Camprubí D, Moreno-García E, Almuedo-Riera A, et al. First imported case of tick-borne encephalitis in Spain—was it alimentary? Travel Med Infect Dis 2020;37:101701. https://doi.org/10.1016/j.tmaid.2020.101701 PMID:32339672
  217. CDC. Biosafety in microbiological and biomedical laboratories. 6th ed. Atlanta, GA: US Department of Health and Human Services, CDC; 2020. https://www.cdc.gov/labs/BMBL.html
  218. Ahmed F. Advisory Committee on Immunization Practices (ACIP) handbook for developing evidence-based recommendations. Version 1.2. Atlanta, GA: US Department of Health and Human Services, CDC; 2013. https://www.cdc.gov/vaccines/acip/recs/grade/downloads/handbook.pdf
  219. CDC. Grading of recommendations, assessment, development, and evaluation for tick-borne encephalitis (TBE) vaccine. Atlanta, GA: US Department of Health and Human Services, CDC; 2022. https://www.cdc.gov/vaccines/acip/recs/grade/table-refs.html
  220. CDC. ACIP evidence to recommendations framework. Atlanta, GA: US Department of Health and Human Services, CDC. https://www.cdc.gov/vaccines/acip/recs/grade/about-grade.html
  221. CDC. ACIP evidence to recommendations framework: tick-borne encephalitis (TBE) vaccination for persons who travel abroad. Atlanta, GA: US Department of Health and Human Services, CDC; 2022. https://www.cdc.gov/vaccines/acip/recs/grade/etr.html
  222. CDC. ACIP evidence to recommendations framework: tick-borne encephalitis (TBE) vaccination for laboratory workers. Atlanta, GA: US Department of Health and Human Services, CDC; 2022. https://www.cdc.gov/vaccines/acip/recs/grade/etr.html
  223. Hills S. Tick-borne encephalitis (TBE) vaccine. Presentation to the Advisory Committee on Immunization Practices. Atlanta, GA: February 23, 2022. https://stacks.cdc.gov/view/cdc/114828
  224. TICOVAC [Package Insert]. New York, NY: Pfizer; 2021. https://www.fda.gov/vaccines-blood-biologics/ticovac
  225. Heinz FX, Holzmann H, Essl A, Kundi M. Field effectiveness of vaccination against tick-borne encephalitis. Vaccine 2007;25:7559–67. https://doi.org/10.1016/j.vaccine.2007.08.024 PMID:17869389
  226. Erber W, Khan F, Zavadska D, et al. Effectiveness of TBE vaccination in southern Germany and Latvia. Vaccine 2022;40:819–25. https://doi.org/10.1016/j.vaccine.2021.12.028 PMID:34952753
  227. Pugh SJ, Moïsi JC, Kundi M, et al. Effectiveness of two doses of tick-borne encephalitis (TBE) vaccine. J Travel Med 2022;29:taab193. https://doi.org/10.1093/jtm/taab193 PMID:34999897
  228. Nygren TM, Pilic A, Böhmer MM, et al. Tick-borne encephalitis vaccine effectiveness and barriers to vaccination in Germany. Sci Rep 2022;12:11706. https://doi.org/10.1038/s41598-022-15447-5 PMID:35810184
  229. Baroutsou V, Zens KD, Sinniger P, Fehr J, Lang P. Analysis of tick-borne encephalitis vaccination coverage and compliance in adults in Switzerland, 2018. Vaccine 2020;38:7825–33. https://doi.org/10.1016/j.vaccine.2020.10.022 PMID:33164805
  230. Kunz C. TBE vaccination and the Austrian experience. Vaccine 2003;21(Suppl 1):S50–5. https://doi.org/10.1016/S0264-410X(02)00813-7 PMID:12628814
  231. Kreil TR, Maier E, Fraiss S, Eibl MM. Neutralizing antibodies protect against lethal flavivirus challenge but allow for the development of active humoral immunity to a nonstructural virus protein. J Virol 1998;72:3076–81. https://doi.org/10.1128/JVI.72.4.3076-3081.1998 PMID:9525632
  232. WHO Publication. Vaccines against tick-borne encephalitis: WHO position paper—recommendations. Vaccine 2011;29:8769–70. https://doi.org/10.1016/j.vaccine.2011.07.024 PMID:21777636
  233. Hayasaka D, Goto A, Yoshii K, Mizutani T, Kariwa H, Takashima I. Evaluation of European tick-borne encephalitis virus vaccine against recent Siberian and far-eastern subtype strains. Vaccine 2001;19:4774–9. https://doi.org/10.1016/S0264-410X(01)00218-3 PMID:11535329
  234. Chiba N, Osada M, Komoro K, Mizutani T, Kariwa H, Takashima I. Protection against tick-borne encephalitis virus isolated in Japan by active and passive immunization. Vaccine 1999;17:1532–9. https://doi.org/10.1016/S0264-410X(98)00360-0 PMID:10195790
  235. Holzmann H, Vorobyova MS, Ladyzhenskaya IP, et al. Molecular epidemiology of tick-borne encephalitis virus: cross-protection between European and Far Eastern subtypes. Vaccine 1992;10:345–9. https://doi.org/10.1016/0264-410X(92)90376-U PMID:1574920
  236. Orlinger KK, Hofmeister Y, Fritz R, et al. A tick-borne encephalitis virus vaccine based on the European prototype strain induces broadly reactive cross-neutralizing antibodies in humans. J Infect Dis 2011;203:1556–64. https://doi.org/10.1093/infdis/jir122 PMID:21592984
  237. Fritz R, Orlinger KK, Hofmeister Y, et al. Quantitative comparison of the cross-protection induced by tick-borne encephalitis virus vaccines based on European and Far Eastern virus subtypes. Vaccine 2012;30:1165–9. https://doi.org/10.1016/j.vaccine.2011.12.013 PMID:22178103
  238. Morozova OV, Bakhvalova VN, Potapova OF, et al. Evaluation of immune response and protective effect of four vaccines against the tick-borne encephalitis virus. Vaccine 2014;32:3101–6. https://doi.org/10.1016/j.vaccine.2014.02.046 PMID:24631082
  239. McAuley AJ, Sawatsky B, Ksiazek T, et al. Cross-neutralisation of viruses of the tick-borne encephalitis complex following tick-borne encephalitis vaccination and/or infection. NPJ Vaccines 2017;2:5. https://doi.org/10.1038/s41541-017-0009-5 PMID:29263866
  240. Chernokhaeva LL, Rogova YV, Vorovitch MF, et al. Protective immunity spectrum induced by immunization with a vaccine from the TBEV strain Sofjin. Vaccine 2016;34:2354–61. https://doi.org/10.1016/j.vaccine.2016.03.041 PMID:27013433
  241. Süss J, Schrader C, Abel U, Bormane A, Duks A, Kalnina V. Characterization of tick-borne encephalitis (TBE) foci in Germany and Latvia (1997–2000). Int J Med Microbiol 2002;291(Suppl 33):34–42. https://doi.org/10.1016/S1438-4221(02)80007-8 PMID:12141755
  242. Karelis G, Bormane A, Logina I, et al. Tick-borne encephalitis in Latvia 1973–2009: epidemiology, clinical features and sequelae. Eur J Neurol 2012;19:62–8. https://doi.org/10.1111/j.1468-1331.2011.03434.x PMID:21615626
  243. Jenkins VA, Hoet B. Considering the market share of vaccines against tick-borne encephalitis reported in Austria. J Travel Med 2022;29:taac027. https://doi.org/10.1093/jtm/taac027 PMID:35234876
  244. Loew-Baselli A, Konior R, Pavlova BG, et al.; FSME-IMMUN study group. Safety and immunogenicity of the modified adult tick-borne encephalitis vaccine FSME-IMMUN: results of two large phase 3 clinical studies. Vaccine 2006;24:5256–63. https://doi.org/10.1016/j.vaccine.2006.03.061 PMID:16624457
  245. Food and Drug Administration. Clinical review memo—TICOVAC. Silver Spring, MD: US Department of Health and Human Services, Food and Drug Administration; 2021. https://www.fda.gov/media/152024/download?attachment
  246. Loew-Baselli A, Poellabauer EM, Pavlova BG, et al. Seropersistence of tick-borne encephalitis antibodies, safety and booster response to FSME-IMMUN 0.5 ml in adults aged 18–67 years. Hum Vaccin 2009;5:551–6. https://doi.org/10.4161/hv.5.8.8571 PMID:19430202
  247. Pöllabauer EM, Fritsch S, Pavlova BG, et al. Clinical evaluation to determine the appropriate paediatric formulation of a tick-borne encephalitis vaccine. Vaccine 2010;28:4558–65. https://doi.org/10.1016/j.vaccine.2010.04.075 PMID:20452432
  248. Poellabauer E, Angermayr R, Behre U, et al. Seropersistence and booster response following vaccination with FSME-IMMUN in children, adolescents, and young adults. Vaccine 2019;37:3241–50. https://doi.org/10.1016/j.vaccine.2019.03.032 PMID:30928173
  249. Wanke K, Von Braun A, Haberli L, et al. Immunogenicity and safety of tick-borne encephalitis vaccination in healthy elderly individuals [Abstract P1048]. Clin Microbiol Infect 2012;18(Suppl 3):246. https://doi.org/10.1111/j.1469-0691.2012.03802.x
  250. Konior R, Brzostek J, Poellabauer EM, Jiang Q, Harper L, Erber W. Seropersistence of TBE virus antibodies 10 years after first booster vaccination and response to a second booster vaccination with FSME-IMMUN 0.5mL in adults. Vaccine 2017;35:3607–13. https://doi.org/10.1016/j.vaccine.2017.03.059 PMID:28545923
  251. Hills S. Tick-borne encephalitis (TBE) vaccine: immunogenicity and safety. Presentation to the Advisory Committee on Immunization Practices. Atlanta, GA, August 29, 2021. https://www.cdc.gov/vaccines/acip/meetings/index.html
  252. Weinberger B, Keller M, Fischer KH, et al. Decreased antibody titers and booster responses in tick-borne encephalitis vaccinees aged 50–90 years. Vaccine 2010;28:3511–5. https://doi.org/10.1016/j.vaccine.2010.03.024 PMID:20332047
  253. Hainz U, Jenewein B, Asch E, Pfeiffer KP, Berger P, Grubeck-Loebenstein B. Insufficient protection for healthy elderly adults by tetanus and TBE vaccines. Vaccine 2005;23:3232–5. https://doi.org/10.1016/j.vaccine.2005.01.085 PMID:15837226
  254. Hombach J, Barrett A, Kollaritsch H. Tick-borne encephalitis vaccines. In: Plotkin SA, Orenstein WA, Offit PA, Edwards KM, eds. Plotkin’s vaccines. 7th ed. Philadelphia, PA: Elsevier; 2018.
  255. Steffen R, Erber W, Schmitt HJ. Can the booster interval for the tick-borne encephalitis (TBE) vaccine ‘FSME-IMMUN’ be prolonged?—a systematic review. Ticks Tick Borne Dis 2021;12:101779. https://doi.org/10.1016/j.ttbdis.2021.101779 PMID:34298356
  256. Loew-Baselli A, Poellabauer E, Fritsch S, et al. Immunogenicity and safety of FSME-IMMUN 0.5 ml using a rapid immunization schedule. Int J Med Microbiol 2006;296:213–4. https://doi.org/10.1016/j.ijmm.2006.01.019
  257. Harrison N, Grabmeier-Pfistershammer K, Graf A, et al. Humoral immune response to tick-borne encephalitis vaccination in allogeneic blood and marrow graft recipients. NPJ Vaccines 2020;5:67. https://doi.org/10.1038/s41541-020-00215-1 PMID:32728481
  258. Jílková E, Vejvalková P, Stiborová I, Skorkovský J, Král V. Serological response to tick-borne encephalitis (TBE) vaccination in the elderly—results from an observational study. Expert Opin Biol Ther 2009;9:797–803. https://doi.org/10.1517/14712590903066711 PMID:19527104
  259. Kantele A, Rombo L, Vene S, Kundi M, Lindquist L, Erra EO. Three-dose versus four-dose primary schedules for tick-borne encephalitis (TBE) vaccine FSME-immun for those aged 50 years or older: a single-centre, open-label, randomized controlled trial. Vaccine 2022;40:1299–305. https://doi.org/10.1016/j.vaccine.2022.01.022 PMID:35101266
  260. Pöllabauer EM, Pavlova BG, Löw-Baselli A, et al. Comparison of immunogenicity and safety between two paediatric TBE vaccines. Vaccine 2010;28:4680–5. https://doi.org/10.1016/j.vaccine.2010.04.047 PMID:20433803
  261. Prymula R, Pöllabauer EM, Pavlova BG, et al. Antibody persistence after two vaccinations with either FSME-IMMUN® Junior or ENCEPUR® Children followed by third vaccination with FSME-IMMUN® Junior. Hum Vaccin Immunother 2012;8:736–42. https://doi.org/10.4161/hv.20058 PMID:22699436
  262. Wittermann C, Schöndorf I, Gniel D. Antibody response following administration of two paediatric tick-borne encephalitis vaccines using two different vaccination schedules. Vaccine 2009;27:1661–6. https://doi.org/10.1016/j.vaccine.2008.10.003 PMID:18940221
  263. Chitimia-Dobler L, Lindau A, Oehme R, et al. Tick-borne encephalitis vaccination protects from alimentary TBE infection: results from an alimentary outbreak. Microorganisms 2021;9:889. https://doi.org/10.3390/microorganisms9050889 PMID:33919318
  264. Sandberg JT, Löfling M, Varnaitė R, et al. Safety and immunogenicity following co-administration of yellow fever vaccine with tick-borne encephalitis or Japanese encephalitis vaccines: results from an open label, non-randomized clinical trial. PLoS Negl Trop Dis 2023;17:e0010616. https://doi.org/10.1371/journal.pntd.0010616 PMID:36758067
  265. Rampa JE, Askling HH, Lang P, et al. Immunogenicity and safety of the tick-borne encephalitis vaccination (2009–2019): a systematic review. Travel Med Infect Dis 2020;37:101876. https://doi.org/10.1016/j.tmaid.2020.101876 PMID:32931931
  266. Pavlova BG, Loew-Baselli A, Fritsch S, et al. Tolerability of modified tick-borne encephalitis vaccine FSME-IMMUN “NEW” in children: results of post-marketing surveillance. Vaccine 2003;21:742–5. https://doi.org/10.1016/S0264-410X(02)00593-5 PMID:12531353
  267. Bühler T, Boos N, Leuppi-Taegtmeyer AB, Berger CT. Febrile illness and bicytopenia within hours after tick-borne encephalitis booster vaccination. NPJ Vaccines 2019;4:52. https://doi.org/10.1038/s41541-019-0152-2 PMID:31871772
  268. Hidalgo H, Kallweit U, Mathis J, Bassetti CL. Post tick-borne encephalitis virus vaccination narcolepsy with cataplexy. Sleep 2016;39:1811–4. https://doi.org/10.5665/sleep.6160 PMID:27397572
  269. Maltezou HC, Effraimidou E, Cassimos DC, et al. Vaccination programs for pregnant women in Europe, 2021. Vaccine 2021;39:6137–43. https://doi.org/10.1016/j.vaccine.2021.08.074 PMID:34462162
  270. Eder G, Kollaritsch H. Antigen dependent adverse reactions and seroconversion of a tick-borne encephalitis vaccine in children. Vaccine 2003;21:3575–83. https://doi.org/10.1016/S0264-410X(03)00422-5 PMID:12922085
  271. Kroger A, Bahta L, Hunter P. General best practice guidelines for immunization: best practices guidance of the Advisory Committee on Immunization Practices (ACIP). Atlanta, GA: US Department of Health and Human Services, CDC. https://www.cdc.gov/vaccines/hcp/acip-recs/general-recs/special-situations.html
  272. Anderson PO. Maternal vaccination and breastfeeding. Breastfeed Med 2019;14:215–7. https://doi.org/10.1089/bfm.2019.0045 PMID:30888205
  273. Shedrawy J, Henriksson M, Hergens MP, Askling HH. Estimating costs and health outcomes of publicly funded tick-born encephalitis vaccination: a cost-effectiveness analysis. Vaccine 2018;36:7659–65. https://doi.org/10.1016/j.vaccine.2018.10.086 PMID:30385058
  274. Desjeux G, Galoisy-Guibal L, Colin C. Cost-benefit analysis of vaccination against tick-borne encephalitis among French troops. PharmacoEconomics 2005;23:913–26. https://doi.org/10.2165/00019053-200523090-00004 PMID:16153134
  275. Šmit R, Postma MJ. Vaccines for tick-borne diseases and cost-effectiveness of vaccination: a public health challenge to reduce the diseases’ burden. Expert Rev Vaccines 2016;15:5–7. https://doi.org/10.1586/14760584.2016.1111142 PMID:26559456
  276. Prelog M, Wilk C, Keller M, et al. Diminished response to tick-borne encephalitis vaccination in thymectomized children. Vaccine 2008;26:595–600. https://doi.org/10.1016/j.vaccine.2007.11.074 PMID:18178293
  277. Panasiuk B, Prokopowicz D, Panasiuk A. Immunological response in HIV-positive patients vaccinated against tick-borne encephalitis. Infection 2003;31:45–6. https://doi.org/10.1007/s15010-002-2020-6 PMID:12590332
  278. Zielinski CC, Stuller I, Dorner F, Pötzi P, Müller C, Eibl MM. Impaired primary, but not secondary, immune response in breast cancer patients under adjuvant chemotherapy. Cancer 1986;58:1648–52. https://doi.org/10.1002/1097-0142(19861015)58:8<1648::AID-CNCR2820580812>3.0.CO;2-E PMID:3019506
  279. Bradt V, Malafa S, von Braun A, et al. Pre-existing yellow fever immunity impairs and modulates the antibody response to tick-borne encephalitis vaccination. NPJ Vaccines 2019;4:38. https://doi.org/10.1038/s41541-019-0133-5 PMID:31508246
Return to your place in the textBOX 1. Clinical manifestations, laboratory findings, investigations, and diagnostic testing for tick-borne encephalitis virus infection

Symptoms and signs

  • First symptoms typically manifest approximately 7–14 days (range = 2–28 days) after tick bite (or if transmission by alimentary route, often at 2–4 days).
  • Acute neurologic illness (e.g., aseptic meningitis, meningoencephalitis, or meningoencephalomyelitis) or febrile illness can occur.
  • Milder neurologic illness (e.g., meningitis) is more common in children (e.g., aged approximately ≤15 years) and severe illness is more common as age increases.
  • Neurologic illness can be monophasic or biphasic (i.e., central nervous system manifestations occur after an initial nonspecific illness and period of remission).
  • Risk factors for severe disease include older age (e.g., ≥60 years), immunocompromise, and infection with the Far Eastern subtype virus (typically found in China, Japan, Mongolia, and the eastern parts of Russia).

Laboratory findings

  • Initial phase: leukopenia, thrombocytopenia, and elevated hepatic enzymes might be detected.
  • Neurologic phase: peripheral leukocytosis, elevated erythrocyte sedimentation rate, and increased C-reactive protein.
  • CSF results: pleocytosis and moderately elevated protein levels; pleocytosis is typically lymphocytic but in early disease neutrophils can predominate.

Neuroimaging and electroencephalogram investigations

  • Sensitivity of MRI is low.
  • Brain MRI changes are most commonly seen in the thalamus, often bilaterally.
  • Spinal MRI can indicate T2-hyperintensities in the anterior horns of the cervical cord in patients with myelitis or radiculitis.
  • Abnormal EEG findings are common and can include diffuse slowing and focal abnormalities.

Laboratory diagnosis

  • Usually based on detection of TBE virus immunoglobulin M antibody in CSF or serum.
  • Plaque reduction neutralization tests can be performed to confirm recent infection.
  • Diagnostic testing can be performed at CDC. Clinicians should contact their state or local health department or the CDC Arboviral Diseases Branch, Division of Vector-Borne Diseases (970-221-6400) for assistance.

Abbreviations: CSF = cerebrospinal fluid; EEG = electroencephalogram; MRI = magnetic resonance imaging.

TABLE 1. Tick-borne encephalitis cases among U.S. civilian travelers, 1979–2021*Return to your place in the text
Case no. Year Age group, yrs Sex Location of potential exposure to TBE virus Month of onset Duration of travel, days Reported activities with risk for tick exposure Outcome
1 1979 0–9 F Hungary NA NA NA Recovered
2 2001 50–59 M Russia June 10 Fishing Mild cognitive impairment
3 2004 20–29 M Russia August 45 NA Recovered
4 2006 40–49 M Sweden July 18 NA Recovered
5 2007 10–19 F China July 23 Hiking Severe neurologic sequelae
6 2008 10–19 M Czech Republic August 69 Extensive outdoor exposure in rural areas Recovered
7 2012 30–39 M Czech Republic August 28 Hiking Recovered, short-term sequelae
8 2012 40–49 M Finland August 16 Camping Recovered, unknown if sequelae
9 2013 70–79 M Russia (including Siberia) July 24 Hiking, picnicking, rural homestay Unknown, discharged from hospital to rehabilitation facility
10 2017 40–49 M Switzerland August 7 NA Recovered, mild sequelae
11 2018 60–69 M Sweden May 16 Trail running, manual work outdoors Recovered, initial sequelae resolved after several months
12 2019 0–9 M Austria, Switzerland June 10 Hiking Recovered

Abbreviations: F = female; M = male; NA = not available; TBE = tick-borne encephalitis.
* Excludes U.S. military personnel and their dependents.
Approximate number of days spent in areas where TBE is endemic.

Return to your place in the textBOX 2. Factors that increase risk for tick-borne encephalitis among U.S. travelers to areas where the disease is endemic

Season

  • TBE virus transmission is primarily during the warmer months of April–November when ticks are most active in the Northern Hemisphere.

Location

  • TBE virus transmission occurs within focal areas of countries in the geographic region extending from western and northern Europe through to northern and eastern Asia.
  • Information on countries with TBE risk areas is available on the CDC website (https://www.cdc.gov/tick-borne-encephalitis/geographic-distribution/index.html). The information should be interpreted cautiously because TBE virus transmission can be highly variable within risk areas and from year to year.
  • Ticks typically are found in woodland habitats including in coniferous or deciduous forests and on the forest edges in the transition zone between forests and grasslands.

Activities and occupations

  • Certain recreational activities can increase the risk for exposure to ticks (e.g., hiking, camping, cycling in woodland areas, hunting, fishing, birdwatching, and collecting mushrooms or berries).
  • Occupational risk might exist for persons with exposure to ticks (e.g., farmers, forestry workers, military personnel, or researchers conducting field work in rural areas).
  • Infection can follow ingestion of unpasteurized milk and milk products from infected goats, sheep, or cattle.

Duration

  • Activities undertaken are more important than time spent abroad. Among U.S. travelers, TBE cases have occurred after travel as short as 7–10 days; however, a longer duration of travel, residence, or repeated travel to areas of where the disease is endemic might increase the likelihood of exposure to TBE virus.

Other considerations

  • Risk for severe disease increases with age, and poor outcomes might be of particular concern for persons aged ≥60 years.
  • Persons with altered immunocompetence can have severe TBE and have a higher risk for a fatal outcome; however, immunocompromise and immunosuppression are precautions for vaccination.

Abbreviation: TBE = tick-borne encephalitis.

TABLE 2. Tick-borne encephalitis cases among U.S. military personnel and their dependents, 2012–2021Return to your place in the text
Case no. Year Age group, yrs Sex State of residence, Germany Month of onset Outcome
1 2012 30–39 M Baden-Württemberg September Recovered
2 2017 40–49 M Bavaria June Moderate sequelae
3 2017 30–39 M Bavaria July Recovered
4 2017 30–39 M Bavaria July Recovered, short-term sequelae
5 2017 30–39 M Bavaria November Moderate sequelae
6 2018 0–9 M Baden-Württemberg April Recovered
7 2018 10–19 M Baden-Württemberg June Recovered, short-term sequelae
8 2018 0–9 F Baden-Württemberg September Moderate sequelae
9 2019 40–49 M NA June or July NA
10 2020 30–39 M Bavaria May, June, or July NA
11 2021 0–9 F NA June or July NA
12 2021 30–39 M NA June or July NA

Abbreviations: F = female; M = male; NA = not available; TBE = tick-borne encephalitis.

Return to your place in the textFIGURE. Decision-making for recommending tick-borne encephalitis vaccination for U.S. travelers to areas where the disease is endemic
A flow chart is illustrated to aid health care providers in making decisions to recommend tick-borne encephalitis vaccine for U.S. travelers to areas where the disease is endemic.

Abbreviation: TBE = tick-borne encephalitis.

TABLE 3. Characteristics and properties of tick-borne encephalitis vaccineReturn to your place in the text
Vaccine characteristic Vaccine property
Trade name Ticovac
Vaccine type Inactivated, whole virus
TBE virus strain Neudoerfl (European TBE virus subtype)
Substrate Chick embryo fibroblast cells
Adjuvant Aluminum hydroxide
Stabilizer Human serum albumin
Preservative None
Other constituents Sodium chloride, dibasic sodium phosphate, and monobasic potassium phosphate
Substances used in manufacturing that might be present in final product Formaldehyde; sucrose; protamine sulfate; trace amounts of neomycin, gentamicin, and chick protein; and DNA from chick embryo fibroblast cells
Final preparation Suspension for injection
Presentation Prefilled syringe
Storage 36°F–46°F (2°C–8°C)
Route Intramuscular
Dose by age group Aged ≥16 years: 0.5 mL (adult presentation)
Aged 1–15 years: 0.25 mL (pediatric presentation)
Primary schedule by age group Aged ≥16 years: 3 doses administered as follows:
Dose 1: On day 0
Dose 2: 14 days–3 months after dose 1
Dose 3: 5–12 months after dose 2
Aged 1–15 years: 3 doses administered as follows:
Dose 1: On day 0
Dose 2: 1–3 months after dose 1
Dose 3: 5–12 months after dose 2
Booster dose All age groups: ≥3 years after completion of the primary series (if ongoing exposure or re-exposure to TBE virus is expected). No Advisory Committee on Immunization Practices recommendations are made on the need for subsequent booster doses.

Abbreviation: TBE = tick-borne encephalitis.


Suggested citation for this article: Hills SL, Poehling KA, Chen WH, Staples JE. Tick-Borne Encephalitis Vaccine: Recommendations of the Advisory Committee on Immunization Practices, United States, 2023. MMWR Recomm Rep 2023;72(No. RR-5):1–29. DOI: http://dx.doi.org/10.15585/mmwr.rr7205a1.

MMWR and Morbidity and Mortality Weekly Report are service marks of the U.S. Department of Health and Human Services.
Use of trade names and commercial sources is for identification only and does not imply endorsement by the U.S. Department of Health and Human Services.
References to non-CDC sites on the Internet are provided as a service to MMWR readers and do not constitute or imply endorsement of these organizations or their programs by CDC or the U.S. Department of Health and Human Services. CDC is not responsible for the content of pages found at these sites. URL addresses listed in MMWR were current as of the date of publication.

All HTML versions of MMWR articles are generated from final proofs through an automated process. This conversion might result in character translation or format errors in the HTML version. Users are referred to the electronic PDF version (https://www.cdc.gov/mmwr) and/or the original MMWR paper copy for printable versions of official text, figures, and tables.

Questions or messages regarding errors in formatting should be addressed to mmwrq@cdc.gov.

View Page In: Article PDF