Viral Encephalitis and Meningitis


Hundreds of viruses exhibit tropism for the central (CNS) and/or peripheral (PNS) nervous systems. In the case of some viruses, involvement of the CNS or PNS is the predominant feature of illness, whereas in others involvement of the nervous system is a rare complication of a more generalized illness. Viral infection of the nervous system can result in a myriad of clinical presentations occurring separately or in combinations including acute or chronic meningitis, encephalitis, myelitis, ganglionitis, and polyradiculitis. Viruses may also incite para- or post-infectious CNS inflammatory or autoimmune syndromes such as acute disseminated encephalomyelitis (ADEM) or encephalitis associated with autoantibodies (see Chapter 80 ). The neurological complications of human immunodeficiency virus (HIV) and human T-cell lymphotropic virus (HTLV) infections are discussed separately (see Chapter 77 ).

Table 78.1 lists the most common viral causes of nervous system disease in North America and the relative propensity of each virus to cause meningitis, encephalitis, post-infectious encephalomyelitis, or myelitis. Infections that occur in residents or travelers to areas outside the United States are listed in Table 78.2 . In the United States, the most common viruses causing meningitis are enteroviruses (EVs), herpes simplex virus type 2 (HSV-2), and arboviruses. The most common identified viral causes of encephalitis are herpesviruses, notably HSV-1, and arboviruses. Even with the best diagnostic efforts, up to 70% of cases of suspected viral encephalitis remain of unknown etiology ( ). However, cases of antibody-mediated autoimmune encephalitis are increasingly recognized as important causes of encephalitis and may represent a substantial portion of the unknown or unidentified encephalitis cases. In an analysis of patients < 30 years of age and enrolled in the California Encephalitis Project (CEP), anti- N -methyl- D -aspartate (NMDA)-receptor encephalitis was as common as viral encephalitis ( ). Worldwide, there are tens of thousands of deaths from rabies each year ( ), and in Asia, nearly 68,000 cases and 13,000–20,000 deaths from Japanese encephalitis virus (JEV; ). Since the original outbreak of West Nile virus (WNV) in 1999, the Centers for Disease Control (CDC) tracks reported cases of WNV disease in the United States https://www.cdc.gov/westnile/statsmaps/index.html From 1999 through 2018, there have been approximately 51,000 cases of WNV disease, including 24,000 cases of WNV neuroinvasive disease, and 2300 deaths.

TABLE 78.1
Primary Causes of Viral Nervous System Infection in North America
Agent Meningitis Encephalitis Postinfectious Acute Disseminated Encephalomyelitis Myelitis
Arboviruses (United States and Canada)
Togaviruses
  • Flavivirus

  • West Nile virus (WNV)

  • St. Louis encephalitis virus (SLE)

  • ∗∗

  • Powassan

  • ∗∗

  • Alphavirus

  • Eastern equine encephalitis (EEE)

  • ∗∗∗

  • Western equine encephalitis (WEE)

  • ∗∗

  • Venezuelan equine encephalitis (VEE)

Reoviridae: orbivirus Colorado tick fever ∗∗
Bunyavirus California (La Crosse) ∗∗
Jamestown Canyon
Snowshoe hare
Herpes viruses Herpes simples virus (HSV)-1 ∗∗
HSV-2 ∗∗ ∗∗
Varicella-zoster virus (VZV) ∗∗ ∗∗
Human
Cytomegalovirus (HCMV)
∗∗ ∗∗
Epstein-Barr virus (EBV) ∗∗
Human herpesvirus (HHV)-6 ∗∗
HHV-7
HHV-8
Herpes B virus ∗∗∗ ∗∗
Lymphocytic choriomeningitis virus (LCMV) ∗∗ ∗∗
Mumps virus ∗∗
Human immunodeficiency virus (HIV) ∗∗
Rabies virus ∗∗∗
Measles virus ∗∗∗
Rubella virus
Enteroviruses ∗∗∗ ∗∗∗
Adenovirus ∗∗
Vaccinia
Influenza
Parainfluenza
Rotavirus
Parvovirus B-19
Coronavirus SARS-CoV2 ∗∗ ∗∗

TABLE 78.2
Additional Causes of Viral Nervous System Infection Resulting from Foreign Exposures
Agent Geographical Distribution
Nipah virus Malaysia, Singapore, India, Bangladesh
Measles virus Europe, Middle East, Asia, Africa, the Pacific
Filovirus Ebola Rainforest Africa
Marburg
Mosquito-borne Eastern equine Caribbean and South America (plus United States)
Venezuelan equine Central and northern South America (plus United States)
St. Louis Caribbean, Central and northern South America (plus United States)
Japanese B Japan, China, Southeast Asia, India, parts of Northern Australia, western Pacific
Kunjin Australia
Murray Valley Australia and New Guinea
West Nile Africa and Middle East, parts of Europe (plus United States)
Ilheus South and Central America
Rocio Brazil
Dengue All tropical areas (plus United States)
Zika Caribbean, Central and South America, Southeast Asia, Pacific Island nations, Africa
Yellow Fever Sub-Saharan Africa, Central and South America
Chikungunya Africa, southern Asia, Oceania, Indian Ocean countries, Caribbean, Central and South America (plus United States)
Sand fly–borne Toscana Italy, Spain, Portugal, France
Tickborne complex Far Eastern (formerly Russian spring-summer) Eastern Russia and neighboring north Asian countries
Siberian Russia and neighboring Asian countries, Scandinavia
Central European Eastern and Central Europe, Scandinavia
Kyasanur Forest India
Louping Ill England, Scotland, and Northern Ireland
Negishi Japan
Bunyavirus Tahyna Czechoslovakia, Yugoslavia, Italy, southern France
Inkoo Finland
Rift Valley East Africa
Rhabdovirus Rabies Asia, Africa, South America, eastern Europe (plus United States)
Enterovirus Poliovirus Afghanistan, Pakistan, India, Nigeria (endemic)
Arenavirus LCMV All continents except Antarctica
Lassa fever virus West Africa
Orthopoxvirus Monkeypox Central and West Africa
LCMV , Lymphocytic choriomeningitis virus.

Although the basic clinical features of most types of viral meningitis and encephalitis are generally similar, specific physical examination findings may help narrow the possible viral etiologies of nervous system disease ( Tables 78.3 and 78.4 ). It is important to recognize that several nonviral diseases can mimic the clinical features of viral CNS infection ( Table 78.5 ) ( ). The treatment, prophylaxis, and immunotherapy of specific viral infections are summarized later in the chapter.

TABLE 78.3
Skin/Mucous Membrane Findings Suggesting Specific Viral Central Nervous System Diseases
Exanthem or Mucous Membrane Change Viral Agent Specific Changes
Vesicular eruption Enterovirus (A71) “Hand, foot, and mouth disease”: Macules/papules/vesicles on palms, soles, buttocks
Herpes simplex Grouped small (3 mm) vesicles on an erythematous base
Varicella-zoster virus Zoster: Vesicles in dermatomal distribution
Primary VZV: Multiple vesicles, papules, pustules in various stages of eruption
Maculopapular eruption Epstein-Barr virus Diffuse maculopapular eruption following ampicillin treatment
Measles Diffuse maculopapular erythematous eruption beginning on face/chest and extending downward
HHV-6 Roseola: Diffuse maculopapular eruption following 4 days of high fever
Colorado tick fever Maculopapular rash in 50%
LCMV Occasionally occurs with lymphadenopathy
WNV, ZIKV Diffuse erythematous maculopapular rash on chest and arms
Erythema multiforme ( Mycoplasma ) Many types of rash
Confluent macular rash Parvovirus Confluent erythema over cheeks (“slapped cheeks”) followed by lacy reticular rash over extremities (late)
Purpura Parvovirus Rare “stocking glove” syndrome: Purpuric lesions on distal extremities
Pharyngitis Enterovirus Herpangina: Vesicles on soft palate
Adenovirus Pharyngitis, conjunctivitis
Conjunctivitis St. Louis encephalitis Conjunctivitis
ZIKV Conjunctivitis
Adenovirus Conjunctivitis with pharyngitis (see above)
HHV-6 , Human herpesvirus type 6; LCMV , lymphocytic choriomeningitis virus; VZV , varicella-zoster virus; WNV , West Nile virus, ZIKV , Zika virus.

TABLE 78.4
Other Specific Findings Associated with Viruses Causing Central Nervous System Disease
Finding Viruses
Alopecia LCMV
Arthritis LCMV, parvovirus, Chikungunya
Biphasic illness LCMV, Colorado tick fever
Lymphadenopathy LCMV, mumps, HIV
Mastitis Mumps
Mononucleosis HCMV, EBV, CMV
Myelitis WNV, St. Louis encephalitis virus, VZV, EBV, HSV-1, CMV, herpes B virus, LCMV, EV-D68, EV-A71
Myocarditis/pericarditis Enterovirus, (mumps, LCMV)
Orchitis/oophoritis Mumps (LCMV, EBV)
Paresthesias Colorado tick fever, LCMV, rabies
Parotitis Mumps (LCMV)
Pneumonia Influenza, parainfluenza, SARS-CoV2
Retinitis HCMV, WNV, ZIKV (congenital)
Tremors, myoclonus Arbovirus (e.g., WNV), EV-A71
Urinary retention St. Louis encephalitis virus, VZV, HCMV, HSV, herpes B virus, LCMV (see myelitis causing viruses)
EBV , Epstein-Barr virus; EV , Enterovirus, HCMV , human cytomegalovirus; HSV , herpes simplex virus type; LCMV , lymphocytic choriomeningitis virus; VZV , varicella-zoster virus; WNV , West Nile virus; ZIKV , Zika virus.

TABLE 78.5
Diseases That Can Masquerade as Viral Nervous System Disease
Etiology Agent Disease Suggestive features
Infectious Bacterial Parameningeal focus (sinusitis, intracranial abscess) Very mild pleocytosis, focal neurological exam
Partially treated bacterial meningitis Prior antibiotic treatment
Lyme disease Tick exposure, arthritis, appropriate geography, erythema migrans
Tuberculosis Very high protein, hypoglycorrhachia
Leptospirosis Conjunctival suffusion, jaundice
Syphilis (primary and tertiary) Chronic
Brucella Farm animal or unpasteurized milk exposure
Whipple disease Gastrointestinal complaints
Bartonella (cat scratch disease) Cat exposure, adenopathy
Listeria Brainstem encephalitis
Typhoid fever Exposure history, bradycardia
Fungal Cryptococcus Usually immunocompromised patient (pt)
Coccidioides Southwestern US exposure, pulmonary symptoms
Histoplasma Pulmonary nodules
Blastomycosis Midwest, pulmonary symptoms
Candida Immunocompromised pt
Nocardia Immunocompromised pt
Parasitic Toxoplasma Retinitis, cat exposure
Cysticercosis Calcified lesions
Amoebic Fresh water: Naegleria
Malaria ( Plasmodium falciparum ) Exposure history, cyclic fevers
Rickettsial Rocky Mountain spotted fever Leukopenia, thrombocytopenia, hyponatremia, petechial rash
Ehrlichia See above
Coxiella burnetii (Q fever) Exposure to sheep, wildlife pulmonary disease
Mycoplasma Precedent pulmonary symptoms
Parainfectious Acute disseminated encephalomyelitis (ADEM) Characteristic MRI findings
Noninfectious Anti-neuronal Antibodies, e.g., anti-NMDA receptor Abs Autoimmune Encephalitis Atypical psychiatric presentations or movement disorders and young age
Connective tissue disorders Systemic lupus erythematosus (SLE) Malar rash, multisystem organ involvement
Sarcoidosis Hilar adenopathy, erythema nodosum
Uveomeningitic syndromes Behçet disease Genital/oral ulcers, uveitis
Intracranial tumors and cysts Recurrent episodes, dermal sinus tract
Drugs NSAIDs, antibiotics, immunomodulators, anticonvulsants Exposure history
Intracranial or subarachnoid hemorrhage
Encephalopathy Toxic or metabolic, mitochondrial disorders, particularly MELAS
MRI , Magnetic resonance imaging; NMDA , N-methyl-D-aspartate; NSAIDs , nonsteroidal antiinflammatory drugs.

Deoxyribonucleic Acid Viruses

Herpesviruses

Multiple members of the herpesvirus family cause neurological disease in humans: HSV-1 and HSV-2, varicella-zoster virus (VZV), human cytomegalovirus (HCMV), Epstein-Barr virus (EBV), human herpesviruses (HHV-6, HHV-7, and HHV-8), and the simian (“monkey”) herpes B virus.

Herpes Simplex Viruses Type 1 and 2

Herpes simplex encephalitis

HSV-1 encephalitis is the most common identified cause of sporadic fatal encephalitis in the United States, accounting for approximately 10% of all cases of encephalitis and occurring with a frequency of about 2–4 cases per 1,000,000 population per year ( ). Early recognition is important because the efficacy of the antiviral drug acyclovir (ACV) in reducing morbidity and mortality decreases as neurological disease progresses ( ). HSV-1 strains cause over 90% of cases of herpes simplex encephalitis (HSE) in adults, with the remainder due to HSV-2. Conversely, HSV-2 is a more common cause of meningitis and neonatal meningoencephalitis than HSV-1. Both HSV types 1 and 2 have been associated with myelitis.

Fever is present in about 75%–80% and headache in about 60%–70% of patients with brain biopsy or cerebral spinal fluid (CSF) polymerase chain reaction (PCR) proven HSE ( ). Other common features include disorientation or altered consciousness (70%–90%), personality change (60%), focal or generalized seizures (55%), memory disturbance (35%), motor deficit (40%), and aphasia (40%–60%) ( ). Immunocompromised patients may have atypical clinical and MRI presentations including the absence of prodromal symptoms, fewer focal findings, and MRI abnormalities outside the “classical” frontotemporal areas ( ). Atypical presentations may also occur in young children ( ).

It is important to recognize that no sets of signs or symptoms are pathognomonic of HSE, and multiple infectious and inflammatory processes can mimic HSE ( ). Definitive diagnosis of HSE is based on amplification of HSV DNA in CSF using an HSV-specific PCR assay or, less commonly, isolation of virus, or detection of viral antigen or viral nucleic acid from brain tissue at biopsy or autopsy (see later discussion).

Examination of CSF is a critical diagnostic test in suspected cases of HSE. CSF is usually under increased pressure, with a median lymphocytic pleocytosis of 25–150 white blood cells (WBC) per μL (range 0–1000). In early studies, over 95% of PCR or biopsy-proven cases of HSE have a CSF pleocytosis ( ), although some more recent reports suggest normocellular CSF can occur in up to 26% of cases (Saraya et al., 2016), and this phenomenon may be more common in patients with HIV-infection or otherwise immunocompromised ( ). A recent retrospective multicenter review of pediatric HSE found that 16% of children with HSE had normal CSF glucose, protein, and cell counts ( ). HSE is often hemorrhagic, and both red blood cells and xanthochromia can be detected in CSF, although neither feature occurs with significantly greater frequency in HSE compared to other causes of focal encephalitis ( ). The presence of red cells in CSF may be associated with a worse prognosis ( ). CSF protein concentration is moderately elevated (65–85 mg/dL) in about 60%–70% of patients and glucose concentration is normal in the majority (75%–90%) patients ( ). Autoantibodies, notably those against the NMDA receptor, may develop as a consequence of HSE in 30% of patients ( ). A retrospective study found that 30% of patients with PCR confirmed HSE had detectable serum NMDAR antibodies ( ). In adult HSE cases the presence of NMDAR antibodies has been associated with delayed recovery and resurgent symptoms suggesting disease relapse, that can respond to appropriate immunomodulatory therapies. In children, NMDAR antibodies may be associated with choreiform and other movement disorders ( ).

CSF PCR is the diagnostic assay of choice in HSE, as virus is cultured from CSF in less than 5% of cases. Amplification of HSV DNA from CSF by PCR testing has a sensitivity of 98% and specificity of 94%–99% for the diagnosis of HSE compared to brain biopsy ( ) ( Table 78.6 ). Surprisingly, CSF HSV genome copy number (“HSV viral load”) is not a reliable predictor of outcome ( ). HSV CSF PCR results must be interpreted considering the pre-test probability that a patient has HSE (Bayesian decision analysis). For example, a negative HSV CSF PCR in a patient with a low (∼5%) prior probability of HSE reduces the post-test likelihood of HSE to approximately 0.2%, whereas a negative PCR in a patient with a high (∼60%) pre-test likelihood of HSE only reduces the post-test likelihood of HSE to around 6% ( ). CSF PCR remains a sensitive technique for the detection of HSE, even in patients who have received up to a week of ACV therapy ( ). Despite the high overall sensitivity of CSF HSV PCR, false-negative results have been reported, most notably in patients in whom CSF was obtained within the first 72 hours of illness onset ( ). For this reason, caution should be used in stopping ACV therapy in patients with suspected HSE on the sole basis of a single negative CSF PCR test obtained within 72 hours of symptom onset, unless a suitable alternative diagnosis has been established.

TABLE 78.6
CSF PCR Diagnosis of Viral Nervous System Disease
Virus Sensitivity Specificity
Adenovirus Unknown
Dengue Unknown
Enterovirus >95% (meningitis), <10% for AFM with EV-D68 or <25% with Neuroinvasive EV-A71 >95%
Herpesviruses
  • HCMV

  • 100% in immunocompromised

  • High

  • >60% in congenital CMV infection

  • High

  • EBV

  • 98.5% as tumor marker in HIV patients with Primary CNS lymphoma

  • Unknown

  • HSV-1 and -2

  • >95%

  • >95%

  • HHV-6

  • Unknown

  • Unknown

  • VZV

  • >95%

  • >95%

HIV HIV RNA present at all stages High
HTLV I and II 75% 98.5%
Influenza Unknown but > culture Unknown
Japanese encephalitis virus Unknown (higher early) High
JC virus 50%–90% in PML, lower copy # in more immunocompetent pts 98%
LCMV Unknown Unknown
Mumps Unknown High
Measles Unknown High
Parvovirus B-19 80% Unknown
Rabies 90% High
WNV 70% (higher early) High
SARS-CoV2 Unknown, likely low (Nasopharyngeal RT-PCR high acutely) High
ZIKV Unknown (congenital ZIKV syndrome) High
AFM, Acute flaccid myelitis; CNS , central nervous system; CSF , cerebral spinal fluid; EBV , Epstein-Barr virus; HCMV , human cytomegalovirus; HHV , human herpesvirus; HSV , herpes simplex virus; HTLV , human T-cell lymphotropic virus; LCMV , lymphocytic choriomeningitis virus; PCR , polymerase chain reaction; PML , progressive multifocal leukoencephalopathy; VZV , varicella-zoster virus; WNV , West Nile virus; ZIKV , Zika virus.

When definitive diagnosis of HSE was dependent on brain biopsy, patients often had significantly depressed levels of consciousness and advanced neurological disease at the time of diagnosis. With routine use of HSV CSF PCR instead of brain biopsy for definitive diagnosis of HSE, patients are often identified at an earlier stage of illness, exhibit less depression of consciousness, and have improved response to therapy. Over 75% of cases with PCR-proven HSE had a Glasgow Coma Score (GCS) above 12 ( ), whereas only 30% of cases with biopsy-proven HSE had a GCS above 10 in the Collaborative Antiviral Study Group (CASG) trial of vidarabine versus ACV therapy ( ).

Magnetic resonance imaging (MRI) is significantly more sensitive than computed tomography (CT) and is the neuroimaging procedure of choice in patients with suspected HSE. Approximately 90% of patients with PCR-proven HSE will have MRI abnormalities involving the temporal lobes ( ) ( Fig. 78.1 ). The electroencephalogram (EEG) may be abnormal early in the course of disease, demonstrating diffuse slowing, focal abnormalities in the temporal regions, or periodic lateralizing epileptiform discharges (PLEDs). EEG abnormalities involving the temporal lobes are seen in approximately 75% of patients with PCR-proven HSE ( ). Brain biopsy is now only rarely performed for diagnosis of HSE. Biopsy is reserved for atypical cases in which the diagnosis remains in question and for those who respond poorly to treatment. Biopsy specimens from patients with HSE show hemorrhagic necrosis, HSV antigen in infected cells, and accumulations of viral particles forming acidophilic intranuclear inclusion bodies in neurons (Cowdry type A inclusions; Fig. 78.2 ).

Fig. 78.1, Herpes Simplex Encephalitis.

Fig. 78.2, Hippocampal Granule Cell Neurons in Herpes Simplex 1 Encephalitis.

Empirical therapy with ACV should be started immediately in acute cases of focal encephalitis of suspected viral etiology ( Table 78.7 ). Delay in the institution of ACV therapy is surprisingly common and occurs more frequently in patients with other severe underlying diseases, chronic alcohol abuse, pleocytosis of less than 10 cells/μL, and delayed initial neuroimaging studies, as these factors may make clinicians less likely to consider HSE in the differential diagnosis ( ). Mortality in untreated cases of HSE is around 70%, but this is reduced to 19%–28% in patients treated with ACV ( ), and to less than 10% in more recent studies ( ). Morbidity due to HSV-1 encephalitis remains high even in patients receiving ACV, with only 37.5% of all patients surviving with no or only mild deficits ( ). In a more recent study 23% had no sequelae and 32% mild sequelae ( ). However, in specific subpopulations of ACV-treated HSE patients, prognosis can be considerably better. For example, 50% (12/24) of patients in whom treatment with ACV was initiated when their GCS exceeded 6 survived with no or only minor sequelae, and more than 60% who were younger than age 30 and had a GCS above 6 survived with no or only minor sequelae ( ). Outcomes have been generally similar in cases of PCR-proven HSE treated with ACV, with 35%–56% of patients returning to normal functional status or having only mild disability or residual sequelae at 6 months follow-up ( ). Survival and prognosis are influenced by several factors including level of consciousness at initiation of therapy (e.g., GCS), patient age, and duration of disease before therapy ( ). In the CASG trial, all (9/9) patients treated with ACV within 4 days of onset of fever, headache, and focal neurological deficits survived, whereas the mortality was 35% in those in whom ACV treatment was initiated when disease was more than 4 days old ( ). In another study, the recovery rate was 50% in patients treated within 5 days of illness onset ( ). In a third study of PCR-proven HSE, 75% of patients with an ultimately favorable outcome had received ACV therapy within 2 days of hospital admission compared to only 30% of those with a poor outcome ( ).

TABLE 78.7
Treatment and Prophylaxis of Viral Infections
Antiviral Class Antiviral Agent Dose Indications Toxicity or Cautions
Nucleoside analogs Acyclovir 10 mg/kg/dose (IV) q 8 h ×14–21 days HSV encephalitis in adults Renal impairment
20 mg/kg/dose (IV) q 8 h ×21 days Neonatal HSV encephalitis
(IV) q 8 h ×21 days
10–12 mg/kg/dose in adults and up to 20 mg/kg/dose in infants) IV q 8 h ×21 days
VZV encephalitis in normal or immunocompromised patient (pt)
15 mg/kg/dose (IV) q 8 h ×21 days Herpes B virus
800 mg (PO) 5×/day ×7 days Dermatomal zoster or primary VZV in immunocompromised pt Low solubility
Famciclovir 200 mg (PO) tid ×7 days Dermatomal zoster or primary VZV in immunocompromised pt Headache, nausea
Valacyclovir 1 g (PO) qid ×7 days Dermatomal zoster or primary VZV in immunocompromised pt Thrombotic thrombocytic purpura/hemolytic uremic syndrome in HIV patients
Ganciclovir 5 mg/kg (IV) q 12 h ×14–21 days HCMV, herpes B virus Bone marrow suppression
Valganciclovir 900 mg (PO) bid ×21 days (induction), then 900 mg (PO) daily (maintenance) HCMV retinitis Bone marrow suppression
Ribavirin 2 g (IV) ×1, then 1 g (IV) q 6 h ×4 days, then 0.5 g (IV) q 8 h ×6 days Lassa fever Hemolytic anemia
20–35 mg/kg/day ×7 days
600 mg/day to 1400 mg/day (PO) divided in 2 doses
Measles virus
Hepatitis C
Cytarabine 2 mg/kg (IV) ×5 days ×4 wk PML Bone marrow suppression
Trifluridine 1% ophthalmic solution Herpetic keratoconjunctivitis
Nucleotide analog cidofovir 5 mg/kg (IV) every 1–2 wk or 1 mg/kg (IV) 3×/wk for 3 wk DNA viruses:
Severe AdV, monkeypox, resistant HCMV
nephrotoxicity
Remdesevir (Gilead, EUA) 200 mg IV x1 then 100mg IV q24h x 4-9d Severe COVID-19
Pyrophosphate analog Foscarnet 90 mg/kg (IV) q 12 h ×14–21 days Acyclovir-resistant HSV/VZV
Ganciclovir-resistant HCMV Hypocalcemia, renal impairment
Neuraminidase inhibitors Oseltamivir 75 mg (PO) bid ×5 days Influenza A and B
Zanamivir 10 mg (inhalation) bid ×5 days Influenza A and B
Cytokine Interferon alfa
Pegylated interferon alfa-2b
3 million U/day (SQ)
1.5 μg/kg/wk (SQ)
PML, acyclovir-resistant VZV, hepatitis C Flulike side effects
10 5 –10 6 U/m 2 body surface (intrathecal) SSPE
Supplements Vitamin A 400,000 IU (IM) Acute measles in vitamin A deficiency
bid , Twice daily; COVID-19, COVID-19 , SARS-CoV2 Disease 2019; EUA, FDA Emergency Use Authorization; HCMV , cytomegalovirus; HSV , herpes simplex virus; IM , intramuscularly; IV , intravenously; PML , progressive multifocal leukoencephalopathy; PO , orally; qid , four times daily; SQ , subcutaneously; SSPE , subacute sclerosing panencephalitis; tid , three times daily; VZV , varicella-zoster virus.

The standard adult dose of ACV for HSE is 10 mg/kg, given intravenously (IV) every 8 hours (20 mg/kg every 8 hours in neonates and children) for 14–21 days. Renal insufficiency is an infrequent, usually reversible side effect of ACV therapy and the risk is reduced by appropriate hydration. Neither higher doses of ACV nor prolonged therapy with oral valacyclovir after a standard intravenous course improved outcomes in adults ( ). ACV dosing should be adjusted appropriately in patients with renal insufficiency. Periodic shortages of intravenous ACV and the cost of intravenous preparations in resource-poor countries have led some to suggest that oral valacyclovir may be an alternative when intravenous ACV is unavailable ( ), but it is critical to emphasize that there are only isolated case reports and no comprehensive studies of the efficacy of oral drugs in HSE and this alternative should only be considered in extraordinary circumstances or settings. When a specific shortage of intravenous formulations of ACV exists, ganciclovir can be used as an alternative. Retrospective studies have suggested that there may be some benefit in adding corticosteroids to ACV treatment ( ), although controlled clinical trials are needed before the role of steroid therapy as an adjunct to ACV can be definitively established. Cases of HSE due to ACV-resistant HSV strains have been reported predominantly in immunocompromised patients and in extraordinarily rare instances in immunocompetent individuals (<1%) who have never been exposed to antiviral drugs ( ). Foscarnet (180 mg/kg IV daily given in two or three divided doses) is an alternative therapy for patients with suspected or proven ACV-resistant strains or with allergy to ACV ( ).

Neonatal herpes simplex virus meningoencephalitis

In contradistinction to adults, in whom HSE is usually caused by HSV-1, HSV-2 is the most common causal agent of meningoencephalitis in neonates (although HSV-1 disease may also occur). Neonates who acquire HSV from the birth canal develop infection of the CNS in 50% of cases ( ). CNS disease occurs as either a component of an overwhelming sepsis-like disseminated disease with multiorgan involvement (in the first week of life) or as isolated CNS disease that usually presents later (weeks 2–10 of life), with or without accompanying vesicular skin, mucous membrane, or conjunctival lesions (skin, eye, mouth disease). The presence of vesicular skin or mucosal lesions in an infant of this age, even in the absence of fever or systemic symptoms, warrants immediate evaluation of CSF for HSV infection, because up to 30% of infants with presumed isolated skin, eye, and mouth disease are subsequently identified as having CNS involvement. Neonates with possible HSV disease should be treated empirically with IV ACV, 20 mg/kg, every 8 hours ( ). Treatment should be continued for 14 days in HSV-infected infants with isolated skin, eye, and mouth disease and 21 days in infants with sepsis or CNS involvement. Relapses of skin, eye, and mouth disease (with the potential for CNS involvement and subsequent neurological deficits) are common in the first year of life following neonatal HSV disease. A recent double-blind, placebo-controlled study found that a 6-month course of oral ACV (300 mg/meter sq. of body surface area 3× daily), after completion of the standard 14- to 21-day course of intravenous ACV, resulted in improved neurodevelopmental outcomes at 12 months after treatment compared to placebo and this has become standard of care ( ).

Herpes simplex virus meningitis

HSV meningitis accounts for approximately 10% of acute viral meningitis in adults. In most series greater than 75% of those affected are women ( ). At the time of their first episode of genital herpes (generally due to HSV-2), approximately 36% of women and 11% of men have symptoms of meningitis including fever, headache, and nuchal rigidity. In a review of the Mayo Clinic experience with HSV-2 meningitis all patients had headache and about half had fever, photophobia, and meningismus, and just over a quarter had nausea or vomiting ( ). Only ∼13%–25% of patients have a history of genital herpes or active lesions ( ). The CSF shows a lymphocytic pleocytosis with an average of ∼500 cells/μL, an elevated protein (average ∼150 mg/dL) and a normal glucose. Neuroimaging is normal in nearly 85% but may show meningeal enhancement in rare cases ( ). Following an initial episode of HSV-2 meningitis, 20%–30% go on to develop recurrent episodes ( ). A randomized double-blind controlled study failed to show any effect of long-term valacyclovir suppressive therapy (500 mg 2×/day) in reducing the frequency of recurrent episodes of meningitis ( ). CSF viral cultures are invariably negative during recurrent episodes of meningitis, although the virus may be isolated during the first (primary) episode. Other neurological complications such as paresthesias, urinary retention, and transverse myelitis have been described with HSV-2 infections.

CSF PCR has identified HSV-2 as the primary etiological agent in patients with benign recurrent lymphocytic meningitis (Mollaret meningitis) ( ). This syndrome may be associated with the presence in the CSF of large cells of monocyte-macrophage lineage (Mollaret cells).

Treatment of HSV-2 meningitis varies considerably across published series and no high-quality randomized controlled trials have been performed ( ). A typical regimen is to use oral valacyclovir (500 mg 3×/day) for 7–10 days. Patients with more severe symptoms may benefit from an initial short course of intravenous ACV (e.g., 500 mg q8h ×3 days) followed by oral valacyclovir.

Varicella-Zoster Virus

Primary infection

At least 95% of the adult population has been infected with VZV, and the lifetime risk of developing shingles is 3%–5%. VZV can involve virtually every part of the CNS and PNS ( ). Primary VZV may produce meningitis or encephalitis in immunocompromised patients. An acute self-limited cerebellar ataxia occurs in about 1 in 4000 children (<15 years of age) during or immediately following primary VZV infection (chickenpox). Postinfectious encephalomyelitis follows an estimated 1 in 2500 cases of primary VZV infection.

Reactivation of herpes zoster can produce meningitis, myelitis, and encephalitis in older children and adults. Zoster “encephalitis” can occur in both immunocompetent and immunocompromised hosts and in adults typically results from reactivation of herpes zoster decades after an initial infection with varicella (chickenpox). It has been suggested that VZV encephalitis may be second only to that caused by HSV as a cause of sporadic encephalitis in adults, and may be the leading cause of sporadic encephalitis in children ( ). VZV encephalitis is in fact a vasculopathy rather than a true encephalitis ( ). In older (>60 years) immunocompetent adults, VZV vasculopathy (previously called granulomatous arteritis ) typically presents with acute focal stroke-like deficits due to inflammatory involvement of large cerebral arteries, typically occurring in the trigeminal distribution. Most cases are monophasic, although rare cases follow a chronic or relapsing-remitting course ( ). CSF shows a lymphocytic pleocytosis. MRI shows a large single infarct, most commonly in the carotid, middle, or anterior cerebral territory. At autopsy, viral particles, antigen, and DNA can be found in the involved artery. Diagnosis can be made by demonstration of VZV DNA in CSF by PCR or by demonstration of VZV immunoglobulin (Ig)M or intrathecal synthesis of VZV IgG in CSF.

In immunocompromised individuals, VZV reactivation produces a multifocal vasculopathy predominantly involving small and medium-sized arteries, resulting in a clinical syndrome of mental status changes, focal deficits, and a CSF mononuclear pleocytosis ( ). The typical rash of zoster may be absent. Neuroimaging shows multifocal hemorrhagic and ischemic cortical and subcortical infarcts. Diagnosis can be established by VZV CSF PCR or by demonstration of CSF VZV IgM or intrathecal VZV-specific IgG antibody synthesis. While most cases of VZV encephalitis present as a vasculopathy on imaging, a recent case series of VZV encephalitis cases found that 14 of 20 patients had a nonvascular, nonspecific or normal neuroimaging result ( ).

VZV can also cause an aseptic meningitis in immunocompetent hosts even in the absence of a characteristic varicella (chickenpox) or zoster (shingles) rash ( ). VZV accounts for up to 8% of viral meningitis cases, making it third in importance behind EVs and HSV-2 ( ).

VZV has recently been associated with giant cell arteritis (GCA). VZV antigen can be detected in up to 75% of temporal artery biopsy specimens from patients with associated evidence of GCA ( ). VZV antigen is also detected in a high percentage (64%) of biopsy specimens of temporal arteries taken for diagnosis of GCA even when histological evidence of GCA is not found. The incidence of VZV in “normal” temporal arteries is reported at 8%–22%. The significance of these findings and their implications for pathogenesis and potentially treatment remain to be established. Although the suggestion that GCA patients be treated with valacyclovir in combination with prednisone has been made ( ), no clinical trials of this strategy have been reported.

There are no controlled randomized clinical trials of antiviral therapy of CNS VZV infection. Intravenous ACV (10 mg/kg every 8 hours) for 7–10 days is generally recommended for treatment of immunocompromised children and adults with chickenpox. Patients with VZV CNS disease including vasculopathy receive IV ACV (10–20 mg/kg or 500 mg/m 2 every 8 hours) for a minimum of 7 ( ) to 14 ( ) days, combined with a steroid pulse for 3–5 days (e.g., prednisone 60–80 mg/day) ( ). HIV-infected individuals with localized (non-disseminated) dermatomal zoster can be treated with oral famciclovir (500 mg 3× daily) or valacyclovir (1000 mg 3× daily) for 7–10 days.

Herpes zoster

Following primary infection, VZV becomes latent in cells of the dorsal root ganglia. Reactivation of endogenous latent virus produces herpes zoster (shingles). The virus can reactivate after injury or trauma to the spine or nerve roots or in response to waning cell-mediated immunity to VZV caused by age or immunosuppression related to HIV infection, cancer, cytotoxic drugs, or systemic illness. Herpes zoster is frequently the first clinical presentation of underlying HIV infection and may present as protracted or multidermatomal disease. The incidence of shingles is up to 25-fold greater in HIV-infected individuals than in the general population. Following universal varicella vaccination for US children, concerns arose that decreases in exposure and boosting of immunity in older adults would result in increased rates of zoster. However, recent work shows that widespread childhood immunization against zoster did not seem to affect the incidence of herpes zoster from 1992 to 2010 ( ).

Herpes zoster typically begins with pain and paresthesias in one or two adjacent spinal or cranial dermatomes ( ). Pain is followed in 3–4 days by a painful pruritic vesicular eruption in the area supplied by the affected root. The eruption typically lasts 10–14 days. Eruption most commonly occurs in the lower thoracic dermatomes but also commonly involves the trigeminal distribution and the cervical or lumbosacral dermatomes. Involvement of the first division of the trigeminal ganglion produces ophthalmic zoster and may be associated with conjunctivitis, keratitis, anterior uveitis, or iridocyclitis. Fortunately, vision loss following herpes zoster ophthalmicus is rare. Involvement of the geniculate ganglion produces otic zoster, or the Ramsay Hunt syndrome—painful facial paresis accompanied by tympanic membrane and external auditory canal vesicular rash. Herpes zoster involving cervical and thoracic levels may be associated with myelitis, and in the lumbosacral region may be accompanied by bladder dysfunction or ileus. Complications of zoster include postherpetic neuralgia, segmental motor atrophy in the affected dermatome, meningitis, myelitis, large-vessel vasculitis (usually involving the carotid or its branches on the side of zoster ophthalmicus), and multifocal leukoencephalitis or encephalitis with generalized cerebral vasculopathy ( ).

Risk factors for postherpetic neuralgia in patients with shingles include age older than 50 years and prodromal sensory symptoms. Treatments are directed toward lessening pain, reducing virus shedding, and shortening healing time. ACV (800 mg orally, 5× daily for 7 days), famciclovir (200 mg orally, 3× daily for 7 days), or valacyclovir (1 g orally, 4× daily for 7 days) accelerates cutaneous healing and decreases acute zoster pain if begun within 72 hours of onset of rash. Whether these agents significantly decrease the incidence, duration, or severity of postherpetic neuralgia is uncertain. In patients without contraindications, a short course of corticosteroids (e.g., 40 mg prednisolone/day, tapered over 3 weeks) may be added to antiviral therapy. Compared with ACV therapy alone, addition of corticosteroids has been shown to improve comfort levels (pain reduction during the acute phase) following herpes zoster, although its efficacy in reducing subsequent risk of postherpetic neuralgia remains uncertain. Currently, zoster vaccination is the primary approach to decreasing zoster incidence and the associated risk of developing post-herpetic neuralgia. A new vaccine (“Shingrix”) containing VZV glycoprotein E and adjuvant has recently been licensed. In phase III trials the vaccine (IM ×2 doses 2 months apart) reduced the incidence rate of shingles from 9.1/1000 person-years to 0.3 per 1000 person years in individuals over the age of 50 followed for a median of ∼3 years ( ). Vaccine efficacy was estimated at between 96.6% and 97.9% for all age groups. The safety profile was excellent, although local injection site and systemic reactions (grade 3) were common (17%). This vaccine is now recommended for all adults over the age of 50 regardless of their prior VZV vaccination history or history of shingles. Because this is not a live attenuated vaccine it can also be used safely in immunocompromised individuals.

Human Cytomegalovirus

HCMV is a ubiquitous virus that causes acute infections with a worldwide distribution resulting in seropositivity in the overwhelming majority of the population. The virus spreads from person to person through direct contact with bodily secretions. Like other herpesviruses, HCMV is never completely cleared from the host and remains latent for the life of the host with periods of persistent or sporadic shedding. Shed virus is an important source of virus transmission. As a rule, infection with HCMV in immunologically normal hosts is clinically silent but can occasionally cause self-limited, febrile, mononucleosis-like illness ( ). Severe HCMV disease is most commonly associated with congenital fetal infection or infection in immunocompromised hosts. When disease does occur, HCMV-related neurological complications include retinitis, encephalitis, polyradiculomyelopathy, neuropathy, and Guillain-Barré syndrome ( ). In immunocompetent hosts, HCMV may rarely cause asymptomatic infection, a mononucleosis syndrome, aseptic meningitis, or the Guillain-Barré syndrome. HCMV encephalitis is rare in immunocompetent hosts beyond the neonatal period but is reported ( ). However, immunocompromised adults and developing fetuses are at high risk of developing CNS disease due to CMV. CMV infection of peripheral nerves, nerve roots, and spinal cord, particularly in patients with AIDS, causes ascending myeloradiculitis ( ).

Congenital human cytomegalovirus

HCMV infection is the most common human congenital infection and can cause severe injury to the infected fetus. Up to 75% of congenital HCMV infections are due to nonprimary maternal infection, implying that better screening procedures are needed to detect and prevent HCMV reactivation and fetal infection ( ). A review of 15 studies published from 1970 to 2004 that screened newborns for HCMV reported that 12.7% of 117,986 newborns with congenital HCMV infection were symptomatic at birth ( ). The mortality for newborns with symptomatic disease is 10%–30%, and up to 90% of survivors will have neurological sequelae. The rate of sequelae in newborns with asymptomatic primary infection has been estimated at roughly 15% ( ), most commonly including microcephaly, poor feeding, lethargy, hypotonia, and seizures. In addition to congenital cases, there are cases that arise in the perinatal period as a consequence of passage through an infected birth canal or following breastfeeding. Persistent high levels of viral replication in the eye and brain of the developing fetus produce encephalitis, ependymitis, and retinitis, a pattern similar to that seen in patients with opportunistic HCMV infection in the setting of HIV infection. Pathologically, encephalitis occurs in a periventricular pattern and may cause polymicrogyria and hydrocephalus ( Fig. 78.3 ). CT scans show characteristic periventricular calcifications in 20%–30% of children with symptomatic HCMV infection. Retinitis with optic atrophy is seen in about 15% of those affected and results in characteristic hyperpigmented retinal scars. As noted, 90% of survivors of congenital HCMV infection have residual neurological sequelae including psychomotor retardation, learning delays, mental retardation, seizures, optic atrophy and retinitis, and hearing loss. Mild or subclinical congenital infections may also manifest later in childhood as sensorineural deafness or developmental delay. Congenital human HCMV infection is the most common, nonheritable cause of hearing loss in the United States.

Fig. 78.3, Human Cytomegalovirus Ventriculitis.

Diagnosis of congenital HCMV is made by identification of HCMV DNA using PCR testing of urine, saliva, or CSF during the immediate postnatal period. Because urinary excretion of HCMV can often persist during the first year of life, isolation of virus or viral DNA from urine may be useful in later diagnosis. HCMV inclusion-bearing cells may also be found in affected organs ( Fig. 78.4 ) and in stained preparations of urinary sediment and saliva. Serological studies may be difficult to interpret owing to transplacental transfer of antibody from the mother. Although detection of virus-specific IgM antibodies has been used to diagnose congenital HCMV, direct detection of viral markers is more accurate and preferred.

Fig. 78.4, Ballooned Cell with Eccentric Nucleus in Human Cytomegalovirus Encephalitis.

In a study evaluating the efficacy of IV ganciclovir therapy (4–6 mg/kg every 12 hours for 6 weeks) in neonates with symptomatic congenital HCMV infection, 69% of treated children, compared to only 39% of untreated children, showed improvement in brainstem auditory evoked potentials. More significantly, none of the ganciclovir-treated children, compared to 42% of those not treated, showed worsening hearing loss over the initial 6 months post infection ( ). In another study, 6 weeks of ganciclovir therapy in neonates with symptomatic congenital CMV infection involving the CMS led to fewer developmental delays at 6 and 12 months as compared to untreated infants ( ). While 6 weeks of therapy was beneficial, it was not known how long infants with congenital CNS HCMV disease should be treated. In a randomized, placebo-controlled study of 96 neonates, infants treated with valganciclovir (16 mg/kg, orally, twice daily) for 6 months were more likely to have normal hearing (73% vs. 57%) at 12- and 24-month follow-up and had improved neurodevelopmental scores when compared to infants treated with valganciclovir for 6 weeks and placebo for 4.5 months ( ). Thus, treating infants diagnosed with congenital HCMV infection for 6 months with valganciclovir may decrease the risk of hearing loss or neurodevelopmental abnormalities.

Cytomegalovirus in immunocompromised adults

HCMV can cause neuroinvasive infections in immunocompromised patients with hematopoietic stem cell transplants, solid organ transplants (SOTs), or in patients with acquired immunodeficiency syndrome (AIDS) ( ).

HCMV is an important pathogen following SOT and hematopoietic stem cell transplantation (HSCT). Risk factors for CNS disease in this group include delayed T-cell recovery, umbilical cord blood transplantation, graft versus host disease, and a history of recurrent HCMV viremia ( ). Antiviral prophylaxis for prevention of primary infection or recurrence is indicated when either or both donor (D+) and recipient (R+) are seropositive for HCMV. A phase 3, double-blind, randomized, placebo-controlled clinical trial recently showed that HSCT patients treated with letermovir prophylaxis (a new antiviral drug that inhibits the CMV-terminase complex) showed reductions in clinically significant CMV disease (37.5%) compared to the placebo group (60.6%) ( ). Use of valganciclovir or ganciclovir is often limited in the HSCT patient population due to the associated myelosuppressive side effects of treatment, so the availability of a less myelosuppressive alternative (letermovir) that can be more widely employed may decrease the risk of CNS disease in this patient population. Prior to the availability of letermovir, a preemptive treatment strategy was used in HSCT patients instead, defined as prevention of development of disease when reactivation has occurred. Patients were monitored for CMV viremia and started on therapy with antiviral treatment to prevent clinical CMV disease and minimize the toxic effects of antiviral drugs to engrafted bone marrow.

Despite recent advances, HCMV infection remains one of the most common complications affecting SOT patients. All patients undergoing SOT who are at risk for CMV disease receive universal prophylaxis, most commonly with valganciclovir starting within 10 days after transplantation and continuing for a period of 3–6 months.

HCMV encephalitis typically occurs in HIV+ patients with a CD4+ cell count of less than 50 cells/mm 3 , although patients with counts below 100/mm 3 are at increased risk of developing HCMV viremia ( ). In HIV patients with low CD4 counts in the era before highly active antiretroviral therapy (HAART), HCMV most commonly caused retinitis, esophagitis, and colitis and less commonly caused encephalitis, peripheral neuropathy, or polyradiculoneuritis. The increased use of HAART for the treatment of HIV has dramatically reduced the occurrence of HCMV viremia and disease.

HCMV encephalitis presents either as a microglial nodular encephalitis with acute onset of confusion and delirium, or as a more slowly progressive ventriculoencephalitis characterized by confusion and cranial nerve palsies. There is a broad pathological spectrum of HCMV infection of the brain, ranging from scattered microglial nodules to widespread necrotizing leukoencephalopathy or focal necrosis deep in the parenchyma.

Diagnosis of HCMV is largely dependent on quantitation of viral load in the blood using PCR and evidence of end organ disease or involvement consistent with HCMV infection. CSF HCMV PCR has a reported sensitivity of 82% and specificity of 99% in AIDS patients with CNS disease due to HCMV ( ). In immunosuppressed patients, a wide spectrum of neuroimaging results are reported, ranging from normal findings to detection of generalized atrophy, periventricular abnormalities, and focal discrete white-matter lesions. The most characteristic MRI finding is periventricular increased signal on T2-weighted images and ependymal enhancement following gadolinium administration on T1-weighted images. In advanced HIV disease, HCMV can manifest as polyradiculitis with diffuse enhancement of cauda equina nerve roots and is often associated with concurrent HCMV infection elsewhere in the body ( ). Many patients with this syndrome have an almost pathognomonic CSF profile of neutrophilic pleocytosis with a low glucose concentration. The diagnosis is confirmed by PCR amplification of HCMV DNA in CSF.

Ganciclovir, foscarnet, and cidofovir all have efficacy against HCMV in vitro and in some clinical settings in vivo . In all immune suppressed patients, immune reconstitution following introduction of HAART or decreasing immune suppressing medications may help control HCMV replication and disease. General recommendations for immunocompromised patients, including those with AIDS, are for initial antiviral therapy with IV ganciclovir (5 mg/kg every 12 hours for 2–3 weeks), followed by maintenance dosing with either IV ganciclovir (5 mg/kg/day, 5 days/wk) or oral valganciclovir (900 mg daily) for at least an additional 4 weeks. For SOT patients, oral valganciclovir or intravenous ganciclovir treatment are associated with similar long-term outcomes based on the VICTOR study conducted in adult renal, liver, heart, and lung transplant recipients ( ). However, patients with severe or life-threatening HCMV disease were excluded from this study, so intravenous ganciclovir is still recommended for CNS disease in this population. A full discussion of CMV resistance and treatment is beyond the scope of this chapter. Briefly, foscarnet should be reserved for treatment of ganciclovir-resistant HCMV because of its nephrotoxicity and IV administration ( ). Foscarnet dosage is 60 mg/kg IV, 3 times daily for 2–3 weeks for initial therapy, then 90–120 mg/kg IV daily for maintenance. There are limited clinical data on the use of cidofovir for treatment of HCMV CNS disease ( Table 78.8 ). In transplant cases, CNS disease is treated with IV ganciclovir, 5–7.5 mg/kg per dose 2 or 3 times daily with the addition of IV foscarnet 90 mg/kg twice daily for refractory cases ( ).

TABLE 78.8
Immunotherapy of Viral Infections
Live vaccines include yellow fever, measles, mumps, rubella, smallpox, polio, and varicella-zoster virus. Killed vaccines include polio, rabies, influenza, arboviruses (Japanese encephalitis, tickborne encephalitis, Kyasanur Forest, Rift Valley, eastern equine encephalitis, western equine encephalitis, and Venezuelan equine encephalitis). Soluble protein includes hepatitis B.
Immunotherapy Class Immunotherapy Dose or Route Indications
Specific hyperimmune globulin VZIG (varicella-zoster) One vial (125 U) per 10 kg of body weight, intramuscularly (IM) Postexposure prophylaxis in hypogammaglobulinemic patient (pt)
RIG (rabies) Human RIG, 20 IU/kg (inject as much as possible in area of wound and remainder IM at site distant from vaccine) Postexposure prophylaxis
Human cytomegalovirus hyperimmunoglobulin Intravenous (IV) Prophylaxis after bone marrow transplantation
Central European encephalitis hyperimmunoglobulin IM Postexposure prophylaxis following multiple tick bites in endemic area
Measles hyperimmunoglobulin IV Treatment of measles inclusion body encephalitis in immunocompromised pt
Polyvalent immune globulin IVIG IV Treatment of chronic enterovirus meningoencephalitis in hypogammaglobulinemic pt
IVIG IV Treatment of CNS parvovirus B19 infection
IVIG IV Treatment of human T-cell leukemia virus I myelopathy
Monoclonal Antibody Bamianivimab (Lily, EUA)
REGN10933/10987 Mab Cocktail (Regeneron, EUA)
IV COVID-19
Cytotoxic T cells Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes Prophylaxis of EBV lymphoproliferative disease in bone marrow transplant recipients
Corticosteroids Dexamethasone PO/IV 6 mg/d x 10d EUA for hospitalized COVID-19 pts
CNS , Central nervous system; EUA , FDA Emergency Use Authorization; IV , intravenous; IVIG , intravenous immunoglobulin.

Epstein–Barr Virus

Primary EBV infection may be asymptomatic, present as a nonspecific febrile illness, or as the infectious mononucleosis syndrome with cervical lymphadenopathy, exudative pharyngitis, and splenomegaly. The pathogenesis of EBV-associated CNS disease remains uncertain because virus, viral antigen, or viral nucleic acid are only rarely isolated directly from CNS tissue in patients with encephalitis or myelitis, raising the possibility that at least some CNS manifestations may be post- or para-infectious immune-mediated phenomena. However, recent studies suggest that patients with EBV-associated neurological disease frequently have EBV DNA that can be amplified from CSF, and that this is frequently associated with amplifiable viral RNA consistent with lytic viral replication ( ).

Nervous system disease occurs in 1%–7% of EBV infectious mononucleosis cases and can manifest as meningitis, encephalitis, acute hemiplegia, Alice in Wonderland syndrome, cerebellitis, cranial neuropathy, transverse myelitis, Guillain–Barré syndrome, or as small-fiber sensory or autonomic neuropathy syndromes ( ). The most common symptomatic EBV-associated CNS infection is meningitis. Cases of asymptomatic laboratory-defined meningitis probably vastly exceed symptomatic cases; up to 25% of patients with acute infectious mononucleosis may have a CSF pleocytosis despite the absence of signs or symptoms of meningitis. There are no unique features of EBV meningitis, although the presence of atypical lymphocytes in CSF may suggest the diagnosis. Diagnosis is typically made by amplification of EBV DNA by CSF PCR. In rare cases, EBV-specific IgM antibodies can be detected in CSF. Attempts to isolate virus from CSF are almost invariably negative. The presence of serum serologies indicative of recent primary infection supports the diagnosis (e.g., IgM antibodies against viral capsid antigen [VCA], presence of antibodies against early antigen [EA], but not Epstein-Barr nuclear antigen [EBNA]).

In rare cases, EBV is associated with frank encephalitis, presenting with altered consciousness including coma, seizures, and focal neurological signs and symptoms. Cases of EBV CNS disease may occur before, during, or after infectious mononucleosis or even in its absence. In one series of 21 cases of EBV encephalitis in children ( ), only one patient had classic infectious mononucleosis; the remainder had a nonspecific prodrome that included fever (81%) and headache (66%). Seizures occurred in 48%, and 57% had EEGs with a diffusely slow background. CSF pleocytosis (81%) and MRI abnormalities (71%) were common. Mortality was 10%, with 80% neurologically normal at follow-up and an additional 10% having mild deficits. EBV encephalitis may mimic HSE, and in the CASG studies, EBV encephalitis accounted for about 8% of the HSV-negative cases of focal encephalitis in which an etiology was established. In a registry of childhood (ages 3–17) encephalitis cases at a large children’s hospital, EBV accounted for 6% of total cases ( ).

EBV myelitis can occur as an isolated syndrome or in association with meningoencephalitis. Most patients have CSF mononuclear pleocytosis and diagnosis is made by amplification of EBV DNA from CSF or by appropriate serological testing (see later discussion). Myelitis typically follows mononucleosis, although in some patients the symptoms of the initial infection may be mild or even absent. A variety of clinical forms have been reported and include transverse myelitis, myeloradiculitis, and a poliomyelitis-like syndrome of acute flaccid paralysis. MRI may show increased T2-weighted intramedullary signal or evidence of cord swelling. Nerve root enhancement has been reported in patients with myeloradiculitis. No controlled treatment trials are available, although isolated cases have been treated with IV ACV and ganciclovir with or without the addition of corticosteroids ( ).

Specific diagnosis of CNS EBV disease requires either amplification of EBV DNA from CSF or serological studies indicative of acute infection. In serum, the presence of EBV VCA IgM antibody is indicative of recently acquired active EBV infection. The presence of EBV VCA IgG antibody and antibody against EA in the absence of antibodies against EBNA antibodies is also indicative of recent infection. The presence of serum IgG VCA and EBNA antibodies indicates remote infection, and these antibodies persist for the lifetime of the infected individual.

CSF PCR for EBV is positive during the acute phase of illness in children with infectious mononucleosis and neurological complications such as transverse myelitis, meningoencephalitis, and aseptic meningitis. CSF PCR is negative in EBV-seropositive individuals in the absence of CNS infection. However, positive EBV PCR may be seen in patients with evidence of other viral or nonviral CNS infection, raising the possibility that these infections may trigger viral reactivation in the absence of EBV-associated disease. EBV has been one of the most frequent agents associated with dual-positive CSF PCR testing and may not always correlate clinically with the presence of CNS infection known to be caused by this virus ( ).

There are no randomized controlled trials for any antiviral or immunosuppressive agent for the treatment of EBV-associated neurological disease. Supportive care is important, and death due to EBV neurological disease is uncommon. ACV inhibits EBV DNA polymerase in vitro , although viral production returns to normal levels after the drug is stopped, even after 11 months of therapy, because ACV does not affect the latent viral burden and latent virus can reactivate and replicate using host-dependent enzymes (Tselis, 2014). There are few studies supporting the use of ACV for EBV encephalitis, and the data supporting efficacy of other antivirals, including ganciclovir and foscarnet, are also limited. Treatment with intravenous immunoglobulin (IVIG) may improve EBV-associated small-fiber sensory or autonomic neuropathies if treatment begins during acute disease.

Human Herpesvirus Type 6

HHV-6 was first isolated in 1986 from human peripheral blood mononuclear cells of patients with lymphoproliferative disorders. Two variants, HHV-6A and HHV-6B, are known. Primary infection with HHV-6 usually occurs during infancy, producing exanthem subitum (or roseola) or a syndrome of generalized lymphadenopathy. HHV-6 infects a broad range of cell types including T-cells, monocytes, dendritic cells, oligodendrocytes, microglia, and astrocytes. Primary HHV-6 infection may cause febrile seizures or acute meningoencephalitis in children. Cases of focal encephalitis in immunocompetent patients have been attributed to HHV-6, as has a syndrome of acute limbic encephalitis occurring in both SOT patients (post-transplant acute limbic encephalitis [PTALE]) and those receiving HSCT ( ) ( Fig. 78.5 ). MRI demonstrates symmetric hyperintensities in the medial temporal lobes. The role, if any, for HHV-6 in inducing medial temporal sclerosis temporal lobe epilepsy ( ) and as a co-factor in other neurological disorders including multiple sclerosis remains unproven.

Fig. 78.5, Concurrent Human Herpesvirus Type 6 and Human Immunodeficiency Virus Disease.

There are no approved therapies for HHV-6 infection. HHV-6 isolates generally resemble HCMV in their in vitro susceptibility to antiviral drugs by exhibiting resistance to ACV and sensitivity to ganciclovir and foscarnet ( ).

Herpes B Virus (Cercopithecine Herpesvirus 1)

Herpes B virus of Old World monkeys is highly pathogenic to humans, and untreated infection has an estimated 70% mortality. Ocular, oral, and genital secretions of monkeys, as well as CNS tissues and CSF, are potentially infectious. Disease is transmitted by direct contact with the virus, usually by animal bite or by virus-containing fomites. One reported fatal case of Herpes B virus infection was due to mucosal splash exposure ( ).

Fever, myalgia, herpetiform rash, meningismus, and early-stage nystagmus or diplopia are followed by an ascending encephalomyelitis causing flaccid paralysis, urinary retention, and signs of CNS involvement including seizures, progressive lethargy, and coma. Diagnosis is made by isolation of virus or viral DNA from wound or contact sites and/or by demonstration of seroconversion between acute and convalescent sera.

Preventive measures for primate workers include use of protective eyewear including side shields and a mask to protect from mucous membrane exposure. Should exposure occur, the most critical period for prevention of infection is within the first few minutes after exposure. Immediate cleansing of the affected skin surface should be undertaken by soaking or scrubbing the contact area with povidone-iodine, chlorhexidine, or detergent soap for 15 minutes. Eyes or mucous membranes should be immediately irrigated with sterile saline solution or water for 15 minutes. Postexposure prophylaxis within 5 days of exposure is recommended with oral valacyclovir, 1 g 3× daily for 2 weeks; oral ACV, 800 mg 5× daily for 2 weeks; or famciclovir, 500 mg 3× daily for 2 weeks. Symptomatic patients without CNS symptoms should be treated with either IV ACV (15 mg/kg every 8 hours) or IV ganciclovir (5 mg/kg every 12 hours). When CNS symptoms are present, most experts advise treatment with IV ganciclovir in preference to ACV, although comparative trials are lacking. Treatment should be continued until symptoms resolve and the results of two sets of cultures are negative for B Herpes virus after being held for 10–14 days. Following IV therapy, therapy is switched to oral valacyclovir, famciclovir, or ACV in the dosages used for postexposure prophylaxis. Because of the potential risk of reactivation of latent virus and development of CNS disease, indefinite maintenance of oral suppressive therapy following documented infection may be warranted.

Polyomaviruses (JC) and Progressive Multifocal Leukoencephalopathy

Progressive multifocal leukoencephalopathy (PML), a subacute demyelinating disease of the CNS, is a result of infection of oligodendrocytes by the polyomavirus, JC virus (JCV) ( ). Specific criteria for establishing the diagnosis have been developed. Definitive diagnosis generally requires either neuropathological confirmation or the presence of consistent radiographic and clinical features with the associated demonstration of JCV DNA in CSF by PCR ( ). Seroepidemiological studies indicate that asymptomatic primary infection with JCV occurs in childhood, and that by adult life roughly 55%–85% of the population is seropositive. Following primary infection, JCV becomes latent in sites including kidney, bone marrow, and tonsil. It is unclear whether CNS latency occurs. Reactivation in immunocompetent hosts usually takes the form of asymptomatic viruria. In the setting of impaired cell-mediated immunity, including immunosuppressive drug treatment, lymphoproliferative disorders, chronic infectious or inflammatory diseases such as tuberculosis and sarcoidosis, and most importantly AIDS, virus can reactivate to produce PML. In most modern series, over 80% of PML patients have underlying HIV infection. Treatment of patients with several immunomodulatory biologicals, as exemplified by natalizumab, has been linked to increased risk of developing PML ( ). The first reports involved patients treated for multiple sclerosis (2 cases) or Crohn disease (1 case) with natalizumab (Tysabri), a humanized monoclonal antibody that blocks lymphocyte binding to α 4 -integrin and inhibits trafficking into the CNS ( ). Over 750 cases of natalizumab-associated PML have now been reported (January, 2018), with a global risk of ∼4.2/1000 treated patients. Key risk factors associated with increased risk of PML development include the presence of JC virus antibodies and higher values of the JCV Ab index, the use of prior immunosuppressive therapy (e.g., mitoxantrone, azathioprine, cyclophosphamide, methotrexate, mycophenolate mofetil), and the duration of natalizumab therapy ( ). Ninety-nine percent (205 of 207) of patients who developed natalizumab-associated PML were JCV seropositive at least 6 months before the onset of PML (Biogen Idec Safety data). The risk of developing PML among natalizumab-treated JCV seronegative individuals is ∼0.07 PML case/1000 patients (95% CI 0.00–0.40). By contrast, in patients who are JCV seropositive but have not received prior immunosuppressive therapy the risk climbs as high as 1.7% (17/1000) in those who have received 72 months of therapy. In patients who are both JCV seropositive and have received prior immunosuppressive therapy, risk increases to 2.7% (27/1000) in those with 72 months of natalizumab exposure (see and Biogen Idec Safety Data).

At least 124 cases of PML have been reported in patients receiving rituximab (Rituxan), an antibody directed against CD20 on B cells. The majority of cases occurred in patients being treated for non-Hodgkin lymphoma, although cases have also been reported in patients receiving rituximab for other diseases including systemic lupus erythematosus, rheumatoid arthritis, and autoimmune hemolytic anemia. Each of these diseases has been associated with PML in the absence of rituximab therapy, and patients were often receiving multiple immunosuppressive medications. A “disproportionality analysis” based on expected versus actual cases reported to the US Adverse Events Reporting System (AERS) suggested that immunosuppressive drugs associated with the highest risk of PML and their associated “reporting odds ratio” (ROR) included rituximab (ROR 73), Mycophenolate mofetil (ROR 23), natalizumab (ROR 22), azathioprine (ROR 22), cyclophosphamide (ROR 18), efalizumab (ROR16), methotrexate (ROR 9), tacrolimus (ROR9), and cyclosporine (ROR 6). The ROR was determined from estimates of number of reported cases and number of exposed individuals ( ). In addition, three or four cases of PML have occurred in patients being treated for chronic plaque psoriasis with efalizumab (Raptiva), a monoclonal antibody directed against the CD11a lymphocyte antigen. The magnitude of the risk was estimated at approximately 1 in 400 treated patients and resulted in this drug being removed from the market.

The likely pathogenesis of PML includes reactivation from an extraneural primary site, dissemination of virus to the CNS through the bloodstream, and subsequent productive lytic infection of oligodendrocytes to induce demyelination ( ). Infection of astrocytes is abortive, although resulting in striking and characteristic bizarre enlarged cells. Neuronal infection does not occur in classic PML but variant forms of JCV encephalitis and JCV cerebellar granule cell infection with distinct clinical presentations have been described ( ). Rearrangements in the regulatory region of the viral genome appear to play a key role in neuropathogenesis insofar as viruses lacking these regulatory region rearrangements have not been isolated from PML brain specimens.

Onset of PML is subacute, with signs and symptoms of multifocal asymmetrical white-matter involvement. In non-AIDS-associated PML, early lesions tend to be in subcortical white matter of the occipital lobes, causing visual-field deficits or cortical blindness. Motor weakness, behavior changes, cognitive impairment, cerebellar ataxia, dysarthria, and sensory abnormalities also are seen, whereas headache, seizures, and extrapyramidal syndromes are rare. The disease progresses to dementia as the number of lesions increases.

CSF cell counts and protein levels are usually normal. Neuroimaging results help suggest the diagnosis. MRI studies show focal or multifocal lesions of subcortical white matter, sometimes involving the cerebellum, brainstem, and spinal cord, without mass effect or contrast enhancement. Fluid-attenuation inversion recovery (FLAIR) sequences, which remove CSF signals, are particularly good for demonstrating paraventricular disease ( Fig. 78.6 ). White-matter lesions are larger and more confluent than those of multifocal leukoencephalitis of VZV. Patients with natalizumab-associated PML typically have large (>3 cm), subcortical, T2 and diffusion hyperintense and T1 hypointense lesions. The lesions have a sharp border on their cortical side and an ill-defined border at their white-matter side. In distinction to the experience with HIV-PML, ∼40% of patients have some contrast enhancement on T1-weighted gadolinium-enhanced images ( ). Lack of CSF pleocytosis and an indolent course distinguish PML from ADEM. Brain biopsy definitively establishes the diagnosis by showing characteristic changes, including demyelination, bizarre astrocytes, and oligodendrocytes with enlarged nuclei that contain inclusion bodies, as well as viral particles and antigen or viral DNA ( ). JCV has never been cultured from CSF, but JCV DNA may be detected in CSF using PCR amplification. Finding JCV DNA in CSF by PCR in the appropriate clinical setting with appropriate imaging abnormalities is also diagnostic of PML ( ) and obviates the need for brain biopsy. Although the specificity of CSF PCR for JCV in the appropriate clinical setting approaches 100%, its sensitivity may be as low as 75%, reflecting variances in the amount of viral load in the CSF in different conditions. For example, 57% of Tysabri-associated PML cases were reported to have fewer than 500 copies of JCV DNA per mL, which is close to the sensitivity limit of some commercial assays ( ).

Fig. 78.6, Progressive Multifocal Leukoencephalopathy Brain Magnetic Resonance Imaging Lesion Patterns.

No specific therapy is available. Isolated reports of benefit from cytarabine were not confirmed in a randomized prospective clinical trial ( ). Cidofovir was also found not to be of significant clinical benefit in HIV-associated PML in a prospective clinical trial ( ). Interferon alfa has been reported to be of benefit in case reports but has not been tested in clinical trials. A study showing that JCV binds the 5HT receptor in cultured cells led to the use of mirtazapine, a serotonin receptor blocker, in patients with PML ( ). However, 12-month follow-up in patients with PML treated with mirtazapine showed no evidence of improved survival ( ). A screen of chemical compounds found that mefloquine, a malarial drug, inhibits JCV replication ( ). A multicenter clinical trial investigating the role of mefloquine treatment of JCV in PML patients did not show significant clinical benefit ( clinicaltrials.gov identifier NCT00746941) ( ). A significant new development in therapeutics has been the use of virus-specific T-cells to treat cases of PML. This typically involves isolating T-cells from an HLA-matched donor, amplifying the JCV antigen-specific cytotoxic T lymphocyte (CTL) population, or the closely related BK-virus-specific CTLs by ex-vivo stimulation of these cells with JCV or BKV proteins or peptide mimics, and adoptive transfer of the amplified cells into a patient. No controlled clinical trials are yet available, although several small studies have shown clinical benefit or disease stabilization, and reduction in viral load in treated patients ( ). Until the effectiveness of adaptive T-cell therapy can be confirmed in controlled clinical trials, the most effective therapy for PML remains treatments which reverse any underlying immunosuppression. Unfortunately, immune reconstitution is often associated with an immune reconstitution inflammatory syndrome (IRIS) that causes paradoxical clinical and radiographic worsening as the host mounts an effective cellular immune response to the JC virus present in PML lesions. No controlled clinical trials of treatment of IRIS are currently available; however, cases are often treated with high-dose intravenous corticosteroids ( ). A single intriguing case report describes successful use of the small-molecule CCR5 antagonist maraviroc rather than corticosteroids ( ) to prevent development of IRIS in an HIV woman with natalizumab-associated PML. It was suggested maraviroc acted by inhibiting trafficking of CCR5+ immune-cell subsets into the CNS and preventing their immunopathological role in disease initiation.

Adenovirus

Adenoviruses cause acute respiratory disease in children and adults in crowded settings such as military recruits, along with conjunctivitis, hemorrhagic cystitis, and gastroenteritis. The most common associated CNS diagnosis is febrile or afebrile seizure in children under 5 years. Meningoencephalitis or unilateral deafness coincident with nasopharyngeal infection are rare complications in normal hosts. Immunosuppression is a risk factor for encephalitis, with fatal meningoencephalitis reported in AIDS and bone marrow transplant patients and encephalomyeloradiculitis reported in an umbilical cord stem cell transplant recipient ( ). Clinical and histopathological features of adenovirus disease may resemble those of HCMV disease, such as adenovirus encephalitis and ependymitis in a child with AIDS (Anders et al., 1990). Diagnosis is by PCR or isolation of virus from extraneural sites, by serology, or by identification of virus or DNA in brain tissue or CSF. Adenovirus-infected neurons and glia have enlarged nuclei with amphophilic inclusions and a thin rim of cytoplasm, referred to as “smudge” cells. Diagnosis of adenoviral infection is complicated by the existence of 51 viral serotypes. Different serotypes have different tissue tropisms. Serotypes 1, 2, 3, 4, 5, 6, 7, 11, 12, 26, 31, 32, and 41 in mixture with 49 have been implicated in CNS infection ( ). Cidofovir is used for severe adenovirus infections; viral clearance and patient survival is aided by lymphocyte reconstitution ( ).

Parvovirus

Acute infection with B19 parvovirus causes the febrile exanthematous illness, fifth disease (erythema infectiosum, or “slapped cheek rash”) in childhood, transient aplastic crises (particularly in immunocompromised and sickle-cell patients), and small-joint arthritis in adults. Parvovirus B19 infections have been described as a potential trigger for various autoimmune disorders including necrotizing vasculitis resembling granulomatosis with polyarteritis in association with meningoencephalitis ( ). According to a systematic review of 129 cases of Parvovirus B19 (PVB19)-associated neurological disorders, 50 (39%) were diagnosed with encephalitis and only 14% were immunocompetent adults ( ). Patients can develop neurological disease in the absence of systemic symptoms such as rash, anemia, and arthropathy (Jun et al., 2017). Neurological manifestations have included encephalitis, chorea, stroke, optic neuropathy abnormal pupillary reflexes, brachial plexitis, and autonomic, sensory, or motor neuropathies and recurrent paresthesias. In CNS disease, PVB19 DNA and less often IgM can be detected in CSF. IVIG speeds clearance of viremia in immunocompromised patients and rescues from rituximab-induced hypogammaglobulinemia and parvovirus infection. IVIG with or without steroids is increasingly used in all CNS disease cases ( ).

Orthopoxviruses

Monkeypox

Monkeypox became a newly emergent poxvirus zoonosis in North America in June 2003 when an outbreak of human monkeypox transmitted from infected exotic pets was identified in the midwestern United States. The source of the US outbreak was traced to native prairie dogs housed with rodent pets from Africa. The index animal source was imported wild rodents from Africa, where the disease is endemic ( ). Human monkeypox was first identified in Zaire, now the Democratic Republic of Congo, in 1970 toward the end of smallpox eradication efforts in Africa. In western and central African countries, where it continues to produce sporadic cases clinically similar to smallpox, spread is by contact with infected animals through biological fluids, bite, bush meat consumption, or by fomite, droplet, or direct contact with infected humans. Suspected reservoir host species are the rope squirrel and Gambian pouched rat. The 37 confirmed human infections in the June 2003 outbreak represented the first time human monkeypox infection was documented in the Western Hemisphere ( ). Most cases were associated with a mild, self-limited febrile rash illness. However, one 6-year-old developed fever, rash, and encephalitis, characterized by seizures, depressed mental status, CSF pleocytosis, normal protein and glucose, and MRI showing diffuse edema, meningeal enhancement, thalamic and parietal cortex signal abnormalities. Monkeypox-specific IgM antibodies were present in serum and CSF. She made a full recovery. In this case, diagnosis was by serum and CSF IgM and IgG, skin lesion viral culture, PCR, and immunohistochemistry (IHC) testing. Viral cultures and PCR in CSF were negative.

Management is strict isolation and supportive care. Cidofovir, other selective antipoxvirus drugs such as oral tecovirimat ( ), and vaccinia immune globulin are investigational treatments. Smallpox vaccine protects against monkeypox both by preventing disease and by decreasing disease severity ( ). For both monkeypox and smallpox, vaccination within 3 days of exposure lessens the severity of symptoms in the majority of individuals. Ultimately, the cases resulted in an importation ban and limitations on movement and sale of African rodents in the United States and restrictions on wild release of these species and prairie dog pets.

Monkeypox has become the most important orthopoxvirus infection of humans worldwide due to population loss of vaccinial protection against smallpox in the smallpox post-eradication era. Early symptoms are fever and lymphadenopathy (a distinguishing feature), followed by a maculopapular rash that begins on the face and evolves through papular, vesicular, pustular, and umbilicated lesion stages. Face, palms, and soles are most commonly affected. Antipox antibodies in an ill unvaccinated individual support infection.

Since 2003, there had been no cases of human monkeypox outside Africa until two cases were diagnosed in the UK in 2018 in travelers from Nigeria. These were the first reports of travel-associated cases diagnosed outside Africa ( ).

Smallpox and Smallpox Vaccination

There are two forms of smallpox, variola major and variola minor, with differing outcomes. Mortality from variola major has been estimated at 30%, and from variola minor approximately 1%. Neurological complications of acute smallpox are uncommon but include stupor, coma, seizure, headache, hemiparesis, incontinence, or flaccid paralysis ( ).

Compulsory vaccination was discontinued in US civilians in 1973 and among military personnel in the late 1980s. The world was declared free of smallpox infection in 1980, but the United States reinstated vaccination against smallpox among military personnel in 2002 and among selected civilian groups in January 2003 as the National Smallpox Vaccination Program, vaccinating military and first responders who could be exposed in terror attacks. Smallpox vaccination has been inoculation with a preparation of vaccinia, a relatively benign orthopoxvirus extracted from calf lymph. Now, Dryvax, made from a heterogenous pool of vaccinia virus clones grown on calf skin, is being replaced by ACAM2000, a cell culture product of single plaque-purified vaccinia virus clone derived from Dryvax for use in the United States. Additional vaccines are in the CDC Strategic National Stockpile. Historically, routine smallpox vaccinations have been 90% effective in preventing smallpox and more than 99% effective in preventing fatalities ( ).

A major complication of vaccination has been post-vaccination CNS disease: post-vaccinal encephalitis (PVE) or post-vaccinal encephalomyelitis (PVEM) ( ), more frequent after primary vaccination or in patients under 1 year of age. After primary vaccination, reported rates vary from 1 in 4000 to 1 in 80,000, and after revaccination, from 1 in 50,000 to 1 in 450,000. Neurological complications, particularly PVEM, account for the majority of vaccination-related deaths; PVEM fatality rates range from 10% to 50%. There are several forms of encephalitic complications: an acute encephalopathy with brain swelling in infants, a perivenular demyelinating disease (ADEM) in vaccinees older than 2 years of age (in which intrathecal vaccinia IgM or IgG production may be found), and a neuroinvasive vaccinial encephalitis or encephalomyelitis in which laboratory evidence of CNS viral replication is found. In the latter, vaccine-strain vaccinia virus is isolated from brain or CSF of some cases of PVEM ( ). Permanent neurological sequelae are estimated in 16% of recovered PVEM cases. Other neurological syndromes following smallpox vaccination have included Guillain-Barré syndrome, Bell palsy, other cranial neuropathies, a poliomyelitis-like syndrome, aseptic meningitis, and transverse myelitis. Newer vaccine efforts through production in cell culture are hoped to provide desired immunity with fewer complications ( ).

Administration of antivaccinia gamma globulin at the time of vaccination is thought to reduce occurrence of PVEM and has been used for treating at least 1 PVEM case. A recently vaccinated patient with rapidly progressive encephalitis diagnosed as severe PVE with ADEM based on orthopoxvirus IgM positive CSF (negative CSF viral cultures) was treated with IVIG, corticosteroids, and 1 dose of 400,000 units IV vaccinia immunoglobulin (VIG) and made a full recovery ( ).

As for safety of vaccination of individuals with known neurological illness, there have been anecdotal reports of worsening of seizures, recurrence of MG, and relapses of MS ( ).

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here