Total Pageviews

Wednesday 11 March 2020

Virus pelentung boleh serang dua kali


Varicella zoster virus infection

2017 Apr 5



Infection with varicella zoster virus (VZV) causes varicella (chickenpox), which can be severe in immunocompromised individuals, infants and adults. Primary infection is followed by latency in ganglionic neurons. During this period, no virus particles are produced and no obvious neuronal damage occurs. Reactivation of the virus leads to virus replication, which causes zoster (shingles) in tissues innervated by the involved neurons, inflammation and cell death — a process that can lead to persistent radicular pain (postherpetic neuralgia). The pathogenesis of postherpetic neuralgia is unknown and it is difficult to treat. Furthermore, other zoster complications can develop, including myelitis, cranial nerve palsies, meningitis, stroke (vasculopathy), retinitis, and gastroenterological infections such as ulcers, pancreatitis and hepatitis. VZV is the only human herpesvirus for which highly effective vaccines are available. After varicella or vaccination, both wild-type and vaccine-type VZV establish latency, and long-term immunity to varicella develops. However, immunity does not protect against reactivation. Thus, two vaccines are used: one to prevent varicella and one to prevent zoster. In this Primer we discuss the pathogenesis, diagnosis, treatment, and prevention of VZV infections, with an emphasis on the molecular events that regulate these diseases. For an illustrated summary of this Primer, visit: http://go.nature.com/14×VI1

Varicella zoster virus (VZV, also known as human herpesvirus 3) is a ubiquitous alphaherpesvirus with a double-stranded DNA genome. VZV only naturally infects humans, with no animal reservoir; its main targets are T lymphocytes, epithelial cells and ganglia. Primary infection causes varicella (chickenpox), during which VZV becomes latent in ganglionic neurons. As cellular immunity to VZV wanes with advancing age or in immunocompromised individuals, VZV reactivates to cause zoster (shingles). Zoster can be complicated by chronic pain (postherpetic neuralgia (PHN)) and other serious neurological and ocular disorders (for example, meningoencephalitis, myelitis, cranial nerve palsies, vasculopathy, keratitis and retinopathy), as well as multiple visceral and gastrointestinal disorders, including ulcers, hepatitis and pancreatitis, (Fig. 1). Antiviral drugs and vaccines against both varicella and zoster are available and are effective in treating and preventing VZV-induced disease.

1

VZV is highly communicable and spreads by the airborne route, with an extraordinarily high transmission rate in temperate countries. Traditionally, the virus was thought to spread to others from the respiratory tract, but such evidence is scant. Instead, most virus comes from skin where it is highly concentrated in vesicles; skin cells and cell-free VZV are frequently shed and are probably the major source of infectious cell-free airborne virus,. Infected children without skin lesions are not contagious to others.
The highly efficient transmission of VZV assured that, before the introduction of the varicella vaccine, most children would contract varicella before 10 years of age. Varicella in children is usually self-limiting, although complications can be unpredictable, and long-lasting immunity follows once the patient recovers. Epidemics are also self-limiting because the high rate of transmission and disease-induced immunity deplete the pool of susceptible individuals. Most older children and adults harbour latent wild-type VZV or vaccinetype VZV (vOka). Sporadic reactivation of VZV causes zoster and provides an evolutionary advantage for the pathogen by providing a source of infection in new, susceptible birth cohorts.
VZV occurs worldwide, but in some developed countries there is less concern for VZV than for other infectious agents, such as influenza virus, Ebola virus and multidrug-resistant staphylococci. However, even in countries where varicella vaccination is routine there has not been an eradication of VZV disease. Import of varicella from countries that do not vaccinate and zoster caused by reactivation of latent wild-type VZV or vOka can occur. Given the increasing number of immunocompromised individuals worldwide, it is important to maintain substantial levels of herd immunity against varicella in developed countries and to extend vaccination to developing countries. Recently, the WHO recommended “routine immunization of children against varicella in countries where varicella has an important public health impact”. In addition, because of the current anti-vaccine movement in some countries, it is also wise to maintain interest in VZV research and to improve current methods to prevent and treat varicella and zoster. In this Primer article, we summarize the diseases and complications of VZV infection, how latency develops, how and when to vaccinate and treat patients, and we highlight open research questions (BOX 1).

Varicella occurs worldwide and is endemic in populations of sufficient size to sustain year-round transmission, with epidemics occurring every 2–3 years. Viral genomic studies have identified five viral clades and their geographical distribution: clades 1, 3 and 5 are of European origin; clade 2 includes Asian strains such as the parental Oka strain, from which varicella and zoster vaccines were derived; and clade 4 contains African strains. Varicella epidemiology and disease burden have been studied primarily in developed countries. Although VZV seroprevalance data are becoming more widely available, additional data are needed on severe disease outcomes and deaths to better characterize the global health burden due to varicella, particularly in regions with high HIV prevalence, such as Africa and India.
Varicella incidence ranges from 13 to 16 cases per 1,000 persons per year, with substantial yearly variation. In temperate climates, age-specific varicella incidence is highest in preschool aged children (1–4 years of age) or children in early elementary school (5–9 years of age) with an annual incidence of greater than 100 per 1,000 children; as a result, >90% of people become infected before adolescence and only a small proportion (<5–10%) of adults remain susceptible,. In tropical climates, acquisition of varicella occurs at a higher overall mean age (for example, at 14.5 years in Sri Lanka), with a higher proportion of cases in adults,. Differences in varicella epidemiology between temperate and tropical climates might be related to the properties of VZV, for example, inactivation by heat and/or humidity, or factors affecting the risk of exposure.


Varicella shows a strong seasonal pattern, with peak incidence during winter and spring or during the cool, dry season,. Outbreaks occur commonly in settings where children congregate, such as childcare centres and schools, but can also occur in other age groups and settings, including hospitals, facilities for institutionalized people, refugee camps and military and correctional facilities.
Although varicella is usually a self-limiting disease, it can result in serious complications and death. In developed countries, ~5 out of 1,000 people with varicella are hospitalized and 2–3 per 100,000 patients die,. Serious varicella complications include bacterial sepsis, pneumonia, encephalitis and haemorrhage. Adults, infants and individuals who are severely immunocompromised are at higher risk of severe complications and death. Varicella acquired in the first two trimesters of pregnancy causes severe congenital defects in the newborn in ~1% of affected pregnancies.


VZV infection and replication

Primary infection

Following transmission to susceptible hosts, VZV proliferates in the oral pharynx (tonsils), infects T cells that enter the circulation and disseminate virus to the skin and possibly other organs; infection is at first controlled by innate immunity,.
VZV can remodel diverse T cell populations to facilitate skin trafficking. VZV DNA can be detected in T cells (viraemia) as early as 10 days prior to the occurrence of a rash and can persist for a week afterwards. Initially, innate immunity delays viral multiplication in the skin, which provides time for adaptive immunity to develop. Eventually, cutaneous innate immune responses are overcome by the virus, and there is substantial viral replication in the skin (and sometimes the viscera), resulting in the characteristic rash of varicella (FIG. 3). High titres of cell-free VZV develop in skin vesicles and transmit VZV to others. Important and unpredictable complications of varicella in previously healthy children include encephalitis, haemorrhagic manifestations, and bacterial superinfections involving skin, blood, bones and lungs.

2

When VZV reactivates, ganglia become necrotic and haemorrhagic. VZV proteins are found in neurons and non-neuronal cells, and ganglionitis is marked by the upregulation of MHC class I and II proteins and the infiltration of CD4+ and CD8+ T cells. Ganglionitis and CD8+ T cell infiltration can persist after zoster. Expression of late viral proteins, such as envelope glycoprotein E, shows that lytic VZV infection has occurred. However, the meaning of the immunocytochemi-cal detection of immediate early and early proteins in neurons during latency is controversial; these proteins have been detected in latently infected neurons. All latency-associated proteins are cytoplasmic, but become nuclear during productive infection. Immediate early and early proteins are also cytoplasmic in latently infected guinea pig neurons and translocate to the nucleus when reactivation is induced,,,.

Neurological complications of zoster

Zoster paresis

Manifestations of zoster paresis include arm or diaphragmatic weakness after cervical zoster, leg weakness after lumbar or sacral zoster and urinary retention after sacral zoster. MRI reveals involvement of both dorsal and ventral roots of spinal nerves. Rarely, cervical zoster paresis extends to the brachial plexus. The prognosis varies, but ~50% of patients recover completely.

Neuralgia

PHN, the most common complication of zoster, is defined as pain that persists for at least 3 months after rash onset. Three non-mutually exclusive theories have been proposed to explain the cause and pathogenesis of PHN. One is that the excitability of ganglionic or spinal cord neurons is altered during recovery. The second is that persistent productive VZV infection exists in ganglia, a notion that is supported by possible chronic ganglionic inflammation in PHN. A third theory is that PHN might be due to gene expression and protein production without virus replication but with disturbance of neuronal physiology.
Recently, certain strains of VZV were postulated to produce PHN by altering voltage-gated sodium channels, leading to altered excitability,. Isolated and then cultured VZV strains from patients with PHN and those with zoster but without PHN were applied to neuroblastoma cells that express fast and slow sodium channels. Voltage-clamp recordings from infected neuroblastoma cells revealed that the amplitude of the sodium current was greater in cells infected with VZV isolated from those with PHN than in cells from those without PHN. Increased sodium current is associated with neuropathic pain; thus, VZV-induced increases in sodium currents could have a role in PHN. These experiments suggest that PHN might be partly attributable to the particular strain causing zoster. This intriguing possibility clearly needs confirmation and further research.
Virological analyses of ganglia from patients with PHN are lacking. One study reported the detection of VZV DNA in peripheral blood mononuclear cells (PBMCs) for up to 8 years after zoster in 11 out of 51 patients with PHN but not in controls. A case report described a correlation between pain and VZV DNA detection in PBMCs in an immunocompetent elderly woman with PHN. After treatment with famciclovir (a guanosine analogue antiviral drug), pain resolved and the PBMCs no longer contained VZV DNA.
If PHN is caused by persistent VZV replication in neurons, antiviral treatment might decrease its severity. Treatment with oral antiviral agents reduces pain associated with acute zoster; however, this acute treatment has not reduced the incidence, severity or duration of chronic pain of immunocompetent patients with PHN. Acyclovir improved symptoms in 1 out of 10 patients with PHN, whereas valaciclovir improved symptoms in 8 out of 15 patients. Proof of a positive effect of antivirals on PHN would require a randomized controlled study of patients with PHN. Most studies, however, have not found antiviral therapy to be effective in the treatment of PHN and regulatory authorities do not recommend antivirals to treat this condition.

VZV meningoencephalitis

Acute VZV infection may present as meningitis or meningoencephalitis with or without rash. Detection of VZV DNA and antibodies in cerebrospinal fluid has confirmed VZV as a cause of aseptic meningitis, meningoradiculitis and cerebellitis.

VZV vasculopathy

A serious complication of VZV reactivation is infection of the cerebral arteries (VZV vasculopathy), which causes ischaemic and haemorrhagic stroke. The incidence of VZV vasculopathy is unknown. In children, up to one-third of ischaemic arteriopathies are associated with varicella. In adults, the risk of stroke is increased by 30% within 1 year of zoster and by 4.5-fold after zoster in the ophthalmic branch of the trigeminal nerve. One large population-based analysis showed that the risk was even higher: stroke was observed within a 1-year of follow-up in 8.1% of people with zoster ophthalmicus compared with only 1.7% in a matched control group. However, no cases of vasculopathy or stroke were observed among 984 patients with documented zoster followed up for >6 months after rash onset, although 86% received antiviral therapy. Indeed, stroke following zoster ophthalmicus is of high clinical importance,. VZV that reactivates in the trigeminal nerve can travel via the ophthalmic sensory nerves to the face and via afferent sensory fibres to the internal carotid artery and its intracranial branches,. Thereafter, the virus establishes infection in the arterial wall, which leads to inflammation, arterial weakening, aneurysm formation, occlusion and stroke. Infected cerebral arteries contain multinucleated giant cells, Cowdry A inclusions (accumulations of viral DNA and protein in the cell nucleus) and herpesvirus particles, as well as VZV DNA and VZV antigens,.
VZV vasculopathy presents with headache, mental status changes and focal neurological deficits. Large and small vessels are involved. Brain MRI frequently reveals lesions at grey-white matter junctions. In more than two-thirds of patients, angiography reveals focal arterial stenosis and occlusion, aneurysm or haemorrhage.

VZV and giant cell arteritis

One of the most exciting recent developments is the detection of VZV antigen, VZV DNA and virus particles in the temporal arteries of patients with giant cell arteritis (inflammatory vasculopathy, most often involving the temporal arteries). Analysis of temporal artery biopsies from healthy individuals aged >50 years and from patients with arteritis revealed VZV antigen in the temporal arteries of 61 out of 82 (74%) of patients with arteritis compared with 1 out of13 (8%) in normal temporal arteries. This discovery, if confirmed, suggests that antiviral treatment might confer additional benefit to corticosteroids in patients with giant cell arteritis.

VZV-induced diseases of the eye

VZV can cause stromal keratitis with corneal anaesthesia, acute retinal necrosis and progressive outer retinal necrosis, particularly in immunocompromised individuals. Patients complain of eye pain and loss of vision. Retinal haemorrhages and whitening with bilateral macular involvement may occur. Zoster, aseptic meningitis, vasculitis or myelitis can precede retinal necrosis. As with neurological disease, VZV-associated ocular disorders can also occur without rash.

Diagnosis

Diagnosis of varicella and zoster is most often made clinically on the basis of the characteristic generalized or unilateral dermatomal vesicular rashes, respectively. Notable exceptions include the following characteristics: atypical rashes, such as disseminated zoster or a minimal or absent dermatomal rash; zosteriform herpes simplex; modified (breakthrough) varicella in vaccinated individuals; and rashes caused by enteroviruses, poxviruses, rickettsia, drug reactions or contact dermatitis; and VZV infection in the absence of a rash. The latter includes, for example, zoster without rash (known as zoster sine herpete, sometimes with or without facial palsy), meningitis,,, stroke,, myelitis and enteric (gastrointestinal) infections,. In these settings, rapid diagnosis is necessary to plan appropriate therapy and public health measures. Diagnosis by measurement of VZV-specific antibodies in serum samples is accurate but does not yield results rapidly enough to be clinically useful because of the time it takes for patients to develop antibodies. Serum antibodies are generally of no help unless anti-VZV IgM is detected and even its presence can be nonspecific.
For laboratory diagnosis of VZV infection, the following approaches are currently most useful: PCR on material from skin vesicles (submitted as swabs, fluid or scabs), saliva,,, and cerebrospinal fluid if neurological symptoms or signs are present,,,. Detection of VZV antigens by direct immunofluorescence from vesicles is also rapid and specific, although less sensitive than PCR. During varicella and zoster, viral DNA can be detected in saliva, and this method is diagnostically useful and specific in symptomatic patients with or without rash,,. PCR along with restriction enzyme digest and sequencing of specific segments of the viral genome can be used to determine whether VZV is resistant to acyclovir.
When symptoms develop in vaccinated individuals that suggest VZV infection, such as rash or meningitis (even without rash), it is important to identify VZV by PCR, and it can be useful to determine whether the virus is wild-type VZV or vOka,. In the United States, testing can be performed free of charge at either the National Virus Laboratory at the Centers for Disease Control and Prevention (CDC) or the Columbia University VZV Identification Program of the Worldwide Adverse Experience System of Merck & Co. The FDA is notified of these results if they are related to complications of vaccination, and clinical information on these vaccinated patients is often published. Other countries have similar testing facilities (for example, the VZV Reference Laboratory in the United Kingdom).
Notably, VZV DNA can be detected transiently in the saliva of severely stressed adults and children in the absence of specific symptoms, indicating subclinical reactivation of the virus. Nevertheless, testing of saliva is remarkably specific because VZV DNA is rarely detected in asymptomatic human volunteers aged <60 years,,. The presence of VZV DNA does not necessarily equate to the presence of infectious virus. During zoster, a single infected cell can contain thousands of copies of VZV DNA and that DNA can persist long after infectious VZV has been cleared. ‘Contained reversions’ (silent reactivation of latent VZV) (FIG. 5) may also be a source of VZV DNA, which makes it problematic to demonstrate that vasculopathy without rash is a complication of zoster or that PHN is associated with continuing VZV replication. Furthermore, despite evidence of asymptomatic shedding of VZV DNA, there is little epidemiological evidence to indicate that asymptomatic individuals transmit VZV infection,. By contrast, most infections with herpes simplex virus result from asymptomatic shedding. Nevertheless, detection of VZV DNA in saliva or blood in immunocompetent individuals with minimal symptoms may permit diagnostic suspicion of early zoster before rash onset and the initiation of antiviral therapy sufficiently early to halt ganglionic infection before the damage responsible for PHN has occurred.

Prevention

Varicella vaccine

A live attenuated varicella vaccine was developed in Japan by Takahashi and colleagues in 1974 (REF ). VZV was isolated from a boy with varicella, and the virus was attenuated by cultivation for 33 serial passages in human and guinea pig fibroblasts, with some passages at low temperature (34°C). Live, attenuated vOka consists of a mixture of distinct VZV genotypes, with 42 single nucleotide polymorphisms distinguishing vOka from the wild-type Oka parent strain,. The exact mechanisms for the attenuation of VZV in live vaccines remain unknown; mutations in ORF62 were identified in vOka and mutations at positions 106262, 107252 and 108111 in ORF62 are important, as is a stop codon mutation at position 560 in ORF0 (REF ). These mutations, particularly the unique mutations at positions 106262 and 108111 in ORF62, can be used to differentiate parental Oka and other wild-type strains of VZV from vOka,.
Initially, the vaccine was highly controversial because of the fear of latent vOka, the possibility that the virus might be oncogenic and the possibility that immunity from vaccination would not be long-lasting. The vaccine was tested in Japan for ~5 years before reaching the rest of the world,. By 1979, there was much interest in the vaccine in the United States because many young children who had been cured of leukaemia died of varicella infection. In a large collaborative trial, >500 children with leukaemia in remission who were still receiving maintenance chemotherapy were vaccinated with vOka. The vaccine was considerably safer than infection with wild-type VZV and protected ~85% of recipients from varicella. Studies in healthy children also demonstrated a high degree of safety and protection against varicella. In 1995, this live attenuated varicella vaccine was licensed in the United States and routine immunization recommended for healthy American children at 1 year of age. In 2007, when a second dose was shown to offer even greater protection than one dose, a two-dose schedule, with the second dose given at aged 4–6 years or at least 3 months after the first dose, was recommended by the CDC. Two doses of the varicella vaccine are also recommended for older children, adolescents and adults without evidence of varicella immunity, including health-care workers. Currently, the varicella vaccine, using one or two doses, is also licensed in Australia, Brazil, Canada, China, Germany, Greece, Israel, Italy, Japan, Qatar, South Korea, Spain, Taiwan and Uruguay.
In countries without varicella vaccination, prevention by passive immunization (injection of VZV-specific antibodies) might be available, which provides temporary protection for several weeks. Antiviral therapy has been used for the prevention of varicella, and zoster, in immunocompromised patients, but is not uniformly accepted as useful. Acyclovir-resistant VZV rarely develops in a small number of treated immunocompromised patients.

Immunity after vaccination

The varicella vaccine is highly effective in preventing varicella, with little decline in immunity over time,. Between 1995 and 2003 the incidence of varicella decreased by 80% in some areas of the United States, and the number of hospitalizations and deaths decreased. In a study of >7,000 vaccine recipients, long-lasting (15 years) immunity was demonstrated, with 90% efficacy in preventing varicella despite 70% of children having received only one dose of the vaccine. At present, there seems to be no need for booster doses of the varicella vaccine because significant waning immunity has not been observed,.
Silent reactivation (contained reversions) of latent VZV (FIG. 5) can boost immunity endogenously and is likely to contribute to long-lasting immunity to varicella and zoster. Exogenous boosting due to exposure to individuals with VZV infections also occurs, (FIG. 5). Not appreciating the potentially important role of asymptomatic reactivation in maintaining immunity to VZV, epidemiologists predicted that widespread vaccination and the resulting lack of exogenous immune stimulation would result in increased incidence of zoster,. This has been the justification for not using the varicella vaccine universally in children in several countries in Europe. The incidence of zoster has been increasing in the United States since the 1950s, long before the varicella vaccine was available, and increases are also reported in other countries without vaccination programmes. Thus, it seems that the varicella vaccine is unlikely to contribute substantially to an increase in the incidence of zoster. The role of subclinical (or mild) endogenous reactivation of latent VZV in maintaining immunity is evidenced by a study performed in France: the rate of zoster (15–16%) in isolated populations over ~30 years was no higher than it was in the general public. Accordingly, latency (with the possibility of silent or minimal viral reactivation) might be an asset rather than a liability for the varicella vaccine.

Vaccine safety

The varicella vaccine is safe and well tolerated. Serious adverse events after vaccination are unusual; ~100 million children and adults have been vaccinated and vaccine-related serious adverse event reactions have been very rarely reported and have occurred almost exclusively in recipients who were immunocompromised but not recognized to be so,. The development of varicella in vaccine recipients (breakthrough varicella) is unusual, and when it occurs it is almost invariably mild.
Vaccinated children who develop a rash in the first month after immunization rarely transmit vOka to close contacts; those who develop varicella due to wild-type VZV are more likely to transmit the virus to others. Transmission is positively correlated with the number of skin lesions,. Rash caused by vOka is unusual following vaccination in children and even if it occurs, vaccinated children usually develop few skin lesions. When wild-type VZV infects vaccinated children, they rarely develop an extensive rash. Transmission of vaccine-type type VZV from vaccinated individuals is rare. The risk of developing zoster after vaccination is lower than after varicella caused by wild-type VZV; at present, 30–50% of cases of zoster in vaccinated children are attributable to wild-type VZV,.

Zoster vaccine

Development of the varicella vaccine paved the way for the development of a vaccine to prevent zoster and its complications. Clinical observations by Hope-Simpson, reported in a landmark publication in 1965 (REF ), provided the rationale for the development of a therapeutic zoster vaccine. Hope-Simpson postulated that primary VZV infection establishes lifelong latency of VZV in sensory ganglia and induces immunity to VZV that prevents the reactivation, replication and spread of latent VZV and, therefore, zoster. He proposed that this immunity gradually decreases, eventually permitting latent VZV to reactivate, multiply and re-emerge as zoster. He further proposed that both exogenous and endogenous exposure to VZV stimulate the host’s immunity (FIG. 5). Noting that second episodes of zoster were uncommon, he proposed that an episode of zoster also stimulates immunity to VZV, essentially immunizing the host against another episode of zoster. Subsequent investigations have validated every component of Hope-Simpson’s prescient hypothesis,.
In the Shingles Prevention Study (SPS), a doubleblind, placebo-controlled trial, 38,546 healthy adults aged ≥60 years (median 69 years) were randomly assigned to receive a single dose of high-potency vOka (14 times greater potency than the varicella vaccine) or placebo,,. Two co-primary end points were investigated: the burden of illness owing to zoster (a severity-by-duration measure representing the total pain and discomfort) and the incidence of clinically significant PHN. The incidence of zoster was also determined. The higher-potency vaccine was required to increase VZV-specific cell-mediated immunity in latently infected older adults,.
A total of 19,270 people who received the zoster vaccine and 19,276 who received placebo were followed up on average for 3.13 years,. There were 957 confirmed evaluable cases of zoster (315 in vaccine recipients and 642 in placebo recipients). In both groups, >93% of the subjects with zoster were positive for wild-type VZV DNA by PCR and none had vOka DNA,.
The vaccine reduced burden of disease by 61.1% (65.5% in people aged 60–69 years and 55.4% in people aged ≥70 years). The duration of pain and discomfort among subjects with zoster was shorter in vaccinated candidates compared with placebo recipients,. The vaccine reduced the incidence of zoster by 51.3% (63.9% in people aged 60–69 years but only 37.6% in people aged ≥70 years). The incidence of clinically significant PHN was reduced by more than 65% for both age groups. Furthermore, the zoster vaccine reduced the percentage of people with zoster who developed PHN by >31%, with most benefit in the group aged ≥70 years, which had the highest risk of developing this complication,. The SPS also demonstrated that the vaccine reduced the adverse impact of zoster on patients’ capacity to perform activities of daily living and health-related quality of life.
The zoster vaccine is safe. Rates of serious adverse events, systemic adverse events, hospitalizations and deaths in the SPS were low in vaccine recipients and comparable with those in placebo recipients,,. During the first 42 days after vaccination, 24 cases of zoster in placebo recipients were reported and 7 cases in the vaccination group, none of which were caused by vOka. In contrast to prophylactic vaccines, such as those against varicella and measles, the zoster vaccine is a therapeutic vaccine intended to prevent reactivation and replication of latent VZV with which the recipient is already infected before vaccination and to which the recipient already has substantial immunity.
The zoster vaccine was licensed by the FDA in 2006 and recommended by the CDC in 2008 for the routine immunization of healthy adults aged ≥60 years for prevention of zoster and its complications, primarily PHN. Post-licensure studies have confirmed the vaccine’s safety and efficacy,,. Unfortunately, uptake of the zoster vaccine has been low, which is probably due to the high cost and general lack of appreciation of the importance of preventing infectious diseases in older adults. The vaccine has now been shown to be safe and effective in healthy individuals aged 50–59 years. At present, the zoster vaccine can be used in this age group but it is not yet officially recommended.
The SPS demonstrated persistent efficacy 4 years after vaccination. In additional substudies,, efficacy for burden of disease persisted for 10 years after vaccination but efficacy for the incidence of zoster persisted only for 8 years. The CDC currently does not recommend booster doses of the zoster vaccine, but it might do in the future.

New subunit vaccine

The development by GlaxoSmithKline of a liposome-based subunit vaccine (HZ/su), which contains the VZV glycoprotein E and the adjuvant ASO1B, promises to change prospects for immunization against zoster and its complications. Phase I and II studies established that two doses of HZ/su containing 50 μg of recombinant VZV glycoprotein E administered 1 or 2 months apart were well tolerated and induced much more robust VZV-specific and VZV glycoprotein E-specific CD4+T cell and antibody responses than vOka,. Two large Phase III placebocontrolled efficacy trials have been recently completed in individuals aged ≥50 years and in individuals aged ≥70 years,. The efficacy against zoster in the younger study group was ~97%. HZ/su has not been tested for its efficacy in preventing varicella, partly because vOka is extremely effective for this purpose. The adjuvanted glycoprotein E vaccine is non-replicating, and can, therefore, be used in immunosuppressed patients who are currently precluded from receiving the live attenuated vOka zoster vaccine.


Severe varicella is characterized by extensive and prolonged viral replication, often associated with fever, continued development of new skin vesicles for >5 days, and/ or involvement of the lungs, liver and/or brain (FIG. 3 illustrates a dense vesicular rash in a febrile infant on day 5). Severe varicella is a feared complication that was a major impetus towards vaccine development. Severe varicella has caused many deaths in individuals with congenital or acquired impairment of cellular immunity, even after the development of antiviral therapy. Children with impaired innate immunity, for example, those with natural killer cell abnormalities, are also at risk for severe varicella, including that caused by vOka. Thus, strenuous measures should be taken to prevent VZV infection in this group, including post-exposure prophylaxis,. Since the 1980s, the main treatment has been antiviral therapy with high-dose intravenous acyclovir together with supportive intensive care. Acyclovir is a guanosine analogue that inhibits the synthesis of viral DNA (FIG. 7) and treatment with acyclovir reduces visceral dissemination of the virus. It is typically given for 7–10 days and can be switched to oral therapy 48 h after the last lesions appear or continued until all lesions are crusted. Oral acyclovir has poor bioavailability; thus, the related drugs valaciclovir and famciclovir, which have excellent absorption from the intestinal tract, produce high blood antiviral activity and have a long half-life, should be used for oral therapy instead. Valaciclovir and famciclovir are prodrugs, which are converted to active guanosine analogues in vivo. As both prodrugs need less frequent administration than acyclovir, patient compliance is improved and they are frequently used in children aged >2 years and in adults. Immunocompromised patients with severe VZV infections still receive intravenous acyclovir, which results in higher blood levels than oral therapy. Alternatively, intravenous foscarnet and cidofovir can be used. However, because of toxicity, these drugs should be only used in immunocompromised individuals with acyclovir-resistant VZV. Notably, intravenous antivirals, including acyclovir, can be nephrotoxic; both acyclovir and foscarnet require dose adjustment in patients with renal impairment.

VZV infection in individuals aged >13 years is associated with an increased risk of severe or fatal outcome, and oral antiviral therapy is recommended, even in otherwise healthy adolescents and adults (FIG. 6). In immunocompetent patients, oral acyclovir, or preferably famciclovir, or valaciclovir need to be started within 24 h of the first skin lesions to shorten the duration of fever and rash; 5 days and 7 days of treatment have comparable efficacy.
Varicella in pregnant women is also problematic: pregnancy increases the risk of severe disease in the mother and VZV can harm the unborn child and lead to congenital abnormalities (congenital varicella syndrome),. Thus, pregnant women with varicella are usually treated with intravenous acyclovir, even though this is a category B drug (that is, animal studies have failed to demonstrate a risk to the fetus but no wellcontrolled studies have been conducted in pregnant women) and not licensed in pregnancy. The effect of treatment on the development of congenital varicella syndrome is unknown. Maternal onset of varicella between 5 days before and 2 days after delivery is associated with a high risk of severe varicella in the newborn, who should receive prophylaxis with VZV-specific immunoglobulin. Newborns with congenital varicella syndrome should receive high-dose intravenous acyclovir every 8 h owing to increased clearance of the drug in this age group. Oral acyclovir is poorly absorbed and should be used cautiously, if at all.
Bacterial superinfection is the most common severe complication of varicella; varicella is a major risk factor for invasive group A streptococcal disease. Superinfection presents with recurrence of fever with or without localized signs of cellulitis (infection of the skin and subcutaneous fat tissue), bone and joint infection or pneumonia. Cellulitic features accompanied by disproportionate pain, fatigue and systemic signs and symptoms can indicate necrotizing fasciitis, even in the absence of fever, and should prompt immediate antibacterial therapy together with resuscitative measures, analgesia and urgent consideration of surgical debridement. Other possible complications of varicella are cerebellar ataxia, ischaemic stroke and acquired protein S deficiency with purpura fulminans and venous thrombosis,,. The effect of antiviral therapy on individual complications is unclear but a pragmatic approach is to treat if there is evidence of ongoing viral replication (such as continuing new vesicle formation and/or persistent PCR positivity in blood or cerebrospinal fluid in a symptomatic patient).

Latency and reactivation

How VZV latency is achieved and maintained is a challenging question. It remains unclear whether VZV latency is a period of relative viral quiescence due to a transient block in complete gene expression or whether the virus is often or constantly in a state of abortive reactivation. Studies of surgical removal of enteric ganglia suggest a block in gene expression,, although recent autopsy studies support the hypothesis of abortive reactivation, but further research is needed. Asymptomatic VZV reactivation is also of great interest. For example, whether some viral genes are translated with or without synthesis of virus particles, and whether these are delivered to skin by axonal transport with or without spread to satellite cells surrounding the neuron, remain unclear. Delivery of virions to skin may not necessarily cause lesions if replication is controlled rapidly by innate immunity and by adaptive immune responses. Further research in this area is important.
The recognition that VZV is a pathogen in the human gut was an unanticipated finding because VZV infections were long associated with cutaneous manifestations, which may not occur when VZV reactivates in enteric neurons. The nature of enteric zoster is, therefore, another issue that requires further study.



.

No comments:

Post a Comment

Note: only a member of this blog may post a comment.