Antiviral Drugs: Treatment of the Fetus and Newborn



Antiviral Drugs: Treatment of the Fetus and Newborn


Richard J. Whitley

Edward P. Acosta

David W. Kimberlin



Introduction

Parenteral therapy of viral infections of the newborn and infant became a reality with the introduction of vidarabine (adenine arabinoside) for the treatment of neonatal herpes simplex virus (HSV) infections in the early 1980s. Since then, acyclovir has become the treatment of choice for neonatal HSV infections, as well as a number of other herpesvirus infections. Similarly, ganciclovir has been established as beneficial for the treatment of congenital cytomegalovirus (CMV) infections that involve the central nervous system (CNS). Although other viruses can infect the newborn, including rubella and parvovirus, this chapter will review the lessons learned from treating neonatal and congenital infections and will consider therapies for respiratory virus infections of infants. Although the natural history of these diseases is reviewed elsewhere in this textbook, a brief summary will precede a detailed discussion of established and alternative therapeutics. The reader is referred to more extensive reviews of antiviral therapy in children and adults (1).


Therapy of Herpes Simplex Virus Infections

Neonatal HSV infections occur in approximately 1 in 3,200 deliveries in the USA (2) and of these, approximately two-thirds develop some form of CNS disease (3). The majority of cases are caused by HSV-2 (4). The risk of transmission is increased with primary maternal infection during the third trimester and can be decreased by cesarean delivery if HSV has been isolated from the cervix or external genitalia near the time of delivery (2). As reviewed by Kimberlin, 85% of neonatal HSV cases occur due to peripartum transmission, 10% occur via postnatal transmission, and only 5% are due to transmission in utero (5).

Infants with intrauterine HSV infection are characterized by the triad of cutaneous (active lesions, scarring), neurologic (microcephaly, hydranencephaly), and eye findings (chorioretinitis, microphthalmia) present at birth. Intrauterine HSV infection has been found to occur with both primary and recurrent maternal HSV infections (6), although the risk from a recurrent infection is less.

HSV infection acquired in the peripartum or postpartum period can be categorized as skin, eye, and/or mouth (SEM) disease, CNS disease, or disseminated disease. By definition, SEM disease does not involve the CNS. Disseminated disease may involve the CNS along with multiple other organ systems including the liver, adrenals, gastrointestinal tract, and the skin, eyes, or mouth. CNS disease may have skin involvement but does not involve other visceral systems. Of all infants with neonatal HSV infections, approximately 33% have CNS disease, while about 25% have disseminated disease (7). Of infants with disseminated disease, approximately 60% to 75% will have CNS involvement (8). Therefore, by combining these statistics, it can be approximated that 50% of infants with neonatal HSV infection will have CNS involvement.

The pathogenesis of CNS involvement in neonatal HSV infections differs depending on whether or not the infection is disseminated. Encephalitis associated with dissemination is due to hematogenous spread, whereas isolated encephalitis or encephalitis associated with only skin involvement likely occurs because of retrograde intraneuronal transport of HSV (3). This corresponds to the clinical presentations of disseminated versus CNS disease in that the blood-borne spread of disseminated disease presents earlier (9 to 11 days of life on average) and causes more diffuse brain involvement with multiple areas of hemorrhagic necrosis, while CNS disease occurring via slower axonal transport presents later (around 16 to 17 days of life) and typically causes more focal CNS involvement (9).

In the pre-antiviral era, infants with neonatal HSV
infections had significant morbidity and mortality. Infants with disseminated disease had an 85% mortality by 1 year of age, and only 50% of survivors had normal neurodevelopmental outcomes; likewise, infants with CNS disease had a mortality rate of 50% by 1 year of age with only 33% of survivors having normal neurodevelopmental outcomes (10). The early era of antiviral therapy for neonatal HSV infection was marked by improved mortality with intravenous vidarabine as well as with standard dose (SD) intravenous acyclovir (30 mg per kg per day in three divided doses). The 1 year mortality for disseminated disease improved to 50% with vidarabine and 61% with SD acyclovir, whereas the 1 year mortality for CNS disease dropped to 14% for both vidarabine and SD acyclovir (11). More recently, high-dose (HD) acyclovir has been shown to further improve upon these mortality figures. Using HD intravenous acyclovir (60 mg per kg per day in three divided doses for 21 days), Kimberlin demonstrated 1 year mortality rates of 29% and 4% for disseminated and CNS diseases, respectively (12). This study also showed that HD acyclovir improves morbidity for infants with disseminated disease (83% of survivors had normal neurodevelopmental outcomes) but not for infants with CNS disease (31% of survivors had normal neurodevelopmental outcomes).

Babies with SEM involvement have the best prognosis and, as noted below, require only 14 days of HD intravenous acyclovir therapy. Importantly, health care providers must exclude an asymptomatic or subclinical CNS infection at presentation by determining whether the cerebrospinal fluid (CSF) is negative for HSV DNA by polymerase chain reaction. With skin recurrences, the management (intravenous vs. oral therapy) of these children must be individualized according to the nature of their illness at the time of recurrence.

Infants can also experience primary gingivostomatitis secondary to HSV-1. Limited controlled clinical trial data are available to define efficacy of therapy, but most experts would recommend treatment, either intravenous or oral, depending upon the severity of illness.


Therapy of Neonatal Varicella Zoster Virus Infections

Women who experience primary varicella zoster virus (VZV) infections within 48 hours of delivery have children at risk for the development of chickenpox within the first 3 weeks of life. Newborns delivered to these women traditionally receive zoster immune globulin (ZIG) to prevent disease. However, with sporadic shortages of ZIG, some newborns may experience neonatal disease. Although no controlled clinical trials have evaluated acyclovir therapy of this disease, experts would recommend this drug for the treatment of neonatal VZV disease.


Available Therapies for HSV and VZV Infections


Acyclovir (Acycloguanosine, Zovirax, ACV)

Acyclovir is the most frequently prescribed antiviral agent for the management of HSV infections of the newborn and infant. It has been available for clinical use for nearly three decades and has demonstrated remarkable safety and efficacy against mild to severe infections caused by HSV and VZV in both normal and immunocompromised patients, including the newborn.


Chemistry and Mechanism of Action

Acyclovir is a deoxyguanosine analogue. After preferential uptake by viral-infected cells, acyclovir is phosphorylated by virus-encoded thymidine kinase (TK). Subsequent di- and triphosphorylation are catalyzed by host cell enzymes. Acyclovir triphosphate prevents viral DNA synthesis by inhibiting viral DNA polymerase (13).


Spectrum and Resistance

Acyclovir is most active against HSV; activity against VZV is also substantial but approximately 10-fold less. Although not related to the therapy of the newborn, Epstein Barr virus (EBV) is only moderately susceptible to acyclovir because EBV has minimal TK activity. Activity against CMV is poor because CMV does not have TK activity and CMV DNA polymerase is poorly inhibited by acyclovir triphosphate (13).

Resistance of HSV and VZV to acyclovir has become an important clinical problem, especially among immunocompromised patients exposed to long-term therapy (14). Resistance has not been considered a problem in the therapy of newborn infections at this time, although it has been reported (15). Viral resistance to acyclovir usually results from mutations in the viral TK gene, although mutations in the viral DNA polymerase gene can also rarely occur. Resistant isolates can cause severe, progressive, debilitating mucosal disease and, rarely, visceral dissemination (16,17). Isolates of HSV resistant to acyclovir have also been reported in normal hosts, most commonly in patients with frequently recurrent genital infection who have been treated with chronic acyclovir (18).


Indications

Acyclovir is effective for the treatment of infections caused by HSV and VZV in both immunocompetent and immunocompromised hosts, including the newborn. For the treatment of neonatal HSV infections, the recommended dose is 20 mg per kg every 8 hours intravenously. The duration of therapy for SEM disease is 14 days, while for those babies with either CNS or disseminated disease, therapy should be extended for 21 days. Several studies are investigating the application of end of treatment polymerase chain reaction detection of viral DNA at the completion of antiviral therapy. If the CSF is positive for HSV DNA, most experts recommend continuing therapy until the CSF is cleared of viral DNA.


Pharmacokinetics

After intravenous doses of 2.5 to 15 mg per kg, steady-state concentrations of acyclovir range from 6.7 to 20.6 μg per mL. Acyclovir is widely distributed; high concentrations are attained in kidneys, lung, liver, heart, and skin vesicles; concentrations in the CSF are about 50% of those in the plasma (19). Acyclovir crosses the placenta and accumulates in breast milk. Protein binding ranges from 9% to 33%, and less than 20% of drug is metabolized to biologically inactive metabolites.

In the absence of compromised renal function, the half-life of acyclovir is 2 to 3 hours in older children and
adults and 2.5 to 5 hours in neonates with normal creatinine clearance. More than 60% of administered drug is excreted in the urine (19). Elimination is prolonged in patients with renal dysfunction; the half-life is approximately 20 hours in persons with end-stage renal disease, necessitating dose modifications for those with creatinine clearance less than 50 mL per minute per 1.73 m2 (20). Acyclovir is effectively removed by hemodialysis but not by continuous ambulatory peritoneal dialysis (13,21).


Adverse Effects

Acyclovir generally is a safe drug. Oral acyclovir sometimes causes mild gastrointestinal upset, rash, and headache. If it extravasates, intravenous acyclovir can cause severe inflammation, phlebitis, and sometimes a vesicular eruption leading to cutaneous necrosis at the injection site. Also, if given by rapid intravenous infusion or to poorly hydrated patients or those with preexisting renal compromise, intravenous acyclovir can cause reversible nephrotoxicity. Renal dysfunction results from obstructive nephropathy caused by the formation of acyclovir crystals precipitating in renal tubules. Occasionally administration of acyclovir by the intravenous route is associated with rash, sweating, nausea, headache, hematuria, and hypotension. High doses of intravenous acyclovir (60 mg per kg per day) in neonates and the prolonged use of oral acyclovir following neonatal disease have been associated with neutropenia in uncontrolled trials (15,12).

The most serious side effect of acyclovir is neurotoxicity, which usually occurs in subjects with compromised renal function who attain high serum concentrations of drug (22). Neurotoxicity is manifest as lethargy, confusion, hallucinations, tremors, myoclonus, seizures, extrapyramidal signs, and changes in state of consciousness developing within the first few days of initiating therapy. These signs and symptoms usually resolve spontaneously within several days of discontinuing acyclovir.

Although acyclovir is mutagenic at high concentrations in some in vitro assays, it is not teratogenic in animals. Limited human data suggest that acyclovir use in pregnant women is not associated with congenital defects or other adverse pregnancy outcomes.


Drug Interactions

Somnolence and lethargy may occur in subjects being treated with both zidovudine and acyclovir. The likelihood of renal toxicity of acyclovir is increased when administered with nephrotoxic drugs such as cyclosporine or amphotericin B. Concomitant administration of probenecid prolongs acyclovir’s half-life, whereas acyclovir can decrease the clearance and prolong the half-life of drugs such as methotrexate that are eliminated by active renal secretion (13).


Untested Therapy for Neonatal HSV and VZV Infections


Valacyclovir (Valtrex)

Valacyclovir has been available for a decade in a tablet formulation. It has a safety and efficacy profile similar to that of acyclovir but offers potential pharmacokinetic advantages. A pediatric liquid formulation has been evaluated (23).


Chemistry, Mechanism of Action, Spectrum, and Resistance

Valacyclovir is a prodrug of acyclovir and as such has the same mechanism of action, antiviral spectrum, and potential for development of resistance (1,13).


Indications

Valacyclovir has the same indications as acyclovir. Although data from controlled clinical trials are limited, because of greater bioavailability, valacyclovir may be advantageous in treating infections caused by viruses relatively less sensitive to acyclovir than HSV (e.g., VZV and CMV). Pharmacokinetics in neonates and very young infants do not support the use of valacyclovir liquid formulation in these populations.


Pharmacokinetics

In contrast to the low bioavailability of acyclovir, the bioavailability of valacyclovir exceeds 50% (13,24). Peak serum concentrations ranging from 0.8 to 8.5 μg per mL following doses of 100 to 2,000 mg are attained about 1.5 hours after a dose. Oral valacyclovir provides plasma acyclovir concentrations comparable to those following a comparable dose of intravenous acyclovir. All other pharmacokinetic characteristics are similar to those of acyclovir (25).


Adverse Effects

The profiles of adverse effects observed with valacyclovir therapy are the same as those observed with acyclovir. In addition, manifestations resembling thrombotic microangiopathy have been described in patients with AIDS receiving high doses of valacyclovir, although the causal relationship to valacyclovir is not absolutely established (26).


Dosage

Valacyclovir dosages in children older than 2 years have recently been approved by the Food and Drug Administration (FDA) as 20 mg per kg administered two to three times daily (27). Adults are treated with 500 to 100 mg per dose, two to three times per day. The higher dose is for the treatment of herpes zoster and the lower dose for the treatment of genital herpes. Suppression of recurrent oral and genital herpes infections has been accomplished with single daily doses of 500 mg and 1,000 mg, respectively.


Famciclovir (Famvir)


Chemistry and Mechanism of Action

Famciclovir is the prodrug of penciclovir. Like acyclovir, penciclovir is a guanosine analogue that has activity against HSV-1, HSV-2, and VZV in vitro. It is similarly phosphorylated by viral TK and subsequently converted to its active form, penciclovir triphosphate. However, its mechanism of
action differs from acyclovir in that, while a competitive inhibitor of DNA polymerase, it does not cause chain termination. Penciclovir has minimal oral bioavailability. Famciclovir is the diacetyl ester prodrug of penciclovir and confers 70% bioavailability (1,13). At the present time, no formulation exists for newborns and infants.


Spectrum and Resistance

The spectrum of activity of the parent drug, penciclovir, is identical to acyclovir. Resistance occurs in a fashion identical to that of acyclovir, as well, with mutation of the viral TK being the most common (1,13).


Indications

Famciclovir is indicated for treatment of herpes zoster infections as well as genital herpes and has similar efficacy to valacyclovir. There have been no studies of famciclovir in the neonate. However, if a pediatric formulation became available, there would be an option for the treatment of neonatal HSV infection involving the skin, eye, and mouth.


Pharmacokinetics

Famciclovir is excreted by the kidney and thus requires dose adjustment in patients with renal insufficiency (13).


Adverse Effects

Famciclovir is tolerated well with minimal side effects with headache and gastrointestinal upset being most common.


Therapy of Congenital Cytomegalovirus Infection

CMV commonly infects humans worldwide, with a seroprevalence of approximately 40% in adolescents and approaching 90% in adults with poor socioeconomic status (28,29). Congenital CMV infection is the most common congenital infection in the developed world and occurs in about 1% of liveborn infants in the United States (30). Congenital CMV infections most commonly occur via intrauterine transmission, but since the virus is shed in body fluids, transmission can also be acquired perinatally during delivery or postnatally through breast milk.

Of all infants born with congenital CMV infection, approximately 7% to 10% have clinically evident disease at birth (31). Clinical characteristics of intrauterine infection include intrauterine growth restriction, hepatosplenomegaly, jaundice, thrombocytopenia, microcephaly, periventricular calcifications, and chorioretinitis. As reviewed by Dollard, true mortality rates are difficult to obtain and have been reported to be as high as 30% for symptomatic infants (32), but more likely average about 5% to 10% (33). Death usually is due to non-CNS manifestations of the infection, such as hepatic dysfunction or bleeding. An estimated 40% to 58% of infants with symptomatic congenital CMV infection have permanent sequelae, whereas infants who are asymptomatic at birth suffer permanent sequelae nearly 14% of the time (32). Sensorineural hearing loss (SNHL), mental retardation, seizures, psychomotor and speech delays, learning disabilities, chorioretinitis, optic nerve atrophy, and defects in dentition are the most common long-term consequences (34). As opposed to intrauterine infections, perinatally acquired CMV infections are not typically associated with long-term sequelae, though acute illness has been reported in premature very low-birth-weight infants (35). In full-term infants, perinatal infections are commonly asymptomatic, but may present with pneumonitis within the first few months of life (36).

Congenital CMV infection is the consequence of maternal-fetal infection, with most symptomatic congenital cases occurring because of asymptomatic maternal infection. The pathogenesis of CNS involvement in congenital CMV infection begins with disseminated viremic spread, including the endothelial cells of the brain and epithelial cells of the choroid plexus. From the endothelial cells of the brain, the virus spreads to contiguous astrocytes. From the choroid plexus, the virus spreads to the ependymal surfaces via the CSF. Once these cells are infected, the virus undergoes continuous replication, which leads to characteristic intranuclear inclusion bodies and cell death. As antibodies are produced in the face of continuous viral replication, immune complexes form as well, leading to further immune-mediated damage (34). Although the specific pathogenesis of CMV-mediated SNHL has not been elucidated, histology has shown evidence of infection in the cells of both the cochlear and vestibular endolabyrinth (37). CMV has also been isolated from the cochlear perilymph upon autopsy of infants with congenital CMV infection (38).

Recent studies of ganciclovir treatment of congenital CMV infections involving the CNS have been promising. Using ganciclovir at doses of 12 mg per kg per day divided every 12 hours for a duration of 6 weeks, improved hearing outcomes were demonstrated in neonates with symptomatic congenital CMV infections involving the CNS (as evidenced by microcephaly, intracranial calcifications, abnormal CSF for age, chorioretinitis, and/or hearing deficits) (39). The primary endpoint was improved brainstem-evoked response (BSER) between baseline and 6-month follow-up (or no deterioration at the 6-month follow-up if the baseline BSER was normal). For total evaluable ears, 69% of patients who received ganciclovir met the primary endpoint as opposed to 39% of the control group. No patients receiving ganciclovir had worsening of their hearing between baseline and 6 months. Ganciclovir recipients also had more rapid resolution of alanine aminotransferase (ALT) abnormalities than did the control group, though they were significantly more likely to become neutropenic. Additional analyses of this randomized controlled trail suggest that ganciclovir may also reduce neurodevelopment delays (40). Improvement of mortality has not yet been shown with ganciclovir therapy.


Treatment of CMV Infections


Ganciclovir (Cytovene)

Ganciclovir was the first antiviral available for the therapy and prevention of infections caused by CMV. It has proved to
be a very valuable drug for immunocompromised patients, particularly hematopoietic stem cell transplant recipients, who suffer substantial morbidity and mortality from CMV infections and, more recently, in children with congenital CMV infection, albeit it is not yet approved by US FDA.


Chemistry and Mechanism of Action

Ganciclovir is structurally similar to acyclovir except for a hydroxymethyl group on its acyclic side chain. The initial phosphorylation step is carried out by pUL97, which is a viral protein kinase. Cellular kinases then phosphorylate the agent two additional times to convert it into its triphosphate derivative, which is able to inhibit the CMV DNA polymerase encoded by UL97 as well as incorporate into and terminate viral DNA.

Ganciclovir triphosphate is a competitive inhibitor of herpesviral DNA polymerases, resulting in cessation of DNA chain elongation (1,13).


Spectrum and Resistance

Ganciclovir has similar activity to acyclovir against HSV-1, HSV-2, and VZV, but, in contrast with acyclovir, its greatest activity is against CMV. Resistance of CMV isolates usually results from mutations in the UL97 gene (41). Mutations in the CMV DNA polymerase gene occur less often.


Indications

Ganciclovir is licensed for several indications outside of the newborn. Reviews summarize these clinical outcomes (42). For the treatment of neonates congenitally infected with CMV, a controlled trial has been performed (39). As noted above, compared with no treatment, ganciclovir therapy prevented hearing deterioration at 2 years, although about two-thirds of treated infants developed neutropenia, often requiring dose modification.


Pharmacokinetics

Oral bioavailability of ganciclovir is poor, with less than 10% of drug being absorbed following oral administration (43,44). The oral formulation of ganciclovir is no longer marketed. Peak serum concentrations of ganciclovir after 6 mg per kg (newborn dose) of intravenously administered drug range from 8 to 11 μg per mL, with concentrations sufficient to inhibit sensitive strains of CMV in aqueous humor, subretinal fluid, CSF, and brain tissue (1). The elimination half-life of ganciclovir is 2 to 3 hours, and most of the drug is eliminated unchanged in the urine. The pharmacokinetics of ganciclovir in the neonatal population is similar to those of adults (45,46). Dose reduction, proportional to the degree of reduction in creatinine clearance, is necessary for persons with impaired renal function. A supplemental dose is recommended after dialysis because it is efficiently removed by hemodialysis (47).


Adverse Effects

Myelosuppression is the most common adverse effect of ganciclovir; dose-related neutropenia (<1,000 WBC per μL) is the most consistent hematologic aberration, with an incidence of about 40% (44). Neutropenia is dose limiting in about one of seven courses and reverses after drug is stopped. Neutropenia is less frequent following oral administration of ganciclovir. Thrombocytopenia (<50,000 platelets per μL) occurs in approximately 20% and anemia in about 2% of ganciclovir recipients. Two percent to 5% of ganciclovir recipients experience headache, confusion, altered mental status, hallucinations, nightmares, anxiety, ataxia, tremors, seizures, fever, rash, and abnormal levels of serum hepatic enzymes, either singly or in some combination.


Dosage

The dose of ganciclovir for therapy of symptomatic congenital CMV infection is 12 mg per kg per day, given by intravenous infusion in two divided daily doses for 6 weeks.


Valganciclovir (Valcyte)

Valganciclovir was approved by the FDA in March, 2001, for the treatment of CMV retinitis. Because it is well absorbed after oral administration, it may represent a favorable option to intravenously administered ganciclovir for the treatment of congenital CMV infection. Currently, it is licensed for the treatment of CMV infections in selected transplant populations and for CMV retinitis in patients with HIV/AIDS.


Chemistry, Mechanism of Action, Spectrum, and Resistance

Valganciclovir is the L-valine ester prodrug of ganciclovir and as such has the same mechanism of action, antiviral spectrum, and potential for development of resistance as ganciclovir (1,13).


Indications

Valganciclovir has similar indications to ganciclovir. However, based upon limited controlled trials published to date, it currently is approved for the induction and maintenance therapy of CMV retinitis and select transplant populations (48). It is currently under investigation for the treatment of congenital CMV infection. This randomized controlled trial, being performed by the National Institute of Allergy and Infectious Diseases (NIAID) Collaborative Antiviral Study Group (CASG), employs SNHL as its primary endpoint with safety and neurodevelopment outcomes as secondary endpoints.


Pharmacokinetics

Valganciclovir is rapidly converted to ganciclovir, with a mean plasma half-life of about 30 minutes (48). The absolute bioavailability of valganciclovir exceeds 60% and actually is enhanced by about 30% with concomitant administration of food (49). The area under the curve of ganciclovir after oral administration of valganciclovir is one-third to one-half of that attained after intravenous administration of ganciclovir. Patients with impaired renal function require
dosage reduction that is roughly proportional to their reduction in creatinine clearance.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Sep 7, 2016 | Posted by in PEDIATRICS | Comments Off on Antiviral Drugs: Treatment of the Fetus and Newborn

Full access? Get Clinical Tree

Get Clinical Tree app for offline access