You are on page 1of 7

Live Attenuated Human Rotavirus Vaccine, Rotarix™

David I. Bernstein, MD, MA

Rotavirus infections are the leading cause of severe gastroenteritis in young children
worldwide. Recently two new rotavirus vaccines have entered the world market. This review
provides a summary of the rationale, development, and evaluation of one of these vaccines,
Rotarix*. Rotarix* is a live oral rotavirus vaccine developed from a single protective human
strain following multiple passages in tissue culture to attenuate the strain. The vaccine is
administered as two oral doses at approximately 2 and 4 months of age. Large safety and
efficacy trials have shown the vaccine is safe, not associated with intussusception, and
effective against the most common circulating human serotypes. Efficacy against severe
rotavirus gastroenteritis and hospitalization have ranged from 85 to 100 percent.
Semin Pediatr Infect Dis 17:188-194 © 2006 Elsevier Inc. All rights reserved.

S ince its discovery in the 1980s and the recognition that


rotavirus infections are the leading cause of severe gastro-
enteritis in infants and young children in developed and de-
only these two are capable of neutralizing the virus. Group A
rotaviruses are further classified by type. Genotypic classifi-
cation is based on nucleic acid sequence analysis, whereas
veloping countries, rotavirus has been a leading target for serotypes are determined by similarities based on neutraliza-
vaccine development. Simply put, almost every child will tion. At least 15 types of the VP7 glycoprotein (G) exist, with
acquire a rotavirus infection before reaching the age of 5 five common in human disease (G1-4 and G9) and three
years. This prevalence leads to at least 50,000 hospitaliza- others (G8, G12, and G5) recently detected. G1 viruses are
tions in the United States,1-3 a number that may represent a detected most often. The genotype and serotypes generally
substantial underestimation,4 and 352,000 to 592,000 deaths are equivalent for VP7, but this is not true for the VP4 or P
around the world, mostly in underdeveloped countries.5,6 (for protease-sensitive) protein. Thus, the nomenclature for
This review provides a summary of the development of VP4 includes both genotypes, based on gene sequencing and
Rotarix™ (GlaxoSmithKline Immunotherapeutics, Need-
serotypes. The P serotype usually is indicated by an open
ham, MA), a live human attenuated rotavirus vaccine that is
number, whereas the genotype is noted with a bracketed
currently available in the European Union and many other
number: for example, the most common type is P1A[8]. At
countries, including most of Latin America. To better under-
least 25 P types exist, with three seen in humans: P1A[8],
stand the rationale for this vaccine, it is important to under-
P1B[4], and P2[6]. P1A and P1B share some cross-reactive
stand the immunologic basis that suggested such an ap-
proach would provide broad protection against the common epitopes, which may account for some cross-protection.8 The
serotypes, a hypothesis that has been confirmed in recent most common combinations are G1P[8], G3P[8], G4P[8],
clinical trials. G9P[8], G9P[6], and G2P[4]. This terminology is similar
to that of the influenza virus nomenclature, with the in-
fluenza H and N types corresponding to the G and P types
Rotavirus Structure of rotavirus. However, cross-protection appears to be
The rotavirus particle is a triple-layered particle with icosa- more common after rotavirus infection than after influenza
hedral symmetry (Fig. 1).7 The outer shell is composed of the virus infection.
VP4 and VP7 proteins. Antibodies to these two proteins and The inner layer of rotavirus is composed of virus protein
VP2, which is surrounded by the middle layer composed of
the most abundant structural virus protein, designated VP6
Division of Infectious Diseases, Department of Pediatrics Cincinnati Chil- (Fig. 1). VP6 is a highly conserved protein among the group
dren’s Hospital Medical Center, University of Cincinnati College of Med- A rotaviruses being discussed. The inner shell surrounds 11
icine, Cincinnati, OH. segments of double-stranded RNA, which code for 11 pro-
Address reprint requests to David I. Bernstein, MD, MA, Cincinnati Children’s
Hospital Medical Center, University of Cincinnati College of Medicine, Di-
teins, of which six are structural proteins (VP1, 2, 3, 4, 6, and
vision of Infectious Diseases, Department of Pediatrics, 3333 Burnet Ave. 7) and five are nonstructural (NSP 1-5). Of interest is the
ML 6014, Cincinnati, OH 45229. E-mail: David.Bernstein@cchmc.org possible role of NSP4 as an enterotoxin.9

188 1045-1870/06/$-see front matter © 2006 Elsevier Inc. All rights reserved.
doi:10.1053/j.spid.2006.08.006
Live attenuated human rotavirus vaccine, Rotarix™ 189

Figure 1 Rotavirus structure illustrating 11 genome segments and the proteins they encode on the left. Schematic
diagram of structure in the middle and computer-generated image on the right.

Rationale for In trials conducted in Cincinnati, a previous rotavirus in-


fection provided significant protection from both asymptom-
Vaccine Development atic and symptomatic reinfections. However, serotype G1
That induction of immune responses occurs at the initial site rotaviruses predominated in all years.15 In a large multicenter
of rotavirus exposure, the mucosal epithelium, is increasingly U.S. vaccine study, the protective efficacy of a previous rota-
being recognized; therefore, almost all of the vaccines devel- virus infection against a symptomatic reinfection was 93 per-
oped against rotavirus disease have been live attenuated oral cent (95% confidence interval 50-99%).16 In this trial, sero-
vaccines. Oral vaccines are used to best induce protection at type G1 infections predominated in year 1 (93% of isolates),
the site of infection and the major site of pathology, the but in year 2, 34 percent of isolates were nonserotype G1,
intestinal mucosa. Replication in intestinal epithelial cells suggesting that heterotypic protection was seen, at least
leads to the primary symptoms of rotavirus infection: fever, based on the VP7 serotype (G type) of the infecting strains. In
diarrhea, and vomiting.10 Although viral antigen and live this trial, symptomatic nonserotype G1 reinfection was not
virus have been found in the blood,11,12 no evidence indicates detected in subjects with natural rotavirus infections in the
that systemic spread plays a major role in the pathogenesis of first year.
the infection. Viremia, however, probably accounts for the Further, in a study in Bangladesh, children who presented
rare complications of extra-intestinal disease. with a clinically significant rotavirus illness had substantially
Rotavirus vaccines are designed to induce a protective re- fewer rotavirus-specific neutralizing antibodies in general
sponse that mimics that induced by natural infection. Thus, than did age-matched well children, but only neutralizing
in an early study, Bishop and coworkers13 reported that neo- antibodies to G heterologous rotavirus strains correlated to
natal infection with rotavirus provided significant protection protection in multivariate logistic regression analysis.17 Sim-
against subsequent disease but not reinfection. In perhaps ilarly, other studies also have failed to detect a correlation
the best study of protection after natural infections reported between G serotype-specific serum neutralizing antibodies
to date, investigators showed that children can be reinfected and protection after vaccination.18-22 In some of these studies,
with rotavirus but that each infection lowers the risk for the a correlation with the presence of rotavirus IgA antibodies
development of both a subsequent infection and a subse- was detected.14,24 However, one should note that second ep-
quent infection with diarrhea.14 In this trial, no child had isodes of severe rotavirus diseases are associated more often
moderate to severe diarrhea due to rotavirus after having two with reinfection with virus of a different serotype than rein-
previous rotavirus infections, whether or not the previous rota- fection with the same serotype.14,24 Further, early vaccine
virus infections were symptomatic. This protection was seen trials with RIT 4237, WC3, and RRV vaccine virus strains
despite the fact that four of the major rotavirus serotypes (G1- showed inconsistent results, with some convincing evidence
G4) circulated during the period of this study. These findings for protection against homologous but not heterologous in-
contributed to the rationale for developing a two-dose vaccine. fections (reviewed in Ward and Franco et al23,25,26).
190 D.I. Bernstein

Immunology antibodies also provided protection in a monkey model of


rotavirus, contradicting other studies.54
This discussion of immunologic mechanisms requires that In summary, vaccine-induced protection could be pro-
one understand that most of the studies evaluating correlates vided by antibodies induced by either shared VP7 or VP4
of protection use serum antibody levels and that the local neutralizing epitopes, by non-neutralizing IgA or IgG anti-
intestinal response could differ (reviewed in Ward and bodies, or by T-cell-mediated mechanisms. Thus, vaccine
Franco et al23,25-27). Also important to understand is that if strategies that induce high levels of IgA antibodies or that
protection is mediated by neutralizing antibodies, these an- induce cross-reactive T cells probably could provide homol-
tibodies may be induced by either the VP7 or VP4 protein, as ogous and heterologous protection. Indeed other researchers
discussed previously.7 Indeed, after a protective natural in- have stated that IgA antibody appears to be the best correlate
fection or immunization has been established, most of the of protection.23
antibodies produced are directed against the VP4 and not the
VP7 protein.28,29 True heterologous protection would be
against a virus that differs in both of these proteins. For Vaccine Development
example, protection induced by the most common serotype
Because of the initial success achieved using naturally atten-
G1P[8] and the one found in Rotarix™ against a G2P[6]
uated animal rotaviruses, such as rhesus rotavirus, as human
inoculation would be an example of heterologous protection,
vaccine candidates, the classical Jennerian technique, the
whereas protection of G1P[8] against other common types,
dominant approach has been to use bovine or monkey rota-
that is, G3P[8], G4P[8], and G9 P[8], would not.
viruses that were modified later to include human rotavirus
The reassortant vaccines RotaTeq™ (see companion arti-
gene segments (discussed in companion article). More re-
cle in this issue) and Rotashield (previously licensed but now
cently, attenuation has been accomplished by repeated pas-
withdrawn) were designed to induce neutralizing antibodies
sage of natural human isolates in tissue culture.
to multiple human serotypes by inclusion of reassortant
strains that include the VP7 and, in the case of Rotateq, also
the G[8] VP4. However, a discrepancy appears to exist in the Development of
high level of protection but modest induction of detectable
serum neutralizing antibodies to these human strains, espe- Rotavirus Vaccine 89-12
cially for Rotashield.30-32 This finding indicates that perhaps The first indication that the 89-12 strain could provide pro-
it is the magnitude of the immune response, rather than tection from subsequent rotavirus infections, especially rota-
specific virus-neutralizing antibody titers, that best correlates virus disease, came from a study conducted in Cincinnati in
with level of protection, or that factors other than serum 1988. Although the WC3 vaccine that was evaluated proved
neutralizing antibodies, such as mucosal IgA, T cells, or cy- to be ineffective, the study provided an opportunity to follow
tokines, can provide protection (reviewed in Ward et al and the subjects prospectively through a subsequent rotavirus
Franco et al22,23,25,26,33). Support for protection by each of season to evaluate protection from the natural infections that
these three immune mechanisms can be found in mouse occurred in the first year. Fortunately, the 1988/89 rotavirus
experiments and gnotobiotic pig experiments, which have season in Cincinnati was unusual in that only one strain of a
been reviewed by other authors.23,25,26,33,34 G1[P8] rotavirus, as identified by electropherotyping, circu-
Studies of experimental rotavirus infection in mice, pigs, and lated that year.55 This occurrence allowed the opportunity to
calves have revealed conflicting results regarding the absolute evaluate the protection afforded by this single strain of rotavi-
role of neutralizing antibodies in protection.24-26,35-39 Some rus.15
studies have favored a role for transcytosed IgA antibodies Protection in the infants infected in the first year was 70
and intracellular neutralization. For example, protection can percent against symptomatic rotavirus infection and 81 per-
be provided by passive administration of a monoclonal anti- cent against any rotavirus reinfection. Curiously, both infants
body to the most abundant rotavirus protein VP6, but this who developed symptoms after reinfection had not devel-
antibody is one that does not possess neutralizing activity in oped a detectable rotavirus antibody response after their pri-
classical in vitro virus neutralization assays. In one study, an mary infection. It appeared, therefore, that the strain circu-
IgA isotype, but not an IgG isotype monoclonal antibody to lating in Cincinnati in 1989 could provide protection from
VP6, provided protection40,41; in another study, an IgA rotavirus even if the initial infection were asymptomatic. Fur-
monoclonal antibody to VP6 provided protection via intra- ther infection with 89-12 induced more neutralizing anti-
cellular neutralization but not immune exclusion.42 In other bodies to the VP4 protein than the VP7 protein,28 with evi-
murine studies, mucosal VP6 immunization with adjuvant or dence of neutralizing antibodies to at least the four major
virus-like particles (VLP) composed of VP2 and VP6 pro- rotavirus serotypes G1-4.56 Recognizing that developing a
vided protection.43-45 The mechanism of this protection is not vaccine from a human rather than an animal rotavirus strain
entirely clear but appears to be mediated by CD4⫹ might have advantages, researchers decided that this strain
T lymphocytes46 and can occur in the absence of IgA anti- would form the basis for a vaccine candidate.
bodies.47 Other evidence suggests that protection can be pro- After several options to attenuate this strain were dis-
vided by CD4⫹ T lymphocytes,48,49 CD8⫹ T cells,48,50,51 and cussed, the decision was made to use multiple cell culture
cytokines.52,53 In a recent study, passively-transferred IgG passages as an attenuation strategy. One isolate from an in-
Live attenuated human rotavirus vaccine, Rotarix™ 191

fected child, designated 89-12, was selected and serially pas- Dose 1
saged 26 times in primary African Green Monkey kidney cells 100
(AGMK), then seven times in a serially passaged AGMK cell 90 RIX4414
line.57 To determine whether the passages in cell culture had 80 Placebo

70
attenuated the virus, researchers conducted studies in adults
60
followed by studies in children with previous rotavirus infec-
50

%
tions.57 After these trials provided evidence of attenuation 40
based on a lack of signs or symptoms of rotavirus disease, the 30

trial proceeded to infants. After the initial safety trial in a 20

10
small number of infants and dose-ranging studies were com-
0
pleted, the evaluation proceeded to a multicenter efficacy Fever ( 38°C) Diarrhea Vomiting Irritability Loss of
Appetite
trial.
In the initial randomized placebo-controlled, double-blind Dose 2

efficacy trial, 213 healthy infants, 10 to 16 weeks of age, were


100
given two oral doses of either 89-12 (1 ⫻ 105 PFU/mL) or 90 RIX4414
placebo after administration of an antacid.58 The vaccine was 80 Placebo
safe, although a mild fever of short duration was detected in 19 70
percent of vaccine recipients compared with 5 percent of pla- 60

cebo recipients after the first dose (P ⬍ 0.001). Rotavirus-spe- 50

%
cific antibody responses developed in 94.4 percent of vaccine 40

recipients, and vaccine virus was detected in 75 percent of stools 30

20
obtained on day 7 after immunization. In the first year after
10
vaccination was given, vaccine efficacy was 89 percent against
0
any rotavirus disease, the highest reported for any multicenter Fever ( 38°C) Diarrhea Vomiting Irritability Loss of appetite

trial. Importantly, vaccine efficacy was 100 percent against very


severe infections and those requiring medical attention. Fol- Figure 2 Percentage of RIX4414 and placebo recipients with solicited
low-up of the children continued for a second year and revealed symptoms during the 15 days after vaccination. (A) After dose one. (B)
After dose two. Reprinted with permission from De Vos et al.60
an efficacy of 76 percent (95% CI 45-85) against any rotavirus
disease and continued to be 100 percent for very severe disease
and those requiring medical intervention.59 epidemiology and disease burden of rotavirus and intussuscep-
tion, as well as the economic impact of rotavirus disease.
Rotarix™ (RIX4414) In a dose-ranging trial conducted in Singapore in 2464
infants, the vaccine was administered concurrently with rou-
Further development of the vaccine, initially named
tine childhood immunizations.62 Again, the vaccine was
RIX4414, was performed by GlaxoSmithKline after they ac-
found to be safe and did not produce an increase in the
quired the rights to the vaccine. The vaccine was developed
incidence of side effects, even at the highest dose of 106.1
further by limiting dilution cloning of 89-12 in Vero cell
fluorescent focus units (ffu). Similar to the results of the
culture and continued passage of the strain. The resulting
Finish study, the vaccine was highly immunogenic, with
vaccine then was formulated as a lyophilized preparation to
“take” rates (defined as seroconversion or vaccine virus shed-
be given after reconstitution with a liquid calcium carbonate
ding) of 97.8 to 100 percent depending on the dose. Inter-
buffer produced by GlaxoSmithKline.60 ference with the other routine childhood vaccines was not
The initial trials of RIX4414 established its safety in adults detected. More safety and immunogenicity trials have been
and previously infected toddlers before a dose-escalation conducted in South Africa, but results are not yet available.
study was performed in infants.61 In this trial, increases were In a multicenter study performed in the United States,
not detected in any solicited symptom, including fever, diar- infants 5 to 15 weeks of age received either placebo or 105.2 or
rhea, or vomiting, at any vaccine dose. Thus, the increased 106.4 ffu of RIX4414 concomitantly with their other child-
passage in tissue culture and/or the clonal selection process hood vaccines.63 The vaccine again did not increase fever,
appeared to produce a vaccine strain that was not associated vomiting, or diarrhea or any unsolicited adverse event (Fig. 2).
with the increase in mild fever seen after inoculation with Vaccine take was detected in 81.5 or 88 percent of the low- or
89-12. The immune response was influenced by the dose, high-dose vaccine recipients, respectively, after receiving two
with seroconversion detected in 73 to 96 percent of infants doses. Further, the rotavirus vaccine did not interfere with
after the second dose. Vaccine virus shedding was detected in the immune response to the other routine immunizations.
38 to 60 percent of recipients after the first dose. In the largest trial conducted to date, safety was evaluated in
Evaluation of RIX4414 began in Asia shortly thereafter, mark- more than 60,000 infants in 11 Latin American countries and
ing the initiation of what would become a global effort to eval- Finland, while efficacy was evaluated in a subgroup of more
uate this vaccine. Thus, Rotarix™ studies have been part of an than 20,000 infants.64 Infants received two doses of either vac-
intense effort, mainly centered in Latin America, to evaluate the cine at a dose of 106.5 or placebo at approximately 2 and 4
192 D.I. Bernstein

Table 1 Efficacy of RotarixTM Against Rotavirus Gastroenteritis importance, efficacy against hospitalization because of gastroen-
in Latin America and Finland teritis of any cause was 75 percent.
Rotarix™ Placebo In conclusion, efficacy of this vaccine is excellent against se-
(n ⴝ 9009) (n ⴝ 8858) Efficacy vere rotavirus disease regardless of the serotype, although effi-
Severe disease 12 77 85
cacy is reduced against strains that do not share either the VP7 or
Hospitalization 9 59 85 VP4 proteins. Efficacy is somewhat increased in the most devel-
Vesikari scale >11 11 71 85 oped countries compared with lesser developed countries, as
Vesikari scale >19 0 16 100 evidenced by comparison of the efficacy in the large trial in Latin
G1 P[8] 3 36 92 America with the one in Europe. Thus, comparison of efficacy in
G3, G4 or G9 and 4 31 87 Europe with any previous vaccine trial shows that Rotarix™
P[8] meets or exceeds previous rates of protection.
G2 P[4] 6 10 41*
*In a large meta-analysis, efficacy was 67%. Intussusception and Rotarix™
Because of the association of the previously licensed
Rotashield™ vaccine with intussusception, large safety trials
months of age. Infants received routine immunizations accord- have been conducted with both Rotateq™ and Rotarix™. As
ing to local regulations, but oral polio was provided at least 2 stated previously, Rotarix™ was evaluated in a randomized
weeks before or after administration of the vaccine. In the safety placebo controlled trial of more than 63,000 infants in Latin
cohort, significantly fewer serious adverse events and hospital- America.64 In that trial, 31,373 infants ages 6 to 17 weeks
izations were reported in the vaccine group than in the placebo were randomized to receive two doses of vaccine and 31,552
group. to receive placebo. The mean age at the administration of the
first dose was 8.2 weeks and at the second dose was 15.8
weeks of age. The primary endpoint of the study was the
Efficacy occurrence of intussusception as defined by the Brighton
collaboration group within 30 days of receiving either dose of
The first efficacy study with RIX4414 was conducted in Fin-
vaccine. A secondary evaluation involved cases occurring
land during two rotavirus epidemic seasons, 2001 to 2002,
during the duration of the study. Cases were captured by
with a relatively low, suboptimal, dose of vaccine virus (104.7
active hospital surveillance and scheduled for visits after each
ffu). In this randomized, double-blind, placebo-controlled
dose. An independent surveillance system also was in place
trial of 405 infants, the vaccine was well tolerated and immu-
to ensure that all cases were identified. Cases were reviewed
nogenic.65 In this trial, efficacy against any episode of rotavi-
by a blinded expert and monitored by an independent data
rus gastroenteritis was 73 percent, whereas efficacy against
monitoring board. Seven cases, two after the first dose was
any severe episode of rotavirus gastroenteritis was 90 percent administered, were identified in the placebo group compared
in the first season and 72 or 85 percent, respectively, over the with six, one following the first dose administered, in the
entire follow-up period. Thus, no decrease in efficacy was vaccine group. No clustering in the initial 1 to 2 weeks was
seen in the second year compared with the first. Thirty-five of identified after the receipt of either dose (Fig. 3). During the
the 38 cases of rotavirus gastroenteritis were caused by virus duration of the study, 16 cases occurred in the placebo group
of the same G1 serotype as the vaccine. compared with nine in the vaccine group. Thus, no evidence
In the large study conducted in Latin America and Finland, was found that Rotarix™ was associated with intussusception.
efficacy was 85 percent against severe rotavirus diarrhea and
hospitalizations.64 Of note, efficacy was high (more than
86%) against severe rotavirus diarrhea caused not only by Current Status
G1P[8] strains but also by the VP4-related G3[8], G4[P8], Rotarix™ currently is available in the European Union, 15
and G9[P8] strains (Table 1). Efficacy against the few G2P[4] Latin American countries, and 29 other countries, including
strains was less, 41.0 percent. However, in several meta-
analysis studies, the efficacy was 67 to 71 percent, with 95
percent CIs of 15 to 87 percent, indicating that the vaccine Table 2 Efficacy of RotarixTM Against Rotavirus Gastroenteritis
will be efficacious against strains that do share VP4 or VP7 in Europe
proteins (personal communications, GSK).66 Efficacy (95% CI)
In the most recent trial conducted in six European countries, Any disease 87% (80-92%)
4274 infants were randomized in a 2:1 ratio to receive Rotarix™ Severe disease 96% (90-99%)
or placebo concomitantly with routine vaccinations.66 Protec- Hospitalization 100% (82-100%)
tion was 87 percent against any rotavirus gastroenteritis, 96 Severe disease
percent against severe disease, and 100 percent against hospi- G1 96 (86-100%)
talization attributable to rotavirus (Table 2). Furthermore, in G3 100% (45-100%)
agreement with the study discussed previously, efficacy against G4 100% (65-100%)
G3, G4, and G9 was similar to that against G1 and exceeded 95 G9 95% (78-99%)
G2 P[4] 75% (ⴚ386 to 89%)
percent, whereas efficacy against G2 strains was 75 percent. Of
Live attenuated human rotavirus vaccine, Rotarix™ 193

Vaccine
First Dose Placebo
Infants with Intussusception

5 10 15 20 25 30 35 40

Second Dose

5 10 15 20 25 30 35 40
Figure 3 Number of cases of intussusception after the administration of Rotarix™ or placebo.

several in Asia. Ongoing discussions with the Food and Drug 7. Bernstein DI, Ward RL: Rotaviruses, in Feigin RD, Cherry JD (eds):
Administration will determine when approval is sought in Textbook of Pediatric Infectious Diseases (ed 5). W.B. Philadelphia, PA,
Saunders, 2004, pp 2110-2133
the United States.
8. Ball JM, Tian P, Zeng CQ, et al: Age-dependent diarrhea induced by a
rotaviral nonstructural glycoprotein. Science 272:101-104, 1996
Summary 9. Gorziglia M, Larralde G, Kapakian AZ, et al: Antigenic relationships
among human rotaviruses as determined by outer capsid protein VP4.
The evidence to date indicates that two oral doses of Rotarix™ is Proc Natl Acad Sci USA 87:7155-7159, 1990
safe and immunization is not associated with either fever or 10. Staat MA, Azimi PH, Berke T, et al: Clinical presentations of rotavirus
infection among hospitalized children. Pediatr Infect Dis J 21:221-227,
gastrointestinal side effects. Most importantly, a large safety trial
2002
showed no association with intussusception. To date, there has 11. Blutt SE, Kirkwood CD, Parreno V, et al: Rotavirus antigenaemia and
been no impact on the immunogenicity of concurrently admin- viraemia: a common event? Lancet 362:1445-1449, 2003
istered vaccines, allowing the vaccine to be administered within 12. Fischer TK, Ashley D, Kerin T, et al: Rotavirus antigenemia in patients
existing immunizations schedules. Efficacy studies conducted with acute gastroenteritis. J Infect Dis 192:913-919, 2005
in Europe and Latin America also have indicated excellent pro- 13. Bishop RF, Barnes GL, Cipriani E, et al: Clinical immunity after neona-
tection especially against severe disease and hospitalizations due tal rotavirus infection. A prospective longitudinal study in young chil-
dren. N Engl J Med 309:72-76, 1983
to rotavirus, including those caused by nonserotype I infections.
14. Velazquez FR, Matson DO, Calva JJ, et al: Rotavirus infection in infants
Results of future trials currently underway will be needed to as protection against subsequent infections. N Engl J Med 335:1022-
determine whether the efficacy detected in the completed stud- 1028, 1996
ies also will be seen in the least developed nations of the world, 15. Bernstein DI, Sander DS, Smith VE, et al: Protection from rotavirus rein-
where rotavirus infections take their highest toll. fection: two year prospective study. J Infect Dis 164:277-283, 1991
16. Ward RL, Bernstein DI: Protection against rotavirus disease after natu-
ral rotavirus infection. US Rotavirus Vaccine Efficacy Group. J Infect
References
Dis 169:900-904, 1994
1. Charles MD, Holman RC, Curns AT, et al: Hospitalizations associated
17. Ward RL, Clemens JD, Knowlton DR, et al: Evidence that protection
with rotavirus gastroenteritis in the United States, 1993-2002. Pediatr
against rotavirus diarrhea after natural infection is not dependent on sero-
Infect Dis J 25:489-493, 2006
type-specific neutralizing antibody. J Infect Dis 166:1251-1257, 1992
2. Malek MA, Curns AT, Holman RC, et al: Diarrhea- and rotavirus-
18. Vesikari T, Isolauri E, Delem A, et al: Immunogenicity and safety of live
associated hospitalizations among children less than 5 years of age:
oral attenuated bovine rotavirus vaccine strain RIT 4237 in adults and
United States, 1997 and 2000. Pediatrics 117:1887-1892, 2006
3. Parashar UD, Holman RC, Clarke MJ, et al: Hospitalizations associated young children. Lancet 2:807-811, 1983
with rotavirus diarrhea in the United States, 1993 through 1995: sur- 19. Vesikari T, Isolauri E, Delem A, et al: Clinical efficacy of the RIT 4237
veillance based on the new ICD-9-CM rotavirus-specific diagnostic live attenuated bovine rotavirus vaccine in infants vaccinated before a
code. J Infect Dis 177:13-17, 1998 rotavirus epidemic. J Pediatr 107:189-194, 1985
4. Hsu VP, Staat MA, Roberts N, et al: Use of active surveillance to validate 20. Madore HP, Christy C, Pichichero M, et al: Field trial or rhesus rotavi-
international classification of diseases code estimates of rotavirus hos- rus or human-rhesus rotavirus reassortant vaccine of VP7 serotype 3 or
pitalizations in children. Pediatrics 115:78-82, 2005 1 specificity in infants. J Infect Dis 166:235-243, 1992
5. Parashar UD, Hummelman EG, Bresee JS, et al: Global illness and deaths 21. Ward RL, Bernstein DI: Lack of correlation between serum rotavirus
caused by rotavirus disease in children. Emerg Infect Dis 9:565-572, 2003 antibody titers and protection following vaccination with reassortant
6. Parashar UD, Gibson CJ, Bresse JS, et al: Rotavirus and severe child- RRV vaccines. Vaccine 13:1226-1232, 1995
hood diarrhea. Emerg Infect Dis 12:304-306, 2006 22. Ward RL, Knowlton DR, Zito ET, et al: Serologic correlates of immunity
194 D.I. Bernstein

in a tetravalent reassortant rotavirus vaccine trial. J Infect Dis protects against viral shedding after challenge with murine rotaviruses.
176:570-577, 1997 Vaccine 20:3310-3321, 2002
23. Friedman MG, Galil A, Sarov B, et al: Two sequential outbreaks of 45. Conner ME, Zarley CD, Hu B, Parsons S, et al: Virus-like particles as a
rotavirus gastroenteritis: Evidence for symptomatic and asymptomatic rotavirus subunit vaccine. J Infect Dis 174:Suppl:S88-S92, 1996
reinfections. J Infect Dis 158:814-822, 1988 46. McNeal MM, VanCott JL, Choi AH-C, et al: CD4 T cells are the only
24. Ward RL, Greenberg HB, Estes MK: Viral gastroenteritis naccines, in lymphocytes needed to protect mice against rotavirus shedding after
Ogra PL, Mestecky MD, Lamm ME, et al (eds): Musosal Immunology intranasal immunization with a chimeric VP6 protein and the adjuvant
(ed 3). Academic Press, San Diego, 2005, pp 887-903 LT(R192G). J Virol 76:560-568, 2002
25. Ward RL: Possible mechanisms of protection elicited by candidate ro- 47. McNeal MM, Stone SC, Basu M, et al: Protection against rotavirus
tavirus vaccines as determined with the adult mouse model. Viral Imu- shedding after intranasal immunization of mice with a chimeric VP6
nol 16L7-24, 2003 protein does not require intestinal IgA. Virology 346:338-347, 2006
26. Franco MA, Angel J, Greenberg HB: Immunity and correlates of pro- 48. McNeal MM, Rae MN, Ward RL: Evidence that resolution of rotavirus
tection for rotavirus vaccines. Vaccine 24:2718-2731, 2006 infection in mice is due to both CD4 and CD8 T cell-dependent activ-
27. Jiang B, Gentsch JR, Glass RI: The role of serum antibodies in the ities. J Virol 71:8735-8742, 1997
protection against rotavirus disease: an overview. Clin Infect Dis 34: 49. Kushnir N, Bos NA, Zuercher AW, et al: B2 but not B1 cells can con-
1351-1361, 2002 tribute to CD⫹ T-cell mediated clearance of rotavirus in SCID mice.
28. Ward RL, McNeal MM, Sander DS, et al: Immunodominance of the VP4 J Virol 75:5482-5490, 2001
neutralization protein of rotavirus in protective natural infections in 50. Jaimes MC, Feng N, Greenberg HB: Characterization of homologous
young children. J Virol 67:464-468, 1993 and heterologous rotavirus-specific T-cell responses in infant and adult
29. Yuan L, Ishida S, Honma S, et al: Homotypic and heterotypic serum mice. J Virol 79:4568-4579, 2005
isotype-specific antibody responses to rotavirus nonstructural protein 51. Franco MA, Tin C, Greenberg HB: CD8⫹ T cells can mediate almost
4 and viral protein (VP) 4, VP6, and VP7 in infants who received complete short-term and partial long-term immunity to rotavirus in
selected live oral rotavirus vaccines. J Infect Dis 189:1833-1845, 2004 mice. J Virol 71:4165-4170, 1997
30. Bernstein DI, Glass RI, Rodgers G, et al: Evaluation of rhesus rotavirus 52. Kaufhold RM, Field JA, Caulfied MJ, et al: Memory T-cell response to
monovalent and tetravalent reassortant vaccines in US children. US rotavirus detected with a gamma interferon enzyme-linked immuno-
Rotavirus Vaccine Efficacy Group. JAMA 273:1191-1196, 1995 spot assay. J Virol 79:5684-569, 2005
31. Rennels MB, Glass RI, Dennehy PH, et al: Safety and efficacy of high- 53. Vancott JL, McNeal MM, Choi AH, et al: The role of interferons in rotavirus
dose rhesus-human reassortant rotavirus vaccines–report of the Na- infections and protection. J Interferon Cytokine Res 23:163-170, 2003
tional Multicenter Trial. United States Rotavirus Vaccine Efficacy 54. Westerman LE, McClure HM, Jiang B, et al: Serum IgG mediates mu-
Group. Pediatrics 97:7-13, 1996 cosal immunity against rotavirus infection. Proc Natl Acad Sci USA
32. Vesikari T, Matson DO, Dennehy P, et al: Safety and efficacy of a 102:7268-7273, 2005
pentavalent human-bovine (WC3) reassortant rotavirus vaccine. 55. Bernstein DI, Smith V, Sander D, et al: Evaluation of WC3 rotavirus
N Engl J Med 354:23-33, 2006 vaccine and correlates of protection in healthy infants. J Infect Dis
33. Ward R, Clark HR, Offit P, et al: Live vaccine strategies to prevent rotavirus 162:1055-1062, 1990
disease, in: Levine, Kaper, Rappuoli, et al (eds): New Generation Vaccines 56. Ward RL, Sander DS, Schiff GM, et al: Effect of vaccination on serotype-
(ed 3). Mercel Dekker, Inc., New York, 1997, pp 607-620 specific antibody responses in infants administered WC3 bovine rota-
34. Yuan L, Saif LJ: Induction of mucosal immune responses and protec- virus before or after a natural rotavirus infection. J Infect Dis 162:1298-
tion against enteric viruses: rotavirus infection of gnotobiotic pigs as a 1303, 1990
model. Vet Immunol Immunopathol 87:147-160, 2002 57. Bernstein DI, Smith VE, Sherwood JR, et al: Safety and immunogenicity of a
35. Wyatt RG, Mebus CA, Yolken RH, et al: Rotaviral immunity in gnoto- live, attenuated human rotavirus vaccine 89-12. Vaccine 16:381-387, 1998
biotic calves: heterologous resistance to human virus induced by bo- 58. Bernstein DI, Sack DA, Rothstein E, et al: Efficacy of live, attenuated,
vine virus. Science 203:548-550, 1979 human rotavirus vaccine 89-12 in infants: a randomised placebo-con-
36. Woode GN, Kelso NE, Simpson TF, et al: Antigenic relationships trolled trial. Lancet 354:287-290, 1999
among some bovine rotaviruses: serum neutralization and cross-pro- 59. Bernstein DI, Sack DA, Reisinger K, et al: Second year follow-up eval-
tection in gnotobiotic calves. J Clin Microbiol 18:358-364, 1983 uation of live, attenuated, human rotavirus 89-12 in healthy infants.
37. Woode GN, Zheng S, Rosen BI, et al: Protection between different J Infect Dis 186:1487-1489, 2002
serotypes of bovine rotavirus in gnotobiotic calves: specificity of serum 60. De Vos B, Vesikari T, Linhares AC, et al: A rotavirus vaccine for pro-
antibody and coproantibody responses. J Clin Microbiol 25:1052- phylaxis of infants against rotavirus gastroenteritis. Pediatr Infect Dis J
1058, 1987 23:S179-S182, 2004 (suppl 10)
38. Bridger JC, Oldham G: Avirulent rotavirus infections protect calves 61. Vesikari T, Karvonen A, Korhonen T, et al: Safety and immunogenicity
from disease with and without inducing high levels of neutralizing of RIX4414 live attenuated human rotavirus vaccine in adults, toddlers
antibody. J Gen Virol 68:2311-2317, 1987 and previously uninfected infants. Vaccine 22:2836-2842, 2004
39. Ward RL, McNeal MM, Sheridan J: Evidence that active protection 62. Phua KB, Quak SH, Lee BW, et al: Evaluation of RIX4414, a live,
following oral immunization of mice with live rotavirus is not depen- attenuated rotavirus vaccine, in a randomized, double-blind, placebo-
dent on neutralizing antibody. Virology 188:57-66, 1992 controlled phase 2 trial involving 2464 Singaporean infants. J Infect Dis
40. Schwartz-Cornil I, Benureau Y, Greenberg H, et al: Heterologous pro- 192:S6-S16, 2005 (suppl 1)
tection induced by the inner capsid proteins of rotavirus requires trans- 63. Dennehy PH, Brady RC, Halperin SA, et al: Comparative evaluation of
cytosis of mucosal immunoglobulins. J Virol 76:8110-8117, 2002 safety and immunogenicity of two dosages of an oral live attenuated
41. Burns JW, Sindat-Pajouh M, Krishnaney AA, et al: Protective effect of human rotavirus vaccine. Pediatr Infect Dis J 24:481-487, 2005
rotavirus VP6-specific IgA monoclonal antibodies that lack neutralizing 64. Ruiz-Palacios GM, Perez-Schael I, Velazquez FR, et al: Safety and effi-
activity. Science 272:104-107, 1996 cacy of an attenuated vaccine against severe rotavirus gastroenteritis.
42. Feng N, Lawton JA, Gilbert J, et al: Inhibition of rotavirus replication by N Engl J Med 354:11-22, 2006
a non-neutralizing, rotavirus VP-6 specific IgA mAb. J Clin Invest 109: 65. Vesikari T, Karvonen A, Puustinen L, et al: Efficacy of RIX4414 live
1203-1213, 2002 attenuated human rotavirus vaccine in Finnish infants. Pediatr Infect
43. Choi AH, Basu M, McNeal MM, et al: Antibody-independent protection Dis J 23:937-943, 2004
against rotavirus infection of mice stimulated by intranasal immuniza- 66. Vesikari T, Karvonen A, Prymula R, et al: Human rotavirus vaccine
tion with chimeric VP4 or VP6 protein. J Virol 73:7574-7581, 1999 Rotarix™ (RIX4414) is highly efficacious in Europe. Abstract of the
44. Choi AH, McNeal MM, Basu M, et al: Intranasal or oral immunization of 24th Annual Meeting of the European Society for Pediatric Infectious
inbred and outbred mice with murine or human rotavirus VP6 proteins Diseases. Basel, Switzerland, 2006

You might also like