Rotavirus Vaccines For Infants - 2019 - Uptodate

Download as pdf or txt
Download as pdf or txt
You are on page 1of 26

Rotavirus vaccines for infants

All topics are updated as new evidence becomes available and our peer review process is complete.
Literature review current through: Apr 2019. | This topic last updated: Apr 01, 2019.

Authors:
David O Matson, MD, PhD
Miguel G O'Ryan, MD
Section Editors:
Morven S Edwards, MD
Teresa K Duryea, MD
Deputy Editor:
Mary M Torchia, MD

Contributor Disclosures

INTRODUCTION — Rotavirus is the most common cause of severe, acute gastroenteritis in infants and
children worldwide [1-3]. In the prevaccine era, rotavirus was estimated to cause approximately 440,000
deaths, 2 million hospitalizations, and 25 million outpatient visits per year worldwide among children <5
years of age [2].

More than 60 percent of countries and more than 85 percent of countries in the Global Alliance for
Vaccines and Immunization have introduced or are planning to introduce rotavirus vaccines into national
or subnational vaccine programs [4].

Rotavirus vaccination of infants will be discussed below. The pathogenesis, clinical presentation, and
diagnosis of rotavirus gastroenteritis are discussed separately, as are general measures to prevent viral
gastroenteritis in children. (See "Clinical manifestations and diagnosis of rotavirus infection" and "Acute
viral gastroenteritis in children in resource-rich countries: Management and prevention".)

MICROBIOLOGY — Rotavirus is a double-stranded RNA virus in the Reoviridae family [3]. The outer
capsid contains two proteins that define rotavirus serotypes: a G protein (VP7) and a P protein (VP4)
(figure 1). Five G-P combinations account for approximately 90 percent of human rotaviruses circulating
worldwide: G1P[8], G2P[4], G3P[8], G4P[8], and G9P[8], but serotype prevalence varies from time to
time and from place to place for reasons that are not understood. The effects of infant immunization on
rotavirus serotype prevalence are discussed below. (See 'Serotype selection' below.)

ROTAVIRUS VACCINES

Vaccine development — Rotavirus vaccines have been developed from animal rotavirus strains, human-
animal rotavirus reassortants (genes from human and animal strains), attenuated human rotaviruses,
subunits of rotavirus virions, and virus-like particles [5-11]. For live virus-based vaccines, reassortants
are necessary because most human rotaviruses grow too poorly in cell culture for production of standard
vaccine lots for large-scale immunization programs. Monovalent vaccines prepared from animal
rotaviruses have not been promising in humans. Assessment of rotavirus subunit vaccine candidates has
reached human studies [12].
Vaccines licensed in the United States — Two live, attenuated oral rotavirus vaccines are licensed for use
in the United States and other countries (table 1). The vaccines have similar efficacy and safety, and no
preference for one over the other vaccine exists, with a few exceptions (eg, latex allergy) [1,3,13,14]. (See
'Contraindications' below and 'Efficacy/effectiveness' below and 'Adverse events and safety' below.)

●Pentavalent human-bovine rotavirus reassortant vaccine (RV5, PRV, RotaTeq) is based upon the
bovine strain, WC3, which is naturally attenuated for humans but not broadly cross-protective. Each
reassortant component contains a single gene derived from a human rotavirus strain encoding a major
outer capsid protein from the most common human serotypes: G1, G2, G3, G4, and P1[8] (figure 2). This
vaccine induces homologous (ie, serotype-specific) protection against the common types of exposure, as
well as the nonvaccine type G9 [15].

●Attenuated human rotavirus vaccine (RV1, HRV, Rotarix) is a monovalent vaccine derived from the
most common human rotavirus serotype combination (G1P[8]) that has been attenuated by serial passage
in cell culture (figure 2) [8]. Observational studies of natural rotavirus infection suggest that infection with
one serotype provides at least partial cross-protection against most other serotypes [16].

Other vaccines

●Human-bovine reassortant vaccine (116E, ROTAVAC) – The 116E rotavirus vaccine is a


nonpathogenic strain (G9P[11]) that occurs naturally in India. It contains a virus reassortant strain in which
one gene was from a rotavirus strain naturally occurring in bovines (P[11]) and 10 genes were from a
rotavirus strain naturally occurring in humans. Phase 3 clinical trials have been completed (demonstrating
efficacy of 54 percent against severe rotavirus gastroenteritis) [10]. The 116E vaccine is licensed for use
in India and has World Health Organization (WHO) prequalification for use in developing countries [17-
19].

●Lamb rotavirus vaccine – A monovalent, G10P[12] oral rotavirus vaccine is licensed in China, where
its effectiveness was demonstrated in two population-based studies [20,21]. However, a mouse study
suggests that monovalent rotavirus vaccines are not sufficiently immunogenic against heterotypic strains
for protection against rotavirus disease in China [18]. Previous studies indicate that strain variation and
induction of antibody response is well correlated in mice and humans [22].

●Oral bovine rotavirus pentavalent vaccine (BRV-PV, ROTASIIL) – BRV-PV is a heat-stable


bovine-human reassortant vaccine that contains serotypes G1, G2, G3, G4, and G9 [23,24]. It is
administered in three doses at 6, 10, and 14 weeks.

A randomized trial compared BRV-PV with placebo in 3508 Nigerian infants [23,24]. Four weeks after
the third dose, infants who received BRV-PV had fewer episodes of laboratory-confirmed, severe
rotavirus gastroenteritis than those who received placebo (31 versus 87 episodes; efficacy of 67 percent,
95% CI 50-78 percent). The rates of adverse events were similar between groups. None of the participants
had confirmed intussusception. In a multicenter randomized trial in 7500 infants in India, the efficacy of
BRV-PV in preventing severe rotavirus gastroenteritis before age two years was 39 percent (95% CI 26-
49 percent) [25].
BRV-PV is less expensive than RV5 and RV1 and may be more suitable for vaccination programs in
remote areas where cold-chain capacity is limited [23]. It is licensed for use in India and has WHO
prequalification for use in developing countries [19].

●Oral human neonatal rotavirus vaccine (RV3-BB) – RV3-BB is a naturally attenuated oral vaccine
developed from a strain initially recovered from an outbreak in a nursery in which infections were
asymptomatic (G3P[6]) [26]. In a phase 2 randomized trial in Indonesia, RV3-BB was efficacious in
preventing severe rotavirus gastroenteritis before age 18 months when administered to neonates (at age 0
to 5 days, 8 weeks, and 14 weeks) and infants (at age 8, 14, and 18 weeks). RV3-BB also appears to be
safe and immunogenic [26,27].

●Tetravalent human-rhesus reassortant vaccine – An oral tetravalent human-rhesus rotavirus


reassortant vaccine (RRV-TV, RotaShield) was licensed in 1998 and recommended for universal
immunization of term infants in the United States, but was withdrawn from the market in 1999 because
of an epidemiologic link to intussusception occurring within two weeks after vaccine administration [28-
32]. (See 'Risk with RRV-TV' below.)

INDICATIONS — We recommend universal immunization of infants against rotavirus, as recommended


by the Centers for Disease Control and Prevention, the World Health Organization, the American
Academy of Pediatrics, the American Academy of Family Physicians, the European Society for Pediatric
Infectious Diseases, and the European Society for Paediatric Gastroenterology, Hepatology, and Nutrition
[1,3,13,14].

In randomized trials and meta-analyses, rotavirus vaccines are highly efficacious in preventing rotavirus
gastroenteritis and rotavirus gastroenteritis-associated hospitalization and health care utilization [33-35].
(See 'Efficacy/effectiveness' below.)

CONTRAINDICATIONS — Rotavirus vaccines are contraindicated in those infants [1,13,14,36-38]:

●Who are allergic to any of the ingredients of the vaccine; attenuated human rotavirus vaccine is
contraindicated in infants with severe (anaphylactic) allergy to latex because the applicator contains latex;
pentavalent human-bovine rotavirus reassortant vaccine can be administered to such infants (see "Allergic
reactions to vaccines", section on 'Latex')

●Who had a severe (anaphylactic) allergic reaction after a previous dose

●With severe combined immunodeficiency (SCID) – Vaccine-acquired rotavirus disease has been
reported in infants who subsequently were diagnosed with SCID [36,39-43] (see "Severe combined
immunodeficiency (SCID): An overview")

●With a history of intussusception – Fatal intussusception after the second dose has been reported in
infants with a history of intussusception after the first dose [37,44]

●Born in the United States (or other countries with evidence of community ["herd"] immunity and good
outcomes after severe rotavirus disease) to women who received a biologic response modifier (eg,
infliximab, adalimumab) during pregnancy [14]; these infants can have detectable drug concentrations for
up to nine months [45-47] (see "Fertility, pregnancy, and nursing in inflammatory bowel disease", section
on 'Anti-tumor necrosis factor agents')

Rotavirus vaccine should be avoided for the first 12 months after the final in utero exposure. Catch-up
vaccination is not recommended given that the maximum age for the first dose is 14 weeks, 6 days [14].

In countries where there is evidence of community immunity and good outcomes after severe rotavirus
disease, the risk of developing vaccine-type rotavirus disease in an infant with prenatal exposure to a
biologic response modifier is greater than the risk of developing severe or fatal wild type rotavirus disease
if unvaccinated.

Although few data exist upon which to base recommendations, the 2013 Infectious Diseases Society of
America guidelines on vaccination of the immunocompromised host consider the following conditions to
also be contraindications to rotavirus vaccination because of the potential risk of serious adverse effects
[38]:

●Combined immunodeficiencies – Di George syndrome and other combined immunodeficiencies with a


CD3 count <500 cells/microL, Wiskott-Aldrich syndrome, X-linked lymphoproliferative disease, and
familial disorders that predispose to hemophagocytic lymphohistiocytosis (eg, Griscelli syndrome,
Chediak-Higashi syndrome, Hermansky-Pudlak syndrome) (see "Combined immunodeficiencies")

●Certain phagocytic cell deficiencies – Leukocyte adhesion deficiency, defects of cytotoxic granule
release (eg, Chediak-Higashi), and other undefined phagocytic cell defects (see "Leukocyte-adhesion
deficiency" and "Chediak-Higashi syndrome")

●Major antibody deficiencies treated with immunoglobulin therapy (see "Primary humoral
immunodeficiencies: An overview")

●Planned or status-post hematopoietic stem cell transplant

●Planned or current receipt of cancer chemotherapy

●Status-post solid organ transplant

●Chronic inflammatory disease treated with immunosuppressive medications (ie, prednisone,


azathioprine, 6-mercaptopurine, biologic agents [eg, tumor necrosis factor antagonists, rituximab])

These conditions are uncommon to rare among infants in the age group for rotavirus vaccine
administration (six weeks to eight months). (See 'Schedule' below.)

The following conditions are not contraindications to rotavirus vaccination [1,14,38]:

●Immunocompromised family member or household member (see 'Immunocompromised household


contact' below)

●Breastfeeding (efficacy trials included breastfeeding infants, with no alteration in normal breastfeeding
patterns) [48,49]
●Pregnant family member or household contact

PRECAUTIONS

●Immunodeficiency other than severe combined immunodeficiency – Decisions regarding


immunization of children with known or suspected immunodeficiency other than severe combined
immunodeficiency (SCID) (which is a contraindication to rotavirus vaccine) should be made on a case-
by-case basis after considering the risks and benefits; however, there are few data regarding the efficacy
or safety of rotavirus vaccine to infants who are potentially immunocompromised.

In this setting, the United States Advisory Committee on Immunization Practices (ACIP) advises
consultation with an immunologist or infectious disease specialist and the American Academy of
Pediatrics advises precaution for administration of rotavirus vaccine for manifestations of altered
immunocompetence other than SCID [1,14,36]. The 2013 Infectious Diseases Society of America (IDSA)
guidelines on vaccination of the immunocompromised host suggest that rotavirus vaccine may be
administered to infants with the following conditions [38]:

•HIV exposure or infection – Rotavirus gastroenteritis may be particularly severe in HIV-infected


children. In a prospective study in South Africa, HIV-infected children were more likely to have a
prolonged hospitalization and had a fourfold increased mortality rate compared with HIV-uninfected
children admitted to the same hospital [50].

Rotavirus vaccine trials that included HIV-infected infants have found no evidence of enhanced disease
in HIV-infected children [38]. In randomized trials, administration of attenuated human rotavirus vaccine
(RV1) and pentavalent human-bovine rotavirus reassortant vaccine (RV5) was safe and immunogenic
when administered to HIV-positive infants in a three-dose schedule [51,52].

Groups other than the IDSA (eg, the Department of Health and Human Services, Centers for Disease
Control and Prevention, American Academy of Pediatrics) suggest that the potential risks and benefits of
rotavirus vaccination be considered before vaccination [53]; however, they suggest that the potential delay
in establishment of definitive diagnosis of HIV infection beyond the recommended age for the first dose
of rotavirus vaccine and considerable attenuation of rotavirus vaccine strains support vaccination of HIV-
exposed and infected infants.

•Primary (congenital) complement deficiencies. (See "Inherited disorders of the complement system".)

•Chronic granulomatous disease. (See "Chronic granulomatous disease: Pathogenesis, clinical


manifestations, and diagnosis".)

•Congenital or cyclic neutropenia. (See "Congenital neutropenia" and "Cyclic neutropenia".)

•Immunoglobulin A deficiency, provided that all other components of the immune system are normal.
(See "Selective IgA deficiency: Clinical manifestations, pathophysiology, and diagnosis".)

•Specific polysaccharide antibody deficiency provided that all other components of the immune system
are normal. (See "Specific antibody deficiency".)
For infants with innate immune defects that result in defects of cytokine generation or response or cellular
activation (eg, defects of the interferon-gamma/interleukin-12 axis), the IDSA recommends consultation
with a specialist before administration of live vaccines [38].

●Other precautions – The ACIP and American Academy of Pediatrics suggest the following conditions
are precautions for the administration of rotavirus vaccines, because the safety and efficacy of the vaccine
were not specifically evaluated in infants with these conditions [1,14]:

•Acute moderate-to-severe gastroenteritis – Immunization of infants with acute, moderate-to-severe


gastroenteritis should be deferred until the illness resolves. A concern is that the antibody response may
be diminished.

•Moderate-to-severe febrile illness – Immunization of infants with moderate to severe illness of any type
should be deferred until the illness resolves. Deferral of the immunization makes it easier to differentiate
possible adverse effects related to the vaccine from manifestations of the underlying disease.

•Pre-existing or acquired chronic gastrointestinal disease excluding intussusception – Infants with pre-
existing or acquired gastrointestinal conditions (eg, congenital malabsorption syndromes, Hirschsprung
disease, short-bowel syndrome, some forms of cystic fibrosis-related gastrointestinal disease) who are not
receiving immunosuppressive therapy should benefit from rotavirus vaccines, and the benefits outweigh
the theoretic risks. In a pilot study, RV1 appeared to be safe and immunogenic in 14 children with early
intestinal failure [54]. In another small study, RV5 appeared to be safe and immunogenic in five infants
with a history of bowel resection [55].

•Spina bifida or bladder exstrophy – Infants with spina bifida or bladder exstrophy have a high risk of
developing latex allergy. To minimize latex exposure, some experts suggest that RV5 be used for such
infants because the applicator is latex-free. If RV5 is not available, the ACIP recommends that RV1 be
used because the benefit of vaccination is greater than the risk of sensitization [1].

SCHEDULE

Routine schedule — The recommended routine schedules for pentavalent human-bovine rotavirus
reassortant vaccine (RV5) and attenuated human rotavirus vaccine (RV1) differ [1,14]:

●RV5 is administered in three oral doses at two, four, and six months of age.

●RV1 is administered in two oral doses at two and four months.

Whenever possible, the rotavirus vaccine series should be completed with the same vaccine product [1];
however, vaccination should not be deferred if the product used for previous doses is not known. A total
of three doses of vaccine should be given to infants who received RV5 for any dose and infants in whom
the vaccine product for previous dose(s) is unknown.

An open-label, multicenter randomized trial confirmed that completion of rotavirus immunization using a
combination of RV5 and RV1 was as immunogenic as completion with RV5 or RV1 and was well
tolerated [56]. In postlicensure surveillance, completion of immunization with a combination of RV5 and
RV1 (eg, two doses of RV5 and one dose of RV1 or two doses of RV1 and one dose of RV5) was 80
percent (95% CI 51-92 percent) effective in preventing rotavirus gastroenteritis [57]. Although mixed
schedules and single formulation schedules were not directly compared, this is similar to published rates
of effectiveness for three doses of RV5 or two doses of RV1 in the same population [58].

Catch-up schedule — The catch-up schedules for RV5 and RV1 differ:

●RV5 – In the United States, the first dose of RV5 should be given between 6 and 15 weeks of age [1,14].
Two subsequent doses are administered with a minimum interval of four weeks between doses. The third
dose should not be administered after eight months, zero days of age.

The vaccine series should not be initiated in infants who are older than 14 weeks, 6 days of age [1]. The
safety of the first dose of rotavirus vaccine in older infants was not studied in the prelicensure trials;
however, for infants in whom the first dose is inadvertently administered at 15 weeks or older, the rest of
the rotavirus immunization series should be completed as described above [1]. The timing of the first dose
should not affect the safety and efficacy of the second and third dose.

In Europe, the first dose of RV5 should be given between 6 and 12 weeks, preferably at 6 to 8 weeks, and
the full schedule completed by 24 weeks of age, but preferably earlier [13].

●RV1 – In the United States, the first dose of RV1 should be given between 6 and 15 weeks of age and
the full schedule completed by eight months, zero days of age [1,14].

In Europe, the first dose of RV1 should be given between 6 and 12 weeks, preferably at 6 to 8 weeks, and
the full schedule completed by 24 weeks of age, but preferably earlier [13].

There is no maximum interval between doses.

Special circumstances

Premature infants — In the United States, rotavirus vaccines can be administered to premature infants
who are clinically stable, at least six weeks old, and are going to be or have been discharged from the
nursery [1,14,59-61]. The European Society for Paediatric Infectious Diseases and the Australian
government recommend rotavirus vaccination of premature infants according to their chronologic age,
whether or not they have been discharged from the nursery, with appropriate precautions to prevent
transmission to high-risk contacts [13,62]. (See 'Shedding and transmission of vaccine virus' below.)

In retrospective reviews, age-appropriate administration of RV5 to preterm enterally-fed infants in the


neonatal intensive care unit was well tolerated and did not appear to be associated with transmission
[63,64]. Subsequent prospective studies support the low risk of transmission [65,66]. The larger
prospective study evaluated rotavirus shedding and transmission among infants age <15 weeks admitted
to an intensive care unit with single or double room assignments of an academic medical center that
permitted RV5 administration during hospitalization [65]. Rotavirus was detected in 13 of 1192 stool
specimens (1.1 percent) collected weekly: one wild-type strain from an unvaccinated infant and 12
vaccine-type strains from nine vaccinated infants. No vaccine-type rotavirus cases were observed among
unvaccinated infants during 1952 days of potential exposure, and no reassortants were identified. These
findings suggest that delaying RV5 vaccination until hospital discharge may be unnecessary in hospitals
with comparable infection control standards. In making decisions for individual infants, attending
clinicians must consider the benefit of vaccination with possible asymptomatic transmission and the risk
of missed opportunity for rotavirus vaccination.

Infants with rotavirus gastroenteritis — The rotavirus vaccine series should be initiated or completed in
infants who have had rotavirus gastroenteritis before receiving the full two- or three-dose series because
natural first infections do not provide complete immunity against subsequent severe disease and multiple
serotypes of rotavirus usually are present in any community [1,14,67].

Receipt of blood products — Rotavirus vaccines may be administered at any time in relation to the receipt
of blood products, including antibody-containing products [1,68].

Hospitalization of vaccinated infant — In the event that an infant requires hospitalization after
administration of rotavirus vaccine, standard precautions should be used to prevent the spread of vaccine
virus in the hospital setting [14]. No additional infection control measures are necessary. Co-rooming with
children with severe combined immunodeficiency or suspected severe immunodeficiency is not
recommended. (See "Infection prevention: Precautions for preventing transmission of infection", section
on 'Standard precautions' and 'Shedding and transmission of vaccine virus' below.)

Immunocompromised household contact — Rotavirus vaccines can be administered to infants living in


households with immunocompromised persons [1,14,38]. (See 'Shedding and transmission of vaccine
virus' below.)

Resource-poor countries with high child mortality — The World Health Organization recommends
routine rotavirus vaccination for all its member countries [24]. Rotavirus vaccine effectiveness varies
among the countries that have introduced a rotavirus vaccine into the routine infant immunization schedule
[69-77]. Although poverty is a surrogate marker for poorer population vaccine effectiveness [78-81],
rotavirus vaccine remains effective in reducing rotavirus-related hospitalization and emergency
department visits in resource-poor countries with high child mortality [82-84]. (See
'Efficacy/effectiveness' below.)

Factors that may explain variation in vaccine effectiveness include:

●Greater portion of missed vaccination opportunities, related to the narrow age range for administration
[75,77]

●Failure to adopt a universal recommendation for rotavirus vaccination (which may prevent or reduce
herd immunity) [85,86]

●Variation in herd effect with increasing interval from initiation of universal vaccination with high uptake
[71,87,88]

●Relatively increased natural, wild-type rotavirus circulation, reducing rotavirus vaccine effectiveness
with increasing interval from initiation of universal vaccination [73,89-91]

●Naturally acquired rotavirus disease in infants too young to be vaccinated [70]


●Attenuation of vaccine response by transplacental maternal antibody [92] or in infants who are
exclusively breast-fed [93]

●Malnutrition or poor diet (which may affect the intestinal biome) [70,94,95]

●Immunogenic competition with simultaneous administration of oral polio vaccine [96] (see
'Administration with other vaccines' below)

●Histo-blood group antigens and secretor status [97,98]

ADMINISTRATION — The licensed rotavirus vaccines are administered orally [1]. Each dose of
pentavalent human-bovine rotavirus reassortant vaccine (RV5) is 2 mL (table 1). Each dose of attenuated
human rotavirus vaccine (RV1) is 1 mL. To avoid loss of a portion of the dose and of splashes into the
eyes of infants, parents, or health care providers, the vaccine should be administered gently inside the
cheek [99].

Dietary restriction, including breastfeeding, is not needed before or after rotavirus vaccine is administered.
Doses that are regurgitated, spit out, or vomited need not be repeated (they were not repeated in
prelicensure studies of efficacy) [1,14,68]. Multiple randomized trials have demonstrated that
breastfeeding does not affect rotavirus vaccine efficacy; normal breastfeeding was not altered in the
prelicensure trials [33,35,48,49]. In randomized trials that specifically evaluated this issue, immune
response was not affected by abstaining from breastfeeding for ≥1 hour before and after each dose of
rotavirus vaccine [100-102].

In the event that rotavirus vaccine is injected rather than administered orally, the dose is not considered
valid and should be repeated within the appropriate age and dosing schedule [99].

ADMINISTRATION WITH OTHER VACCINES — Rotavirus vaccines can be administered at the same
visit as the other routine infant immunizations [1,14,103,104].

Simultaneous administration of oral polio vaccine (OPV) may be associated with decreased immune
response to the first dose of rotavirus vaccine, but this interference does not persist after subsequent doses
[3,96,105-108].

The United States Advisory Committee on Immunization Practices suggests that rotavirus vaccine can be
administered simultaneously or at any interval before or after other injectable or intranasal live vaccines
[68]. The American Academy of Pediatrics suggests that rotavirus vaccine can be administered
concurrently with other childhood vaccines [14]. In countries where OPV continues to be used, the
European Society for Pediatric Infectious Disease and the European Society for Paediatric
Gastroenterology, Hepatology, and Nutrition suggest that rotavirus vaccines and OPV not be administered
at the same visit [13].

EFFICACY/EFFECTIVENESS

Rotavirus gastroenteritis — The protection against rotavirus gastroenteritis provided by the two licensed
vaccines is similar to that observed following natural infection [67].
●Overall effectiveness – The effectiveness of rotavirus immunization is difficult to determine because
many children with rotavirus gastroenteritis do not seek medical attention; however, the number of stool
samples sent for rotavirus testing and the number of positive samples can serve as a marker of vaccine
effectiveness. Such laboratory surveillance indicates that during each rotavirus season since reintroduction
of rotavirus vaccine in the United States in 2006, rotavirus activity was delayed in onset and diminished
in magnitude compared with the 2000-2006 rotavirus seasons (figure 3) [109-111]. A pattern of alternating
reduced and markedly reduced seasonality has occurred from 2007-2008 through the 2013-2014 rotavirus
seasons.

In a systematic review of 57 observational studies from 27 countries, rotavirus immunization reduced


rotavirus-related hospitalization and emergency department visits among children <5 years by 67 percent
[82].

●Efficacy and effectiveness of RV5 – In randomized trials and meta-analyses, pentavalent human-bovine
rotavirus reassortant vaccine (RV5) was efficacious in preventing rotavirus gastroenteritis, rotavirus
gastroenteritis hospitalization, and rotavirus gastroenteritis-associated health care utilization in infants
[33,34,112,113]. In the largest trial comparing RV5 with placebo (the Rotavirus Efficacy and Safety
Trial), 68,038 healthy infants from 11 countries were randomly assigned to receive three doses of vaccine
or placebo at 4- to 10-week intervals [33]. The following results were reported:

•In the first rotavirus season after immunization, efficacy against severe G1-G4 rotavirus gastroenteritis
(defined by an established clinical score based upon intensity and duration of symptoms [9]) was 98
percent (95% CI 88-100 percent) and against G1-G4 rotavirus gastroenteritis of any severity was 74
percent (95% CI 67-80 percent).

•RV5 reduced hospitalization for G1-G4 rotavirus gastroenteritis by 96 percent (95% CI 90-98 percent)
and for any gastroenteritis by 59 percent (95% CI 52-65 percent).

•RV5 reduced emergency department visits related to G1-G4 rotavirus gastroenteritis by 94 percent (95%
CI 89-96 percent) and clinic visits by 86 percent (95% CI 74-93 percent).

•In the second rotavirus season after immunization, the efficacy against severe rotavirus gastroenteritis
was 88 percent (95% CI 49-99 percent) and against rotavirus gastroenteritis of any severity was 63 percent
(95% CI 44-75 percent).

•In an extension study of 20,736 original participants followed for up to 3.1 years after the last dose of
RV5, the efficacy against hospitalization and emergency department visits related to rotavirus
gastroenteritis of any serotype was 94 percent (95% CI 91-96 percent) [114].

In a systematic review of rotavirus vaccine effectiveness from 2006 to 2016, the median effectiveness of
RV5 in preventing rotavirus hospitalizations, emergency department visits, and outpatient visits was 90
percent (range 63 to 100 percent) in 20 studies from countries with low child mortality and 45 percent
(range 43 to 92 percent) in seven studies from countries with high child mortality [83]. RV5 protection
appears to be sustained through the first four years of life [58,115-117]. (See 'Resource-poor countries
with high child mortality' above.)
●Efficacy and effectiveness of RV1 – In randomized trials and meta-analyses, attenuated human
rotavirus vaccine (RV1) is efficacious in preventing rotavirus gastroenteritis, rotavirus gastroenteritis
hospitalizations, and rotavirus-gastroenteritis-associated health care utilization in infants
[34,35,84,113,118-123]. In the largest trial comparing RV1 with placebo, 63,225 healthy infants from 11
countries in Latin America and Finland were randomly assigned to receive RV1 or placebo at two and
four months of age [35]. Episodes of severe gastroenteritis were identified by active surveillance of 20,169
infants from two weeks after the second dose until one year of age. Severe gastroenteritis was defined by
the passage of ≥3 loose or watery stools within a 24-hour period, with or without vomiting, that required
overnight hospitalization or rehydration therapy in a medical facility. The following results were noted:

•The efficacy against severe rotavirus gastroenteritis was 85 percent (95% CI 72-92 percent) and against
severe gastroenteritis from any cause was 40 percent (95% CI 28-50 percent).

•The efficacy against gastroenteritis caused by G1P[8] strains was 92 percent (95% CI 74-98 percent) and
against strains sharing only the P[8] antigen (G3P[8], G4P[8], G9P[8]) was 87 percent.

•The efficacy against hospitalization for rotavirus gastroenteritis was 85 percent (95% CI 70-94 percent)
and against hospitalization for gastroenteritis from any cause was 42 percent (95% CI 29-53 percent).

The efficacy of RV1 during the first two years of life was assessed in a subgroup of patients and found to
be similar to that during the first year [123].

In a systematic review of rotavirus vaccine effectiveness from 2006 to 2016, the median effectiveness of
RV1 in preventing rotavirus hospitalizations, emergency department visits, and outpatient visits was 84
percent (range 19 to 97 percent) in 13 studies from countries with low child mortality, 75 percent (range
-2 to 94 percent) in eight studies from countries with medium child mortality, and 57 percent (range 18 to
69 percent) in nine studies from countries with high child mortality [83]. Protection appears to be sustained
through the first four years of life [58,83,116,117,124,125]. (See 'Resource-poor countries with high child
mortality' above.)

RV1 also appears to provide protection against G2P[4], which shares neither the G1 nor the P[8] antigen
[124].

●Incomplete immunization – A systematic review of postlicensure studies (2006 to 2016) found that
incomplete immunization with RV5 or RV1 was effective in preventing rotavirus health care utilization
but less effective than complete immunization [83].

Reduced risk of seizures — Rotavirus vaccine appears to reduce the risk of seizures [126-130]. In a cohort
of >1.7 million commercially insured children in the United States, complete rotavirus vaccination was
associated with a decreased risk of hospitalization for seizure before age five years compared with no
vaccination (adjusted hazard ratio 0.76, 95% CI 0.67-0.87) [130]. The differential effects of febrile versus
afebrile seizures could not be determined because of rarity of seizure hospitalization (estimated five-year
risk of 0.35 percent).

Community ("herd") immunity — Rotavirus immunization in infants is associated with reduced rotavirus
morbidity among unvaccinated neonates and young infants too young for vaccination, older children, and
adults (ie, indirect protection or community ["herd"] immunity) [131-141]. Seventy percent vaccine
uptake by 2010 significantly altered natural rotavirus disease peaks in most of the United States [109,132].
Even partial (approximately 50 percent) uptake of RV5 under a recommendation for universal
immunization of infants with rotavirus vaccine was associated with reduced rotavirus disease in
unvaccinated older children and adults [133-137]. As an example, compared with 2006, rates of
hospitalization for rotavirus infection in 2008 were reduced among children younger than three years,
whether or not they were vaccinated [134]. An 87 percent reduction occurred in the 6- to 11-month age
group (with vaccine coverage 77 percent), a 96 percent reduction occurred in the 12- to 23-month age
group (vaccine coverage 46 percent), and a 92 percent reduction occurred in the 24- to 35-month age
group (vaccine coverage 1 percent). Similar results were observed at several and widely dispersed sites in
North America [132-134,137,138]. Protection in older nonvaccinated children indicating community
immunity has also been reported in England [142].

ADVERSE EVENTS AND SAFETY

Overview — Rotavirus immunization is safe. In prelicensure studies, the rates of death (<0.1 percent) and
serious adverse events (approximately 2.5 percent) were similar among vaccine and placebo recipients
[33,35]. Vaccine and placebo recipients also reported similar rates of solicited events, including of fever
(approximately 42 percent), vomiting (approximately 13 percent), and diarrhea (approximately 19
percent), all of which were mild [33].

Intussusception

Risk with RV5 and RV1 — Intussusception is a rare potential adverse effect of oral rotavirus vaccination,
estimated to occur in approximately 1 in 20,000 to 1 in 100,000 vaccine recipients in high- and middle-
income countries [143-149]. A history of intussusception is a contraindication to rotavirus vaccination
[37,150], yet for infants without a history of intussusception, the risk of intussusception after rotavirus
vaccination is much lower than the risk of severe rotavirus gastroenteritis in children who do not receive
rotavirus vaccine [151-155].

Parents should contact their child's health care provider if the child develops signs of intussusception (ie,
stomach pain, vomiting, diarrhea, blood in the stool, or change in bowel habits) any time after vaccination,
but especially within the first 14 days after a dose was given [156]. (See "Intussusception in children",
section on 'Clinical manifestations'.)

Prelicensure studies of pentavalent human-bovine rotavirus reassortant vaccine (RV5) and attenuated
human rotavirus vaccine (RV1) found no increased risk of intussusception among vaccine recipients
compared with placebo recipients [33,35]; however, postlicensure studies suggest a rare association
between RV5 and RV1 vaccination and intussusception within 21 days of the first dose in high- and middle
income countries [143-149,157,158]. In active surveillance, the risk of intussusception following RV1
administration was not increased in lower-income sub-Saharan African countries [159].

Despite the small risk of intussusception, the absolute number of estimated rotavirus hospitalizations
prevented by rotavirus vaccines far exceeds that of cases of intussusception associated with rotavirus
vaccine (eg, 65,000 hospitalizations prevented and 40 to 120 cases of intussusception per year in the
United States) [151]. The Centers for Disease Control and Prevention and World Health Organization
Global Advisory Committee on Vaccine Safety continue to recommend universal rotavirus vaccine for
infants [160,161].
Risk with RRV-TV — In 1999, just over a year after human-rhesus rotavirus reassortant vaccine (RRV-
TV, RotaShield) was licensed, it was withdrawn from the market because of a strong epidemiologic link
to intussusception [29-32]. The increased risk was estimated to be approximately 22-fold over the
background risk within five to seven days of vaccination and overall approximately one excess case for
every 10,000 to 12,000 infants vaccinated [31,162].

The mechanism of this association is unclear. One hypothesis is that vaccination triggered intussusception
in infants who were likely to develop intussusception with any enteric infection, based upon the
observation that rates of intussusception were actually lower among vaccine recipients than nonvaccinees
in the period 4 to 12 weeks after vaccination [32]. Thus, RRV-TV may have caused intussusception in
infants who otherwise would not have experienced intussusception, but it also may have protected against
natural rotavirus infection-induced intussusception in others.

Rotavirus strain differences appear to make a difference in intussusception risk, as demonstrated by the
reduced intussusception risk after RV5 or RV1 compared with RRV, and supported by results in a mouse
model [163].

Kawasaki disease — Cases of Kawasaki disease were reported during clinical trials of rotavirus vaccines
and in postmarketing surveillance [150,164,165]; however, the number of cases of Kawasaki disease
reported after administration of rotavirus vaccine did not exceed the number of cases that would be
expected to occur in infants who did not receive rotavirus vaccine, and there is no known cause-and-effect
relationship between rotavirus vaccine and Kawasaki disease [150,165,166].

Parents should contact their child's health care provider if the child develops signs of Kawasaki disease
(eg, fever, conjunctivitis, erythema of the lips and oral mucosa, rash, swelling of the hands and feet,
cervical lymphadenopathy), whether or not the child recently received rotavirus vaccine. (See "Kawasaki
disease: Clinical features and diagnosis".)

Shedding and transmission of vaccine virus — Rotavirus shedding in the stool peaks within approximately
seven days of administration and is most common after the first dose [150,167-170]. Viral shedding may
be prolonged in infants with immunodeficiency [40,171].

Transmission of vaccine virus resulting in symptomatic gastroenteritis is known from three case reports
[172-174]; separately, zero of 100 participants in a placebo-controlled twin study experienced
symptomatic transmission of RV1 [175].

To minimize the risk of vaccine-derived rotavirus infections transmitted by the fecal-oral route,
individuals who care for infants should wash their hands after changing a diaper. Particular care should
be taken with this precaution for at least one week after the first dose. Highly immunocompromised
individuals should avoid handling diapers of infants who have received rotavirus vaccine for at least four
weeks after vaccination [38]. Highly immunocompromised individuals include (but are not limited to)
those with combined primary immunodeficiency; receiving cancer chemotherapy; within two months of
solid organ transplant; with HIV infection and CD4 count <200 cells/microL (adults and adolescents) or
CD4 percentage <15 percent (infants and children); receiving daily glucocorticoid therapy for ≥14 days
at a dose equivalent to prednisone ≥20 mg/day or >2 mg/kg per day, if they weigh <10 kg.
In the phase 3 prelicensure trial of RV5, 9 percent of subjects who were evaluated had fecal shedding of
vaccine virus four to six days after the first dose [167,169]. None and 0.3 percent of recipients shed vaccine
virus four to six days after the second and third dose, respectively. In studies of RV1, 50 to 80 percent of
infants shed vaccine virus approximately one week and 24 percent at approximately one month after the
first dose [176]. After the second dose, between 4 and 18 percent of recipients shed virus at one week and
1.2 percent at approximately one month.

Transmission of vaccine virus has not been well studied; it appears to occur more frequently among
recipients of RV1 than RV5 [168]; however, it rarely results in symptoms. In a randomized trial, in which
one twin in each of 100 twin pairs received two doses of RV1 and the other twin received placebo,
transmission of vaccine virus occurred in 15 of 80 evaluable cases (18.8 percent) but was not associated
with symptomatic gastroenteritis [175]. In an observational study in Malawi, RV1 fecal shedding was
detected in 68 percent of 60 vaccinated infants but only 1.4 percent of 147 household contacts, indicating
that horizontal transmission of vaccine virus is also uncommon [177]. Asymptomatic transmission may
contribute to community ("herd") immunity [168]. (See 'Community ("herd") immunity' above.)

Serotype selection — RV5 was first introduced into a national vaccine program in 2006. After
introduction of RV5 (and subsequently of RV1), surveillance of rotavirus genotypes and rotavirus disease
prevalence in countries with and without rotavirus vaccine programs has not demonstrated sustained
selection of one or more serotypes due to vaccine selective pressure [178], although transient increases of
a given serotype and decreased incidence of simultaneous infections with multiple rotavirus types (ie,
"mixed infections") have been documented [76,179-181]. Continued surveillance is necessary to
understand the epidemiologic significance of potential genetic/antigenic modifications in novel circulating
strains for which licensed rotavirus vaccines may not be effective [75,76,181-187]. (See 'Microbiology'
above.)

Reporting adverse events — In the United States, any clinically significant or unexpected adverse events
that occur after administration of rotavirus vaccine (including intussusception and Kawasaki disease)
should be reported to the Vaccine Adverse Event Reporting System (telephone number 1-800-822-7967)
[1]. (See "Standard immunizations for children and adolescents: Overview", section on 'Reporting adverse
events'.)

Porcine circovirus contamination — In 2010, an academic group using a novel technique found
components of porcine circovirus (PCV1) in RV1 and the US Food and Drug Administration (FDA)
temporarily suspended use of RV1 [188]. Additional review by the FDA and vaccine manufacturers
determined that PCV1 components were present from the early stages of RV1 development, including
during the prelicensure clinical trials, and that RV5 contained components of PCV1 and PCV2 [188,189].

Given that PCV1 and PCV2 are not known to cause illness in humans, and the absence of any evidence
that millions of rotavirus vaccine recipients suffered adverse effects related to PCV, the FDA
recommended resumption of use of RV1 and continued use of RV5 [189,190]. In a subsequent
observational study, PCV1 did not appear to replicate in RV1 recipients [191].

The known benefits of the oral vaccination outweigh the theoretic risk related to PCV1 or PCV2 [192].

RESOURCES — Resources related to immunization in infants include:


●The American Academy of Pediatrics

●The Centers for Disease Control and Prevention

●The Immunization Action Coalition

●The Vaccine Information Statement for Rotavirus vaccine

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected
countries and regions around the world are provided separately. (See "Society guideline links:
Immunizations in children and adolescents".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The
Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the
5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a
given condition. These articles are best for patients who want a general overview and who prefer short,
easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and
more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients
who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or email
these topics to your patients. (You can also locate patient education articles on a variety of subjects by
searching on "patient education" and the keyword[s] of interest.)

●Basics topic (see "Patient education: Rotavirus infection (The Basics)")

●Beyond the Basics topic (see "Patient education: Vaccines for infants and children age 0 to 6 years
(Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

●Rotavirus is the most common cause of severe gastroenteritis in infants and children around the world.
(See 'Introduction' above.)

●Two oral vaccines are available for the prevention of rotavirus disease: pentavalent human-bovine
reassortant rotavirus vaccine (RV5, PRV, RotaTeq) and attenuated human rotavirus vaccine (RV1, HRV,
Rotarix) (table 1 and figure 2). The vaccines have similar efficacy and safety, and neither vaccine is
preferred over the other. (See 'Rotavirus vaccines' above.)

●We recommend universal immunization of infants against rotavirus (Grade 1A). Rotavirus vaccines are
highly effective in preventing rotavirus gastroenteritis and rotavirus gastroenteritis-associated
hospitalization and health care utilization. (See 'Indications' above and 'Efficacy/effectiveness' above.)

●Contraindications to rotavirus vaccine include allergy to any of the vaccine ingredients, severe allergic
reaction (anaphylaxis) to a previous dose, severe combined immunodeficiency (SCID), certain other
primary and secondary immunodeficiencies, and history of intussusception. RV1 is contraindicated in
infants with a history of severe allergic reaction to latex, but RV5 may be administered to such infants.
(See 'Contraindications' above.)

●Conditions that are precautions for administration of rotavirus vaccine include immunodeficiency other
than SCID, acute moderate or severe illness, certain pre-existing or acquired gastrointestinal conditions
(eg, congenital malabsorption syndromes, Hirschsprung disease, short-bowel syndrome, previous bowel
surgery), and spina bifida or bladder exstrophy. (See 'Precautions' above.)

●The recommended dose and schedule for RV5 and RV1 differ (table 1). Whenever possible, the vaccine
series should be completed with the same product; however, vaccination should not be deferred if the
product used for previous doses is not known. (See 'Schedule' above.)

●Intussusception is a rare potential adverse effect of oral rotavirus vaccination in some settings; however,
the risk of intussusception after rotavirus vaccination is much lower than the risk of severe rotavirus
gastroenteritis in children who do not receive rotavirus vaccine. (See 'Adverse events and safety' above.)

●Continued surveillance of rotavirus genotypes and rotavirus disease prevalence in countries with and
without rotavirus vaccine programs is necessary to understand the epidemiologic significance of potential
genetic/antigenic modifications in novel circulating strains for which licensed rotavirus vaccines may not
be effective. (See 'Serotype selection' above.)

Use of UpToDate is subject to the Subscription and License Agreement.

REFERENCES
1. Cortese MM, Parashar UD, Centers for Disease Control and Prevention (CDC). Prevention of
rotavirus gastroenteritis among infants and children: recommendations of the Advisory Committee
on Immunization Practices (ACIP). MMWR Recomm Rep 2009; 58:1.
2. Parashar UD, Hummelman EG, Bresee JS, et al. Global illness and deaths caused by rotavirus
disease in children. Emerg Infect Dis 2003; 9:565.
3. World Health Organization. Rotavirus vaccines. WHO position paper - January 2013.
http://www.who.int.pbidi.unam.mx:8080/wer/2013/wer8805.pdf (Accessed on February 19,
2013).
4. International Vaccine Access Center. Available at: http://view-hub.org.
5. Cunliffe NA, Bresee JS, Hart CA. Rotavirus vaccines: development, current issues and future
prospects. J Infect 2002; 45:1.
6. Conner ME, Matson DO, Estes MK. Rotavirus vaccines and vaccination potential. Curr Top
Microbiol Immunol 1994; 185:285.
7. Vesikari T. Rotavirus vaccines: development and use for the prevention of diarrhoeal disease. Ann
Med 1999; 31:79.
8. Glass RI, Parashar UD. The promise of new rotavirus vaccines. N Engl J Med 2006; 354:75.
9. Clark HF, Bernstein DI, Dennehy PH, et al. Safety, efficacy, and immunogenicity of a live,
quadrivalent human-bovine reassortant rotavirus vaccine in healthy infants. J Pediatr 2004;
144:184.
10. Bhandari N, Rongsen-Chandola T, Bavdekar A, et al. Efficacy of a monovalent human-bovine
(116E) rotavirus vaccine in Indian infants: a randomised, double-blind, placebo-controlled trial.
Lancet 2014; 383:2136.
11. Changotra H, Vij A. Rotavirus virus-like particles (RV-VLPs) vaccines: An update. Rev Med
Virol 2017; 27.
12. Groome MJ, Koen A, Fix A, et al. Safety and immunogenicity of a parenteral P2-VP8-P[8] subunit
rotavirus vaccine in toddlers and infants in South Africa: a randomised, double-blind, placebo-
controlled trial. Lancet Infect Dis 2017; 17:843.
13. Vesikari T, Van Damme P, Giaquinto C, et al. European Society for Paediatric Infectious Diseases
consensus recommendations for rotavirus vaccination in Europe: update 2014. Pediatr Infect Dis
J 2015; 34:635.
14. American Academy of Pediatrics. Rotavirus infections. In: Red Book: 2018 Report of the
Committee on Infectious Diseases, 31st ed, Kimberlin DW, Brady MT, Jackson MA, Long SS
(Eds), American Academy of Pediatrics, Itasca, IL 2018. p.700.
15. RotaTeq (Rotavirus vaccine, live, oral, pentavalent). United States Prescribing Information.
Revised February 2017, US Food & Drug Administration. Available online at:
https://www.fda.gov/BiologicsBloodVaccines/Vaccines/ApprovedProducts/ucm094063.htm
(Accessed on March 07, 2017).
16. De Vos B, Han HH, Bouckenooghe A, et al. Live attenuated human rotavirus vaccine, RIX4414,
provides clinical protection in infants against rotavirus strains with and without shared G and P
genotypes: integrated analysis of randomized controlled trials. Pediatr Infect Dis J 2009; 28:261.
17. Santosham M, Steele D. Rotavirus Vaccines - A New Hope. N Engl J Med 2017; 376:1170.
18. Mi K, Ou X, Guo L, et al. Comparative analysis of the immunogenicity of monovalent and
multivalent rotavirus immunogens. PLoS One 2017; 12:e0172156.
19. Kirkwood CD, Steele AD. Rotavirus Vaccines in China: Improvement Still Required. JAMA Netw
Open 2018; 1:e181579.
20. Zhen SS, Li Y, Wang SM, et al. Effectiveness of the live attenuated rotavirus vaccine produced by
a domestic manufacturer in China studied using a population-based case-control design. Emerg
Microbes Infect 2015; 4:e64.
21. Fu C, Dong Z, Shen J, et al. Rotavirus gastroenteritis infection among children vaccinated and
unvaccinated with rotavirus vaccine in southern China: A population-based assessment. JAMA
Network Open 2018; 1:e181382.
22. Desselberger U, Huppertz HI. Immune responses to rotavirus infection and vaccination and
associated correlates of protection. J Infect Dis 2011; 203:188.
23. Isanaka S, Guindo O, Langendorf C, et al. Efficacy of a Low-Cost, Heat-Stable Oral Rotavirus
Vaccine in Niger. N Engl J Med 2017; 376:1121.
24. Loharikar A, Dumolard L, Chu S, et al. Status of New Vaccine Introduction - Worldwide,
September 2016. MMWR Morb Mortal Wkly Rep 2016; 65:1136.
25. Kulkarni PS, Desai S, Tewari T, et al. A randomized Phase III clinical trial to assess the efficacy
of a bovine-human reassortant pentavalent rotavirus vaccine in Indian infants. Vaccine 2017;
35:6228.
26. Bines JE, At Thobari J, Satria CD, et al. Human Neonatal Rotavirus Vaccine (RV3-BB) to Target
Rotavirus from Birth. N Engl J Med 2018; 378:719.
27. Bines JE, Danchin M, Jackson P, et al. Safety and immunogenicity of RV3-BB human neonatal
rotavirus vaccine administered at birth or in infancy: a randomised, double-blind, placebo-
controlled trial. Lancet Infect Dis 2015; 15:1389.
28. Recommended childhood immunization schedule-United States, January-December 1999.
American Academy of Pediatrics Committee on Infectious Diseases. Pediatrics 1999; 103:182.
29. Peter G, Myers MG, National Vaccine Advisory Committee, National Vaccine Program Office.
Intussusception, rotavirus, and oral vaccines: summary of a workshop. Pediatrics 2002; 110:e67.
30. Centers for Disease Control and Prevention (CDC). Intussusception among recipients of rotavirus
vaccine--United States, 1998-1999. MMWR Morb Mortal Wkly Rep 1999; 48:577.
31. Murphy TV, Gargiullo PM, Massoudi MS, et al. Intussusception among infants given an oral
rotavirus vaccine. N Engl J Med 2001; 344:564.
32. Murphy BR, Morens DM, Simonsen L, et al. Reappraisal of the association of intussusception
with the licensed live rotavirus vaccine challenges initial conclusions. J Infect Dis 2003; 187:1301.
33. Vesikari T, Matson DO, Dennehy P, et al. Safety and efficacy of a pentavalent human-bovine
(WC3) reassortant rotavirus vaccine. N Engl J Med 2006; 354:23.
34. Soares-Weiser K, Maclehose H, Bergman H, et al. Vaccines for preventing rotavirus diarrhoea:
vaccines in use. Cochrane Database Syst Rev 2012; 11:CD008521.
35. Ruiz-Palacios GM, Pérez-Schael I, Velázquez FR, et al. Safety and efficacy of an attenuated
vaccine against severe rotavirus gastroenteritis. N Engl J Med 2006; 354:11.
36. Centers for Disease Control and Prevention (CDC). Addition of severe combined
immunodeficiency as a contraindication for administration of rotavirus vaccine. MMWR Morb
Mortal Wkly Rep 2010; 59:687.
37. Centers for Disease Control and Prevention (CDC). Addition of history of intussusception as a
contraindication for rotavirus vaccination. MMWR Morb Mortal Wkly Rep 2011; 60:1427.
38. Rubin LG, Levin MJ, Ljungman P, et al. 2013 IDSA clinical practice guideline for vaccination of
the immunocompromised host. Clin Infect Dis 2014; 58:e44.
39. Patel NC, Hertel PM, Estes MK, et al. Vaccine-acquired rotavirus in infants with severe combined
immunodeficiency. N Engl J Med 2010; 362:314.
40. Werther RL, Crawford NW, Boniface K, et al. Rotavirus vaccine induced diarrhea in a child with
severe combined immune deficiency. J Allergy Clin Immunol 2009; 124:600.
41. Bakare N, Menschik D, Tiernan R, et al. Severe combined immunodeficiency (SCID) and rotavirus
vaccination: reports to the Vaccine Adverse Events Reporting System (VAERS). Vaccine 2010;
28:6609.
42. Kaplon J, Cros G, Ambert-Balay K, et al. Rotavirus vaccine virus shedding, viremia and clearance
in infants with severe combined immune deficiency. Pediatr Infect Dis J 2015; 34:326.
43. Morillo-Gutierrez B, Worth A, Valappil M, et al. Chronic Infection with Rotavirus Vaccine Strains
in UK Children with Severe Combined Immunodeficiency. Pediatr Infect Dis J 2015; 34:1040.
44. MedWatch. The FDA safety information and adverse event reporting program. Drug safety
labeling changes. Rotarix (rotavirus vaccine, live, oral) oral suspension.
http://www.fda.gov/Safety/MedWatch/SafetyInformation/ucm230397.htm (Accessed on April
18, 2011).
45. Nguyen GC, Seow CH, Maxwell C, et al. The Toronto Consensus Statements for the Management
of Inflammatory Bowel Disease in Pregnancy. Gastroenterology 2016; 150:734.
46. Mahadevan U, Wolf DC, Dubinsky M, et al. Placental transfer of anti-tumor necrosis factor agents
in pregnant patients with inflammatory bowel disease. Clin Gastroenterol Hepatol 2013; 11:286.
47. Julsgaard M, Christensen LA, Gibson PR, et al. Concentrations of Adalimumab and Infliximab in
Mothers and Newborns, and Effects on Infection. Gastroenterology 2016; 151:110.
48. Goveia MG, DiNubile MJ, Dallas MJ, et al. Efficacy of pentavalent human-bovine (WC3)
reassortant rotavirus vaccine based on breastfeeding frequency. Pediatr Infect Dis J 2008; 27:656.
49. Vesikari T, Prymula R, Schuster V, et al. Efficacy and immunogenicity of live-attenuated human
rotavirus vaccine in breast-fed and formula-fed European infants. Pediatr Infect Dis J 2012;
31:509.
50. Groome MJ, Madhi SA. Five-year cohort study on the burden of hospitalisation for acute
diarrhoeal disease in African HIV-infected and HIV-uninfected children: potential benefits of
rotavirus vaccine. Vaccine 2012; 30 Suppl 1:A173.
51. Steele AD, Madhi SA, Louw CE, et al. Safety, Reactogenicity, and Immunogenicity of Human
Rotavirus Vaccine RIX4414 in Human Immunodeficiency Virus-positive Infants in South Africa.
Pediatr Infect Dis J 2011; 30:125.
52. Levin MJ, Lindsey JC, Kaplan SS, et al. Safety and immunogenicity of a live attenuated
pentavalent rotavirus vaccine in HIV-exposed infants with or without HIV infection in Africa.
AIDS 2017; 31:49.
53. Department of Health and Human Services. Panel on Opportunistic Infections in HIV-Exposed
and HIV-Infected Children. Guidelines for the prevention and treatment of opportunistic infections
in HIV-exposed and HIV-infected children. Available at:
aidsinfo.nih.gov/contentfiles/lvguidelines/oi_guidelines_pediatrics.pdf (Accessed on November
20, 2013).
54. Javid PJ, Sanchez SE, Jacob S, et al. The Safety and Immunogenicity of Rotavirus Vaccination in
Infants With Intestinal Failure. J Pediatric Infect Dis Soc 2014; 3:57.
55. McGrath EJ, Thomas R, Duggan C, Asmar BI. Pentavalent rotavirus vaccine in infants with
surgical gastrointestinal disease. J Pediatr Gastroenterol Nutr 2014; 59:44.
56. Libster R, McNeal M, Walter EB, et al. Safety and Immunogenicity of Sequential Rotavirus
Vaccine Schedules. Pediatrics 2016; 137:e20152603.
57. Payne DC, Sulemana I, Parashar UD, New Vaccine Surveillance Network. Evaluation of
Effectiveness of Mixed Rotavirus Vaccine Course for Rotavirus Gastroenteritis. JAMA Pediatr
2016; 170:708.
58. Payne DC, Selvarangan R, Azimi PH, et al. Long-term Consistency in Rotavirus Vaccine
Protection: RV5 and RV1 Vaccine Effectiveness in US Children, 2012-2013. Clin Infect Dis 2015;
61:1792.
59. Goveia MG, Rodriguez ZM, Dallas MJ, et al. Safety and efficacy of the pentavalent human-bovine
(WC3) reassortant rotavirus vaccine in healthy premature infants. Pediatr Infect Dis J 2007;
26:1099.
60. Omenaca F, Sarlangue J, Szenborn L, et al. Safety, reactogenicity and immunogenicity of the
human rotavirus vaccine in preterm European Infants: a randomized phase IIIb study. Pediatr
Infect Dis J 2012; 31:487.
61. Dahl RM, Curns AT, Tate JE, Parashar UD. Effect of Rotavirus Vaccination on Acute Diarrheal
Hospitalizations Among Low and Very Low Birth Weight US Infants, 2001-2015. Pediatr Infect
Dis J 2018; 37:817.
62. Rotavirus. In Australian Immunisation Handbook, 10th edition. Available at:
http://www.immunise.health.gov.au/internet/immunise/publishing.nsf/Content/Handbook10-
home~handbook10part4~handbook10-4-17#4.17.6 (Accessed on January 08, 2018).
63. Monk HM, Motsney AJ, Wade KC. Safety of rotavirus vaccine in the NICU. Pediatrics 2014;
133:e1555.
64. Thrall S, Doll MK, Nhan C, et al. Evaluation of pentavalent rotavirus vaccination in neonatal
intensive care units. Vaccine 2015; 33:5095.
65. Hofstetter AM, Lacombe K, Klein EJ, et al. Risk of Rotavirus Nosocomial Spread After Inpatient
Pentavalent Rotavirus Vaccination. Pediatrics 2018; 141.
66. Hiramatsu H, Suzuki R, Nagatani A, et al. Rotavirus Vaccination Can Be Performed Without Viral
Dissemination in the Neonatal Intensive Care Unit. J Infect Dis 2018; 217:589.
67. Velázquez FR, Matson DO, Calva JJ, et al. Rotavirus infection in infants as protection against
subsequent infections. N Engl J Med 1996; 335:1022.
68. Kroger AT, Duchin J, Vázquez M. General best practice guidelines for immunization. Best
practices guidance of the Advisory Committee on Immunization Practices (ACIP). Available at:
https://www.cdc.gov/vaccines/hcp/acip-recs/general-recs/index.html (Accessed on April 27,
2017).
69. Abeid KA, Jani B, Cortese MM, et al. Monovalent Rotavirus Vaccine Effectiveness and Impact
on Rotavirus Hospitalizations in Zanzibar, Tanzania: Data From the First 3 Years After
Introduction. J Infect Dis 2017; 215:183.
70. Narváez J, Osorio MB, Castañeda-Orjuela C, et al. Is Colombia reaching the goals on infant
immunization coverage? A quantitative survey from 80 municipalities. Vaccine 2017; 35:1501.
71. Bennett A, Bar-Zeev N, Cunliffe NA. Measuring indirect effects of rotavirus vaccine in low
income countries. Vaccine 2016; 34:4351.
72. Madhi SA, Cunliffe NA, Steele D, et al. Effect of human rotavirus vaccine on severe diarrhea in
African infants. Malawi Med J 2016; 28:108.
73. Velázquez RF, Linhares AC, Muñoz S, et al. Efficacy, safety and effectiveness of licensed
rotavirus vaccines: a systematic review and meta-analysis for Latin America and the Caribbean.
BMC Pediatr 2017; 17:14.
74. Gruber JF, Hille DA, Liu GF, et al. Heterogeneity of Rotavirus Vaccine Efficacy Among Infants
in Developing Countries. Pediatr Infect Dis J 2017; 36:72.
75. Wylie KM, Stanley KM, TeKippe EM, et al. Resurgence of Rotavirus Genotype G12 in St. Louis
During the 2014-2015 Rotavirus Season. J Pediatric Infect Dis Soc 2017; 6:346.
76. Muhsen K, Kassem E, Rubenstein U, et al. Incidence of rotavirus gastroenteritis hospitalizations
and genotypes, before and five years after introducing universal immunization in Israel. Vaccine
2016; 34:5916.
77. Lo Vecchio A, Liguoro I, Dias JA, et al. Rotavirus immunization: Global coverage and local
barriers for implementation. Vaccine 2017; 35:1637.
78. Tissera MS, Cowley D, Bogdanovic-Sakran N, et al. Options for improving effectiveness of
rotavirus vaccines in developing countries. Hum Vaccin Immunother 2017; 13:921.
79. Al-Aidaroos AYA, Standaert B, Meszaros K, Shibl AM. Economic assessment of rotavirus
vaccination in Saudi Arabia. J Infect Public Health 2017; 10:564.
80. Hill HA, Elam-Evans LD, Yankey D, et al. Vaccination Coverage Among Children Aged 19-35
Months - United States, 2015. MMWR Morb Mortal Wkly Rep 2016; 65:1065.
81. Gosselin V, Petit G, Gagneur A, Généreux M. Trends in severe gastroenteritis among young
children according to socio-economic characteristics before and after implementation of a
rotavirus vaccination program in Quebec. Can J Public Health 2016; 107:e161.
82. Burnett E, Jonesteller CL, Tate JE, et al. Global Impact of Rotavirus Vaccination on Childhood
Hospitalizations and Mortality From Diarrhea. J Infect Dis 2017; 215:1666.
83. Jonesteller CL, Burnett E, Yen C, et al. Effectiveness of Rotavirus Vaccination: A Systematic
Review of the First Decade of Global Postlicensure Data, 2006-2016. Clin Infect Dis 2017; 65:840.
84. Mujuru HA, Burnett E, Nathoo KJ, et al. Monovalent rotavirus vaccine effectiveness against
rotavirus hospitalizations among children in Zimbabwe. Clin Infect Dis 2018.
85. Loganathan T, Jit M, Hutubessy R, et al. Rotavirus vaccines contribute towards universal health
coverage in a mixed public-private healthcare system. Trop Med Int Health 2016; 21:1458.
86. Santos VS, Marques DP, Martins-Filho PR, et al. Effectiveness of rotavirus vaccines against
rotavirus infection and hospitalization in Latin America: systematic review and meta-analysis.
Infect Dis Poverty 2016; 5:83.
87. Holubar M, Stavroulakis MC, Maldonado Y, et al. Impact of vaccine herd-protection effects in
cost-effectiveness analyses of childhood vaccinations. A quantitative comparative analysis. PLoS
One 2017; 12:e0172414.
88. Markkula J, Hemming-Harlo M, Salminen MT, et al. Rotavirus epidemiology 5-6 years after
universal rotavirus vaccination: persistent rotavirus activity in older children and elderly. Infect
Dis (Lond) 2017; 49:388.
89. da Silva MF, Fumian TM, de Assis RM, et al. VP7 and VP8* genetic characterization of group A
rotavirus genotype G12P[8]: Emergence and spreading in the Eastern Brazilian coast in 2014. J
Med Virol 2017; 89:64.
90. Asada K, Kamiya H, Suga S, et al. Rotavirus vaccine and health-care utilization for rotavirus
gastroenteritis in Tsu City, Japan. Western Pac Surveill Response J 2016; 7:28.
91. Rogawski ET, Platts-Mills JA, Colgate ER, et al. Quantifying the Impact of Natural Immunity on
Rotavirus Vaccine Efficacy Estimates: A Clinical Trial in Dhaka, Bangladesh (PROVIDE) and a
Simulation Study. J Infect Dis 2018; 217:861.
92. Mwila K, Chilengi R, Simuyandi M, et al. Contribution of Maternal Immunity to Decreased
Rotavirus Vaccine Performance in Low- and Middle-Income Countries. Clin Vaccine Immunol
2017; 24.
93. Bautista-Marquez A, Velasquez DE, Esparza-Aguilar M, et al. Breastfeeding linked to the
reduction of both rotavirus shedding and IgA levels after Rotarix® immunization in Mexican
infants. Vaccine 2016; 34:5284.
94. Becker-Dreps S, Vilchez S, Bucardo F, et al. The Association Between Fecal Biomarkers of
Environmental Enteropathy and Rotavirus Vaccine Response in Nicaraguan Infants. Pediatr Infect
Dis J 2017; 36:412.
95. Harris VC, Armah G, Fuentes S, et al. Significant Correlation Between the Infant Gut Microbiome
and Rotavirus Vaccine Response in Rural Ghana. J Infect Dis 2017; 215:34.
96. Ramani S, Mamani N, Villena R, et al. Rotavirus Serum IgA Immune Response in Children
Receiving Rotarix Coadministered With bOPV or IPV. Pediatr Infect Dis J 2016; 35:1137.
97. Armah GE, Cortese MM, Dennis FE, et al. Rotavirus Vaccine Take in Infants Is Associated With
Secretor Status. J Infect Dis 2019; 219:746.
98. Kazi AM, Cortese MM, Yu Y, et al. Secretor and Salivary ABO Blood Group Antigen Status
Predict Rotavirus Vaccine Take in Infants. J Infect Dis 2017; 215:786.
99. Hibbs BF, Miller ER, Shimabukuro T, Centers for Disease Control and Prevention (CDC). Notes
from the field: rotavirus vaccine administration errors--United States, 2006-2013. MMWR Morb
Mortal Wkly Rep 2014; 63:81.
100. Groome MJ, Moon SS, Velasquez D, et al. Effect of breastfeeding on immunogenicity of
oral live-attenuated human rotavirus vaccine: a randomized trial in HIV-uninfected infants in
Soweto, South Africa. Bull World Health Organ 2014; 92:238.
101. Rongsen-Chandola T, Strand TA, Goyal N, et al. Effect of withholding breastfeeding on
the immune response to a live oral rotavirus vaccine in North Indian infants. Vaccine 2014; 32
Suppl 1:A134.
102. Ali A, Kazi AM, Cortese MM, et al. Impact of withholding breastfeeding at the time of
vaccination on the immunogenicity of oral rotavirus vaccine--a randomized trial. PLoS One 2015;
10:e0127622.
103. Rodriguez ZM, Goveia MG, Stek JE, et al. Concomitant use of an oral live pentavalent
human-bovine reassortant rotavirus vaccine with licensed parenteral pediatric vaccines in the
United States. Pediatr Infect Dis J 2007; 26:221.
104. Ciarlet M, He S, Lai S, et al. Concomitant use of the 3-dose oral pentavalent rotavirus
vaccine with a 3-dose primary vaccination course of a diphtheria-tetanus-acellular pertussis-
hepatitis B-inactivated polio-Haemophilus influenzae type b vaccine: immunogenicity and
reactogenicity. Pediatr Infect Dis J 2009; 28:177.
105. Tregnaghi MW, Abate HJ, Valencia A, et al. Human rotavirus vaccine is highly efficacious
when coadministered with routine expanded program of immunization vaccines including oral
poliovirus vaccine in Latin America. Pediatr Infect Dis J 2011; 30:e103.
106. Ciarlet M, Sani-Grosso R, Yuan G, et al. Concomitant use of the oral pentavalent human-
bovine reassortant rotavirus vaccine and oral poliovirus vaccine. Pediatr Infect Dis J 2008; 27:874.
107. Steele AD, De Vos B, Tumbo J, et al. Co-administration study in South African infants of
a live-attenuated oral human rotavirus vaccine (RIX4414) and poliovirus vaccines. Vaccine 2010;
28:6542.
108. Zaman K, Sack DA, Yunus M, et al. Successful co-administration of a human rotavirus and
oral poliovirus vaccines in Bangladeshi infants in a 2-dose schedule at 12 and 16 weeks of age.
Vaccine 2009; 27:1333.
109. Aliabadi N, Tate JE, Haynes AK, et al. Sustained decrease in laboratory detection of
rotavirus after implementation of routine vaccination—United States, 2000-2014. MMWR Morb
Mortal Wkly Rep 2015; 64:337.
110. Leshem E, Tate JE, Steiner CA, et al. Acute gastroenteritis hospitalizations among US
children following implementation of the rotavirus vaccine. JAMA 2015; 313:2282.
111. Kaufman HW, Chen Z. Trends in Laboratory Rotavirus Detection: 2003 to 2014. Pediatrics
2016; 138.
112. Grant LR, Watt JP, Weatherholtz RC, et al. Efficacy of a pentavalent human-bovine
reassortant rotavirus vaccine against rotavirus gastroenteritis among American Indian children.
Pediatr Infect Dis J 2012; 31:184.
113. Lamberti LM, Ashraf S, Walker CL, Black RE. A Systematic Review of the Effect of
Rotavirus Vaccination on Diarrhea Outcomes Among Children Younger Than 5 Years. Pediatr
Infect Dis J 2016; 35:992.
114. Vesikari T, Karvonen A, Ferrante SA, et al. Sustained efficacy of the pentavalent rotavirus
vaccine, RV5, up to 3.1 years following the last dose of vaccine. Pediatr Infect Dis J 2010; 29:957.
115. Payne DC, Boom JA, Staat MA, et al. Effectiveness of pentavalent and monovalent
rotavirus vaccines in concurrent use among US children <5 years of age, 2009-2011. Clin Infect
Dis 2013; 57:13.
116. Leshem E, Moritz RE, Curns AT, et al. Rotavirus vaccines and health care utilization for
diarrhea in the United States (2007-2011). Pediatrics 2014; 134:15.
117. Getachew HB, Dahl RM, Lopman BA, Parashar UD. Rotavirus Vaccines and Health Care
Utilization for Diarrhea in US Children, 2001 to 2015. Pediatr Infect Dis J 2018; 37:943.
118. Vesikari T, Karvonen A, Puustinen L, et al. Efficacy of RIX4414 live attenuated human
rotavirus vaccine in Finnish infants. Pediatr Infect Dis J 2004; 23:937.
119. Salinas B, Pérez Schael I, Linhares AC, et al. Evaluation of safety, immunogenicity and
efficacy of an attenuated rotavirus vaccine, RIX4414: A randomized, placebo-controlled trial in
Latin American infants. Pediatr Infect Dis J 2005; 24:807.
120. De Vos B, Vesikari T, Linhares AC, et al. A rotavirus vaccine for prophylaxis of infants
against rotavirus gastroenteritis. Pediatr Infect Dis J 2004; 23:S179.
121. Ruiz-Palacios GM, Guerrero ML, Bautista-Márquez A, et al. Dose response and efficacy
of a live, attenuated human rotavirus vaccine in Mexican infants. Pediatrics 2007; 120:e253.
122. Vesikari T, Karvonen A, Prymula R, et al. Efficacy of human rotavirus vaccine against
rotavirus gastroenteritis during the first 2 years of life in European infants: randomised, double-
blind controlled study. Lancet 2007; 370:1757.
123. Linhares AC, Velázquez FR, Pérez-Schael I, et al. Efficacy and safety of an oral live
attenuated human rotavirus vaccine against rotavirus gastroenteritis during the first 2 years of life
in Latin American infants: a randomised, double-blind, placebo-controlled phase III study. Lancet
2008; 371:1181.
124. Cortese MM, Immergluck LC, Held M, et al. Effectiveness of monovalent and pentavalent
rotavirus vaccine. Pediatrics 2013; 132:e25.
125. Immergluck LC, Parker TC, Jain S, et al. Sustained Effectiveness of Monovalent and
Pentavalent Rotavirus Vaccines in Children. J Pediatr 2016; 172:116.
126. Payne DC, Baggs J, Zerr DM, et al. Protective association between rotavirus vaccination
and childhood seizures in the year following vaccination in US children. Clin Infect Dis 2014;
58:173.
127. Pardo-Seco J, Cebey-López M, Martinón-Torres N, et al. Impact of Rotavirus Vaccination
on Childhood Hospitalization for Seizures. Pediatr Infect Dis J 2015; 34:769.
128. Sheridan SL, Ware RS, Grimwood K, Lambert SB. Febrile Seizures in the Era of Rotavirus
Vaccine. J Pediatric Infect Dis Soc 2016; 5:206.
129. Pringle KD, Burke RM, Steiner CA, et al. Trends in Rate of Seizure-Associated
Hospitalizations Among Children <5 Years Old Before and After Rotavirus Vaccine Introduction
in the United Sates, 2000-2013. J Infect Dis 2018; 217:581.
130. Burke RM, Tate JE, Dahl RM, et al. Rotavirus Vaccination Is Associated With Reduced
Seizure Hospitalization Risk Among Commercially Insured US Children. Clin Infect Dis 2018;
67:1614.
131. Atchison CJ, Stowe J, Andrews N, et al. Rapid Declines in Age Group-Specific Rotavirus
Infection and Acute Gastroenteritis Among Vaccinated and Unvaccinated Individuals Within 1
Year of Rotavirus Vaccine Introduction in England and Wales. J Infect Dis 2016; 213:243.
132. Tate JE, Haynes A, Payne DC, et al. Trends in national rotavirus activity before and after
introduction of rotavirus vaccine into the national immunization program in the United States,
2000 to 2012. Pediatr Infect Dis J 2013; 32:741.
133. Lopman BA, Curns AT, Yen C, Parashar UD. Infant rotavirus vaccination may provide
indirect protection to older children and adults in the United States. J Infect Dis 2011; 204:980.
134. Payne DC, Staat MA, Edwards KM, et al. Direct and indirect effects of rotavirus
vaccination upon childhood hospitalizations in 3 US Counties, 2006-2009. Clin Infect Dis 2011;
53:245.
135. Dey A, Wang H, Menzies R, Macartney K. Changes in hospitalisations for acute
gastroenteritis in Australia after the national rotavirus vaccination program. Med J Aust 2012;
197:453.
136. Lopman BA, Payne DC, Tate JE, et al. Post-licensure experience with rotavirus vaccination
in high and middle income countries; 2006 to 2011. Curr Opin Virol 2012; 2:434.
137. Anderson EJ, Shippee DB, Weinrobe MH, et al. Indirect protection of adults from rotavirus
by pediatric rotavirus vaccination. Clin Infect Dis 2013; 56:755.
138. Gastañaduy PA, Curns AT, Parashar UD, Lopman BA. Gastroenteritis hospitalizations in
older children and adults in the United States before and after implementation of infant rotavirus
vaccination. JAMA 2013; 310:851.
139. Cortese MM, Dahl RM, Curns AT, Parashar UD. Protection against gastroenteritis in US
households with children who received rotavirus vaccine. J Infect Dis 2015; 211:558.
140. Mast TC, Wang FT, Su S, Seeger JD. Evidence of herd immunity and sustained impact of
rotavirus vaccination on the reduction of rotavirus-related medical encounters among infants from
2006 through 2011 in the United States. Pediatr Infect Dis J 2015; 34:615.
141. Prelog M, Gorth P, Zwazl I, et al. Universal Mass Vaccination Against Rotavirus: Indirect
Effects on Rotavirus Infections in Neonates and Unvaccinated Young Infants Not Eligible for
Vaccination. J Infect Dis 2016; 214:546.
142. Thomas SL, Walker JL, Fenty J, et al. Impact of the national rotavirus vaccination
programme on acute gastroenteritis in England and associated costs averted. Vaccine 2017;
35:680.
143. Patel MM, López-Collada VR, Bulhões MM, et al. Intussusception risk and health benefits
of rotavirus vaccination in Mexico and Brazil. N Engl J Med 2011; 364:2283.
144. Velázquez FR, Colindres RE, Grajales C, et al. Postmarketing surveillance of
intussusception following mass introduction of the attenuated human rotavirus vaccine in Mexico.
Pediatr Infect Dis J 2012; 31:736.
145. Haber P, Patel M, Pan Y, et al. Intussusception after rotavirus vaccines reported to US
VAERS, 2006-2012. Pediatrics 2013; 131:1042.
146. Carlin JB, Macartney KK, Lee KJ, et al. Intussusception risk and disease prevention
associated with rotavirus vaccines in Australia's National Immunization Program. Clin Infect Dis
2013; 57:1427.
147. Weintraub ES, Baggs J, Duffy J, et al. Risk of intussusception after monovalent rotavirus
vaccination. N Engl J Med 2014; 370:513.
148. Yih WK, Lieu TA, Kulldorff M, et al. Intussusception risk after rotavirus vaccination in
U.S. infants. N Engl J Med 2014; 370:503.
149. Leino T, Ollgren J, Strömberg N, Elonsalo U. Evaluation of the Intussusception Risk after
Pentavalent Rotavirus Vaccination in Finnish Infants. PLoS One 2016; 11:e0144812.
150. Rotarix (Rotavirus vaccine, live, oral) prescribing information, 2011.
http://us.gsk.com/products/assets/us_rotarix.pdf (Accessed on April 21, 2011).
151. Centers for Disease Control and Prevention. Vaccine Safety. Rotavirus.
http://www.cdc.gov/vaccinesafety/vaccines/rotavsb.html (Accessed on September 04, 2013).
152. Centers for Disease Control and Prevention. Rotarix® rotavirus vaccine: Rare side effect
possible. Questions and answers for parents and caregivers. http://www.cdc.gov/vaccines/vpd-
vac/rotavirus/Vac-label-parents.htm (Accessed on September 23, 2010).
153. Desai R, Cortese MM, Meltzer MI, et al. Potential intussusception risk versus benefits of
rotavirus vaccination in the United States. Pediatr Infect Dis J 2013; 32:1.
154. Glass RI, Parashar UD. Rotavirus vaccines--balancing intussusception risks and health
benefits. N Engl J Med 2014; 370:568.
155. Buttery JP, Standish J, Bines JE. Intussusception and rotavirus vaccines: consensus on
benefits outweighing recognized risk. Pediatr Infect Dis J 2014; 33:772.
156. U.S. Food and Drug Administration. Information on Rotarix--Labeling revision pertaining
to intussusception.
http://www.fda.gov/BiologicsBloodVaccines/Vaccines/ApprovedProducts/ucm226690.htm
(Accessed on September 23, 2010).
157. Shui IM, Baggs J, Patel M, et al. Risk of intussusception following administration of a
pentavalent rotavirus vaccine in US infants. JAMA 2012; 307:598.
158. Tate JE, Yen C, Steiner CA, et al. Intussusception Rates Before and After the Introduction
of Rotavirus Vaccine. Pediatrics 2016; 138.
159. Tate JE, Mwenda JM, Armah G, et al. Evaluation of Intussusception after Monovalent
Rotavirus Vaccination in Africa. N Engl J Med 2018; 378:1521.
160. Robinson CL, Bernstein H, Romero JR, Szilagyi P. Advisory Committee on Immunization
Practices Recommended Immunization Schedule for Children and Adolescents Aged 18 Years or
Younger - United States, 2019. MMWR Morb Mortal Wkly Rep 2019; 68:112.
161. Global Advisory Committee on Vaccine Safety, 11–12 December 2013. Wkly Epidemiol
Rec 2014; 89:53.
162. DeStefano F, Vaccine Safety Datalink Research Group. The Vaccine Safety Datalink
project. Pharmacoepidemiol Drug Saf 2001; 10:403.
163. Warfield KL, Blutt SE, Crawford SE, et al. Rotavirus infection enhances
lipopolysaccharide-induced intussusception in a mouse model. J Virol 2006; 80:12377.
164. RotaTeq® [Rotavirus Vaccine, Live, Oral, Pentavalent] product label
http://www.merck.com/product/usa/pi_circulars/r/rotateq/rotateq_pi.pdf (Accessed on August 02,
2011).
165. Information pertaining to labeling revision for RotaTeq
http://www.fda.gov/BiologicsBloodVaccines/Vaccines/ApprovedProducts/ucm142393.htm
(Accessed on August 02, 2011).
166. Loughlin J, Mast TC, Doherty MC, et al. Postmarketing evaluation of the short-term safety
of the pentavalent rotavirus vaccine. Pediatr Infect Dis J 2012; 31:292.
167. Dennehy PH, Goveia MG, Dallas MJ, Heaton PM. The integrated phase III safety profile
of the pentavalent human-bovine (WC3) reassortant rotavirus vaccine. Int J Infect Dis 2007; 11
Suppl 2:S36.
168. Anderson EJ. Rotavirus vaccines: viral shedding and risk of transmission. Lancet Infect
Dis 2008; 8:642.
169. Matson DO, Vesikari T, Dennehy P, et al. Analysis by rotavirus gene 6 reverse
transcriptase-polymerase chain reaction assay of rotavirus-positive gastroenteritis cases observed
during the vaccination phase of the Rotavirus Efficacy and Safety Trial (REST). Hum Vaccin
Immunother 2014; 10:2267.
170. Ye S, Whiley DM, Ware RS, et al. Multivalent Rotavirus Vaccine and Wild-type Rotavirus
Strain Shedding in Australian Infants: A Birth Cohort Study. Clin Infect Dis 2018; 66:1411.
171. Uygungil B, Bleesing JJ, Risma KA, et al. Persistent rotavirus vaccine shedding in a new
case of severe combined immunodeficiency: A reason to screen. J Allergy Clin Immunol 2010;
125:270.
172. Payne DC, Edwards KM, Bowen MD, et al. Sibling transmission of vaccine-derived
rotavirus (RotaTeq) associated with rotavirus gastroenteritis. Pediatrics 2010; 125:e438.
173. Hemming M, Vesikari T. Detection of rotateq vaccine-derived, double-reassortant
rotavirus in a 7-year-old child with acute gastroenteritis. Pediatr Infect Dis J 2014; 33:655.
174. Sakon N, Miyamoto R, Komano J. An infant with acute gastroenteritis caused by a
secondary infection with a Rotarix-derived strain. Eur J Pediatr 2017; 176:1275.
175. Rivera L, Peña LM, Stainier I, et al. Horizontal transmission of a human rotavirus vaccine
strain--a randomized, placebo-controlled study in twins. Vaccine 2011; 29:9508.
176. Centers for Disease Control and Prevention. Rotavirus. In: Epidemiology and Prevention
of Vaccine-Preventable Diseases. The Pink Book: Course Textbook, 13th ed, Hamborsky J,
Kroger A, Wolfe S, (Eds). Public Health Foundation, Washington, DC 2015.
http://www.cdc.gov/vaccines/pubs/pinkbook/index.html (Accessed on July 09, 2015).
177. Bennett A, Pollock L, Jere KC, et al. Infrequent Transmission of Monovalent Human
Rotavirus Vaccine Virus to Household Contacts of Vaccinated Infants in Malawi. J Infect Dis
2019.
178. Leshem E, Lopman B, Glass R, et al. Distribution of rotavirus strains and strain-specific
effectiveness of the rotavirus vaccine after its introduction: a systematic review and meta-analysis.
Lancet Infect Dis 2014; 14:847.
179. Roczo-Farkas S, Kirkwood CD, Cowley D, et al. The Impact of Rotavirus Vaccines on
Genotype Diversity: A Comprehensive Analysis of 2 Decades of Australian Surveillance Data. J
Infect Dis 2018; 218:546.
180. Reyes JF, Wood JG, Beutels P, et al. Beyond expectations: Post-implementation data shows
rotavirus vaccination is likely cost-saving in Australia. Vaccine 2017; 35:345.
181. Bowen MD, Mijatovic-Rustempasic S, Esona MD, et al. Rotavirus Strain Trends During
the Postlicensure Vaccine Era: United States, 2008-2013. J Infect Dis 2016; 214:732.
182. Bernstein DI. Rotavirus Vaccines: Mind Your Ps and Gs. J Infect Dis 2018; 218:519.
183. Hungerford D, Allen DJ, Nawaz S, et al. Impact of rotavirus vaccination on rotavirus
genotype distribution and diversity in England, September 2006 to August 2016. Euro Surveill
2019; 24.
184. Al-Ayed MS, Asaad AM, Qureshi MA, Hawan AA. Epidemiology of group A rotavirus
infection after the introduction of monovalent vaccine in the National Immunization Program of
Saudi Arabia. J Med Virol 2017; 89:429.
185. Zeller M, Heylen E, Tamim S, et al. Comparative analysis of the Rotarix™ vaccine strain
and G1P[8] rotaviruses detected before and after vaccine introduction in Belgium. PeerJ 2017;
5:e2733.
186. Mukhopadhya I, Murdoch H, Berry S, et al. Changing molecular epidemiology of rotavirus
infection after introduction of monovalent rotavirus vaccination in Scotland. Vaccine 2017;
35:156.
187. Roczo-Farkas S, Kirkwood CD, Bines JE, and the Australian Rotavirus Surveillance
Group. Australian Rotavirus Surveillance Program annual report, 2015. Commun Dis Intell Q Rep
2016; 40:E527.
188. US Food and Drug Administration. Components of extraneous virus detected in Rotarix
vaccine; no known safety risk
www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm205625.htm (Accessed on
March 22, 2010).
189. U.S. Food and Drug Administration. Update on recommendations for the use of rotavirus
vaccines. www.fda.gov/BiologicsBloodVaccines/Vaccines/ApprovedProducts/ucm212140.htm
(Accessed on May 17, 2010).
190. Victoria JG, Wang C, Jones MS, et al. Viral nucleic acids in live-attenuated vaccines:
detection of minority variants and an adventitious virus. J Virol 2010; 84:6033.
191. Mijatovic-Rustempasic S, Immergluck LC, Parker TC, et al. Shedding of porcine circovirus
type 1 DNA and rotavirus RNA by infants vaccinated with Rotarix®. Hum Vaccin Immunother
2017; 13:928.
192. Kuehn BM. FDA: Benefits of rotavirus vaccination outweigh potential contamination risk.
JAMA 2010; 304:30.

Topic 6023 Version 63.0

You might also like