2,185
Views
25
CrossRef citations to date
0
Altmetric
Product Review

Product review of the rotavirus vaccines ROTASIIL, ROTAVAC, and Rotavin-M1

, , , & ORCID Icon
Pages 1223-1234 | Received 06 Apr 2020, Accepted 28 Jul 2020, Published online: 29 Oct 2020

ABSTRACT

Rotavirus is the leading cause of severe dehydrating gastroenteritis and death due to diarrhea among children under 5, causing over 180,000 under-5 deaths annually. Safe, effective rotavirus vaccines have been available for over a decade and are used in over 98 countries. In addition to the globally available, WHO-prequalified ROTARIX (GSK) and RotaTeq (Merck), several new rotavirus vaccines have attained national licensure – ROTAVAC (Bharat Biotech) and ROTASIIL (Serum Institute of India), licensed and manufactured in India and now WHO-prequalified, and Rotavin-M1 (PolyVac), licensed and manufactured in Vietnam. In this review, we summarize the available clinical trial and post-introduction evidence for these three new orally administered rotavirus vaccines. All three vaccines have demonstrated safety and efficacy against rotavirus diarrhea, although publicly available preclinical data are limited in some cases. This expanding product landscape presents a range of options to optimize immunization programs, and new presentations of each vaccine are currently under development.

Rotavirus and rotavirus gastroenteritis

Diarrhea is a leading cause of child mortality around the world, annually responsible for over 400,000 deaths among children under five years of ageCitation1. Among this age group, rotavirus is one of the most common causes of diarrheal deaths, accounting for over 180,000 under-five diarrheal deaths each year,Citation2,Citation3 and the most common cause of severe dehydrating gastroenteritis, with a particularly heavy burden in developing countries.Citation4–7 The virus is highly transmissible, with nearly all children in unvaccinated populations experiencing at least one rotavirus infection in the first five years of life.Citation8 Experts estimate that, globally, for each rotavirus death in 2017, an additional 12 patients were hospitalized and 2,000 patients contracted rotavirus diarrhea.Citation2

Rotavirus, of the Reoviridae family, is a highly communicable double-stranded RNA virus with an incubation period typically lasting 48 hours, during which time virus can be shed in the stool of an asymptomatic patient.Citation4 The virus is primarily transmitted through the fecal-oral route, but person-to-person and fomite transmission are also important.Citation4 Many, but not all, settings see seasonal patterns in rotavirus infection.Citation4

Rotavirus targets and damages enterocytes lining the small intestine, leading to symptoms ranging from mild watery diarrhea to more severe illness, with vomiting, fever, and dehydration.Citation9 Mild cases can be treated in the community with oral rehydration solution (ORS) and zinc supplements,Citation10 but access to these simple interventions is limited for the poorest children; severe cases require intravenous fluids and resuscitation and can lead to death.Citation11 Providing clean water and improving sanitation reduces overall diarrheal illness but, on its own, is insufficient to control rotavirus; rotavirus diarrhea continues to have a significant burden in populations without access to vaccination.Citation12,Citation13

In the community, the most commonly available diagnostic testing to confirm rotavirus infection is through enzyme immunoassay (EIA) of stool samples.Citation1 Culture and other techniques including reverse transcription-polymerase chain reaction (RT-PCR) are common in research settings.Citation14

Vaccine background

Safe, effective rotavirus vaccines (RVVs) have been available for over a decade. Building upon its original 2007 RVV position paper, in 2009 the WHO recommended that all countries include RVV in their national programs and schedules;Citation15 to date, the vaccines are included in the national immunization programs of more than 98 countries worldwide.Citation16 RVVs promote the immune response to rotavirus infection by stimulating the production of neutralizing antibodies to viral protein (VP) antigens, VP7 (G-type) and VP4 (P-type),Citation4,Citation17,Citation18 which are common to multiple, diverse strains of rotavirus.Citation19,Citation20 With studies demonstrating that the globally available vaccines have comparable efficacy despite different strain composition, questions have emerged about the mediating role of non-neutralizing antibodies against other VP antigens.Citation20–22 As an oral agent given in multiple doses, currently available RVVs are thought to selectively activate IgA production in the gastrointestinal tract, where infection occurs.Citation1

Although vaccination clearly protects against rotavirus disease, protection has notbeen demonstrated to correspond to a given level of neutralizing antibodies, thus, no immune correlate of protection is defined for RVV;Citation20,Citation23 however, rotavirus-specific IgA is the most frequently used metric in clinical evaluations of vaccines. Clinical trial efficacy is typically evaluated on the basis of clinical outcomes, such as gastroenteritis and its related signs and symptoms, hospitalization, or death.Citation20

ROTARIX® (GlaxoSmithKline, licensed in 2004) and RotaTeq® (Merck, licensed in 2006) are 80–90% efficacious in preventing severe rotavirus diarrhea in settings with relatively high income and low or very low mortality (across Europe and Latin America),Citation24,Citation25 and 40–60% efficacious in settings with relatively low income and high or very high mortality (particularly across Asia and Africa).Citation26 Various studies also show cross-protection against rotavirus strains not included in the vaccine.Citation27

Common, short-term side effects of rotavirus vaccine include irritability and mild, short-lived diarrhea or vomiting.Citation28 Large, controlled trials of the first RVV global market entrants, ROTARIX and RotaTeq, both live attenuated vaccines, have shown no difference in serious adverse event rates between RVV and placebo.Citation24,Citation25 However, some post-marketing studies have shown a small increased risk for intussusception – one to seven additional cases per 100,000 vaccinated infants – mainly after the first dose.Citation24,Citation25,Citation29-38 Intussusception is a rare, naturally occurring condition involving a “telescoping” of the intestines over themselves, which can damage the tissue; the mechanism of intussusception remains poorly understood.Citation39 The benefits of widespread RVV in preventing severe illness, hospitalizations, and deaths far outweigh the small risk of intussusception.Citation26 However, the WHO continues to conduct and review regional surveillance of intussusception among populations vaccinated with RVV.Citation40 Post-licensure studies of ROTARIX in Africa showed no increased risk of intussusception, confirmed with a recent analysis of active surveillance data from seven African countries.Citation38 Recent post-licensure analysis of surveillance data from India, specifically states that have introduced ROTAVAC, has also found no increased risk of intussusception; these reports are summarized below.Citation41,Citation42

Product characteristics

In addition to ROTARIX and RotaTeq, three newer RVV products are available nationally (Rotavin-M1) or internationally (ROTAVAC, ROTASIIL), with varying presentations ().

Table 1. Product characteristics of Rotavin-M1, ROTAVAC, and ROTASIIL (9, 47, 48, 50, 93−96).

Rotavin-M1, a liquid-frozen, monovalent rotavirus vaccine manufactured by Polyvac in Vietnam, was licensed in Vietnam in 2012.Citation43 It is not currently prequalified by WHO. Rotavin-M1 contains a single, live attenuated human rotavirus G1P[8] strain (KH0118-2003) isolated from a child in Vietnam.Citation44 Rotavin-M1 is available in single-dose vials that can be stored at −25°C to −15°C for two years and then at 2–8°C for two months after it has been thawed.Citation9,Citation43 Rotavin-M1 is administered orally in a two-dose schedule, with the first dose at 6 weeks or later and the second dose 60 d after the first.Citation44,Citation45 Data on the cold chain footprint required for storage at central and peripheral levels have not yet been made publicly available. A non-inferiority trial is currently underway to assess the immunogenicity of a liquid, ready-to-use formulation of Rotavin against the currently licensed form.Citation46

ROTAVAC ®, a liquid-frozen, monovalent rotavirus vaccine manufactured by Bharat Biotech in India, was licensed in India in 2014 and received WHO prequalification in January 2018.Citation45 ROTAVAC contains a single, live attenuated human rotavirus G9P[11] strain, isolated from asymptomatic neonates at the All India Institute of Medical Sciences, New Delhi.Citation45 Each glass vaccine vial contains either one, five, or ten doses of the frozen vaccine.Citation47 In the frozen form, it can be stored in central cold chain facilities at −20°C for up to five years, and can be refrigerated for up to 6 months between 2°C and 8°C; it can be thawed and refrozen up to six times.Citation47 ROTAVAC vials are labeled with VVM2 markers, a heat-sensitive visual marker that becomes darker when exposed to heat as the vaccine moves through the supply chain.Citation48 ROTAVAC is administered orally in a three-dose schedule with DTP-1, 2, and 3. The cold chain storage requirements for the three doses required to fully immunize one child range from 9.6 cm3 for the 10-dose vial to 45 cm3 for the 1-dose vial ().Citation9 ROTAVAC 5 CM (now ROTAVAC 5D), a liquid formulation of ROTAVAC, has completed clinical trials and is under consideration for WHO prequalification.

ROTASIIL®, a freeze-dried/lyophilized, pentavalent rotavirus vaccine manufactured by Serum Institute of India, Pvt. Ltd., was licensed in India in 2017 and received WHO prequalification in September 2018. ROTASIIL contains five live attenuated human-bovine rotavirus reassortant strains, G1, G2, G3, G4, and G9, produced in vero cells.Citation14 Each vaccine vial contains either one or two doses of the lyophilized vaccine; the diluent is stored in a separate vial that can withstand ambient temperature fluctuations.Citation47 In its freeze-dried form, the vaccine can be stored up to 30 months, refrigerated at 2–8°C; the antacid diluent may be stored up to 60 months at ambient temperature or 2–8°C.Citation47 Lyophilization reduces the risk of virus destabilization through aqueous processes, imparting ROTASIIL with a longer shelf-life over other forms of drying.Citation49 ROTASIIL has demonstrated thermostability at temperatures up to 25°C for up to 36 months, between 37°C and 40°C for 18 months, and for short time periods over 55°C.Citation50 However, the lyophilizing process can damage virus components, and there are limited measurement and analytic tools to accurately detect destabilization associated with this process.Citation49 ROTASIIL vials are labeled with a vaccine vial monitor (VVM30)Citation48,Citation51 Once reconstituted, the vaccine can be stored up to 6 hours at 2–8°C.Citation47,Citation52 ROTASIIL is administered orally in a three-dose schedule, with DTP-1, 2 and 3. Excluding the diluent, the cold chain footprint required for the three doses required to fully immunize one child is 52.7 cm3 for one-dose vials and 31.6 cm3 for two-dose vials.Citation9 A new liquid formulation of ROTASIIL has also been developed, with demonstrated immunological non-inferiority to lyophilized ROTASIIL and a similar safety profile. It is under consideration for WHO prequalification.Citation53,Citation54

Preclinical studies

Preclinical study results for Rotavin-M1 are publicly available on a limited basis (). Preclinical testing of Rotavin-M1 involved the development of in vitro and in vivo assays for the rotavirus working seed and master seed, consistent with WHO’s guidelines for attenuated rotavirus vaccine development. Testing was approved by Vietnam’s national regulatory authority, and each working and master seed lot was tested for safety, toxicity, and immunogenicity in several animals. Immunogenicity was demonstrated by a three-fold increase of antibody titers in baby monkeys.Citation77

Table 2. Summary of clinical trials for Rotavin-M1, ROTAVAC, and ROTASIIL

Descriptions of the results of preclinical studies of ROTASIIL are likewise somewhat limited in availability. ROTASIIL was produced in compliance with India’s current Good Manufacturing Practices (GMP), which provides quality assurance of pharmaceuticals manufactured in-country.Citation69 Clinical lot testing demonstrated both consistency and stability between lots.Citation69 Toxicity testing was performed in rabbits and rats for single and multiple oral doses, with no significant adverse effects observed in the test animals.Citation69 Immunogenicity data in the studied animals are not publically available.Citation69

The authors were not able to identify any publicly available preclinical study results for ROTAVAC.

Clinical studies

The clinical studies described below consider immunogenicity, safety, and efficacy of the three new RVV products ().

Rotavin-M1

Phase I

A 2009 two-stage study conducted by the National Institute of Hygiene and Epidemiology in Vietnam evaluated the safety of Rotavin-M1 produced by the Center for Research and Production of Vaccines and Biologicals (POLYVAC).Citation55 The first stage enrolled 29 healthy adult volunteers ages 18–40 y and administered two doses of Rotavin-M1 with a one-month interval between doses; no adverse events were reported.Citation42,Citation55 The second stage, a phase I and II adaptive trial, enrolled 200 healthy infants, assessing the reactogenicity of Rotavin-M1 by looking at immediate reactions (30 minutes) to administration of each dose and changes in blood cell counts, serum transaminase, and urea nitrogen concentration after vaccination.Citation42,Citation55 Researchers found that Rotavin-M1 was immunogenic and safe, and did not lead to an increased rate of fever, diarrhea, vomiting, or irritability in comparison to ROTARIX.Citation44 They also found that the two-dose schedule given weeks apart performed better than the three-dose schedule in terms of reactogenicity and rates of seroconversion. Rates of seroconversion were similar to infants in the ROTARIX group, but infants receiving Rotavin-M1 had less shedding of vaccine strain after subsequent doses.Citation44 Later studies intend to assess the efficacy of Rotavin-M1 with a larger sample size.Citation44

Phase II

A Phase II study conducted by the National Institute of Hygiene and Epidemiology in Vietnam in 2009 (completed in 2010) evaluated the safety and immunogenicity of Rotavin-M1 in infants.Citation56 Additionally, this study evaluated dosage and scheduling in order to determine the optimum regimen for the administration of Rotavin-M1.Citation56 This study enrolled 200 healthy infants ages 6–12 weeks at enrollment and randomly assigned them to either a 3-dose schedule with a 1-month interval between doses or a 2-dose schedule with a 2-month interval between doses.Citation56 This study also assessed the safety and reactogenicity of Rotavin-M1 compared to GSK’s ROTARIX. The duration of the immune response was assessed by analyzing antibody titers after one year.Citation56 Finally, this study evaluated the presence and shedding of rotavirus in stool.Citation56 No elevation of levels of serum transaminase, blood urea, or blood cell counts were observed.Citation44 The highest rotavirus IgA seroconversion rate (73%, 95% CI (58–88%)) was achieved in group 2 H (2 doses–10(6.3)FFU/dose, 2 months apart).Citation44 The 2-dose schedules performed slightly better than the three dose schedules and the higher titer doses performed slightly better than the lower titer doses.Citation44 These rates of seroconversion were similar to that of the ROTARIX group (58%, 95% CI (42–73%)).Citation44 However, more infants who received ROTARIX (65%) shed virus in their stool after the first dose than those who received Rotavin-M1 (44–48%) (p < .05), although the percent shedding decreased after subsequent doses of either vaccine.Citation44 These data demonstrate that Rotavin-M1 is safe and immunogenic in infants, and is not associated with an increased rate of fever, diarrhea, vomiting, or irritability in comparison to ROTARIX; no intussusception events were detected.Citation44 Beyond initial Phase II studies, there are no publicly available immunogenicity, safety, efficacy, or risk of intussusception data. Data also demonstrate that the two-dose schedule performed better than the three-dose schedule.Citation44

Phase II/III

A 2010 Phase IIb study conducted by the National Institute of Hygiene and Epidemiology in Vietnam evaluated the safety and immunogenicity of Rotavin-M1, including an assessment of the vaccine schedule of 2 doses with a 2-month interval at two study sites, Thanh Son-Phu Tho and Thai Binh city.Citation57 Researchers enrolled 799 healthy infants, ages 6 to 12 weeks at time of enrollment.Citation57 Participants were randomized to either placebo (cell culture medium in absence of virus) or active (Rotavin-M1 vaccine) groups.Citation57 Primary outcome measures included anti-rotavirus IgA antibody responses one month after vaccination. Secondary outcome measures included RV-IgA antibody responses to Rotavin-M1 one year after first dose, anti-rotavirus IgG antibody responses one month after vaccination, safety, and reactogenicity of each dose, and anti-RV IgG responses one year after the first dose.Citation57 The results of this study have not yet been published.

Phase III

A Phase III study registered in 2019 will be conducted by the Center for Research and production of Vaccines and Biologicals in Vietnam, to examine the non-inferiority of the immunogenicity of the liquid form of Rotavin compared to the frozen formulation.Citation46 The study will also assess the reactogenicity and safety of the vaccine.Citation46 This study will recruit 825 healthy infants from ages 60 to 91 d, and participants will be randomized to either the liquid or frozen formulation of Rotavin-M1.Citation46 The study will compare geometric mean concentration (GMC) of serum anti-rotavirus IgA antibodies between the liquid and frozen formulations, after each vaccine dose.Citation46 The study will also examine seroconversion, seropositivity, immediate adverse events, solicited and unsolicited adverse and severe adverse events, including intussusception.Citation46 Enrollment for this study has not yet begun.

Phase IV

The authors did not identify any Phase IV Rotavin-M1 studies published in the literature.

New presentations

The 2019 Phase III study described above will provide data to compare a new liquid formulation versus the existing frozen formulation.

ROTAVAC

Phase I/II

A 2005 Phase I study assessed the safety and immunogenicity of ROTAVAC and I321 (a live attenuated Indian rotavirus vaccine candidate strain) in healthy infants, using a randomized design with a placebo comparison group.Citation58 The study recruited 90 healthy infants age 8 weeks at enrollment, who received a single dose of ROTASIIL, I321, or placebo.Citation58 The study found that ROTAVAC induced a more robust immune response than either I321 or placebo.Citation58 There was no significant difference in the number of adverse events between the experimental and placebo groups.Citation58 This study found that ROTAVAC is attenuated, safe, and immunogenic with a single dose.Citation58

A 2006 (completed 2008) study assessed the safety of ROTAVAC in comparison to placebo in healthy infants.Citation59 The Phase I/II study was a double-blind, placebo-controlled, dose-escalation trial in India, in which 187 participants received a dose of 1 × 104 focus-forming units (ffu) while 182 received a dose of 1 × 105 ffu in a 1:1 randomization with placebo participants.Citation59 The study found no significant difference in adverse events or toxicity between groups; zero vaccine-related adverse events (including intussusception) were observed.Citation59 A significant difference in immunogenicity between the two experimental groups and the placebo group was observed; the study found that the 1 × 105 ffu dose resulted in a robust immune response, compared to the 1 × 104 ffu dose, and was selected for further trials.Citation59 The study also reported viral shedding in approximately 20% of enrolled infants; shedding rate was lower than with ROTARIX but greater than with RotaTeq.Citation59

A 2014 study of data collected in 2006 (completed in 2008) assessed the immune response and seroconversion of ROTAVAC in healthy infants age 6 weeks at enrollment.Citation60 The Phase Ia/IIb study was randomized, double-blinded, and placebo-controlled, and enrolled 369 infants.Citation60 The study reported interference of maternal antibody on ROTAVAC vaccine immunogenicity in infants, noting that ROTAVAC is able to overcome this interference when administered at a higher dose.Citation60

Phase III

A 2011 (completed 2014) study conducted by Bharat Biotech in India evaluated the efficacy of a three-dose schedule of ROTAVAC in comparison to placebo.Citation62 The primary outcome was efficacy against severe rotavirus gastroenteritis, defined as number of cases of non-vaccine rotavirus in subjects from 14 d after the third dose to age 2 y.Citation62 The secondary outcomes were efficacy at preventing hospitalization, use of supervised rehydration therapy, and safety compared to the placebo group. This study enrolled 6,800 healthy infants beginning in 2011, ages 6–7 weeks in 2011.Citation62 It used a double-blind, placebo-controlled design and randomized participants in a 2 to 1 allocation to (1) oral ROTAVAC vaccine, or (2) placebo. Participants were given three doses at four-week intervals co-administered with other routine childhood vaccines.Citation62 ROTAVAC was effective in preventing cases of severe rotavirus gastroenteritis and was well tolerated in Indian infants; the study further noted that ROTAVAC offered protection against a range of commonly circulating genotypes, including G1P[8], G2P[4], G12P[6], G12P[8] and G9P[4].Citation29 Vaccine efficacy against severe rotavirus gastroenteritis was 53.6% (95% CI, 35.0–66.9%; P < .001) and 56.4% (95% CI, 36.6–70.1%; P < .001) in the first year of life.Citation29 Vaccine efficacy in the second year of life was 48.9% (95% CI 17.4 to 68.4; p = .0056).Citation63 Prevalence of immediate, solicited, and serious adverse events was similar in both groups.Citation29 In the initial period, there were six cases of intussusception in the vaccine group and two in the placebo group, and in the second year there were eight cases in the vaccine group and three in the placebo group; all intussusception cases in the vaccine group occurred more than 100 d after the third dose and were not associated with vaccination.Citation29 No deaths were found to be related to the study vaccine.Citation29,Citation63

A study completed in 2015 assessed possible ROTAVAC interference with the immune response to coadministered vaccines.Citation65 Healthy infants aged 6 weeks at enrollment were randomized to either a control or placebo ground (where they would also receive coadministered childhood vaccines).Citation65 Coadministered vaccines included oral polio vaccine, diptheria, pertussis, tetanus, hepatitis b, and Haemophilus influenza type b.Citation65 The study showed that ROTAVAC can be safely administered with childhood vaccines without interfering with the immune response to the antigens contained in these vaccines.Citation65

A 2018 (completed 2019) study conducted by the Institute of Clinical Research and Clinical Trial Support for Vaccine and Biological Products in Thái Bình, Vietnam, evaluated the immunogenicity, safety, and reactogenicity of ROTAVAC as a three-dose series in 360 healthy infants, ages 6–8 weeks.Citation63 This study used an open-label, single-group assignment model and administered three doses of ROTAVAC at a four-week interval with the first dose given at age 6–8 weeks.Citation64 Primary outcome measures were the frequency and rate of adverse events after vaccination, including immediate adverse events within 30 minutes of vaccination, and adverse events 7 d and 28 d after each vaccination.Citation64 The results of this study have not been published at the time of writing.

A 2019 study evaluated the impact of coinfections on the efficacy of the ROTAVAC vaccine.Citation66 The study recruited 1,169 healthy infants who were tested for the presence of coinfections.Citation66 This study found that vaccine efficacy increased from 49.3% to 60.6% in the absence of coinfections (difference, 11.3%, 95% CI, −10.3% to 30.2%).Citation66 In other words, accounting for coinfections could help to explain variance in efficacy amongst populations; this nonsignificant difference may merit further evaluation in larger studies.

Phase IV

A 2014 Phase IV, multi-center, single-blind, randomized study assessed the immunogenicity and safety of ROTAVAC when administered to healthy infants with and without the buffering agent.Citation67 The study enrolled and randomized to three arms 900 infants 6–8 weeks of age: Group I received the buffer five minutes before receiving ROTAVAC, Group II received no buffer, and Group III received the buffer simultaneously with ROTAVAC.Citation67 Eighteen serious adverse events were reported, but none were attributable to the vaccine.Citation67 ROTAVAC administration without a buffer was shown to be well tolerated and immunogenic.Citation67

A 2015 (completed 2016) Phase IV study assessed the antibody response of two different dosing schedules of ROTAVAC in healthy infants.Citation68 This study also assessed the non-inferiority of ROTAVAC to ROTARIX for GMC in infants.Citation68 The study was a multicenter, open-label, randomized design with two experimental groups (Group I, 3 doses; Group II, 2 doses).Citation68 It showed that the GMC ratio met the non-inferiority criteria (0.82; 95% CI: 0.64, 1.05) and that there was no substantive difference between ROTAVAC and Rotarix.Citation68

Surveillance

From 2015 to 2019, passive surveillance was conducted at 35 health facilities across three Indian states – Himachal Pradesh, Maharashtra, and Tamil Nadu – to better understand and estimate the risk of intussusception among infants vaccinated with ROTAVAC.Citation41 A total of 151 intussusception cases meeting Brighton Level 1 criteria were identified; of these, 104 were vaccinated with ROTAVAC and 47 were unvaccinated.Citation41 Attributable risk 1–21 d after the first and second doses (combined) was 0.11 (95% CI: 0.0–0.25) per 100,000 doses, and 0.42 (95% CI: 0.0-.70) per 100,000 doses for all three doses combined; no clustering of cases was seen after any dose.Citation41 The analysis found no increased risk of intussusception during the first 21 d following any of the three doses of ROTAVAC or all three doses combined, compared to the control period (d 22–365).Citation41

As reported at the December 2019 meeting of the Global Advisory Committee on Vaccine Safety, active sentinel site surveillance was conducted across 28 hospitals in nine states in India following the introduction of ROTAVAC, with analysis of cases meeting Brighton Level 1 criteria to assess possible association of ROTAVAC vaccination and intussusception.Citation42 A total of 589 children with known ROTAVAC vaccination status (0, 1, 2, or 3 doses) were included in the analysis; incidence rate ratios for seven d after the first, second, and third doses were compared to the period from 28 d to 1 year of age.Citation42 The incident rate ratios showed no statistically significant difference from no association for d 1–7, and no significant difference was found for the risk window of 8–21 d after vaccination, with any dose.Citation42

New presentations

A 2019 Phase IIb, single-center, randomized, controlled, open-label immunogenicity study conducted by the Center for Infectious Disease Research in Zambia enrolled 450 healthy infant volunteers, comparing 3 doses with the frozen formulation of ROTAVAC, 3 doses with the liquid formulation of ROTAVAC 5 CM (now ROTAVAC 5D), and 2 doses of ROTARIX.Citation61 The study evaluated immunogenicity at 28 d post-dose-3, as well as the reactogenicity and safety of the two ROTAVAC formulations compared against each other and compared against ROTARIX, at one week after each dose.Citation61 The primary outcome measure was GMC of serum-anti-rotavirus IgA antibodies.Citation61 This study has been completed, but results have not yet been published.

ROTASIIL

Phase I/II

A 2014 multi-phased study assessed the toxicity of ROTASIIL in adults, toddlers, and infants.Citation69 The Phase I component was a randomized, double-blind, placebo-controlled study that examined the safety, immunogenicity, and shedding of the ROTASIIL vaccine in healthy adults, toddlers, and infants.Citation69 ROTASIIL was found to be safe and well tolerated and there were no reports of severe adverse events.Citation69 There were a few adverse event reports, including nausea, loss of appetite, diarrhea, and vomiting, but none of these were severe.Citation69 No shedding was seen in stool samples.Citation69

The Phase IIa component was a double-blind, placebo-controlled study that assessed the safety, immunogenicity, and shedding of the vaccine virus contained in ROTASIIL, in healthy infants.Citation69 Two cases of severe adverse events were reported in post-vaccination follow-up, one urinary tract infection, and one septicemia; both were found to be unrelated to the ROTASIIL vaccine.Citation69 The vaccine was found to be safe and well tolerated, and three doses of the vaccine were found to be immunogenic.Citation69 No shedding was seen in stool samples.Citation69

A randomized, double-blind, placebo-controlled Phase IIb study examined the immunogenicity of ROTASIIL in healthy infants.Citation69 The main difference between the Phase IIa and Phase IIb trials was the dose: Phase IIa used a dose of 1 × 105.2 ffu/serotype while Phase IIb used a dose of 1 × 105.6 ffu/serotype.Citation69 Vaccine and placebo were administered as three doses with a minimum four-week gap between doses, administered in a 1:1 ratio.Citation69 The vaccine was found to be safe, immunogenic in infants, well tolerated, and no severe adverse events were reported.Citation69

Phase III

A 2014 (completed 2017) study conducted by Serum Institute of India used a multicenter, randomized, double-blind, placebo-controlled design to examine efficacy and safety of a three-dose series of ROTASIIL, administered orally with a four-week interval to healthy Indian infants, ages 6–8 weeks at enrollment.Citation71 Seven thousand five hundred participants were randomized to either the experimental vaccine arm or the placebo group with a 1:1 allocation.Citation71 The primary outcome was the occurrence of severe rotavirus gastroenteritis up to two years of age.Citation71 Secondary outcomes were safety, including solicited post-vaccination reactions, severe adverse events, unsolicited adverse events, intussusception, and death.Citation71 At the time of primary analysis, vaccine efficacy against severe rotavirus gastroenteritis was 36% (95% CI 11.7, 53.6, p = .0067) for the per-protocol analysis and 41.9% (21.1, 57.3, p = .0005) for the intention-to-treat (ITT) analysis; at the end of the follow-up period (until two years of age), vaccine efficacy was 39.5% using a per-protocol analysis and 38.8% using ITT analysis.Citation30 Vaccine efficacy against very severe rotavirus cases was 60.5% (95% CI 17.7, 81, p = .0131) at the time of the primary analysis and 54.7% (95% CI 29.7, 70.8, p = .0004) over the complete follow-up period in the per-protocol analysis.Citation30 The incidence of solicited, unsolicited, and serious adverse events was comparable between vaccine and placebo groups.Citation30 ROTASIIL was found to be effective, safe, and well tolerated by Indian infants.Citation30

A 2014 (completed 2017) double-blinded, randomized, placebo-controlled study conducted by Epicenter in Niger evaluated the efficacy and safety of ROTASIIL in infants followed until they were young children.Citation72,Citation73Researchers recruited 4,092 participants ages 6 weeks and followed participants out to 2 y.Citation72,Citation73 Participants were randomized to experimental vaccine and placebo groups using a 1:1 allocation.Citation72,Citation73Three doses of the vaccine or placebo were administered at four-week intervals, with the first dose occurring at age 6–8 weeks.Citation72 The primary outcome measures were episodes of severe rotavirus gastroenteritis after the final dose.Citation72 Secondary outcome measures were rotavirus gastroenteritis, hospitalization due to rotavirus gastroenteritis, hospitalization of any cause (adverse effects or rotavirus gastroenteritis), serious adverse events, and anti-rotavirus IgA sero-response rates and geometric mean titers.Citation72–74 The study found an efficacy of 66.7% (95% CI: 49.9, 77.9) against severe rotavirus gastroenteritis.Citation73 Only one case of intussusception was reported, 542 d after receipt of the third dose.Citation74 The vaccine group experienced 395 serious adverse events, compared to 419 serious adverse events in the placebo group; this difference was not significant.Citation74 Adverse events between the two groups were comparable, with 1474 (72.1%) participants receiving ROTASIIL and 1456 (71.1%) participants receiving placebo experiencing at least one adverse event (p = .49) in the follow-up period; ROTASIIL was found to be safe.Citation74 Limitations of this study included the complexity of accurately diagnosing participants in a low-resource environment where overlapping clinical features and pathologies can complicate classification.Citation74

A 2015 (completed 2017) open-label, randomized study conducted by the Serum Institute of India recruited 1,500 healthy volunteers, ages 6–8 weeks.Citation75 Participants were randomized to four experimental arms: (1) ROTASIIL Lot A; (2) ROTASIIL Lot B; (3) ROTASIIL Lot C; or (4) ROTARIX.Citation75 This study evaluated equivalence in specific anti-rotavirus IgA antibodies across the three production lots and examined potential interference of the vaccine with other standard infant vaccines given concurrently.Citation75 Primary outcome measures included immunogenicity of the rotavirus vaccine in each group at four weeks after the third dose, and immunogenicity of universal immunization program vaccines, four weeks after the third dose of vaccination. Secondary outcome measures included immediate, solicited, and unsolicited adverse events (reactogenicity), within 30 minutes after vaccination and seven d after each vaccination, respectively.Citation75 Antibody responses between ROTASIIL and ROTARIX were comparable.Citation76 ROTASIIL was not found to interfere with the immunogenicity of concurrently administered vaccines: more than 97% of participants demonstrated seroprotective antibodies against diphtheria, tetanus, hepatitis B, and polio type 1 and 3.Citation76

Phase IV

The authors did not identify any Phase IV ROTASIIL studies in preparation, ongoing, or published in the literature.

New presentations

A 2017 (completed 2018) study conducted by the Serum Institute of India in New Delhi, India, evaluated the liquid formulation of ROTASIIL in comparison to the lyophilized, or freeze-dried, formulation.Citation70 This non-inferiority study compared the induction of anti-rotavirus IgA antibodies across three different production lots.Citation70 Researchers recruited 1,500 healthy infants, ages 6–8 weeks at enrollment.Citation70 This study also evaluated consistency in the manufacturing of ROTASIIL by comparing anti-rotavirus IgA antibodies across three different production lots.Citation70 Participants were randomized to four different study arms; (1) liquid formulation Lot A; (2) liquid formulation Lot B; (3) liquid formulation Lot C; and (4) lyophilized formulation.Citation70 The incidence of adverse events was similar across all four study arms.Citation53 There was only one vaccine-related serious adverse event of gastroenteritis in the ROTASIIL group.Citation53 The liquid formulation was found to be non-inferior from an immunologic and safety perspective compared to the lyophilized formulation, and the study demonstrated lot-to-lot consistency.Citation53

Vaccine impact on circulating rotavirus strains

No published studies have yet focused on the impact of Rotavin-M1, ROTASIIL, or ROTAVAC on circulating rotavirus strains. However, multiple studies have documented genotype changes in circulating strains following the introduction of other licensed rotavirus vaccines. In a systematic review on circulating rotavirus strains in the Middle East region, studies saw elevated proportions of non-vaccine strains G9P[8], G2P[4], and G9P[4] in Yemen, Saudi Arabia, and Morocco in post-vaccination periods with RotaTeq (G1, G2, G3, G4, and P[8]) and ROTARIX (G1P[8]).Citation78 A systematic review of rotavirus strains at the global level six years after RotaTeq and ROTARIX licensure found no significant shifts in strain prevalence, but recommended continued monitoring of G2P[4] prevalence in regions using ROTARIX.Citation79 Despite this scholarly interest in detecting any shifts in prevalence after population-wide introduction of the vaccine, the potential clinical relevance of such a trend is incompletely understood, as currently available oral, live attenuated RVV products demonstrate cross-protection against non-vaccine strains.Citation80,Citation81 Continuing regionally representative surveillance after wider-spread introduction of the three new products described in this review will be useful to detect the emergence of potentially pathogenic strains.Citation79

Schedule and integration with routine infant vaccines

Strong safety data support the concurrent administration of both ROTARIX and RotaTeq with the routine infant DTP dosing schedule;Citation65,Citation82 the schedule for ROTAVIN-M1 differs slightly, as described above. Although no published studies have assessed the impact on immunogenicity, safety, or efficacy of the simultaneous co-administration of Rotavin-M1 with other routine infant vaccines, there is no evidence of appreciable interference of ROTAVAC and ROTASIIL with other simultaneously administered routine childhood vaccines.

Cost-effectiveness

Some data have suggested greater cost-effectiveness of ROTARIX compared to ROTAVAC and ROTASIIL,Citation83 however, a model in the Palestinian setting suggests ROTAVAC is cost-saving compared to ROTARIX;Citation84 increasing scale of use and changes in other parameters can easily shift results of such modeling in the opposite direction.Citation85 Future economic and cost-effectiveness studies in low- and low-middle income countries may highlight further economic benefits of these three new vaccines.

Interchangeability

As more products become available, the likelihood increases that a child might receive a mixed schedule or be mid-series when a country switches from one product to another. The WHO recommends that children complete the vaccine product series on which they began, since no studies on interchangeability have been performed to demonstrate equivalent efficacy and immunogenicity with multiple vaccine products.Citation86 However, in settings where additional doses with the initial product are unavailable, substitution of other rotavirus vaccine products offers protection over an incomplete vaccine series.Citation86 A multicenter-randomized study of rotavirus vaccine schedules in children showed that mixed vaccine schedules (ROTARIX and RotaTeq) were noninferior in the induced immune response.Citation87 A retrospective study of children with gastrointestinal symptoms in the New Vaccine Surveillance Network found that a 3-dose regimen of mixed RotaTeq and ROTARIX vaccines showed 80% vaccine effectiveness against rotavirus infection.Citation88 Studies of interchangeability between ROTAVAC and ROTASIIL are underway in India, where the two vaccines have been introduced on a state-by-state basis.Citation89

New formulations and future developments

While the liquid-frozen and lyophilized formulations of ROTAVAC and ROTASIIL, respectively, are now in use in several countries, and Rotavin-M1 is available on the private market, all three manufacturers continue to develop new formulations to better meet the needs of a range of settings and markets. These aim to mitigate potential barriers to use of the current presentations in some settings – for example, a fully liquid ROTAVAC will eliminate the need for freezer (−20°C) storage for the vaccine – and improve their suitability for immunization programs by shifting toward ready-to-use presentations, while also gathering additional clinical trial and post-licensure data.

Studies are underway to elicit necessary, more widely applicable clinical trial data for Rotavin-M1. Once these additional clinical trials are complete and data available, it is anticipated that Rotavin-M1 will be eligible for consideration for licensure by other national regulatory authorities, beyond Vietnam’s private market, and potentially for WHO prequalification.

New formulations of both ROTAVAC and ROTASIIL have also been prioritized by manufacturers and are expected to be considered for WHO prequalification. ROTAVAC 5D – a fully liquid, ready-to-use version of the WHO-prequalified liquid-frozen ROTAVAC – is available in single- and multi-dose vials with VVM7, and can be stored at 2–8°C for up to 24 months.Citation90 It was commercially licensed in India in mid-2019; as discussed above, clinical trials for ROTAVAC 5D are completed and the vaccine will likely receive WHO prequalification status soon. A fully liquid formulation of ROTASIIL – which is currently available only as a lyophilized vaccine, requiring reconstitution – was launched in early 2020 and is undergoing clinical trials. A heat-stable presentation of ROTASIIL using a VVM250 is also anticipated; prior studies have demonstrated the current, lyophilized formulation of ROTASIIL retains its stability at temperatures up to 25°C for up to 36 months, between 37°C and 40°C for 18 months, and for short time periods over 55°C.Citation50 Using the newly WHO-approved VVM250 will ensure accurate tracking of ROTASIIL excursions from viable temperature ranges when storing the product outside the cold chain.

Market context

The global market for rotavirus vaccines has expanded significantly over the last 10 y, and demand is expected to continue increasing over the coming decade.Citation91 To reach the global market, ROTAVAC and ROTASIIL must be exported from India.Citation48 Given their WHO prequalification status, both can be procured by UN agencies for use in Gavi-eligible countries.Citation52,Citation92-94 Rotavin-M1 would also require exportation from Vietnam to reach the global market, and pre-qualification to be eligible for UN agency procurement.

Conclusion

Data supports the safety and efficacy of these three new products – Rotavin-M1, ROTAVAC, and ROTASIIL – for preventing severe rotavirus disease. Post-licensure data on current formulations and clinical trial data on new formulations will be critical to inform country introduction decisions and market uptake. Continued research also is needed to understand the individual clinical, epidemiologic, and programmatic implications of using two RVV products simultaneously in the same population or switching from one product to another. Likewise, little is known about the potential implications of using a ‘mixed’ schedule for the individual child.

Economic questions remain as well. Although ROTAVAC and ROTASIIL are available for global procurement, exportation and shipment from India may incur additional costs. Operational research is needed to understand the feasibility of technology transfer agreements and similar mechanisms to allow localized manufacturing. Likewise, implementation research is needed to evaluate the implications of product characteristics on real-world access and uptake of vaccine. Frozen vaccines may offer lower cost and longer shelf life, but require an investment in cold chain expansion; lyophilized vaccines offer thermostability, but the requirement for reconstitution may introduce human error, or present complicated storage considerations. Further study should consider if the health workforce and training program can adequately prepare for complicated reconstitution workflows, as well as if projected vaccination costs are representative of the total cost of procuring and delivering one of these new vaccines. As the evidence continues to accumulate for these three products, studies answering these programmatic and implementation questions will be of similar importance as studies providing post-marketing safety and impact data. As Rotavin-M1, ROTAVAC, and ROTASIIL become more widely used, it will be important to consider global and national policies regarding their use, and how these policies can best reflect the body of emerging evidence to guarantee widespread vaccine access and high-quality program implementation.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

References

  • World Health Organization. Number of deaths by region, Diarrhoeal diseases. World Health Organization Global Health Observatory. [Accessed March 9, 2020]. http://apps.who.int/gho/data/view.main.CM1002015REG6-CH3?lang=en.
  • Global Burden of Disease Collaborative Network. Global burden of disease study 2017 (GBD 2017) results. [Accessed February 14, 2020]. http://ghdx.healthdata.org/gbd-results-tool.
  • Tate JE, Burton AH, Boschi-Pinto C, Parashar UD. World health organization–coordinated global rotavirus surveillance network. Global, regional, and national estimates of rotavirus mortality in children <5 Years of Age, 2000-2013. Clin Infect Dis Off Publ Infect Dis Soc Am. 2016;62(Suppl 2):S96–S105. doi:10.1093/cid/civ1013.
  • Centers for Disease Control and Prevention. Rotavirus. centers for disease control and prevention. [Published September 25, 2019; accessed February 23, 2020]. https://www.cdc.gov/vaccines/pubs/pinkbook/rota.html.
  • Kotloff KL, Nataro JP, Blackwelder WC, Nasrin D, Farag TH, Panchalingam S, Wu Y, Sow SO, Sur D, Breiman RF, et al. Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): a prospective, case-control study. Lancet Lond Engl. 2013;382(9888):209–22. doi:10.1016/S0140-6736(13)60844-2.
  • Liu J, Platts-Mills JA, Juma J, Kabir F, Nkeze J, Okoi C, Operario DJ, Uddin J, Ahmed S, Alonso PL, et al. Use of quantitative molecular diagnostic methods to identify causes of diarrhoea in children: a reanalysis of the GEMS case-control study. Lancet Lond Engl. 2016;388(10051):1291–301. doi:10.1016/S0140-6736(16)31529-X.
  • ROTA Council at International Vaccine Access Center (IVAC), Johns Hopkins Bloomberg school of public health. Rotavirus disease and immunization: the epidemiology and disease burden of rotavirus. [Published 2019; accessed March 9, 2020]. http://rotacouncil.org/wp-content/uploads/2019/05/ROTA-Brief3-Burden-SP-1.pdf.
  • Kotloff KL. The Burden and Etiology of Diarrheal Illness in Developing Countries. Pediatr Clin North Am. 2017;64(4):799–814. doi:10.1016/j.pcl.2017.03.006.
  • Gavi Secretariat and Partners. Gavi- supported rotavirus vaccine profiles to support country decision making. [Published July 2019; accessed March 31, 2020]. https://www.gavi.org/sites/default/files/document/rotavirus-vaccine-profilespdf.pdf.
  • Bhatnagar S, Bahl R, Sharma PK, Kumar GT, Saxena SK, Bhan MK. Zinc With Oral Rehydration Therapy Reduces Stool Output and Duration of Diarrhea in Hospitalized Children: A Randomized Controlled Trial. J Pediatr Gastroenterol Nutr. 2004;38(1):34–40. doi:10.1097/00005176-200401000-00010.
  • Santosham M, Chandran A, Fitzwater S, Fischer-Walker C, Baqui AH, Black R. Progress and barriers for the control of diarrhoeal disease. Lancet. 2010;376(9734):63–67. doi:10.1016/S0140-6736(10)60356-X.
  • Yen C, Healy K, Tate JE, Parashar UD, Bines J, Neuzil K, Santosham M, Steele AD. Rotavirus vaccination and intussusception - Science, surveillance, and safety: A review of evidence and recommendations for future research priorities in low and middle income countries. Hum Vaccines Immunother. 2016;12(10):2580–89. doi:10.1080/21645515.2016.1197452.
  • Cook SM, Glass RI, LeBaron CW, Ho MS. Global seasonality of rotavirus infections. Bull World Health Organ. 1990;68:171–77.
  • World Health Organization. Rotavirus. [Published September 5, 2018; accessed January 29, 2020]. https://www.who.int/immunization/monitoring_surveillance/burden/vpd/WHO_SurveillanceVaccinePreventable_19_Rotavirus_R2.pdf?ua=1.
  • World Health Organization. 2009. Rotavirus Vaccines: an Update. World Health Organization. 533–40.
  • VIEW-hub. [Accessed February 3, 2020]. https://view-hub.org/viz/?YXBwaWQ9MSZpbmRpY2F0b3JpZD02MCZvdmVybGF5aWQ9NA==.
  • Marwick C. Rotavirus Vaccine a Boon to Children. JAMA. 1998;279(7):489–90. doi:10.1001/jama.279.7.489.
  • Schwartz JL. The First Rotavirus Vaccine and the Politics of Acceptable Risk: first Rotavirus Vaccine and Politics of Acceptable Risk. Milbank Q. 2012;90(2):278–310. doi:10.1111/j.1468-0009.2012.00664.x.
  • Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P. Innate Immunity. Mol Biol Cell. 4th ed. 2002 [Published; Accessed January 29]. https://www.ncbi.nlm.nih.gov/books/NBK26846/
  • Desselberger U, Huppertz H-I. Immune Responses to Rotavirus Infection and Vaccination and Associated Correlates of Protection. J Infect Dis. 2011;203(2):188–95. doi:10.1093/infdis/jiq031.
  • Ogden KM, Tan Y, Akopov A, Stewart LS, McHenry R, Fonnesbeck CJ, Bhinnata P, Carter MH, Fedorova NB, Halpin RA, et al. Multiple Introductions and Antigenic Mismatch with Vaccines May Contribute to Increased Predominance of G12P[8] Rotaviruses in the United States. López S, ed. J Virol. 2018;93(1):e01476–18. /jvi/93/1/JVI.01476-18.atom. doi:10.1128/JVI.01476-18.
  • Lappalainen S, Pastor AR, Tamminen K, López-Guerrero V, Esquivel-Guadarrama F, Palomares LA, Vesikari T, Blazevic V. Immune responses elicited against rotavirus middle layer protein VP6 inhibit viral replication in vitro and in vivo. Hum Vaccines Immunother. 2014;10(7):2039–47. doi:10.4161/hv.28858.
  • Angel J, Steele AD, Franco MA. Correlates of protection for rotavirus vaccines: possible alternative trial endpoints, opportunities, and challenges. Hum Vaccines Immunother. 2014;10(12):3659–71. doi:10.4161/hv.34361.
  • Ruiz-Palacios GM, Pérez-Schael I, Velázquez FR, Abate H, Breuer T, Clemens SC, Cheuvart B, Espinoza F, Gillard P, Innis BL, et al. Safety and efficacy of an attenuated vaccine against severe rotavirus gastroenteritis. N Engl J Med. 2006;354(1):11–22. doi:10.1056/NEJMoa052434.
  • Vesikari T, Matson DO, Dennehy P, Van Damme P, Santosham M, Rodriguez Z, Dallas MJ, Heyse JF, Goveia MG, Black SB, et al. Safety and Efficacy of a Pentavalent Human–Bovine (WC3) Reassortant Rotavirus Vaccine. N Engl J Med. 2006;354(1):23–33. doi:10.1056/NEJMoa052664.
  • World Health Organization. Rotavirus vaccines. WHO position paper – january 2013. Releve Epidemiol Hebd. 2013;88(5):49–64.
  • Patel MM, Steele D, Gentsch JR, Wecker J, Glass RI, Parashar UD. Real-world Impact of Rotavirus Vaccination. Pediatr Infect Dis J. 2011;30(1):S1. doi:10.1097/INF.0b013e3181fefa1f.
  • Centers for Disease Control and Prevention. Vaccine Adverse Event Reporting System (VAERS) | monitoring | ensuring Safety | vaccine Safety | CDC. [Published December 11, 2019; accessed February 3, 2020]. https://www.cdc.gov/vaccinesafety/ensuringsafety/monitoring/vaers/index.html.
  • Bhandari N, Rongsen-Chandola T, Bavdekar A, John J, Antony K, Taneja S, Goyal N, Kawade A, Kang G, Rathore SS, et al. Efficacy of a Monovalent Human-Bovine (116E) Rotavirus Vaccine in Indian Infants: A Randomised Double Blind Placebo Controlled Trial. Lancet Lond Engl. 2014;383(9935):2136–43. doi:10.1016/S0140-6736(13)62630-6.
  • Kulkarni PS, Desai S, Tewari T, Kawade A, Goyal N, Garg BS, Kumar D, Kanungo S, Kamat V, Kang G, 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(45):6228–37. doi:10.1016/j.vaccine.2017.09.014.
  • Weintraub ES, Baggs J, Duffy J, Vellozzi C, Belongia EA, Irving S, Klein NP, Glanz JM, Jacobsen SJ, Naleway A, et al. Risk of Intussusception after Monovalent Rotavirus Vaccination. N Engl J Med. 2014;370(6):513–19. doi:10.1056/NEJMoa1311738.
  • Patel MM, López-Collada VR, Bulhões MM, De Oliveira LH, Márquez AB, Flannery B, Esparza-Aguilar M, Montenegro Renoiner EI, Luna-Cruz ME, Sato HK, et al. Intussusception risk and health benefits of rotavirus vaccination in Mexico and Brazil. N Engl J Med. 2011;364(24):2283–92. doi:10.1056/NEJMoa1012952.
  • Velázquez FR, Colindres RE, Grajales C, Hernández MT, Mercadillo MG, Torres FJ, Cervantes-Apolinar M, DeAntonio-Suarez R, Ortega-Barria E, Blum M, et al. Postmarketing surveillance of intussusception following mass introduction of the attenuated human rotavirus vaccine in Mexico. Pediatr Infect Dis J. 2012;31(7):736–44. doi:10.1097/INF.0b013e318253add3.
  • Stowe J, Andrews N, Ladhani S, Miller E. The risk of intussusception following monovalent rotavirus vaccination in England: A self-controlled case-series evaluation. Vaccine. 2016;34(32):3684–89. doi:10.1016/j.vaccine.2016.04.050.
  • Haber P, Patel M, Pan Y, Baggs J, Haber M, Museru O, Yue X, Lewis P, DeStefano F, Parashar UD, et al. Intussusception after rotavirus vaccines reported to US VAERS, 2006-2012. Pediatrics. 2013;131(6):1042–49. doi:10.1542/peds.2012-2554.
  • Haber P, Parashar UD, Haber M, DeStefano F. Intussusception after monovalent rotavirus vaccine-United States, Vaccine Adverse Event Reporting System (VAERS), 2008-2014. Vaccine. 2015;33(38):4873–77. doi:10.1016/j.vaccine.2015.07.054.
  • Carlin JB, Macartney KK, Lee KJ, Quinn HE, Buttery J, Lopert R, Bines J, McIntyre PB. Intussusception risk and disease prevention associated with rotavirus vaccines in Australia’s National Immunization Program. Clin Infect Dis Off Publ Infect Dis Soc Am. 2013;57(10):1427–34. doi:10.1093/cid/cit520.
  • Tate JE, Mwenda JM, Lopman Armah BAG, Jani B, Omore R, Ademe A, Mujuru H, Mpabalwani E, Ngwira B, Cortese MM, et al. Evaluation of Intussusception after Monovalent Rotavirus Vaccination in Africa. N Engl J Med. 2018;378(16):1521–28. doi:10.1056/NEJMoa1713909.
  • Mayo Clinic. Intussusception. [Published 2020]. https://www.mayoclinic.org/diseases-conditions/intussusception/symptoms-causes/syc-20351452.
  • World Health Organization. Rotavirus vaccine safety update. World Health Organization. [Accessed February 3, 2020]. http://www.who.int/vaccine_safety/committee/topics/rotavirus/rotarix_and_rotateq/dec_2017/en/.
  • Early Rollout of ROTAVAC® India Network. Assessment of risk of intussusception after pilot rollout of rotavirus vaccine in the Indian public health system. Vaccine. 2020;38(33):5241–48. doi:10.1016/j.vaccine.2020.05.093.
  • World Health Organization. Global Advisory Committee on Vaccine Safety, 4–5 December 2019. Wkly Epidemiol Rec. 2020;95(4):25–36.
  • Riveros X Overview of characteristics of RV vaccines licensed and/or in clinical evaluation phase. Presented at the: Rotavirus scoping meeting. October 2, 2017. [Accessed February 13, 2020] https://www.who.int/immunization/research/meetings_workshops/3_RV_vaccines_RX_oct17.pdf.
  • Dang DA, Nguyen VT, Vu DT, Anh NTH, Mao ND, Wang Y, Jiang B, Hien ND, Luan LT. A dose-escalation safety and immunogenicity study of a new live attenuated human rotavirus vaccine (Rotavin-M1) in Vietnamese children. Vaccine. 2012;30(Suppl 1):A114–121. doi:10.1016/j.vaccine.2011.07.118.
  • World Health Organization. About WHO prequalification of vaccines. WHO. [Accessed February 24, 2020]. http://www.who.int/medicines/regulation/prequalification/prequal-vaccines/about/en/.
  • ClinicalTrials.gov. Identifier NCT03703336, Phase III study of liquid formulation of ROTAVIN. NIH U.S. National Library of Medicine. [Published January 25, 2019; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT03703336.
  • ROTA Council at International Vaccine Access Center (IVAC), Johns Hopkins Bloomberg school of public health. Current and Upcoming Rotavirus Vaccines. ROTA Council:7. [Accessed February 24, 2020]. http://rotacouncil.org/wp-content/uploads/2019/05/ROTA-Brief2-ProductLandscape-SP-1-3.pdf.
  • Deen J, Lopez AL, Kanungo S, Wang X-Y, Anh DD, Tapia M, Grais RF. Improving rotavirus vaccine coverage: can newer-generation and locally produced vaccines help? Hum Vaccines Immunother. 2018;14(2):495–99. doi:10.1080/21645515.2017.1403705.
  • Hansen LJJ, Daoussi R, Vervaet C, Remon J-P, De Beer TRM. Freeze-drying of live virus vaccines: A review. Vaccine. 2015;33(42):5507–19. doi:10.1016/j.vaccine.2015.08.085.
  • Naik SP, Zade JK, Sabale RN, Pisal SS, Menon R, Bankar SG, Gairola S, Dhere RM. Stability of heat stable, live attenuated Rotavirus vaccine (ROTASIIL®). Vaccine. 2017;35(22):2962–69. doi:10.1016/j.vaccine.2017.04.025.
  • World Health Organization. What is VVM and how does it work? [Accessed February 24, 2020]. https://www.who.int/immunization_standards/vaccine_quality/What%20is%20VVM%20and%20how%20does%20it%20work.pdf?ua=1.
  • Serum Institute of India Pvt. Ltd. Rotavirus vaccine live attenuated, oral (Freeze-Dried). Serum Institute of India Pvt. Ltd. [Accessed March 31, 2020]. https://www.seruminstitute.com/product_viral_rotasiil.php.
  • Kawade A, Babji S, Kamath V, Raut A, Kumar CM, Kundu R, Venkatramanan P, Lalwani SK, Bavdekar A, Juvekar S, et al. Immunogenicity and lot-to-lot consistency of a ready to use liquid bovine-human reassortant pentavalent rotavirus vaccine (ROTASIIL - Liquid) in Indian infants. Vaccine. 2019;37(19):2554–60. doi:10.1016/j.vaccine.2019.03.067.
  • Anil K, Desai S, Bhamare C, Dharmadhikari A, Madhusudhan RL, Patel J, Kulkarni PS. Safety and tolerability of a liquid bovine rotavirus pentavalent vaccine (LBRV-PV) in adults. Vaccine. 2018;36(12):1542–44. doi:10.1016/j.vaccine.2018.02.024.
  • ClinicalTrials.gov. Identifier NCT01375907, Safety study of a rotavirus vaccine (Rotavin-M1) among healthy adults. NIH U.S. National Library of Medicine. [Published July 4, 2016; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT01375907.
  • ClinicalTrials.gov. Identifier NCT01377571, A dose-escalating study to evaluate the immunogenicity and safety of Rotavin-M1 vaccine in healthy infants. NIH U.S. National Library of Medicine. [Published July 4, 2016; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT01377571.
  • ClinicalTrials.gov. Identifier NCT01502969, safety and immunogenicity assessment of Rotavin-M1 in vietnamese children. NIH U.S. National Library of Medicine. [published January 2, 2012; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT01502969.
  • Bhandari N, Sharma P, Glass RI, Ray P, Greenberg H, Taneja S, Saksena M, Rao CD, Gentsch JR, Parashar U, et al. Safety and immunogenicity of two live attenuated human rotavirus vaccine candidates, 116E and I321, in infants: results of a randomised controlled trial. Vaccine. 2006;24(31–32):5817–23. doi:10.1016/j.vaccine.2006.05.001.
  • Bhandari N, Sharma P, Taneja S, Kumar T, Rongsen‐Chandola T, Appaiahgari M, Mishra A, Singh S, Vrati S. A Dose-Escalation Safety and Immunogenicity Study of Live Attenuated Oral Rotavirus Vaccine 116E in Infants: A Randomized, Double-Blind, Placebo‐Controlled Trial. J Infect Dis. 2009;200(3):421–29. doi:10.1086/600104.
  • Appaiahgari MB, Glass R, Singh S, Taneja S, Rongsen-Chandola T, Bhandari N, Mishra S, Vrati S. Transplacental rotavirus IgG interferes with immune response to live oral rotavirus vaccine ORV-116E in Indian infants. Vaccine. 2014;32(6):651–56. doi:10.1016/j.vaccine.2013.12.017.
  • ClinicalTrials.gov. Identifier NCT03602053, Study of BBIL’s ROTAVAC® and ROTAVAC 5CM Vaccines in Zambia. NIH U.S. National Library of Medicine. [Published February 20, 2020; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT03602053.
  • ClinicalTrials.gov. Identifier NCT01305109, A Phase III clinical trial to evaluate the protective efficacy of three doses of oral rotavirus vaccine (ORV) 116E. NIH U.S. National Library of Medicine. Ppublished June 12, 2012; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT01305109.
  • Bhandari N, Rongsen-Chandola T, Bavdekar A, John J, Antony K, Taneja S, Goyal N, Kawade A, Kang G, Rathore SS, et al. Efficacy of a monovalent human-bovine (116E) rotavirus vaccine in Indian children in the second year of life. Vaccine. 2014;32:A110–A116. doi:10.1016/j.vaccine.2014.04.079.
  • ClinicalTrials.gov. Evaluate immunogenicity, safety, and reactogenicity of Rotavac® in healthy infants aged between 6-8 weeks in Vietnam - full text view - ClinicalTrials.gov. NIH U.S. National Library of Medicine. [Published March 25, 2019; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT03367559.
  • Chandola TR, Taneja S, Goyal N, Antony K, Bhatia K, More D, Bhandari N, Cho I, Mohan K, Prasad S, et al. ROTAVAC® ROTAVAC ® does not interfere with the immune response to childhood vaccines in Indian infants: A randomized placebo controlled trial. Heliyon. 2017;3(5):e00302. doi:10.1016/j.heliyon.2017.e00302.
  • Praharaj I, Platts-Mills JA, Taneja S, Antony K, Yuhas K, Flores J, Cho I, Bhandari N, Revathy R, Bavdekar A, et al. Diarrheal Etiology and Impact of Coinfections on Rotavirus Vaccine Efficacy Estimates in a Clinical Trial of a Monovalent Human–Bovine (116E) Oral Rotavirus Vaccine, Rotavac, India. Clin Infect Dis. 2019;69(2):243–50. doi:10.1093/cid/ciy896.
  • Ella R, Bobba R, Muralidhar S, Babji S, Vadrevu KM, Bhan MK, Phase A. 4, multicentre, randomized, single-blind clinical trial to evaluate the immunogenicity of the live, attenuated, oral rotavirus vaccine (116E), ROTAVAC®, administered simultaneously with or without the buffering agent in healthy infants in India. Hum Vaccines Immunother. 2018;14(7):1791–99. doi:10.1080/21645515.2018.1450709.
  • Ella R, Babji S, Ciarlet M, Blackwelder WC, Vadrevu KM. A randomized, open-labelled, non-inferiority phase 4 clinical trial to evaluate the immunogenicity and safety of the live, attenuated, oral rotavirus vaccine, ROTAVAC® in comparison with a licensed rotavirus vaccine in healthy infants. Vaccine. 2019;37(31):4407–13. doi:10.1016/j.vaccine.2019.05.069.
  • Zade JK, Kulkarni PS, Desai SA, Sabale RN, Naik SP, Dhere RM. Bovine rotavirus pentavalent vaccine development in India. Vaccine. 2014;32(Suppl 1):A124–128. doi:10.1016/j.vaccine.2014.03.003.
  • ClinicalTrials.gov. Identifier NCT03474055, study on liquid bovine rotavirus pentavalent vaccine (LBRV-PV) to evaluate lot-to-lot consistency and to compare non-inferiority with ROTASIIL (Lyophilized BRV-PV) in healthy infants in India. NIH U.S. National Library of Medicine. [Published September 10, 2018; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT03474055.
  • ClinicalTrials.gov. Identifier NCT02133690, a clinical trial to study the effect and safety of rotavirus vaccine against severe rotavirus gastroenteritis in healthy Indian infants. NIH U.S. National Library of Medicine. [Published September 10, 2018; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT02133690.
  • ClinicalTrials.gov. Identifier NCT02145000, efficacy and safety of a pentavalent rotavirus vaccine (BRV-PV) against severe rotavirus gastroenteritis in Niger. NIH U.S. National Library of Medicine. [Published August 14, 2019; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT02145000.
  • Isanaka S, Guindo O, Langendorf C, Matar Seck A, Plikaytis BD, Sayinzoga-Makombe N, McNeal MM, Meyer N, Adehossi E, Djibo A, et al. Efficacy of a Low-Cost, Heat-Stable Oral Rotavirus Vaccine in Niger. N Engl J Med. 2017;376(12):1121–30. doi:10.1056/NEJMoa1609462.
  • Coldiron ME, Guindo O, Makarimi R, Soumana I, Matar Seck A, Garba S, Macher E, Isanaka S, Grais RF. Safety of a heat-stable rotavirus vaccine among children in Niger: data from a phase 3, randomized, double-blind, placebo-controlled trial. Vaccine. 2018;36(25):3674–80. doi:10.1016/j.vaccine.2018.05.023.
  • ClinicalTrials.gov. Identifier NCT02584816, Phase III study on rotavirus vaccine to evaluate lot-to-lot consistency and interference with routine UIP immunization. NIH U.S. National Library of Medicine. [Published September 11, 2018; accessed March 31, 2020]. https://clinicaltrials.gov/ct2/show/NCT02584816.
  • Desai S, Rathi N, Kawade A, Venkatramanan P, Kundu R, Lalwani SK, Dubey AP, Venkateswara Rao J, Narayanappa D, Ghildiyal R, et al. Non-interference of Bovine-Human reassortant pentavalent rotavirus vaccine ROTASIIL® with the immunogenicity of infant vaccines in comparison with a licensed rotavirus vaccine. Vaccine. 2018;36(37):5519–23. doi:10.1016/j.vaccine.2018.07.064.
  • Le LT, Nguyen TV, Nguyen PM, Huong NT, Huong NT, Huong NTM, Hanh TB, Ha DN, Anh DD, Gentsch JR, et al. Development and characterization of candidate rotavirus vaccine strains derived from children with diarrhoea in Vietnam. Vaccine. 2009;27:F130–F138. doi:10.1016/j.vaccine.2009.08.086.
  • Almalki SSR. Circulating rotavirus G and P strains post rotavirus vaccination in Eastern Mediterranean Region. Saudi Med J. 2018;39(8):755–56. doi:10.15537/smj.2018.6.21394.
  • Dóró R, László B, Martella V, Leshem E, Gentsch J, Parashar U, Bányai K. Review of global rotavirus strain prevalence data from six years post vaccine licensure surveillance: is there evidence of strain selection from vaccine pressure? Infect Genet Evol. 2014;28:446–61. doi:10.1016/j.meegid.2014.08.017.
  • Steele AD, Neuzil KM, Cunliffe NA, Madhi SA, Bos P, Ngwira B, Witte D, Todd S, Louw C, Kirsten M, et al. Human rotavirus vaccine RotarixTM provides protection against diverse circulating rotavirus strains in African infants: a randomized controlled trial. BMC Infect Dis. 2012;12:213. doi:10.1186/1471-2334-12-213.
  • Payne DC, Boom JA, Staat MA, Edwards KM, Szilagyi PG, Klein EJ, Selvarangan R, Azimi PH, Harrison C, Moffatt M, et al. Effectiveness of pentavalent and monovalent rotavirus vaccines in concurrent use among US children <5 years of age, 2009-2011. Clin Infect Dis Off Publ Infect Dis Soc Am. 2013;57(1):13–20. doi:10.1093/cid/cit164.
  • Soares-Weiser K Rotavirus Vaccines Schedules: A systematic review of safety and efficacy from randomized controlled trials and observational studies of childhood schedules using RV1 and RV5 vaccines:140.
  • Pecenka C, Debellut F, Bar-Zeev N, Bhandari N, Rongsen-Chandola T, Bavdekar A, John J, Antony K, Taneja S, Goyal N. Re-evaluating the cost and cost-effectiveness of rotavirus vaccination in Bangladesh, Ghana, and Malawi: A comparison of three rotavirus vaccines. Vaccine. 2018;36(49):7472–78. doi:10.1016/j.vaccine.2018.10.068.
  • Debellut F, Jaber S, Bouzya Y, Sabbah J, Barham M, Abu-Awwad F, Hjaija D, Ramlawi A, Pecenka C, Clark A, et al. Introduction of rotavirus vaccination in Palestine: an evaluation of the costs, impact, and cost-effectiveness of ROTARIX and ROTAVAC. PLoS ONE. 2020;15(2):2. doi:10.1371/journal.pone.0228506.
  • Pecenka C, Debellut F, Bar-Zeev N, Anwari P, Nonvignon J, Clark A. Cost-effectiveness analysis for rotavirus vaccine decision-making: how can we best inform evolving and complex choices in vaccine product selection? Vaccine. 2020;38(6):1277–79. doi:10.1016/j.vaccine.2019.12.014.
  • World Health Organization. WHO programmatic guidance note use of more than one rotavirus vaccine product to complete the vaccination series. [Accessed February 25, 2020]. https://www.who.int/immunization/diseases/rotavirus/Rota_mixed_schedules_interchangeability.pdf?ua=1.
  • Libster R, McNeal M, Walter EB, Shane AL, Winokur P, Cress G, Berry AA, Kotloff KL, Sarpong K, Turley CB, et al. Safety and Immunogenicity of Sequential Rotavirus Vaccine Schedules. Pediatrics. 2016;137(2):e20152603. doi:10.1542/peds.2015-2603.
  • Payne DC, Sulemana I, Parashar UD. Evaluation of effectiveness of mixed rotavirus vaccine course for rotavirus gastroenteritis. JAMA Pediatr. 2016;170(7):708–10. doi:10.1001/jamapediatrics.2016.0014.
  • Kanungo S, Datta S Rotavirus vaccine interchangeability trial begins enrolling participants in Kolkata, India. [accessed March 22, 2020]. http://www.niced.org.in/news/documents/2019/RVICS-trail-23Mar2019.pdf.
  • Bharat Biotech. Bharat biotech launches ROTAVAC 5D® - Lowest dose volume rotavirus vaccine in the world. [accessed April 3, 2020]. https://www.bharatbiotech.com/images/press/Bharat-biotech-launches-ROTAVAC5D-lowest-dose-volume-rotavirus-vaccine-in-the-world.pdf.
  • UNICEF. Rotavirus vaccine: supply and demand update. [published Nov 2018; accessed March 9, 2020]. https://www.unicef.org/supply/files/RV_Supply_Update.pdf.
  • World Health Organization. WHO prequalified vaccines: rotasiil. WHO prequalified vaccines. [published Feb 25, 2020; accessed Feb 24, 2020]. https://extranet.who.int/gavi/PQ_Web/PreviewVaccine.aspx?nav=0&ID=323.
  • World Health Organization. WHO prequalified vaccines: rotavac. WHO Prequalified Vaccines. [published Feb 25, 2020; accessed Feb 24, 2020]. https://extranet.who.int/gavi/PQ_Web/PreviewVaccine.aspx?nav=0&ID=314.
  • Bharat Biotech. ROTAVAC multi dose product insert. [published March 2018; accessed April 5, 2020]. https://www.bharatbiotech.com/images/rotavac/Rotavac-multi-dose-package-Insert.pdf.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.