Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Rafael De La Barrera is active.

Publication


Featured researches published by Rafael De La Barrera.


Nature | 2016

Vaccine protection against Zika virus from Brazil

Rafael A. Larocca; Peter Abbink; Jean Pierre Schatzmann Peron; Paolo Marinho de Andrade Zanotto; M. Justin Iampietro; Alexander Badamchi-Zadeh; Michael Boyd; David Ng’ang’a; Marinela Kirilova; Ramya Nityanandam; Noe B. Mercado; Zhenfeng Li; Edward T. Moseley; Christine A. Bricault; Erica N. Borducchi; Patricia B. Giglio; David Jetton; George H. Neubauer; Joseph P. Nkolola; Lori F. Maxfield; Rafael De La Barrera; Richard G. Jarman; Kenneth H. Eckels; Nelson L. Michael; Stephen J. Thomas; Dan H. Barouch

Zika virus (ZIKV) is a flavivirus that is responsible for the current epidemic in Brazil and the Americas. ZIKV has been causally associated with fetal microcephaly, intrauterine growth restriction, and other birth defects in both humans and mice. The rapid development of a safe and effective ZIKV vaccine is a global health priority, but very little is currently known about ZIKV immunology and mechanisms of immune protection. Here we show that a single immunization with a plasmid DNA vaccine or a purified inactivated virus vaccine provides complete protection in susceptible mice against challenge with a strain of ZIKV involved in the outbreak in northeast Brazil. This ZIKV strain has recently been shown to cross the placenta and to induce fetal microcephaly and other congenital malformations in mice. We produced DNA vaccines expressing ZIKV pre-membrane and envelope (prM-Env), as well as a series of deletion mutants. The prM-Env DNA vaccine, but not the deletion mutants, afforded complete protection against ZIKV, as measured by absence of detectable viraemia following challenge, and protective efficacy correlated with Env-specific antibody titers. Adoptive transfer of purified IgG from vaccinated mice conferred passive protection, and depletion of CD4 and CD8 T lymphocytes in vaccinated mice did not abrogate this protection. These data demonstrate that protection against ZIKV challenge can be achieved by single-shot subunit and inactivated virus vaccines in mice and that Env-specific antibody titers represent key immunologic correlates of protection. Our findings suggest that the development of a ZIKV vaccine for humans is likely to be achievable.


Science | 2016

Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys

Peter Abbink; Rafael A. Larocca; Rafael De La Barrera; Christine A. Bricault; Edward T. Moseley; Michael Boyd; Marinela Kirilova; Zhenfeng Li; David Ng’ang’a; Ovini Nanayakkara; Ramya Nityanandam; Noe B. Mercado; Erica N. Borducchi; Arshi Agarwal; Amanda L. Brinkman; Crystal Cabral; Abishek Chandrashekar; Patricia B. Giglio; David Jetton; Jessica Jimenez; Benjamin C. Lee; Shanell Mojta; Katherine Molloy; Mayuri Shetty; George H. Neubauer; Kathryn E. Stephenson; Jean Pierre Schatzmann Peron; Paolo Marinho de Andrade Zanotto; Johnathan Misamore; Brad Finneyfrock

Zika virus (ZIKV) is responsible for a major ongoing epidemic in the Americas and has been causally associated with fetal microcephaly. The development of a safe and effective ZIKV vaccine is therefore an urgent global health priority. Here we demonstrate that three different vaccine platforms protect against ZIKV challenge in rhesus monkeys. A purified inactivated virus vaccine induced ZIKV-specific neutralizing antibodies and completely protected monkeys against ZIKV strains from both Brazil and Puerto Rico. Purified immunoglobulin from vaccinated monkeys also conferred passive protection in adoptive transfer studies. A plasmid DNA vaccine and a single-shot recombinant rhesus adenovirus serotype 52 vector vaccine, both expressing ZIKV premembrane and envelope, also elicited neutralizing antibodies and completely protected monkeys against ZIKV challenge. These data support the rapid clinical development of ZIKV vaccines for humans.


Science | 2016

Rapid development of a DNA vaccine for Zika virus

Kimberly A. Dowd; Sung-Youl Ko; Kaitlyn M. Morabito; Eun Sung Yang; Rebecca S. Pelc; Christina R. DeMaso; Leda R. Castilho; Peter Abbink; Michael Boyd; Ramya Nityanandam; David N. Gordon; John R. Gallagher; Xuejun Chen; John-Paul Todd; Yaroslav Tsybovsky; Audray K. Harris; Yan-Jang S. Huang; Stephen Higgs; Dana L. Vanlandingham; Hanne Andersen; Mark G. Lewis; Rafael De La Barrera; Kenneth H. Eckels; Richard G. Jarman; Martha Nason; Dan H. Barouch; Mario Roederer; Wing-Pui Kong; John R. Mascola; Theodore C. Pierson

A DNA vaccine candidate for Zika The ongoing Zika epidemic in the Americas and the Caribbean urgently needs a protective vaccine. Two DNA vaccines composed of the genes that encode the structural premembrane and envelope proteins of Zika virus have been tested in monkeys. Dowd et al. show that two doses of vaccine given intramuscularly completely protected 17 of 18 animals against Zika virus challenge. A single low dose of vaccine was not protective but did reduce viral loads. Protection correlated with serum antibody neutralizing activity. Phase I clinical trials testing these vaccines are already ongoing. Science, this issue p. 237 DNA-vaccine–induced neutralizing antibodies largely protect monkeys after experimental challenge by virus infection. Zika virus (ZIKV) was identified as a cause of congenital disease during the explosive outbreak in the Americas and Caribbean that began in 2015. Because of the ongoing fetal risk from endemic disease and travel-related exposures, a vaccine to prevent viremia in women of childbearing age and their partners is imperative. We found that vaccination with DNA expressing the premembrane and envelope proteins of ZIKV was immunogenic in mice and nonhuman primates, and protection against viremia after ZIKV challenge correlated with serum neutralizing activity. These data not only indicate that DNA vaccination could be a successful approach to protect against ZIKV infection, but also suggest a protective threshold of vaccine-induced neutralizing activity that prevents viremia after acute infection.


American Journal of Tropical Medicine and Hygiene | 2008

Safety and Immunogenicity of a Tetravalent Live-attenuated Dengue Vaccine in Flavivirus Naive Children

Sriluck Simasathien; Stephen J. Thomas; Veerachai Watanaveeradej; Ananda Nisalak; Célia Barberousse; Bruce L. Innis; Wellington Sun; J. Robert Putnak; Kenneth H. Eckels; Yanee Hutagalung; Robert V. Gibbons; Chunlin Zhang; Rafael De La Barrera; Richard G. Jarman; Wipa Chawachalasai; Mammen P. Mammen

A Phase I/II observer-blind, randomized, controlled trial evaluated the safety and immunogenicity of a dengue virus (DENV) vaccine candidate in healthy Thai infants (aged 12-15 months) without measurable pre-vaccination neutralizing antibodies to DENV and Japanese encephalitis virus. Fifty-one subjects received two doses of either DENV (N = 34; four received 1/10th dose) or control vaccine (N = 17; dose 1, live varicella; dose 2, Haemophilus influenzae type b). After each vaccine dose, adverse events (AEs) were solicited for 21 days, and non-serious AEs were solicited for 30 days; serious AEs (SAEs) were recorded throughout the study. Laboratory safety assessments were performed at 10 and 30 days; neutralizing antibodies were measured at 30 days. The DENV vaccine was well-tolerated without any related SAEs. After the second dose, 85.7% of full-dose DENV vaccinees developed at least trivalent and 53.6% developed tetravalent neutralizing antibodies ≥ 1:10 to DENV (control group = 0%). This vaccine candidate, therefore, warrants continued development in this age group (NCT00322049; clinicaltrials.gov).


American Journal of Tropical Medicine and Hygiene | 2013

A Phase II, Randomized, Safety and Immunogenicity Study of a Re-Derived, Live-Attenuated Dengue Virus Vaccine in Healthy Adults

Stephen J. Thomas; Kenneth H. Eckels; Isabelle Carletti; Rafael De La Barrera; Francis Dessy; Stefan Fernandez; Robert Putnak; Jean-François Toussaint; Wellington Sun; Kristen Bauer; Robert V. Gibbons; Bruce L. Innis

Two formulations of a new live tetravalent dengue virus (DENV) vaccine produced using re-derived master seeds from a precursor vaccine and that same precursor vaccine as a control were compared in a placebo-controlled, randomized, observer-blind, phase II trial of 86 healthy adults. Two vaccine doses were administered 6 months apart; a third dose was offered to a subset. Symptoms and signs of dengue-like illness reported after vaccination were mild to moderate, transient, and occurred with similar frequency among recipients of the new DENV vaccine and placebo, except for rash. Neither dengue nor vaccine-related serious adverse events were reported. The first DENV vaccine dose was moderately immunogenic; the second dose increased the potency and breadth of the neutralizing antibody response. Tetravalent response rates to the new formulations were 60% and 66.7% in unprimed subjects. A third dose did not increase tetravalent antibody rates. The new DENV vaccine candidates merit additional evaluation.


Cell | 2017

Zika Virus Persistence in the Central Nervous System and Lymph Nodes of Rhesus Monkeys

Malika Aid; Peter Abbink; Rafael A. Larocca; Michael Boyd; Ramya Nityanandam; Ovini Nanayakkara; Amanda J. Martinot; Edward T. Moseley; Eryn Blass; Erica N. Borducchi; Abishek Chandrashekar; Amanda L. Brinkman; Katherine Molloy; David Jetton; Lawrence J. Tartaglia; Jinyan Liu; Katharine Best; Alan S. Perelson; Rafael De La Barrera; Mark G. Lewis; Dan H. Barouch

Zika virus (ZIKV) is associated with severe neuropathology in neonates as well as Guillain-Barré syndrome and other neurologic disorders in adults. Prolonged viral shedding has been reported in semen, suggesting the presence of anatomic viral reservoirs. Here we show that ZIKV can persist in cerebrospinal fluid (CSF) and lymph nodes (LN) of infected rhesus monkeys for weeks after virus has been cleared from peripheral blood, urine, and mucosal secretions. ZIKV-specific neutralizing antibodies correlated with rapid clearance of virus in peripheral blood but remained undetectable in CSF for the duration of the study. Viral persistence in both CSF and LN correlated with upregulation of mechanistic target of rapamycin (mTOR), proinflammatory, and anti-apoptotic signaling pathways, as well as downregulation of extracellular matrix signaling pathways. These data raise the possibility that persistent or occult neurologic and lymphoid disease may occur following clearance of peripheral virus in ZIKV-infected individuals.


PLOS Pathogens | 2017

Impact of prior flavivirus immunity on Zika virus infection in rhesus macaques

Michael K. McCracken; Gregory D. Gromowski; Heather Friberg; Xiaoxu Lin; Peter Abbink; Rafael De La Barrera; Kenneth H. Eckles; Lindsey S Garver; Michael Boyd; David Jetton; Dan H. Barouch; Matthew C. Wise; Bridget S. Lewis; Jeffrey R. Currier; Kayvon Modjarrad; Mark Milazzo; Michelle Liu; Anna B. Mullins; J. Robert Putnak; Nelson L. Michael; Richard G. Jarman; Stephen J. Thomas

Studies have demonstrated cross-reactivity of anti-dengue virus (DENV) antibodies in human sera against Zika virus (ZIKV), promoting increased ZIKV infection in vitro. However, the correlation between in vitro and in vivo findings is not well characterized. Thus, we evaluated the impact of heterotypic flavivirus immunity on ZIKV titers in biofluids of rhesus macaques. Animals previously infected (≥420 days) with DENV2, DENV4, or yellow fever virus were compared to flavivirus-naïve animals following infection with a Brazilian ZIKV strain. Sera from DENV-immune macaques demonstrated cross-reactivity with ZIKV by antibody-binding and neutralization assays prior to ZIKV infection, and promoted increased ZIKV infection in cell culture assays. Despite these findings, no significant differences between flavivirus-naïve and immune animals were observed in viral titers, neutralizing antibody levels, or immune cell kinetics following ZIKV infection. These results indicate that prior infection with heterologous flaviviruses neither conferred protection nor increased observed ZIKV titers in this non-human primate ZIKV infection model.


The Lancet | 2017

Preliminary aggregate safety and immunogenicity results from three trials of a purified inactivated Zika virus vaccine candidate: phase 1, randomised, double-blind, placebo-controlled clinical trials

Kayvon Modjarrad; Leyi Lin; Sarah L. George; Kathryn E. Stephenson; Kenneth H. Eckels; Rafael De La Barrera; Richard G. Jarman; Erica Sondergaard; Janice Tennant; Jessica L Ansel; Kristin Mills; Michael Koren; Merlin L. Robb; Jill Barrett; Jason Thompson; Alison E Kosel; Peter Dawson; Andrew J. Hale; C Sabrina Tan; Stephen R. Walsh; Keith Meyer; James D. Brien; Trevor A Crowell; Azra Blazevic; Karla Mosby; Rafael A. Larocca; Peter Abbink; Michael Boyd; Christine A. Bricault; Michael S. Seaman

BACKGROUND A safe, effective, and rapidly scalable vaccine against Zika virus infection is needed. We developed a purified formalin-inactivated Zika virus vaccine (ZPIV) candidate that showed protection in mice and non-human primates against viraemia after Zika virus challenge. Here we present the preliminary results in human beings. METHODS We did three phase 1, placebo-controlled, double-blind trials of ZPIV with aluminium hydroxide adjuvant. In all three studies, healthy adults were randomly assigned by a computer-generated list to receive 5 μg ZPIV or saline placebo, in a ratio of 4:1 at Walter Reed Army Institute of Research, Silver Spring, MD, USA, or of 5:1 at Saint Louis University, Saint Louis, MO, USA, and Beth Israel Deaconess Medical Center, Boston, MA, USA. Vaccinations were given intramuscularly on days 1 and 29. The primary objective was safety and immunogenicity of the ZPIV candidate. We recorded adverse events and Zika virus envelope microneutralisation titres up to day 57. These trials are registered at ClinicalTrials.gov, numbers NCT02963909, NCT02952833, and NCT02937233. FINDINGS We enrolled 68 participants between Nov 7, 2016, and Jan 25, 2017. One was excluded and 67 participants received two injections of Zika vaccine (n=55) or placebo (n=12). The vaccine caused only mild to moderate adverse events. The most frequent local effects were pain (n=40 [60%]) or tenderness (n=32 [47%]) at the injection site, and the most frequent systemic reactogenic events were fatigue (29 [43%]), headache (26 [39%]), and malaise (15 [22%]). By day 57, 52 (92%) of vaccine recipients had seroconverted (microneutralisation titre ≥1:10), with peak geometric mean titres seen at day 43 and exceeding protective thresholds seen in animal studies. INTERPRETATION The ZPIV candidate was well tolerated and elicited robust neutralising antibody titres in healthy adults. FUNDING Departments of the Army and Defense and National Institute of Allergy and Infectious Diseases.


American Journal of Tropical Medicine and Hygiene | 2015

An Adjuvanted, Tetravalent Dengue Virus-Purified Inactivated Vaccine Candidate Induces Long-Lasting and Protective Antibody Responses Against Dengue Challenge in Rhesus Macaques

Stefan Fernandez; Stephen J. Thomas; Rafael De La Barrera; Rawiwan Imerbsin; Richard G. Jarman; Benoı̂t Baras; Jean-François Toussaint; Sally Mossman; Bruce L. Innis; Alexander C. Schmidt; Marie-Pierre Malice; Pascale Festraets; Lucile Warter; J. Robert Putnak; Kenneth H. Eckels

The immunogenicity and protective efficacy of a candidate tetravalent dengue virus purified inactivated vaccine (TDENV PIV) formulated with alum or an Adjuvant System (AS01, AS03 tested at three different dose levels, or AS04) was evaluated in a 0, 1-month vaccination schedule in rhesus macaques. One month after dose 2, all adjuvanted formulations elicited robust and persisting neutralizing antibody titers against all four dengue virus serotypes. Most of the formulations tested prevented viremia after challenge, with the dengue serotype 1 and 2 virus strains administered at 40 and 32 weeks post-dose 2, respectively. This study shows that inactivated dengue vaccines, when formulated with alum or an Adjuvant System, are candidates for further development.


Science Translational Medicine | 2017

Durability and correlates of vaccine protection against Zika virus in rhesus monkeys

Peter Abbink; Rafael A. Larocca; Kittipos Visitsunthorn; Michael Boyd; Rafael De La Barrera; Gregory D. Gromowski; Marinela Kirilova; Rebecca Peterson; Zhenfeng Li; Ovini Nanayakkara; Ramya Nityanandam; Noe B. Mercado; Erica N. Borducchi; Abishek Chandrashekar; David Jetton; Shanell Mojta; Priya Gandhi; Jake LeSuer; Shreeya Khatiwada; Mark G. Lewis; Kayvon Modjarrad; Richard G. Jarman; Kenneth H. Eckels; Stephen J. Thomas; Nelson L. Michael; Dan H. Barouch

Not all vaccines afford robust protection against ZIKV challenge in rhesus monkeys at 1 year after vaccination. Patience pays off As an individual may not encounter the pathogen for years after they have been vaccinated, efficacious vaccines typically require induction of long-lasting immunity. Abbink and colleagues vaccinated nonhuman primates with one of several candidate Zika virus vaccines and then waited an entire year before conducting a viral challenge. These vaccines had all shown promising results in previous experiments with a more immediate challenge, but here, one vaccine faltered, likely due to waning antibodies. The researchers performed more experiments to suggest that circulating antibodies are mediating protection for these vaccines. These results are useful for Zika virus vaccine development and instructive for vaccine development in general. An effective Zika virus (ZIKV) vaccine will require long-term durable protection. Several ZIKV vaccine candidates have demonstrated protective efficacy in nonhuman primates, but these studies have typically involved ZIKV challenge shortly after vaccination at peak immunity. We show that a single immunization with an adenovirus vector–based vaccine, as well as two immunizations with a purified inactivated virus vaccine, afforded robust protection against ZIKV challenge in rhesus monkeys at 1 year after vaccination. In contrast, two immunizations with an optimized DNA vaccine, which provided complete protection at peak immunity, resulted in reduced protective efficacy at 1 year that was associated with declining neutralizing antibody titers to subprotective levels. These data define a microneutralization log titer of 2.0 to 2.1 as the threshold required for durable protection against ZIKV challenge in this model. Moreover, our findings demonstrate that protection against ZIKV challenge in rhesus monkeys is possible for at least 1 year with a single-shot vaccine.

Collaboration


Dive into the Rafael De La Barrera's collaboration.

Top Co-Authors

Avatar

Kenneth H. Eckels

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Stephen J. Thomas

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Richard G. Jarman

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Abbink

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michael Boyd

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dan H. Barouch

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rafael A. Larocca

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

David Jetton

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Erica N. Borducchi

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge