Network


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

Hotspot


Dive into the research topics where Erica N. Borducchi is active.

Publication


Featured researches published by Erica N. Borducchi.


Nature | 2013

Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys

Dan H. Barouch; James B. Whitney; Brian Moldt; Florian Klein; Thiago Y. Oliveira; Jinyan Liu; Kathryn E. Stephenson; Hui-Wen Chang; Karthik Shekhar; Sanjana Gupta; Joseph P. Nkolola; Michael S. Seaman; Kaitlin M. Smith; Erica N. Borducchi; Crystal Cabral; Jeffrey Y. Smith; Stephen Blackmore; Srisowmya Sanisetty; James R. Perry; Matthew Beck; Mark G. Lewis; William Rinaldi; Arup K. Chakraborty; Pascal Poignard; Michel C. Nussenzweig; Dennis R. Burton

Human immunodeficiency virus type 1 (HIV-1)-specific monoclonal antibodies with extraordinary potency and breadth have recently been described. In humanized mice, combinations of monoclonal antibodies have been shown to suppress viraemia, but the therapeutic potential of these monoclonal antibodies has not yet been evaluated in primates with an intact immune system. Here we show that administration of a cocktail of HIV-1-specific monoclonal antibodies, as well as the single glycan-dependent monoclonal antibody PGT121, resulted in a rapid and precipitous decline of plasma viraemia to undetectable levels in rhesus monkeys chronically infected with the pathogenic simian–human immunodeficiency virus SHIV-SF162P3. A single monoclonal antibody infusion afforded up to a 3.1 log decline of plasma viral RNA in 7 days and also reduced proviral DNA in peripheral blood, gastrointestinal mucosa and lymph nodes without the development of viral resistance. Moreover, after monoclonal antibody administration, host Gag-specific T-lymphocyte responses showed improved functionality. Virus rebounded in most animals after a median of 56 days when serum monoclonal antibody titres had declined to undetectable levels, although, notably, a subset of animals maintained long-term virological control in the absence of further monoclonal antibody infusions. These data demonstrate a profound therapeutic effect of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys as well as an impact on host immune responses. Our findings strongly encourage the investigation of monoclonal antibody therapy for HIV-1 in humans.


Nature | 2014

Rapid seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys

James B. Whitney; Alison L. Hill; Srisowmya Sanisetty; Pablo Penaloza-MacMaster; Jinyan Liu; Mayuri Shetty; Lily Parenteau; Crystal Cabral; Jennifer Shields; Stephen Blackmore; Jeffrey Y. Smith; Amanda L. Brinkman; Lauren Peter; Sheeba Mathew; Kaitlin M. Smith; Erica N. Borducchi; Daniel I. S. Rosenbloom; Mark G. Lewis; Jillian Hattersley; Bei Li; Joseph Hesselgesser; Romas Geleziunas; Merlin L. Robb; Jerome H. Kim; Nelson L. Michael; Dan H. Barouch

The viral reservoir represents a critical challenge for human immunodeficiency virus type 1 (HIV-1) eradication strategies. However, it remains unclear when and where the viral reservoir is seeded during acute infection and the extent to which it is susceptible to early antiretroviral therapy (ART). Here we show that the viral reservoir is seeded rapidly after mucosal simian immunodeficiency virus (SIV) infection of rhesus monkeys and before systemic viraemia. We initiated suppressive ART in groups of monkeys on days 3, 7, 10 and 14 after intrarectal SIVMAC251 infection. Treatment with ART on day 3 blocked the emergence of viral RNA and proviral DNA in peripheral blood and also substantially reduced levels of proviral DNA in lymph nodes and gastrointestinal mucosa as compared with treatment at later time points. In addition, treatment on day 3 abrogated the induction of SIV-specific humoral and cellular immune responses. Nevertheless, after discontinuation of ART following 24 weeks of fully suppressive therapy, virus rebounded in all animals, although the monkeys that were treated on day 3 exhibited a delayed viral rebound as compared with those treated on days 7, 10 and 14. The time to viral rebound correlated with total viraemia during acute infection and with proviral DNA at the time of ART discontinuation. These data demonstrate that the viral reservoir is seeded rapidly after intrarectal SIV infection of rhesus monkeys, during the ‘eclipse’ phase, and before detectable viraemia. This strikingly early seeding of the refractory viral reservoir raises important new challenges for HIV-1 eradication strategies.


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.


Cell | 2013

Protective Efficacy of a Global HIV-1 Mosaic Vaccine against Heterologous SHIV Challenges in Rhesus Monkeys

Dan H. Barouch; Kathryn E. Stephenson; Erica N. Borducchi; Kaitlin M. Smith; Kelly Stanley; Anna McNally; Jinyan Liu; Peter Abbink; Lori F. Maxfield; Michael S. Seaman; Anne-Sophie Dugast; Galit Alter; Melissa Ferguson; Wenjun Li; Patricia L. Earl; Bernard Moss; Elena E. Giorgi; James Szinger; Leigh Anne Eller; Erik Billings; Mangala Rao; Sodsai Tovanabutra; Eric Sanders-Buell; Mo Weijtens; Maria G. Pau; Hanneke Schuitemaker; Merlin L. Robb; Jerome H. Kim; Bette T. Korber; Nelson L. Michael

The global diversity of HIV-1 represents a critical challenge facing HIV-1 vaccine development. HIV-1 mosaic antigens are bioinformatically optimized immunogens designed for improved coverage of HIV-1 diversity. However, the protective efficacy of such global HIV-1 vaccine antigens has not previously been evaluated. Here, we demonstrate the capacity of bivalent HIV-1 mosaic antigens to protect rhesus monkeys against acquisition of infection following heterologous challenges with the difficult-to-neutralize simian-human immunodeficiency virus SHIV-SF162P3. Adenovirus/poxvirus and adenovirus/adenovirus vector-based vaccines expressing HIV-1 mosaic Env, Gag, and Pol afforded a significant reduction in the per-exposure acquisition risk following repetitive, intrarectal SHIV-SF162P3 challenges. Protection against acquisition of infection correlated with vaccine-elicited binding, neutralizing, and functional nonneutralizing antibodies, suggesting that the coordinated activity of multiple antibody functions may contribute to protection against difficult-to-neutralize viruses. These data demonstrate the protective efficacy of HIV-1 mosaic antigens and suggest a potential strategy for the development of a global HIV-1 vaccine. PAPERCLIP:


Science | 2015

Protective Efficacy of Adenovirus/Protein Vaccines Against SIV Challenges in Rhesus Monkeys

Dan H. Barouch; Galit Alter; Thomas A. Broge; Caitlyn Linde; Margaret E. Ackerman; Eric P. Brown; Erica N. Borducchi; Kaitlin M. Smith; Joseph P. Nkolola; Jinyan Liu; Jennifer Shields; Lily Parenteau; James B. Whitney; Peter Abbink; David Ng’ang’a; Michael S. Seaman; Christy L. Lavine; James R. Perry; Wenjun Li; Arnaud D. Colantonio; Mark G. Lewis; Bing Chen; Holger Wenschuh; Ulf Reimer; Michael Piatak; Jeffrey D. Lifson; Scott A. Handley; Herbert W. Virgin; Marguerite Koutsoukos; Clarisse Lorin

To defeat SIV, add a protein boost Despite 30 years of effort, no HIV-1 vaccine exists. Barouch et al. evaluated one promising strategy in rhesus macaques, a preclinical model commonly used to test potential HIV-1 vaccine candidates. They immunized monkeys with adenovirus-36 vectors engineered to express SIV (simian immunodeficiency virus) genes and then boosted them with a recombinant gp120 envelope glycoprotein (Env) from SIV. This regimen afforded greater protection than a strategy that instead used a viral vector–based boost. A parallel trial using a SHIV (simian/human immunodeficiency virus)–based vaccine and challenge model produced similar results. Whether this particular approach will be equally successful in humans remains to be tested. Science, this issue p. 320 A viral vector–recombinant envelope glycoprotein–based HIV-1 vaccine strategy protected 50% of monkeys from infection. Preclinical studies of viral vector–based HIV-1 vaccine candidates have previously shown partial protection against neutralization-resistant virus challenges in rhesus monkeys. In this study, we evaluated the protective efficacy of adenovirus serotype 26 (Ad26) vector priming followed by purified envelope (Env) glycoprotein boosting. Rhesus monkeys primed with Ad26 vectors expressing SIVsmE543 Env, Gag, and Pol and boosted with AS01B-adjuvanted SIVmac32H Env gp140 demonstrated complete protection in 50% of vaccinated animals against a series of repeated, heterologous, intrarectal SIVmac251 challenges that infected all controls. Protective efficacy correlated with the functionality of Env-specific antibody responses. Comparable protection was also observed with a similar Ad/Env vaccine against repeated, heterologous, intrarectal SHIV-SF162P3 challenges. These data demonstrate robust protection by Ad/Env vaccines against acquisition of neutralization-resistant virus challenges in rhesus monkeys.


Journal of Virology | 2012

Adenovirus Serotype 5 Neutralizing Antibodies Target both Hexon and Fiber following Vaccination and Natural Infection

Ritu R. Bradley; Diana M. Lynch; Mark J. Iampietro; Erica N. Borducchi; Dan H. Barouch

ABSTRACT The immunogenicity of adenovirus serotype 5 (Ad5) vectors has been shown to be suppressed by neutralizing antibodies (NAbs) directed primarily against the hexon hypervariable regions (HVRs). However, the role of NAbs directed against other capsid components, particularly the adenovirus fiber, remains unclear. Here we show that Ad5 NAbs target both hexon and fiber following vaccination and natural infection. Utilizing neutralization assays with capsid chimeric vectors, we observed that NAb responses to hexon appeared dominant and NAb responses against fiber were subdominant in sera from vaccinated mice, vaccinated humans, and naturally exposed humans. A novel chimeric Ad5 vector in which both the hexon HVRs and the fiber knob were exchanged nearly completely evaded Ad5-specific NAbs both in vitro and in vivo.


Science | 2016

Antibody-mediated protection against SHIV challenge includes systemic clearance of distal virus

Jinyan Liu; Khader Ghneim; Devin Sok; William J. Bosche; Yuan Li; Elizabeth Chipriano; Brian Berkemeier; Kelli Oswald; Erica N. Borducchi; Crystal Cabral; Lauren Peter; Amanda L. Brinkman; Mayuri Shetty; Jessica Jimenez; Jade Mondesir; Benjamin C. Lee; Patricia B. Giglio; Abishek Chandrashekar; Peter Abbink; Arnaud D. Colantonio; Courtney Gittens; Chantelle Baker; Wendeline Wagner; Mark G. Lewis; Wenjun Li; Rafick-Pierre Sekaly; Jeffrey D. Lifson; Dennis R. Burton; Dan H. Barouch

HIV-1–specific broadly neutralizing antibodies (bNAbs) can protect rhesus monkeys against simian-human immunodeficiency virus (SHIV) challenge. However, the site of antibody interception of virus and the mechanism of antibody-mediated protection remain unclear. We administered a fully protective dose of the bNAb PGT121 to rhesus monkeys and challenged them intravaginally with SHIV-SF162P3. In PGT121-treated animals, we detected low levels of viral RNA and viral DNA in distal tissues for seven days following challenge. Viral RNA–positive tissues showed transcriptomic changes indicative of innate immune activation, and cells from these tissues initiated infection after adoptive transfer into naïve hosts. These data demonstrate that bNAb-mediated protection against a mucosal virus challenge can involve clearance of infectious virus in distal tissues.


Journal of Virology | 2012

Adenovirus Serotype 5-Specific Neutralizing Antibodies Target Multiple Hexon Hypervariable Regions

Ritu R. Bradley; Lori F. Maxfield; Diana M. Lynch; Mark J. Iampietro; Erica N. Borducchi; Dan H. Barouch

ABSTRACT The immunogenicity of adenovirus serotype 5 (Ad5) vectors has been shown to be suppressed by neutralizing antibodies (NAbs) directed primarily against the hexon hypervariable regions (HVRs). We previously reported that replacing all seven HVRs with those from the rare serotype virus Ad48 resulted in a chimeric Ad5HVR48(1-7) vector that largely evaded preexisting Ad5 immunity in mice and rhesus monkeys. In this study, we evaluated the extent to which Ad5-specific NAbs are directed against various HVRs. We constructed partial HVR-chimeric Ad5 vectors with only a subset of HVRs exchanged, and we utilized these vectors in both NAb assays and murine immunogenicity studies with and without baseline Ad5 immunity. Our results demonstrate that Ad5-specific NAbs target multiple HVRs, suggesting that replacing all HVRs is required to optimize evasion of anti-Ad5 immunity. These data have important implications for the development of novel vectors for both vaccines and gene therapy.


Journal of Virology | 2013

Alternative serotype adenovirus vaccine vectors elicit memory T cells with enhanced anamnestic capacity compared to Ad5 vectors

Pablo Penaloza-MacMaster; Nicholas M. Provine; Joshua Ra; Erica N. Borducchi; Anna McNally; Nathaniel L. Simmons; Mark J. Iampietro; Dan H. Barouch

ABSTRACT The failure of the adenovirus serotype 5 (Ad5) vector-based human immunodeficiency virus type 1 (HIV-1) vaccine in the STEP study has led to the development of adenovirus vectors derived from alternative serotypes, such as Ad26, Ad35, and Ad48. We have recently demonstrated that vaccines using alternative-serotype Ad vectors confer partial protection against stringent simian immunodeficiency virus (SIV) challenges in rhesus monkeys. However, phenotypic differences between the T cell responses elicited by Ad5 and those of alternative-serotype Ad vectors remain unexplored. Here, we report the magnitude, phenotype, functionality, and recall capacity of memory T cell responses elicited in mice by Ad5, Ad26, Ad35, and Ad48 vectors expressing lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP). Our data demonstrate that memory T cells elicited by Ad5 vectors were high in magnitude but exhibited functional exhaustion and decreased anamnestic potential following secondary antigen challenge compared to Ad26, Ad35, and Ad48 vectors. These data suggest that vaccination with alternative-serotype Ad vectors offers substantial immunological advantages over Ad5 vectors, in addition to circumventing high baseline Ad5-specific neutralizing antibody titers.

Collaboration


Dive into the Erica N. Borducchi's collaboration.

Top Co-Authors

Avatar

Dan H. Barouch

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter Abbink

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

Nicholas M. Provine

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Pablo Penaloza-MacMaster

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kathryn E. Stephenson

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mark G. Lewis

Southern Research Institute

View shared research outputs
Top Co-Authors

Avatar

Jinyan Liu

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joseph P. Nkolola

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lily Parenteau

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge