Network


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

Hotspot


Dive into the research topics where Abigail B. Ventura is active.

Publication


Featured researches published by Abigail B. Ventura.


Nature | 2011

Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine

Scott G. Hansen; Julia C. Ford; Matthew S. Lewis; Abigail B. Ventura; Colette M. Hughes; Lia Coyne-Johnson; Nathan Whizin; Kelli Oswald; Rebecca Shoemaker; Tonya Swanson; Alfred W. Legasse; Maria J. Chiuchiolo; Christopher L. Parks; Michael K. Axthelm; Jay A. Nelson; Michael A. Jarvis; Michael Piatak; Jeffrey D. Lifson; Louis J. Picker

The acquired immunodeficiency syndrome (AIDS)-causing lentiviruses human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) effectively evade host immunity and, once established, infections with these viruses are only rarely controlled by immunological mechanisms. However, the initial establishment of infection in the first few days after mucosal exposure, before viral dissemination and massive replication, may be more vulnerable to immune control. Here we report that SIV vaccines that include rhesus cytomegalovirus (RhCMV) vectors establish indefinitely persistent, high-frequency, SIV-specific effector memory T-cell (TEM) responses at potential sites of SIV replication in rhesus macaques and stringently control highly pathogenic SIVMAC239 infection early after mucosal challenge. Thirteen of twenty-four rhesus macaques receiving either RhCMV vectors alone or RhCMV vectors followed by adenovirus 5 (Ad5) vectors (versus 0 of 9 DNA/Ad5-vaccinated rhesus macaques) manifested early complete control of SIV (undetectable plasma virus), and in twelve of these thirteen animals we observed long-term (≥1 year) protection. This was characterized by: occasional blips of plasma viraemia that ultimately waned; predominantly undetectable cell-associated viral load in blood and lymph node mononuclear cells; no depletion of effector-site CD4+ memory T cells; no induction or boosting of SIV Env-specific antibodies; and induction and then loss of T-cell responses to an SIV protein (Vif) not included in the RhCMV vectors. Protection correlated with the magnitude of the peak SIV-specific CD8+ T-cell responses in the vaccine phase, and occurred without anamnestic T-cell responses. Remarkably, long-term RhCMV vector-associated SIV control was insensitive to either CD8+ or CD4+ lymphocyte depletion and, at necropsy, cell-associated SIV was only occasionally measurable at the limit of detection with ultrasensitive assays, observations that indicate the possibility of eventual viral clearance. Thus, persistent vectors such as CMV and their associated TEM responses might significantly contribute to an efficacious HIV/AIDS vaccine.


Nature | 2013

Immune clearance of highly pathogenic SIV infection

Scott G. Hansen; Michael Piatak; Abigail B. Ventura; Colette M. Hughes; Roxanne M. Gilbride; Julia C. Ford; Kelli Oswald; Rebecca Shoemaker; Yuan Li; Matthew S. Lewis; Awbrey N. Gilliam; Guangwu Xu; Nathan Whizin; Benjamin J. Burwitz; Shannon L. Planer; John M. Turner; Alfred W. Legasse; Michael K. Axthelm; Jay A. Nelson; Klaus Früh; Jonah B. Sacha; Jacob D. Estes; Brandon F. Keele; Paul T. Edlefsen; Jeffrey D. Lifson; Louis J. Picker

Established infections with the human and simian immunodeficiency viruses (HIV and SIV, respectively) are thought to be permanent with even the most effective immune responses and antiretroviral therapies only able to control, but not clear, these infections. Whether the residual virus that maintains these infections is vulnerable to clearance is a question of central importance to the future management of millions of HIV-infected individuals. We recently reported that approximately 50% of rhesus macaques (RM; Macaca mulatta) vaccinated with SIV protein-expressing rhesus cytomegalovirus (RhCMV/SIV) vectors manifest durable, aviraemic control of infection with the highly pathogenic strain SIVmac239 (ref. 5). Here we show that regardless of the route of challenge, RhCMV/SIV vector-elicited immune responses control SIVmac239 after demonstrable lymphatic and haematogenous viral dissemination, and that replication-competent SIV persists in several sites for weeks to months. Over time, however, protected RM lost signs of SIV infection, showing a consistent lack of measurable plasma- or tissue-associated virus using ultrasensitive assays, and a loss of T-cell reactivity to SIV determinants not in the vaccine. Extensive ultrasensitive quantitative PCR and quantitative PCR with reverse transcription analyses of tissues from RhCMV/SIV vector-protected RM necropsied 69–172 weeks after challenge did not detect SIV RNA or DNA sequences above background levels, and replication-competent SIV was not detected in these RM by extensive co-culture analysis of tissues or by adoptive transfer of 60 million haematolymphoid cells to naive RM. These data provide compelling evidence for progressive clearance of a pathogenic lentiviral infection, and suggest that some lentiviral reservoirs may be susceptible to the continuous effector memory T-cell-mediated immune surveillance elicited and maintained by cytomegalovirus vectors.


Science | 2013

Cytomegalovirus vectors violate CD8+ T cell epitope recognition paradigms

Scott G. Hansen; Jonah B. Sacha; Colette M. Hughes; Julia C. Ford; Benjamin J. Burwitz; Isabel Scholz; Roxanne M. Gilbride; Matthew S. Lewis; Awbrey N. Gilliam; Abigail B. Ventura; Daniel Malouli; Guangwu Xu; Rebecca Richards; Nathan Whizin; Jason S. Reed; Katherine B. Hammond; Miranda Fischer; John M. Turner; Alfred W. Legasse; Michael K. Axthelm; Paul T. Edlefsen; Jay A. Nelson; Jeffrey D. Lifson; Klaus Früh; Louis J. Picker

Introduction CD8+ T cell responses focus on a small fraction of total pathogen-encoded peptides, which are similar among individuals with shared major histocompatibility complex (MHC) alleles. This focus can limit immune control of genetically flexible pathogens, such as HIV and SIV, because CD8+ T cells in most infected subjects do not target sequences required for pathogen fitness, resulting in viral escape. Although a vaccine capable of broadening or redirecting CD8+ T cell epitope targeting to prevent viral escape would be highly advantageous, it remains unclear whether this targeting can be diverted from its default pattern during priming. Fibroblast-adapted RhCMV/gag vectors elicit MHC class II–restricted CD8+ T cells, greatly expanding the breadth of the response. (Top) Differential inhibition of SIVgag-specific CD8+ T cells from SIV+, fibroblast-adapted RhCMV/gag vector–vaccinated, and tropism-repaired RhCMV/gag vector–vaccinated rhesus macaques by MHC-I versus MHC-II blockade. (Bottom) Responses to consecutive SIVgag 15mer peptides in the indicated animals, classified by sensitivity to MHC-I versus MHC-II blockade. Methods We used intracellular cytokine analysis to compare the epitope targeting of SIV-specific CD8+ T cell responses in rhesus macaques with controlled SIV infection or after vaccination with either conventional SIV vaccines or rhesus cytomegalovirus (RhCMV) vectors. RhCMV vectors have been associated with stringent control of SIV challenge in the absence of protective MHC alleles. Results Fibroblast-adapted RhCMV/SIV vectors elicited SIV-specific CD8+ T cells that failed to target any canonical epitopes associated with SIV infection or conventional SIV vaccination. Instead, they recognized distinct epitopes characterized by extraordinary breadth (greater than that of conventional vaccines by a factor of >3), MHC class II (MHC-II) restriction (63% of epitopes), and high promiscuity (epitopes common to most or all responses in vaccinated macaques). These unconventionally targeted CD8+ T cell responses recognized autologous SIV-infected cells, indicating that processing and presentation of the unconventional epitopes is CMV-independent. However, CMV gene expression was responsible for directing epitope specificity of CD8+ T cells during priming. The induction of canonical SIV epitope–specific CD8+ T cell responses was specifically suppressed by expression of the Rh189/US11 gene, and the promiscuous MHC-I– and MHC-II–restricted CD8+ T cell responses occurred only in the absence of the Rh157.4–.6/UL128–131 genes involved in CMV tropism for nonfibroblasts. Discussion These findings suggest that CD8+ T cell recognition is more flexible than had been thought, and that the focused epitope recognition profiles of conventional CD8+ T cell responses may be primarily restricted by immunoregulation during priming (which can be subverted by CMV) rather than by intrinsic limitations in antigen processing/presentation or in T cell receptor repertoire. The ability of CMVs with different genetic modifications to differentially elicit CD8+ T cell responses with divergent patterns of epitope recognition raises the possibility of a CMV vector–based vaccine platform with programmable CD8+ T cell epitope targeting, including vectors that can selectively elicit CD8+ T cell responses targeting conventional or unconventional epitopes. Because the latter would be unaffected by escape mutations arising during natural infection, these vectors would be well suited for therapeutic vaccine applications. CMV Breaks All the Rules One vaccine strategy being pursued against HIV is to generate protection that is dependent on cell-mediated, rather than humoral, immune responses. A cytomegalovirus (CMV)–vectored vaccine that expresses simian immunodeficiency virus (SIV) antigens exhibits stringent and durable viral control upon SIV challenge in approximately half of vaccinated rhesus macaques. Hansen et al. (10.1126/science.1237874, see the Perspective by Goonetilleke and McMichael) sought to determine the basis for the protection and discovered that the CD8+ T cell response in vaccinated monkeys does not target canonical SIV epitopes, which SIV is known to escape, but rather generates a broad, promiscuous response. A vaccine that uses one virus to deliver components of a second virus elicits T cells that recognize noncanonical epitopes. [Also see Perspective by Goonetilleke and McMichael] CD8+ T cell responses focus on a small fraction of pathogen- or vaccine-encoded peptides, and for some pathogens, these restricted recognition hierarchies limit the effectiveness of antipathogen immunity. We found that simian immunodeficiency virus (SIV) protein–expressing rhesus cytomegalovirus (RhCMV) vectors elicit SIV-specific CD8+ T cells that recognize unusual, diverse, and highly promiscuous epitopes, including dominant responses to epitopes restricted by class II major histocompatibility complex (MHC) molecules. Induction of canonical SIV epitope–specific CD8+ T cell responses is suppressed by the RhCMV-encoded Rh189 gene (corresponding to human CMV US11), and the promiscuous MHC class I– and class II–restricted CD8+ T cell responses occur only in the absence of the Rh157.5, Rh157.4, and Rh157.6 (human CMV UL128, UL130, and UL131) genes. Thus, CMV vectors can be genetically programmed to achieve distinct patterns of CD8+ T cell epitope recognition.


Science | 2010

Evasion of CD8+ T Cells Is Critical for Superinfection by Cytomegalovirus

Scott G. Hansen; Colin Powers; Rebecca Richards; Abigail B. Ventura; Julia C. Ford; Don C. Siess; Michael K. Axthelm; Jay A. Nelson; Michael A. Jarvis; Louis J. Picker; Klaus Früh

Cytomegalovirus Immune Evasion Strategy Cytomegalovirus (CMV) infects a large percentage of the worlds population. Most of those infected are asymptomatic; however, CMV is a substantial public health concern for immunocompromised individuals and neonates. CMV is unusual in that it can superinfect: it re-infects hosts who are already infected with the virus, even in the presence of a strong, specific immune response. Hansen et al. (p. 102; see the Perspective by Hengel and Koszinowski) now find that in rhesus macaques, a good model for human CMV superinfection, CMV establishes superinfections by evading the immune response mediated by CD8+ T cells. A series of viral mutants deficient in expression of the US2-11 glycoproteins, which regulate antigen presentation to CD8+ T cells, revealed that, although able to establish the initial infection, these viral mutants were unable to superinfect. Depletion of CD8+ T cells from the monkeys allowed infection by the mutant viruses. These results highlight the difficulties in developing an effective protective vaccine against CMV itself, but suggest that CMV-based vectors may be useful in other vaccine efforts such as those against HIV. Cytomegalovirus monkeys can reinfect an already-infected host by evading the CD8+ T cell–mediated immune response. Cytomegalovirus (CMV) can superinfect persistently infected hosts despite CMV-specific humoral and cellular immunity; however, how it does so remains undefined. We have demonstrated that superinfection of rhesus CMV–infected rhesus macaques (RM) requires evasion of CD8+ T cell immunity by virally encoded inhibitors of major histocompatibility complex class I (MHC-I) antigen presentation, particularly the homologs of human CMV US2, 3, 6, and 11. In contrast, MHC-I interference was dispensable for primary infection of RM, or for the establishment of a persistent secondary infection in CMV-infected RM transiently depleted of CD8+ lymphocytes. These findings demonstrate that US2-11 glycoproteins promote evasion of CD8+ T cells in vivo, thus supporting viral replication and dissemination during superinfection, a process that complicates the development of preventive CMV vaccines but that can be exploited for CMV-based vector development.


Science | 2016

Broadly targeted CD8+ T cell responses restricted by major histocompatibility complex-E

Scott G. Hansen; Helen L. Wu; Benjamin J. Burwitz; Colette M. Hughes; Katherine B. Hammond; Abigail B. Ventura; Jason S. Reed; Roxanne M. Gilbride; Emily Ainslie; David W. Morrow; Julia C. Ford; Andrea N. Selseth; Reesab Pathak; Daniel Malouli; Alfred W. Legasse; Michael K. Axthelm; Jay A. Nelson; Geraldine Gillespie; Lucy C. Walters; Simon Brackenridge; Hannah R. Sharpe; Cesar A. López; Klaus Früh; Bette T. Korber; Andrew J. McMichael; S. Gnanakaran; Jonah B. Sacha; Louis J. Picker

An unconventional route to protection One promising approach toward an HIV-1 vaccine involves infecting people with cytomegalovirus engineered to express proteins from HIV-1. This approach, which works by eliciting virus-killing CD8+ T cells, provides robust protection in nonhuman primate models. Hansen et al. have found out why this approach is so effective. Normally, peptide antigens presented by major histocompatibility complex-1a (MHC-Ia) activate CD8+ T cells. In vaccinated monkeys, however, CD8+ T cells reacted to peptide antigens presented by MHC-E molecules instead. Moreover, MHC-E could present a much wider range of peptides than MHC-Ia. Science, this issue p. 714 Nonclassical major histocompatibility complex E molecules can present highly diverse peptide epitopes to CD8+ T cells. Major histocompatibility complex E (MHC-E) is a highly conserved, ubiquitously expressed, nonclassical MHC class Ib molecule with limited polymorphism that is primarily involved in the regulation of natural killer (NK) cells. We found that vaccinating rhesus macaques with rhesus cytomegalovirus vectors in which genes Rh157.5 and Rh157.4 are deleted results in MHC-E–restricted presentation of highly varied peptide epitopes to CD8αβ+ T cells, at ~4 distinct epitopes per 100 amino acids in all tested antigens. Computational structural analysis revealed that MHC-E provides heterogeneous chemical environments for diverse side-chain interactions within a stable, open binding groove. Because MHC-E is up-regulated to evade NK cell activity in cells infected with HIV, simian immunodeficiency virus, and other persistent viruses, MHC-E–restricted CD8+ T cell responses have the potential to exploit pathogen immune-evasion adaptations, a capability that might endow these unconventional responses with superior efficacy.


Journal of Clinical Investigation | 2014

Cytomegalovirus pp65 limits dissemination but is dispensable for persistence

Daniel Malouli; Scott G. Hansen; Ernesto S. Nakayasu; Emily Marshall; Colette M. Hughes; Abigail B. Ventura; Roxanne M. Gilbride; Matthew S. Lewis; Guangwu Xu; Craig N. Kreklywich; Nathan Whizin; Miranda Fischer; Alfred W. Legasse; Kasinath Viswanathan; Don C. Siess; David G. Camp; Michael K. Axthelm; Christoph A. Kahl; Victor R. DeFilippis; Richard D. Smith; Daniel N. Streblow; Louis J. Picker; Klaus Früh

The most abundantly produced virion protein in human cytomegalovirus (HCMV) is the immunodominant phosphoprotein 65 (pp65), which is frequently included in CMV vaccines. Although it is nonessential for in vitro CMV growth, pp65 displays immunomodulatory functions that support a potential role in primary and/or persistent infection. To determine the contribution of pp65 to CMV infection and immunity, we generated a rhesus CMV lacking both pp65 orthologs (RhCMVΔpp65ab). While deletion of pp65ab slightly reduced growth in vitro and increased defective particle formation, the protein composition of secreted virions was largely unchanged. Interestingly, pp65 was not required for primary and persistent infection in animals. Immune responses induced by RhCMVΔpp65ab did not prevent reinfection with rhesus CMV; however, reinfection with RhCMVΔUS2-11, which lacks viral-encoded MHC-I antigen presentation inhibitors, was prevented. Unexpectedly, induction of pp65b-specific T cells alone did not protect against RhCMVΔUS2-11 challenge, suggesting that T cells targeting multiple CMV antigens are required for protection. However, pp65-specific immunity was crucial for controlling viral dissemination during primary infection, as indicated by the marked increase of RhCMVΔpp65ab genome copies in CMV-naive, but not CMV-immune, animals. Our data provide rationale for inclusion of pp65 into CMV vaccines but also demonstrate that pp65-induced T cell responses alone do not recapitulate the protective effect of natural infection.


Nature Medicine | 2018

Prevention of tuberculosis in rhesus macaques by a cytomegalovirus-based vaccine

Scott G. Hansen; Guangwu Xu; Julia C. Ford; Emily Marshall; Daniel Malouli; Roxanne M. Gilbride; Colette M. Hughes; Abigail B. Ventura; Emily Ainslie; Kurt T Randall; Andrea N. Selseth; Parker Rundstrom; Lauren Herlache; Matthew S. Lewis; Haesun Park; Shannon L. Planer; John M. Turner; Miranda Fischer; Christina Armstrong; Robert C Zweig; Joseph Valvo; Jackie Braun; Smitha Shankar; Lenette L. Lu; Andrew W. Sylwester; Alfred W. Legasse; Martin Messerle; Michael A. Jarvis; Lynn M. Amon; Alan Aderem

Despite widespread use of the bacille Calmette–Guérin (BCG) vaccine, tuberculosis (TB) remains a leading cause of global mortality from a single infectious agent (Mycobacterium tuberculosis or Mtb). Here, over two independent Mtb challenge studies, we demonstrate that subcutaneous vaccination of rhesus macaques (RMs) with rhesus cytomegalovirus vectors encoding Mtb antigen inserts (hereafter referred to as RhCMV/TB)—which elicit and maintain highly effector-differentiated, circulating and tissue-resident Mtb-specific CD4+ and CD8+ memory T cell responses—can reduce the overall (pulmonary and extrapulmonary) extent of Mtb infection and disease by 68%, as compared to that in unvaccinated controls, after intrabronchial challenge with the Erdman strain of Mtb at ∼1 year after the first vaccination. Fourteen of 34 RhCMV/TB-vaccinated RMs (41%) across both studies showed no TB disease by computed tomography scans or at necropsy after challenge (as compared to 0 of 17 unvaccinated controls), and ten of these RMs were Mtb-culture-negative for all tissues, an exceptional long-term vaccine effect in the RM challenge model with the Erdman strain of Mtb. These results suggest that complete vaccine-mediated immune control of highly pathogenic Mtb is possible if immune effector responses can intercept Mtb infection at its earliest stages.


PLOS Pathogens | 2016

Natural Killer Cell Evasion Is Essential for Infection by Rhesus Cytomegalovirus.

Elizabeth R. Sturgill; Daniel Malouli; Scott G. Hansen; Benjamin J. Burwitz; Seongkyung Seo; Christine L. Schneider; Jennie L. Womack; Marieke C. Verweij; Abigail B. Ventura; Amruta Bhusari; Krystal M. Jeffries; Alfred W. Legasse; Michael K. Axthelm; Amy W. Hudson; Jonah B. Sacha; Louis J. Picker; Klaus Früh

The natural killer cell receptor NKG2D activates NK cells by engaging one of several ligands (NKG2DLs) belonging to either the MIC or ULBP families. Human cytomegalovirus (HCMV) UL16 and UL142 counteract this activation by retaining NKG2DLs and US18 and US20 act via lysomal degradation but the importance of NK cell evasion for infection is unknown. Since NKG2DLs are highly conserved in rhesus macaques, we characterized how NKG2DL interception by rhesus cytomegalovirus (RhCMV) impacts infection in vivo. Interestingly, RhCMV lacks homologs of UL16 and UL142 but instead employs Rh159, the homolog of UL148, to prevent NKG2DL surface expression. Rh159 resides in the endoplasmic reticulum and retains several NKG2DLs whereas UL148 does not interfere with NKG2DL expression. Deletion of Rh159 releases human and rhesus MIC proteins, but not ULBPs, from retention while increasing NK cell stimulation by infected cells. Importantly, RhCMV lacking Rh159 cannot infect CMV-naïve animals unless CD8+ cells, including NK cells, are depleted. However, infection can be rescued by replacing Rh159 with HCMV UL16 suggesting that Rh159 and UL16 perform similar functions in vivo. We therefore conclude that cytomegaloviral interference with NK cell activation is essential to establish but not to maintain chronic infection.


PLOS Pathogens | 2016

Cross-Species Rhesus Cytomegalovirus Infection of Cynomolgus Macaques.

Benjamin J. Burwitz; Daniel Malouli; Benjamin N. Bimber; Jason S. Reed; Abigail B. Ventura; Meaghan H. Hancock; Luke S. Uebelhoer; Amruta Bhusari; Katherine B. Hammond; Renee G. Espinosa Trethewy; Alex Klug; Alfred W. Legasse; Michael K. Axthelm; Jay A. Nelson; Byung Park; Daniel N. Streblow; Scott G. Hansen; Louis J. Picker; Klaus Früh; Jonah B. Sacha

Cytomegaloviruses (CMV) are highly species-specific due to millennia of co-evolution and adaptation to their host, with no successful experimental cross-species infection in primates reported to date. Accordingly, full genome phylogenetic analysis of multiple new CMV field isolates derived from two closely related nonhuman primate species, Indian-origin rhesus macaques (RM) and Mauritian-origin cynomolgus macaques (MCM), revealed distinct and tight lineage clustering according to the species of origin, with MCM CMV isolates mirroring the limited genetic diversity of their primate host that underwent a population bottleneck 400 years ago. Despite the ability of Rhesus CMV (RhCMV) laboratory strain 68–1 to replicate efficiently in MCM fibroblasts and potently inhibit antigen presentation to MCM T cells in vitro, RhCMV 68–1 failed to productively infect MCM in vivo, even in the absence of host CD8+ T and NK cells. In contrast, RhCMV clone 68–1.2, genetically repaired to express the homologues of the HCMV anti-apoptosis gene UL36 and epithelial cell tropism genes UL128 and UL130 absent in 68–1, efficiently infected MCM as evidenced by the induction of transgene-specific T cells and virus shedding. Recombinant variants of RhCMV 68–1 and 68–1.2 revealed that expression of either UL36 or UL128 together with UL130 enabled productive MCM infection, indicating that multiple layers of cross-species restriction operate even between closely related hosts. Cumulatively, these results implicate cell tropism and evasion of apoptosis as critical determinants of CMV transmission across primate species barriers, and extend the macaque model of human CMV infection and immunology to MCM, a nonhuman primate species with uniquely simplified host immunogenetics.


Nature | 2017

Addendum: Immune clearance of highly pathogenic SIV infection

Scott G. Hansen; Michael Piatak; Abigail B. Ventura; Colette M. Hughes; Roxanne M. Gilbride; Julia C. Ford; Kelli Oswald; Rebecca Shoemaker; Yuan Li; Matthew S. Lewis; Awbrey N. Gilliam; Guangwu Xu; Nathan Whizin; Benjamin J. Burwitz; Shannon L. Planer; John M. Turner; Alfred W. Legasse; Michael K. Axthelm; Jay A. Nelson; Klaus Früh; Jonah B. Sacha; Jacob D. Estes; Brandon F. Keele; Paul T. Edlefsen; Jeffrey D. Lifson; Louis J. Picker

This corrects the article DOI: 10.1038/nature12519

Collaboration


Dive into the Abigail B. Ventura's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alfred W. Legasse

Oregon National Primate Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael K. Axthelm

Oregon National Primate Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge