Network


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

Hotspot


Dive into the research topics where Alejandra Ramos is active.

Publication


Featured researches published by Alejandra Ramos.


Nature | 2011

Broad neutralization coverage of HIV by multiple highly potent antibodies

Laura M. Walker; Michael Huber; Katie J. Doores; Emilia Falkowska; Robert Pejchal; Jean-Philippe Julien; Sheng-Kai Wang; Alejandra Ramos; Po-Ying Chan-Hui; Matthew Moyle; Jennifer L. Mitcham; Phillip W. Hammond; Ole A. Olsen; Pham Phung; Steven P. Fling; Chi-Huey Wong; Sanjay Phogat; Terri Wrin; Melissa Simek; Protocol G. Principal Investigators; Wayne C. Koff; Ian A. Wilson; Dennis R. Burton; Pascal Poignard

Broadly neutralizing antibodies against highly variable viral pathogens are much sought after to treat or protect against global circulating viruses. Here we probed the neutralizing antibody repertoires of four human immunodeficiency virus (HIV)-infected donors with remarkably broad and potent neutralizing responses and rescued 17 new monoclonal antibodies that neutralize broadly across clades. Many of the new monoclonal antibodies are almost tenfold more potent than the recently described PG9, PG16 and VRC01 broadly neutralizing monoclonal antibodies and 100-fold more potent than the original prototype HIV broadly neutralizing monoclonal antibodies. The monoclonal antibodies largely recapitulate the neutralization breadth found in the corresponding donor serum and many recognize novel epitopes on envelope (Env) glycoprotein gp120, illuminating new targets for vaccine design. Analysis of neutralization by the full complement of anti-HIV broadly neutralizing monoclonal antibodies now available reveals that certain combinations of antibodies should offer markedly more favourable coverage of the enormous diversity of global circulating viruses than others and these combinations might be sought in active or passive immunization regimes. Overall, the isolation of multiple HIV broadly neutralizing monoclonal antibodies from several donors that, in aggregate, provide broad coverage at low concentrations is a highly positive indicator for the eventual design of an effective antibody-based HIV vaccine.


Science | 2011

A potent and broad neutralizing antibody recognizes and penetrates the HIV glycan shield.

Robert Pejchal; Katie J. Doores; Laura M. Walker; Reza Khayat; Po-Ssu Huang; Sheng-Kai Wang; Robyn L. Stanfield; Jean-Philippe Julien; Alejandra Ramos; Matthew Crispin; Rafael S. Depetris; Umesh Katpally; Andre J. Marozsan; Albert Cupo; Sebastien Maloveste; Yan Liu; Ryan McBride; Yukishige Ito; Rogier W. Sanders; Cassandra Ogohara; James C. Paulson; Ten Feizi; Christopher N. Scanlan; Chi-Huey Wong; John P. Moore; William C. Olson; Andrew B. Ward; Pascal Poignard; William R. Schief; Dennis R. Burton

An HIV antibody achieves potency and breadth by binding simultaneously to two conserved glycans on the viral envelope protein. The HIV envelope (Env) protein gp120 is protected from antibody recognition by a dense glycan shield. However, several of the recently identified PGT broadly neutralizing antibodies appear to interact directly with the HIV glycan coat. Crystal structures of antigen-binding fragments (Fabs) PGT 127 and 128 with Man9 at 1.65 and 1.29 angstrom resolution, respectively, and glycan binding data delineate a specific high mannose-binding site. Fab PGT 128 complexed with a fully glycosylated gp120 outer domain at 3.25 angstroms reveals that the antibody penetrates the glycan shield and recognizes two conserved glycans as well as a short β-strand segment of the gp120 V3 loop, accounting for its high binding affinity and broad specificify. Furthermore, our data suggest that the high neutralization potency of PGT 127 and 128 immunoglobulin Gs may be mediated by cross-linking Env trimers on the viral surface.


Immunity | 2014

Broadly Neutralizing HIV Antibodies Define a Glycan-Dependent Epitope on the Prefusion Conformation of gp41 on Cleaved Envelope Trimers

Emilia Falkowska; Khoa Le; Alejandra Ramos; Katherine Doores; Jeong Hyun Lee; Claudia Blattner; Alejandro Ramirez; Ronald Derking; Marit J. van Gils; Chi-Hui Liang; Ryan McBride; Benjamin von Bredow; Sachin S. Shivatare; Chung-Yi Wu; Po-Ying Chan-Hui; Yan Liu; Ten Feizi; Michael B. Zwick; Wayne C. Koff; Michael S. Seaman; Kristine Swiderek; John P. Moore; David T. Evans; James C. Paulson; Chi-Huey Wong; Andrew B. Ward; Ian A. Wilson; Rogier W. Sanders; Pascal Poignard; Dennis R. Burton

Broadly neutralizing HIV antibodies are much sought after (a) to guide vaccine design, both as templates and as indicators of the authenticity of vaccine candidates, (b) to assist in structural studies, and (c) to serve as potential therapeutics. However, the number of targets on the viral envelope spike for such antibodies has been limited. Here, we describe a set of human monoclonal antibodies that define what is, to the best of our knowledge, a previously undefined target on HIV Env. The antibodies recognize a glycan-dependent epitope on the prefusion conformation of gp41 and unambiguously distinguish cleaved from uncleaved Env trimers, an important property given increasing evidence that cleavage is required for vaccine candidates that seek to mimic the functional HIV envelope spike. The availability of this set of antibodies expands the number of vaccine targets on HIV and provides reagents to characterize the native envelope spike.


PLOS Pathogens | 2013

Broadly neutralizing antibody PGT121 allosterically modulates CD4 binding via recognition of the HIV-1 gp120 V3 base and multiple surrounding glycans.

Jean-Philippe Julien; Devin Sok; Reza Khayat; Jeong Hyun Lee; Katherine Doores; Laura M. Walker; Alejandra Ramos; Devan Diwanji; Robert Pejchal; Albert Cupo; Umesh Katpally; Rafael S. Depetris; Robyn L. Stanfield; Ryan McBride; Andre J. Marozsan; James C. Paulson; Rogier W. Sanders; John P. Moore; Dennis R. Burton; Pascal Poignard; Andrew B. Ward; Ian A. Wilson

New broad and potent neutralizing HIV-1 antibodies have recently been described that are largely dependent on the gp120 N332 glycan for Env recognition. Members of the PGT121 family of antibodies, isolated from an African donor, neutralize ∼70% of circulating isolates with a median IC50 less than 0.05 µg ml−1. Here, we show that three family members, PGT121, PGT122 and PGT123, have very similar crystal structures. A long 24-residue HCDR3 divides the antibody binding site into two functional surfaces, consisting of an open face, formed by the heavy chain CDRs, and an elongated face, formed by LCDR1, LCDR3 and the tip of the HCDR3. Alanine scanning mutagenesis of the antibody paratope reveals a crucial role in neutralization for residues on the elongated face, whereas the open face, which accommodates a complex biantennary glycan in the PGT121 structure, appears to play a more secondary role. Negative-stain EM reconstructions of an engineered recombinant Env gp140 trimer (SOSIP.664) reveal that PGT122 interacts with the gp120 outer domain at a more vertical angle with respect to the top surface of the spike than the previously characterized antibody PGT128, which is also dependent on the N332 glycan. We then used ITC and FACS to demonstrate that the PGT121 antibodies inhibit CD4 binding to gp120 despite the epitope being distal from the CD4 binding site. Together, these structural, functional and biophysical results suggest that the PGT121 antibodies may interfere with Env receptor engagement by an allosteric mechanism in which key structural elements, such as the V3 base, the N332 oligomannose glycan and surrounding glycans, including a putative V1/V2 complex biantennary glycan, are conformationally constrained.


Journal of Virology | 2006

Human Immunodeficiency Virus Type 1 Coreceptor Switching: V1/V2 Gain-of-Fitness Mutations Compensate for V3 Loss-of-Fitness Mutations

Cristina Pastore; Rebecca Nedellec; Alejandra Ramos; Suzanne E. Pontow; Lee Ratner; Donald E. Mosier

ABSTRACT Human immunodeficiency virus type 1 (HIV-1) entry into target cells is mediated by the virus envelope binding to CD4 and the conformationally altered envelope subsequently binding to one of two chemokine receptors. HIV-1 envelope glycoprotein (gp120) has five variable loops, of which three (V1/V2 and V3) influence the binding of either CCR5 or CXCR4, the two primary coreceptors for virus entry. Minimal sequence changes in V3 are sufficient for changing coreceptor use from CCR5 to CXCR4 in some HIV-1 isolates, but more commonly additional mutations in V1/V2 are observed during coreceptor switching. We have modeled coreceptor switching by introducing most possible combinations of mutations in the variable loops that distinguish a previously identified group of CCR5- and CXCR4-using viruses. We found that V3 mutations entail high risk, ranging from major loss of entry fitness to lethality. Mutations in or near V1/V2 were able to compensate for the deleterious V3 mutations and may need to precede V3 mutations to permit virus survival. V1/V2 mutations in the absence of V3 mutations often increased the capacity of virus to utilize CCR5 but were unable to confer CXCR4 use. V3 mutations were thus necessary but not sufficient for coreceptor switching, and V1/V2 mutations were necessary for virus survival. HIV-1 envelope sequence evolution from CCR5 to CXCR4 use is constrained by relatively frequent lethal mutations, deep fitness valleys, and requirements to make the right amino acid substitution in the right place at the right time.


Journal of Virology | 2004

Intrinsic Obstacles to Human Immunodeficiency Virus Type 1 Coreceptor Switching

Cristina Pastore; Alejandra Ramos; Donald E. Mosier

ABSTRACT The natural evolution of human immunodeficiency virus type 1 infection often includes a switch in coreceptor preference late in infection from CCR5 to CXCR4, a change associated with expanded target cell range and worsened clinical prognosis. Why coreceptor switching takes so long is puzzling, since it requires as few as one to two mutations. Here we report three obstacles that impede the CCR5-to-CXCR4 switch. Coreceptor switch variants were selected by target cell replacement in vitro. Most switch variants showed diminished replication compared to their parental R5 isolate. Transitional intermediates were more sensitive to both CCR5 and CXCR4 inhibitors than either the parental R5 virus or the final R5X4 (or rare X4) variant. The small number of mutations in viruses selected for CXCR4 use were distinctly nonrandom, with a dominance of charged amino acid substitutions encoded by G-to-A transitions, changes in N-linked glycosylation sites, and isolate-specific mutation patterns. Diminished replication fitness, less-efficient coreceptor use, and unique mutational pathways may explain the long delay from primary infection until the emergence of CXCR4-using viruses.


Science Translational Medicine | 2014

Promiscuous Glycan Site Recognition by Antibodies to the High-Mannose Patch of gp120 Broadens Neutralization of HIV

Devin Sok; Katie J. Doores; Bryan Briney; Khoa Le; Karen L. Saye-Francisco; Alejandra Ramos; Daniel W. Kulp; Jean-Philippe Julien; Sergey Menis; Lalinda Wickramasinghe; Michael S. Seaman; William R. Schief; Ian A. Wilson; Pascal Poignard; Dennis R. Burton

HIV broadly neutralizing monoclonal antibodies targeting the high-mannose patch of Env can use alternate glycan sites for neutralization. Neutralizing Antibodies with a Sweet Tooth Sugar can be quite tempting—as anyone who’s seen a kid rip into birthday cake can attest. Yet, antibodies can also have a sweet tooth, targeting glycan modifications on the surface of proteins. Indeed, some antibodies that neutralize multiple HIV strains—broadly neutralizing monoclonal antibodies (bnmAbs)—target a high-mannose patch on the HIV protein Env. Although this high-mannose patch is centered around the glycan at position 332 (N332), it has remained unclear if the N332 glycan is absolutely required for neutralization and, if not, why not. Sok et al. found that these mannose patch–targeting antibodies can bind alternate glycans in the absence of N332, which helps to explain their ability to neutralize many strains of HIV. Specifically, some bnmAbs can bind to the N334 site when that replaces the N332 site and some can form more interactions with other glycans, particularly complex-type glycans on variable loops, if the N332 sugar is absent. These data also suggest that mannose patch–targeting bnmAbs can work in combination to neutralize a wider range of different strains than single bnmAbs. The promiscuity of glycan binding by these sugar-loving antibodies is important to consider for both vaccine and therapeutic antibody development. Broadly neutralizing monoclonal antibodies (bnmAbs) that target the high-mannose patch centered around the glycan at position 332 on HIV Env are promising vaccine leads and therapeutic candidates because they effectively protect against mucosal SHIV challenge and strongly suppress SHIV viremia in established infection in macaque models. However, these antibodies demonstrate varying degrees of dependency on the N332 glycan site, and the origins of their neutralization breadth are not always obvious. By measuring neutralization on an extended range of glycan site viral variants, we found that some bnmAbs can use alternate N-linked glycans in the absence of the N332 glycan site and therefore neutralize a substantial number of viruses lacking the site. Furthermore, many of the antibodies can neutralize viruses in which the N332 glycan site is shifted to the 334 position. Finally, we found that a combination of three antibody families that target the high-mannose patch can lead to 99% neutralization coverage of a large panel of viruses containing the N332/N334 glycan site and up to 66% coverage for viruses that lack the N332/N334 glycan site. The results indicate that a diverse response against the high-mannose patch may provide near-equivalent coverage as a combination of bnmAbs targeting multiple epitopes. Additionally, the ability of some bnmAbs to use other N-linked glycan sites can help counter neutralization escape mediated by shifting of glycosylation sites. Overall, this work highlights the importance of promiscuous glycan binding properties in bnmAbs to the high-mannose patch for optimal antiviral activity in either protective or therapeutic modalities.


Journal of Virology | 2012

PGV04, an HIV-1 gp120 CD4 Binding Site Antibody, Is Broad and Potent in Neutralization but Does Not Induce Conformational Changes Characteristic of CD4

Emilia Falkowska; Alejandra Ramos; Yu Feng; Tongqing Zhou; Stephanie Moquin; Laura M. Walker; Xueling Wu; Michael S. Seaman; Terri Wrin; Peter D. Kwong; Richard T. Wyatt; John R. Mascola; Pascal Poignard; Dennis R. Burton

ABSTRACT Recently, several broadly neutralizing monoclonal antibodies (bnMAbs) directed to the CD4-binding site (CD4bs) of gp120 have been isolated from HIV-1-positive donors. These include VRC01, 3BNC117, and NIH45-46, all of which are capable of neutralizing about 90% of circulating HIV-1 isolates and all of which induce conformational changes in the HIV-1 gp120 monomer similar to those induced by the CD4 receptor. In this study, we characterize PGV04 (also known as VRC-PG04), a MAb with potency and breadth that rivals those of the prototypic VRC01 and 3BNC117. When screened on a large panel of viruses, the neutralizing profile of PGV04 was distinct from those of CD4, b12, and VRC01. Furthermore, the ability of PGV04 to neutralize pseudovirus containing single alanine substitutions exhibited a pattern distinct from those of the other CD4bs MAbs. In particular, substitutions D279A, I420A, and I423A were found to abrogate PGV04 neutralization. In contrast to VRC01, PGV04 did not enhance the binding of 17b or X5 to their epitopes (the CD4-induced [CD4i] site) in the coreceptor region on the gp120 monomer. Furthermore, in contrast to CD4, none of the anti-CD4bs MAbs induced the expression of the 17b epitope on cell surface-expressed cleaved Env trimers. We conclude that potent CD4bs bnMAbs can display differences in the way they recognize and access the CD4bs and that mimicry of CD4, as assessed by inducing conformational changes in monomeric gp120 that lead to enhanced exposure of the CD4i site, is not uniquely correlated with effective neutralization at the site of CD4 binding on HIV-1.


PLOS Pathogens | 2016

Broadly Neutralizing Antibody Responses in a Large Longitudinal Sub-Saharan HIV Primary Infection Cohort.

Elise Landais; Xiayu Huang; Colin Havenar-Daughton; Ben Murrell; Matthew Price; Lalinda Wickramasinghe; Alejandra Ramos; Charoan B. Bian; Melissa Simek; Susan Allen; Etienne Karita; William Kilembe; Shabir Lakhi; Mubiana Inambao; Anatoli Kamali; Eduard J. Sanders; Omu Anzala; Vinodh Edward; Linda-Gail Bekker; Jianming Tang; Jill Gilmour; Sergei L. Kosakovsky-Pond; Pham Phung; Terri Wrin; Shane Crotty; Adam Godzik; Pascal Poignard

Broadly neutralizing antibodies (bnAbs) are thought to be a critical component of a protective HIV vaccine. However, designing vaccines immunogens able to elicit bnAbs has proven unsuccessful to date. Understanding the correlates and immunological mechanisms leading to the development of bnAb responses during natural HIV infection is thus critical to the design of a protective vaccine. The IAVI Protocol C program investigates a large longitudinal cohort of primary HIV-1 infection in Eastern and South Africa. Development of neutralization was evaluated in 439 donors using a 6 cross-clade pseudo-virus panel predictive of neutralization breadth on larger panels. About 15% of individuals developed bnAb responses, essentially between year 2 and year 4 of infection. Statistical analyses revealed no influence of gender, age or geographical origin on the development of neutralization breadth. However, cross-clade neutralization strongly correlated with high viral load as well as with low CD4 T cell counts, subtype-C infection and HLA-A*03(-) genotype. A correlation with high overall plasma IgG levels and anti-Env IgG binding titers was also found. The latter appeared not associated with higher affinity, suggesting a greater diversity of the anti-Env responses in broad neutralizers. Broadly neutralizing activity targeting glycan-dependent epitopes, largely the N332-glycan epitope region, was detected in nearly half of the broad neutralizers while CD4bs and gp41-MPER bnAb responses were only detected in very few individuals. Together the findings suggest that both viral and host factors are critical for the development of bnAbs and that the HIV Env N332-glycan supersite may be a favorable target for vaccine design.


Cell | 2016

Tailored Immunogens Direct Affinity Maturation toward HIV Neutralizing Antibodies.

Bryan Briney; Devin Sok; Joseph G. Jardine; Daniel W. Kulp; Patrick Skog; Sergey Menis; Ronald Jacak; Oleksandr Kalyuzhniy; Natalia de Val; Fabian Sesterhenn; Khoa Le; Alejandra Ramos; Meaghan Jones; Karen L. Saye-Francisco; Tanya R. Blane; Skye Spencer; Erik Georgeson; Xiaozhen Hu; Gabriel Ozorowski; Yumiko Adachi; Michael Kubitz; Anita Sarkar; Ian A. Wilson; Andrew B. Ward; David Nemazee; Dennis R. Burton; William R. Schief

Summary Induction of broadly neutralizing antibodies (bnAbs) is a primary goal of HIV vaccine development. VRC01-class bnAbs are important vaccine leads because their precursor B cells targeted by an engineered priming immunogen are relatively common among humans. This priming immunogen has demonstrated the ability to initiate a bnAb response in animal models, but recall and maturation toward bnAb development has not been shown. Here, we report the development of boosting immunogens designed to guide the genetic and functional maturation of previously primed VRC01-class precursors. Boosting a transgenic mouse model expressing germline VRC01 heavy chains produced broad neutralization of near-native isolates (N276A) and weak neutralization of fully native HIV. Functional and genetic characteristics indicate that the boosted mAbs are consistent with partially mature VRC01-class antibodies and place them on a maturation trajectory that leads toward mature VRC01-class bnAbs. The results show how reductionist sequential immunization can guide maturation of HIV bnAb responses.

Collaboration


Dive into the Alejandra Ramos's collaboration.

Top Co-Authors

Avatar

Dennis R. Burton

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Pascal Poignard

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ian A. Wilson

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Andrew B. Ward

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Devin Sok

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Bryan Briney

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Khoa Le

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Laura M. Walker

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Michael S. Seaman

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge